Next Article in Journal
Experimental In-Field Transfer and Survival of Escherichia coli from Animal Feces to Romaine Lettuce in Salinas Valley, California
Next Article in Special Issue
Oncogenic Virome Benefits from the Different Vaginal Microbiome-Immune Axes
Previous Article in Journal
Opinion: Are Organoids the End of Model Evolution for Studying Host Intestinal Epithelium/Microbe Interactions?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dendritic Cells/Macrophages-Targeting Feature of Ebola Glycoprotein and its Potential as Immunological Facilitator for Antiviral Vaccine Approach

1
Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
2
Centre de Recherche en Infectiologie de l’ Université Laval/Centre Hospitalier de l’ Université Laval (CHUL), Québec, QC G1V 4G2, Canada
*
Author to whom correspondence should be addressed.
Microorganisms 2019, 7(10), 402; https://doi.org/10.3390/microorganisms7100402
Submission received: 7 August 2019 / Revised: 19 September 2019 / Accepted: 26 September 2019 / Published: 29 September 2019
(This article belongs to the Special Issue Virus-Host Interaction: From Physiology to Pathology)

Abstract

:
In the prevention of epidemic and pandemic viral infection, the use of the antiviral vaccine has been the most successful biotechnological and biomedical approach. In recent times, vaccine development studies have focused on recruiting and targeting immunogens to dendritic cells (DCs) and macrophages to induce innate and adaptive immune responses. Interestingly, Ebola virus (EBOV) glycoprotein (GP) has a strong binding affinity with DCs and macrophages. Shreds of evidence have also shown that the interaction between EBOV GP with DCs and macrophages leads to massive recruitment of DCs and macrophages capable of regulating innate and adaptive immune responses. Therefore, studies for the development of vaccine can utilize the affinity between EBOV GP and DCs/macrophages as a novel immunological approach to induce both innate and acquired immune responses. In this review, we will discuss the unique features of EBOV GP to target the DC, and its potential to elicit strong immune responses while targeting DCs/macrophages. This review hopes to suggest and stimulate thoughts of developing a stronger and effective DC-targeting vaccine for diverse virus infection using EBOV GP.

Graphical Abstract

1. Introduction

1.1. Dendritic cell (DC)-targeting Vaccines

The development of the antiviral vaccine has been the most successful biotechnological and biomedical approach against epidemic and pandemic viral infections [1]. Qualities of an ideal vaccine include safety (even in an immunocompromised patient), high effectivity in inducing immune responses [2], cost effectivity, and high stability and durability state [2]. There have been successful productions of many antiviral vaccines using different strategies, including live attenuated vaccines for yellow fever, smallpox, poliovirus, measles, mumps, rubella, adenovirus, varicella, and rotavirus; inactivated vaccines for poliovirus, influenza virus, hepatitis A virus, Japanese encephalitis; and virus-like particle (VLP) vaccines for hepatitis B and human papillomavirus (Table 1) [1,3]. In recent years, the impact of the new advent of technology in gene delivery and expression, adjuvants, the convergence of human monoclonal antibody isolation, structural biology, and high throughput sequencing, among others, has greatly influenced biotechnological approach for the development of new vaccines [4].
Despite the progress that has been recorded so far in vaccine development for viral infection, limitations such as the narrow-spectrum effect of vaccine and low immune response call for a new approach in the event of vaccines development [26]. Attention has thus been shifted to the abilities of dendritic cells (DCs)/macrophages to induce potent immune responses [27], and their usage is in the pipeline for the development of vaccines against cancer [28], adenovirus [29], and yellow fever [30], among others. A DC-targeting vaccine approach depends on the ability of DCs to target specific antigens by recognizing pathogen-associated molecular patterns (PAMPs) of the antigen, and further stimulate innate, humoral, and corresponding cellular immune responses [31]. Hawiger et al. showed that an antigen delivery system targeting the DEC-205, which is a DC-restricted endocytic receptor, using monoclonal antibody of DC induced a high magnitude of T cell responses [32]. Zaneti et al. also demonstrated recently that a DNA vaccine consisting of plasmid encoding single-chain Fv antibody (ScFv) αDEC205 fused with dengue virus (DENV) envelope domain III (EDIII) induced a strong anti-EDIII IgG titer and CD4+ capable of inhibiting DENV2 infection when intramuscularly injected into Balb/c mice followed by electroporation [33]. Table 2 shows a summary of other different strategies that have been used to target DCs for the development of vaccines.
However, there are some limitations. For instance, Cheng et al. showed that the mechanism of targeting DCs using recombinant adenovirus (rAd) vector vaccine is associated with toxicity related to ‘prior human gene therapy fatality’ [34]. Also, a study by Boudewijns et al. revealed the toxicity profile associated with DC vaccination in stage III and stage IV melanoma patients. In this case, melanoma patients were vaccinated with DCs loaded with antigens associated with the tumor. About 84% of the patients had adverse events related to the treatment, including reaction at the injection site, and symptoms such as flu, while about 3% of the patients experienced grade 3 toxicity [35]. However, some of the adverse effects noted are considered to be normal clinical or immunological responses [35].
Hawiger et al. also showed that the T cell activated by DC-targeted antigen could not be polarized to produce T helper cells. Hawiger et al. further demonstrated that T cells severely reduced after seven days and were not responsive to systemic antigen challenge [32]. Almand et al. also confirmed that the production of immature myeloid cells might cause dysfunction of DCs and can lead to immunosuppression of T cells [36]. However, Apostolico et al. demonstrated the induction of long-lived T cells against HIV using a DC-targeting approach with a heavy-chain αDEC205 (αDECHIVBr8) in the presence of TLR3 agonist [37].

1.2. Dendritic Cells (DCs)/Macrophages and Immune Responses

DCs are antigen-presenting cells (APCs) capable of initiating and directing innate and adaptive immune responses [44]. The intricate properties of DCs that account for their roles in the immune system are: Unique mechanisms for antigen presentation, the ability to migrate to a particular site in lymphoid organs for immune response stimulation, and their rapid differentiation or maturation in response to a variety of stimuli ranging from Toll-like receptor (TLR) ligands to many other non-microbial factors [45]. Briefly, after exposure to the foreign material, the DCs mature and migrate to the lymphoid organ, where the DCs induce a cellular immune response (T cells) and humoral immune responses (B cells) [46]. Targeting peptides to DCs can also induce an innate immune response by activating natural killer cells and natural killer T cells [47]. DCs also function by producing protective cytokines—like interleukin (IL)-12, IL-6 [48], and type I interferons [49], which influence distinct steps in the adaptive immune response of lymphocytes—and the activation and expansion of innate lymphocytes [45,49,50].
The presence of specialized surface receptors, known as pattern recognition receptors (PRRs), on DCs facilitate the functions of DCs. These PRRs are named as follows: Toll-like receptors (TLRs), NOD (Nucleotide-binding oligomerization domain)-like receptors (NLRs), C-lectin type receptors (CLRs), RIG-1 like receptors (RLRs), and helicases recognize pathogens associated molecular patterns (PAMPS) [51,52]. DCs play an essential role in conferring protection against pathogens and commensal microorganisms [53].
The TLRs, known as ‘sensors that detect infection’, were the first discovered PRRs [54]. TLRs are innate immune receptors with a full length of a membrane that can use pattern recognition processing of ligands to detect a variety of molecules that insinuate tissue damage, and a wide range of human pathogens including bacteria, viruses, protozoans, and nematodes [54,55]. The conserved pathogen recognition features of TLRs have led to the stimulation of several immune cells, not excluding proinflammatory cytokines, antimicrobial molecules, phagosomal maturation, and costimulatory molecules [56]. There are thirteen (13) known TLRs that can recognize a wide range of microbial pathogens, but differ in their specificity for microbial patterns. For instance, to recognize microbial cell walls and membranes unique to pathogens, TLRs 1, 2, 4, 5, and 6 are much employed; TLR4 recognizes lipopolysaccharides (LPS), while heterodimers of TLR2/1 and TLR2/6 recognize lipopeptides and TLR5 recognizes flagellin; TLR9 recognizes DNA unmethylated CpG motifs, various forms of RNA by TLRs 3, 7, 8 and 13; and TLR11 recognizes profilin and flagellin of Salmonella. Additionally, Fukuda et al. demonstrated that TLR9 that has an affinity for bacterial DNA ligands plays a crucial role by activating proinflammatory cytokines of macrophages, leading to the development of vascular inflammation and atherogenesis [57], while Koblansky et al. reported that the previously uncharacterized TLR12 can recognize Toxoplasma gondii profilin by plasmacytoid dendritic cells (pDCs) [58]. TLRs are localized intracellularly. As already reviewed, TLRs 3, 4, 7, and 9 have their transmembrane domain localized intracellularly [59]. Also, Raetz et al. reported that TLR11 and TLR12 are both intracellularly localized, where they both bind with T. gondii and lead to the signaling of MyD88- and UNC93B1-dependent signaling cascade [60].
Importantly, TLRs coordinate both the innate and adaptive immune responses [54,55,61]. Innate immune responses are activated via recognition of microbial products by TLRs, leading to the stimulation of proinflammatory cytokines maturation of DCs for antigen presentation. Also, the activation of DCs via TLRs can increase the level of proinflammatory cytokines, chemokines, and co-stimulatory molecules produced, thus modulating adaptive immune responses, including T regulatory cells [61]. More importantly, TLR4 can also recognize the EBOV glycoprotein (GP). Okumura et al. revealed that the sensor for EBOV GP is the host TLR4, which leads to the production of proinflammatory cytokines. Their study proved that EBOV GP could stimulate the expression NF-κB in vitro in a TLR4-dependent manner [62]. Moreover, Lai et al. pretreated mice with TLR4 antagonist (ultrapure lipopolysaccharide from the bacterium Rhodobacter sphaeroides (LPS-RS)) to inhibit the production of GP-induced cytokines [63]. Their study gave an in vivo evidence that the early stimulation of proinflammatory cytokines during EBOV infection is via the TLR4 pathway.
The RLRs are also host PRRs which are involved in the regulation of innate immune responses by recognizing the pathogen-specific 5′ di or triphosphate non-self RNA in bacteria and viruses, consequently leading to the transcription of IFN-β [64,65]. They can also recognize bacterial mRNA that is uncapped and is 5′ triphosphorylated in the cytosol [66]. He et al. demonstrated that EBOV VP24 inhibits both IFN-induced antiviral responses and type III IFN-γ1 gene expression by inhibiting the RIG-1 pathway responsible for the IRF3 activation [67], indicating the role played by RLRs during EBOV infection. Whereas, NLRs which also regulate the innate immune responses by triggering NF-κB signaling for expression of innate immune responses genes and hydrolyzing viral RNA using activated RNase, do so by recognizing 2′, 5′ -oligoadenylate synthetase type 2 (OAS2) in bacteria and viruses [65,68,69].
The primary role of DCs is to mediate cellular immune response (CD8+ T cells and CD4+ T cells) and humoral immune responses (B cells) [70,71,72], which are of great importance in developing vaccines. In the development of vaccines, DCs are targeted to elicit innate and acquired immune responses by capturing antigens or foreign material at their initial location in the peripheral tissues, processing and presenting antigens on major histocompatibility complex I and II (MHC I and II) [73]. DCs can also be used as adjuvants for DNA vaccines to elicit immune responses [28,52,74,75].
Meanwhile, macrophages are myeloid progenitor immune cells that are characterized by avid phagocytosis because they ingest and degrade dead cells, debris, and foreign material and orchestrate inflammatory processes in the body tissues [44]. They originated from either embryonic development or circulating monocytes [76] and are found all over the body in tissues by adhering to mucosal surfaces and can also be further classified based on their microenvironment [77]. Macrophages serve as the vital component of the innate immune system and also function as professional antigen-presenting cells [78,79]. Besides their role as APCs for the stimulation of specific cellular and humoral immune cells, macrophages also critically regulate the innate immune system by eliciting proinflammatory cytokines and chemokines such as interleukin-6 (IL-6) and tumor necrotic factor (TNF), as well as anti-inflammatory cytokines such as IL-10 [63]. Deficiency of macrophages in mice has been demonstrated to significantly reduce the protection ability of opsonizing antibodies, suggesting its crucial impact on the induction of immune response [80].
In the immune system, a relationship exists between DCs and macrophages. DCs stimulate autoimmune responses to induce specific T cells, that consequently leads to the proliferation of macrophages, which damages the tissue [79]. However, macrophages are involved in the homeostasis of tissues and repair, which helps to prevent tissue damage [79]. The DCs and macrophages connect innate immunity with adaptive immunity. DCs and macrophages are activated during infection for protection by recognizing pathogen-associated molecular patterns (PAMPs) via their PRRs [81]. Upon activation, the matured DCs migrate to the lymph nodes and display the processed peptides on their MHC I or II to trigger T cytotoxic cells (CD8+) or T helper cells (CD4+), respectively [82].

1.3. EBOV Infection and Immune Responses

Ebola virus causes hemorrhagic viral infection by primarily infecting the macrophages and the DCs upon contact with the mucous membrane, and replicate efficiently. Furthermore, the Ebola virus can impede interferon production in DCs, macrophages, and monocytes by protein VP24 and VP35 [83,84,85]. The EBOV glycoprotein (GP) enhances the entry of the Ebola virus to DCs/ macrophages by the presence of C-type lectin-like receptors (CLRs) present on the DCs [86,87]. As described elsewhere, EBOV can also evade the immune system by vitiating both the humoral and cellular adaptive immunity [84]. Although the mechanisms by which EBOV mitigate the host humoral and adaptive immune responses are poorly understood, the depletion of T-cells during EBOV infection has been hypothetically implicated with the deficient signaling events needed during the induction and maintaining the transition of T cells to memory cells and partial clearance of APCs [88]. Furthermore, Lubaki et al. demonstrated that the IFN-inhibiting domains (IIDs) in VP24 and VP35 also contribute to the depletion of the immune system by inhibiting the T cell receptor binding and are also responsible for deficient matured DC [89]. Lubaki et al. also recently revealed that the IIDs in VP24 and VP35 could vitiate humoral immune responses by inhibiting the human B cells differentiation and activation [90]. Whereas, the association of EBOV GP with DCs facilitates the ability of the EBOV GP to modify immune responses by modulating both innate and adaptive immune responses [82]. Interestingly, Groseth et al. demonstrated that although EBOV GP is involved in EBOV infection, EBOV GP alone is not sufficient to cause a lethal effect on the host [91].
The evasion of the immune system consequently affects the vascular system to cause coagulopathy, leading to shock, failure of circulation, bleeding, and death. Other complications are defective inflammations associated with mild secretion of IL-6 and TNFα and a very high level of secretion of IL-1, IL-10 [92], and flawed immune responses such as enormous apoptosis of T cells and the inhibition of the production of specific antibodies [84,93].
To further elucidate the relationship between EBOV GP and APCs, Lüdke et al. showed that a subset of DCs reduced significantly among patients that had acute EBOV in Guinea, while the survivors had activated CD16+ during recovery [94]. The study of Lüdke et al. further showed that EBOV primarily infects DCs, and patients still require DCs to fight and clear EBOV infection. Also, using a chimeric mouse characterized by a competent hematopoietic-immunity, the same authors demonstrated that EBOV primarily infects CD11b+ DCs in both the lymphoid tissues and non-lymphoid tissues which can lead to the depletion of CD8 and CD4 T cells [95]. Although there are reports that showed that some of DCs subsets, including CD141+ DCs, are not prone to viral infection by RAB15, which is expressed on CD141+ DCs and serves as a vesicle-trafficking protein [95,96], the population of DCs that are primarily infected by EBOV is enough for the modulation of both the innate and adaptive immune responses. Moreover, Silvin et al. showed that CD141+ DCs can still act as APCs for the regulation of adaptive immune responses by depending on the viral antigen from bystander cells [96].
Studies have demonstrated that EBOV infection can trigger macrophages to induce innate immune responses, such as inflammatory cytokines and chemokines (e.g., tumor necrosis factor, IL-6, IL-1β, etc.) [97]. EBOV GP is involved in the stimulation of both innate and adaptive immune responses. A study showed that immunization of mice with liposome-encapsulated irradiated Ebola virus induces immune response against Ebola virus via Ebola GP [98]. More recently, a group of scientists proved that Ebola GP can elicit an innate immune response, such as proinflammatory cytokines including IL-6, TNF-α, and anti-inflammatory cytokines and IL-10 alone without adjuvant, which depends solely on the internalization of the EBOV GP by macrophages [63]. They further elucidated that the efficacy of the current vaccine for Ebola virus largely depends on the innate immune response induced by EBOV GP through the toll-like receptor-4 (TLR4) pathway. Ayithan et al. also demonstrated that the induction of chemokines by EBOV GP is via the TLR4 pathway [99]. The role played by EBOV GP in the stimulation of immune response has thus been considered as a significant platform for generating a vaccine for EBOV infection [100].

2. EBOV GP: Bane or Benefit

The synthesis of 676-residue transmembrane of EBOV Glycoprotein (GP) results from the transcriptomic editing of the fourth of the eight (8) genes in the genome of EBOV [101,102]. The EBOV GP is responsible for targeting cell and virus entry by mediating receptor binding and membrane fusion [103]. GP is the only surface protein on EBOV, and it is cleaved by furin to produce disulphide-linked GP1 and GP2 subunits [104]. The endosomal entry of EBOV is by GP1, while the low pH membrane fusion is coordinated by GP2 using Neimann-Pick C1 protein (NPC1), and thus implicated as major pathogenic determinants for infection [101] and the main target for the development of a vaccine for Ebola virus [105].
As previously described, GP1 is a membrane surface protein that comprises three main subunits, including the base, composed of β sheets and Cys53, that may be responsible for the intermolecular bridge with Cys609 of GP2 subunit. The second subunit of GP1 is the head, located between the base and glycan cap. The glycan cap is the third subunit, characterized by the presence of N-linked glycans [101,106]. The recent description of GP1 revealed that EBOV GP has three subdomains, including the receptor-binding domain (RBD) (approximately 149-residue), mucin-like domain (MLD), and the glycan cap (approximately 108-residue) [107,108] (Figure 1A). The MLD is also another highly glycosylated domain on EBOV GP. Unlike the glycan cap, which has only N-linked glycans, MLD has both N-linked glycans and O-linked glycans [108]. Lennemann et al. showed that the removal of all the 15 N-glycosylation of EBOV GP using site-directed mutagenesis significantly increases the pseudovirion transduction of EBOV in Vero cells. However, the removal of the N-glycosylation also favors the recognition of the EBOV GP by antibodies, resulting in the production of neutralizing antibodies [108].
The MLD is found on the variable region of GP1 (C-terminal) and increases the permeability of EBOV into the blood vessels, and also masks the cell from the innate immune response by obstructing access to the epitope of GP [117,118]. MLD can also shield the cellular surface protein sterically, causing cell damage, leakage of an explanted blood vessel, rounding and detachment of cell, and loss of physiological functions [119,120,121]. A study showed that the MLD blocks access to the surface MHC I and II, which leads to decrease in CD 8+ cells and consequently leads to cell rounding (cytopathic effect), while the removal of MLD uncovers the epitope of GP to induce neutralizing antibodies [120,122]. Another study also checked for the impact of EBOV GP without MLD (EBOV GP ΔMLD) on the stimulation of anti-GP and neutralizing antibodies; this study revealed that EBOV GP ΔMLD elicits more anti-EBOV GP antibody than EBOV GP VLP, with moderate stimulation of neutralizing antibodies [107], indicating that MLD is dispensable for EBOV attachment. Our study also showed that the removal of MLD from EBOV facilitates the cell entry efficiently more than the wild type; however, EBOV GP wild-type stimulated NFκB than EBOV GP with deleted MLD [115]. Moreover, in the development of drugs for EBOV, EBOV GP is an important target to be considered. A study showed that the inhibition of GP1 binding by toremifene (an antiviral drug against EBOV) could lead to the premature release of GP2, and thus prevent fusion of the endosome membranes and the virus [123].
On the other hand, GP2 contains the heptad repeated regions 1 and 2 (HR1 and HR2) and internal fusion loop (FL) lacking a free N-terminus (511–556-residue), which display a hydrophobic fusion peptide by utilizing an antiparallel β (Figure 1) [101]. During proteolytic cleavage and endosomal binding of GP1, GP2 undergoes a conformational rearrangement which exposes FL for fusion [124]. Lee et al. recently presented the membrane-proximal external region (MPER) as the missing part of GP2 that is connected to the transmembrane using NMR and EPR spectroscopy. They further showed that MPER consist of ‘helix-turn-helix architecture.’ Lee et al. also revealed the role MPER played by mutating the aromatic neighboring, and the results revealed that the MPER region interacts with EBOV FL through aromatic residues and the mutation of these aromatic residues decreases the fusion and viral entry of EBOV [124]. Although GP2 contains two N-glycosylation sites, Asn563 and Asn618, Wang et al. demonstrated that EBOV GP expression does not depend on GP2. However, their study highlights the functions of N-glycosylation sites on GP2, which includes regulation of GP processing, oligomerization, demannosylation, conformation, and facilitation of the incorporation of EBOV-like particles and pseudovirions of HIV type 1 (HIV-1) for the determination of viral transduction efficiency [125].
Moreover, Lee et al. described a 364-residue of non-structural secreted glycoprotein (pre-sGP) that contains the gene product of EBOV, which is also emitted during the transcriptional editing of the fourth gene of EBOV [101,102] and results from the unedited mRNA [126]. Briefly, sGP is produced from the post-translational cleavage of pre-sGP at the C-terminus by furin—a cellular protease. The proteolytic cleavage produces Δ-peptide and sGP, and while we know little about the receptors and role played by Δ-peptide during EBOV infection, a study has demonstrated that Δ-peptide competes with the binding of EBOV GP when interacting with the host permissive cells for EBOV [127]. On the other hand, sGP forms homodimer by the linking of its monomers at residue Cys53 and Cys306 [128] and just like the GP1, sGP is N-linked glycosylated [126]. Several studies have been done to highlight the role played by sGP during EBOV infection and their effect on immune responses (see review [126]). Research has also recently demonstrated the functions of sGP during the pathogenesis of EBOV. In their study, Wahl-Jensen et al. showed that sGP could not induce production macrophages [129]. Also, Monath et al. showed that the construction of rVSVΔG-ZEBOV-GP lacking sGP produces more neutralizing antibodies against EBOV GP [130], because sGP can vitiate the neutralizing of EBOV GP by acting as a decoy for the antibody responsible for the neutralization of EBOV GP [126,130,131,132].
The GP and sGP are identical in the N-terminal, with 295 amino acids, but are different at the C-terminal. At the C-terminal, sGP has about 65 amino acids, while GP has 381 amino acids, including the mucin-like domain [107,133,134,135]. The similarity in the N-terminal of GP and sGP has been suggested to be responsible for a term called antigenic subversion by sGP, which prevents an immune response to GP by diverting it away. Mohan et al. demonstrated in mice immunized with GP1,2 and sGP, that sGP competes efficiently for anti-GP1,2 antibodies by refocusing the host antibody response to the membrane-bound, thus underscoring the robust immune response responsible for clearing Ebola virus in the system [136]. Therefore, in developing VLPs for EBOV, the GP1,2 must be essential components because of their ability to induce immune response, while sGP might not be included in the EBOV VLP as it has low immunogenicity [129].
Nonetheless, EBOV GP has some right sides in mediating immunity. The EBOV GP can facilitate the maturation of DCs and activates T cells, as well as B cells [89], and can also induce changes in secondary target cells [105]. EBOV GP can upregulate the expression of costimulatory molecules in bone marrow-derived macrophages (BMDM), suggesting its capability to enhance APC stimulatory capacity, which is very important for the induction of potent antigen-specific adaptive immunity [63]. The recent development of Ebola vaccine (rVSV-EBOV Vaccine) is solely dependent on the immunogenicity of EBOV GP. The rVSV-EBOV vaccine was constructed using a recombinant form of vesicular stomatitis virus expressing the EBOV-glycoprotein (GP) on the surface (rVSV/ZEBOV-GP) [137]. Agnandji et al. conducted the phase 1 clinical trial of rVSV/ZEBOV-GP vaccine and reported that this vaccine could induce stable neutralizing antibodies against EBOV GP with a very mild side effect, such as fever [138]. The induction of the neutralizing antibodies is of no doubt associated with the targeting of DCs/macrophages by EBOV GP. This claim is in line with the study done by Marzi et al., which indicated the importance of antibodies for the protection against Ebola virus using a non-human primate [139]. Aside from the utilization of Ebola GP with rVSV for the development of a vaccine against Ebola virus, another study has shown that EBOV GP expressed on adenoviruses virus-like particles (VLPs) also stimulates immune responses [140]. Takada et al. reviewed that the ability of EBOV GP to induce both innate and adaptive immune responses, which could be via complement antibody-dependent enhancement (ADE) (see extensive review in [141]). Therefore, in this review, we will further describe the use of EBOV GP to induce immune response in the subsequent paragraphs.

3. The Interaction Between EBOV GP and DCs/Macrophages Can Induce Robust Innate and Adaptive Immune Responses

During exposure of EBOV to the susceptible cell, the main first line of defense is the innate immune response. The innate immune response is not specific, unlike the adaptive immune response, and is active during the critical hour of infection before the recruitment of adaptive immune responses. Phagocytic cells regulate innate immune responses by inducing inflammatory cytokines, and recognizing conserved features among many pathogens called pathogen-associated immunostimulants [142].
EBOV GP has also been investigated to induce inflammatory cytokines and chemokines, including interleukin (IL)-6, tumor necrosis factor (TNF)-α and IL-1β, interferon (IFN)-γ, IL-2, IL-5, IL-4, IL-12 p20, and IL-10, among others [97], through the Toll-like receptor (TLR)-4 pathway like many other GPs [63]. DCs and macrophages express TLRs, which is essential in the activation of MyD88-dependent and independent signaling pathways and consequently leads to the activation of transcription factor interferon regulatory factor 3/7 (IRF3/7), nuclear factor κB (NF-κB), and activator protein-1 (AP-1) to induce downstream genes [82].
Moreover, Lai et al. showed that immunization with EBOV GP without adjuvants can stimulate a significantly high amount of innate immune response. Lai et al. (2017) [63] further established that EBOV GP can induce a robust innate immune response even after 2 h of treatment, which was built upon the findings of Henao-Restrepo et al. (2015), who revealed that the rVSV-EBOV GP vaccine candidate induces innate immune response within six (6) days of immunization [143]. Furthermore, in contrast to other pathogens with recognizable PAMPS by the PRRs on the monocytes, EBOV GP causes the release of cytokine and chemokines in a manner leading to the recruitment of more DCs and macrophages to the site of infection [82,86].
EBOV GP also plays an essential role in mediating the interaction between innate and adaptive immune response. Although there are studies that have demonstrated that EBOV GP could impair the ability of CD8+ to recognize peptide on the MHC Class I [120], analysis from a 2013–2016 outbreak on the induction of CD8+ T cells revealed that about 40% of the populace examined stimulated CD8+ T cells by EBOV GP [144]. A comprehensive study on the cellular immune response by EBOV infection showed that CD8+ T cell is the predominantly induced T cell, but is relatively low [145]. However, the usage of adjuvant with EBOV GP can produce CD8+ T cells that are enough to protect against lethal Ebola virus challenge [146]. Generally, EBOV GP can trigger CD8+ T cell and CD4+ T cell by interacting with the DCs and macrophages. Thus, EBOV GP can be used to recruit more T cells. Targeting antigens towards DC-specific endocytic receptor, together with other relevant antibodies or ligands, can elicit durable and robust T cell responses against viral pathogens [31].

4. EBOV GP’s Affinity for Dendritic Cells and Macrophages in Ebola Virus Infection: An Insight for Vaccine Development

EBOV infects the macrophages and DCs by binding its GP with the CLRs on DCs/macrophages [86,87]. C-type lectins (CLRs) present on DCs interact with N- and O-linked glycans on GP1 (RBD, MLD, and glycan cap) (Figure 1) to facilitate viral entry. Unlike HIV-1 GP, EBOV GP has a variety of receptors on DCs and macrophages, making them an efficient and better stimulator of antigen-presenting cells. There are four members of CLECs that have been identified as Ebola GP receptors, namely, CLEC4G/LSECtin, dendritic cell-specific ICAM-3-grabbing non-integrin (DC-SIGN), liver/lymph node-specific ICAM-3 grabbing non-integrin (L-SIGN), asialoglycoprotein receptor 1 (ASGPRI) and human macrophage galactose- and acetylgalactosamine-specific C-type lectin (hMGL) [147,148]. CLECs are on the liver, alveolar macrophages, and epithelial cells [117,149,150]. Besides, the expressions of Nieman-Pick C1 (NPC1), integrin αV, and Mer have been reported to be essential for the infection of macrophages by EBOV GP. Although, some other receptors have also been previously reported for EBOV GP, including TAM receptor tyrosine kinases (Axl and Tyro3), T cell immunoglobulin and mucin domain (TIM proteins) [151], recent findings have demonstrated that TAM and TIM do not contribute to the EBOV GP-driven transduction of macrophages [152]. We therefore describe the three (3) major receptors on the DCs specific for EBOV GP below:
DC-SIGN: These are type II membrane protein and are expressed primarily on immature DCs [110]. DC-SIGN is involved in the initial mediation of immune responses by coordinating the DC interaction with T-lymphocytes and endothelial cells [110]. Other viruses such as measles [153], HIV [154,155], influenza virus [156], and HPV L1 [157] have specific receptors for DC-SIGN, which also help to internalize the virus into the DC for processing. The virulence of different species of Ebola virus, ranging from Reston Ebola Virus (REBOV) to Zaire Ebola Virus, depends on the differences in the N-glycan composition of their glycoprotein [158,159]. Thus, the large proportion of the high mannose N-glycans allows EBOV GP to interact with DC-SIGN and further leads to the induction of immune responses. L-SIGN, a homolog of DC-SIGN expressed on the endothelial cells in the placental villi, lymph node sinuses, and liver also has high mannose N-glycans for binding with EBOV GP. Development of an efficient vaccine depends on the antigenic or virulent factor of the invading pathogen; thus, studies for development of a DC-targeting vaccine can employ the modification of the N-glycans that target DC-SIGN(R) [160].
LSECtin: LSECtin, which is also known as CLEC4G, also mediates EBOV GP–DCs interaction to stimulate inflammatory responses. Liver and lymph nodes, sinusoidal endothelial cells express LSECtin [161], and Domínguez-Soto has also reported the expression of LSECtin in DCs and macrophages [162]. LSECtin also plays a vital role in the pathogenicity of the EBOV by serving as a receptor for GP1 for EBOV internalization [105,156,158]. Zhao et al. demonstrated that LSECtin can induce TNF-α and IL-6 production in DCs, suggesting that LSECtin can aid GP in inducing inflammatory responses [109]. In contrast to DC-SIGN and other glycan-binding receptors, the antibody-induced internalization by LSECtin on myeloid cells is not in clathrin-mediated endocytosis, but could aid the antigen capturing and presentation by DCs and macrophages [111,163]. Unlike most lectins, LSECtin does not interact with many viruses, but has a strong affinity for EBOLA GP and not HIV-1 GP. Gramberg et al. (2008) also showed that LSECtin and DC-SIGN act differently in the ways they capture pathogen, and even in the lectin biological functions [163].
hMGLs: Human macrophage galactose-type C-type lectins (hMGLs, CD301) are also transmembrane II proteins and play a critical role in the pathogenesis of EBOV. They are expressed on the DCs and macrophages and enhance adhesion of cells, internalization, and hematopoiesis [113]. The hMGLs have two homologs of MGL: MGL 1 (CD301a) and MGL 2 (CD301b) [112]; however, MGL 1 and MGL 2 have an affinity for Lewis trisaccharide (Galβ1–4[Fucα1–3]GlcNAc) and N-acetylgalactosamine, respectively [164]. hMGLs expressed on the monocyte-derived immature dendritic cells (MDDCs) and macrophages function as an endocytic receptor for galactosylated GP antigens [165]. In the affinity of hMGLs and EBOV GP, the highly glycosylated mucin-like-domain must be present for efficient interaction [165]. Usami et al. demonstrated that EBOV GP2 interacts with hMGLs of macrophages and DCs via the N-acetylgalactosamine for cell entry of the virus and to initiate infection [166].
In all, the N-glycan moieties and N-acetylgalactosamine present on the EBOV GP1 are essential features for the binding to the CLRs on the macrophages and DCs. Further modification of the binding sites on EBOV GP can influence the binding efficiency of EBOV with lectin receptors and other cellular factors to facilitate the activation of APCs.
Moreover, another possible receptor on DCs has been identified to have an affinity for EBOV GP. This receptor is known to be a hydrophobic Neimann-Pick C1 (NPC-1) receptor-binding pocket. Bornholdt et al. showed that EBOV GP could bind with the endosomal (NPC-1) receptor on DCs, initially by interacting electrostatically with the NPC-1 by the hydrophilic crest on GP1, while hydrophobic trough exposure on GP1 facilitates specific interactions due to their ability to migrate to lymph nodes where they can interact with DCs [114]. Their results revealed that mutation of the GP1 to block the hydrophilic and hydrophobic sites on the GP1 inhibit infectivity and binding of GP1 with NPC-1. They also demonstrated that the observed interaction leads to the stimulation of monoclonal neutralizing antibodies. This important finding is significant in developing a DC-targeting vaccine using EBOV GP. The hydrophilic crest and the hydrophobic trough of EBOV GP1 can be used heterogeneously with other viral protein to direct these peptides to DCs.

5. Ebola GP-Targeting DCs Can Facilitate Immune Responses for an Antiviral Vaccine Approach

An essential feature of antigenic agents is the ability to induce innate and adaptive immune response, as well as humoral and cellular immune responses. It is interesting to find out that the affinity of EBOV GP with DCs and macrophages can not only induce an adaptive immune response by recruitment of DCs/macrophages and facilitation of the maturation of DCs /macrophages [63], but can also induce innate immune responses which can serve as protection against other viruses. In the development of Ebola virus vaccine, EBOV GP has been shown to play a significant role, as both the VLP and vector-based approach depend so much of Ebola virus. [167]. Briefly, we will elucidate how EBOV GP can be used to stimulate DCs and macrophages for vaccine production.

5.1. EBOV GP-Coated Virus-Like Particle Vaccine Approach (VLP)

EBOV GP can be incorporated into VLPs to enhance the stimulation of DCs and macrophages, which, in turn, function in inducing adaptive immunity and interact with innate immune cells (Figure 1B) [168]. The efficiency of VLPs is undoubtful as it has succeeded in the induction of immune responses against several viruses, such as Rotavirus, among others [169]. Considering the high immunological characteristics of EBOV GP, it was co-expressed with matrix protein (VP40) to produce VLP. Warfield et al. showed that EBOV GP VLPs are immunogenic by facilitating the maturation of macrophages and DCs to induce the secretion of IL-10, IL-6, tumor necrosis factor α, and macrophage inflammatory protein (MIP)-1α [169]. This immunogenic property of EBOV GP VLP suggests that EBOV GP VLP is a promising tool for the development of a vaccine for Ebola. Moreover, EBOV GP is relevant as a tool to develop a universal vaccine against other viruses due to its ability to induce innate immune responses. There are ongoing clinical trials to test the efficiency of EBOV GP VLP vaccine against EBOV [140]. A recent study showed that EBOV GP VLP (consisting of VP40, NP, and GP) enhances the stimulation of DCs and macrophages [170]. Also, Venezuelan Equine Encephalitis (VEE), virus-like replicon particles with the replacement of VEE virus structural genes by EBOV GP or NP, has been demonstrated to have full protection against Ebola virus challenge [171]. Interestingly, our recent study showed that the incorporation of EBOV GP into the HIV VLP induces a more effective immune response against HIV-1 [115]. We showed that the presence of EBOV GP enhances the ability of the HIV VLP to target MDMs and MDDCs. Also, we revealed that EBOV GP-pseudotyped HIV VLP induces a significantly stronger humoral immune response than that of HIV VLP alone. Furthermore, macrophages inflammatory cytokines (MIP-1α) is significantly induced in the spleen by EBOV GP-pseudotyped HIV VLP more than HIV VLPs [115]. The heterogenic induction of immune responses by EBOV GP suggests that the immunogenicity of EBOV GP is not only beneficial in the development of a vaccine for EBOV, but can also be used to develop a vaccine for some other infectious diseases. Also, Wong et al. incorporated HA of H5N1 into the VSVΔG-ZGP (a previously described vaccine for EBOV) to form a bivalent vaccine, VSVΔG-HA-ZGP, which protects against both EBOV and H5N1 lethal challenge [116]. In this study, the presence of EBOV GP targeted the peptides of influenza HA to DCs/macrophages, which processed and presented the HA peptide, on MHC I or II for the eliciting of T-cells specific for influenza H5. [116]. Also, Chahal et al. also demonstrated that an adjuvant free dendrimer nanoparticle vaccine has broad protection against Ebola virus, influenza H1N1, and Toxoplasma gondii [172]. The eliciting of immune responses by this vaccine depends on dendrimer nanoparticle vaccine platform in which a dendrimer nanoparticle is encapsulated with mRNA replicons to generate specific CD8+ T cell antibody responses.

5.2. EBOV GP and Vector-Based Vaccine

Different vector-based vaccines are also an effective platform for the development of a vaccine for EBOV, ranging from vaccinia virus-based vaccines expressing ZEBOV GP, VP24, VP35, and VP40 [173], adenovirus-based vaccines having ZEBOV GP (AdHu5-ZGP), and combination with ZEBOV NP, SEBOV GP, and ICEBOV GP as a DNA vaccine [167] and Vesicular stomatitis virus (VSV)-based candidate vaccines [167,174]. The use of recombinant VSV to develop a candidate vaccine induces a strong humoral and cellular immune response and gives 100% protection in an animal model [167,174]. VSV used as an expressing vector for foreign proteins has a small amenable genome feature for genetic manipulation; thus, it is suitable for vaccine development [174]. Furthermore, the efficacy of chimpanzee adenovirus three vectored vaccine expressing EBOV GP has been demonstrated both in monovalent and bivalent forms in clinical trials in the UK, Europe, the USA, Nigeria, and Mali [140,175,176]. Also, in a clinical trial, a modified vaccinia Ankara vectored quadrivalent vaccine consisting of GPs of EBOV, Sudan Ebola virus, and Marburg virus and NP from the Tai forest strain boosted the humoral and cellular adaptive immune system, including neutralizing antibodies [170]. And recently, Zhu et al. showed that recombinant human adenovirus-vectored vaccine (rAd5—vectored vaccine) encoding GP is safe, with very high immunogenicity among adults in Sierra Leone and China with the requirement of high dose [177]. The efficacy of VSV–EBOV has been demonstrated in cynomolgus macaques to give 100% protection [21]. Also, human phase 1–3 trials have revealed the effectiveness of VSV–EBOV GP in inducing an immune response against EBOV [100,117]. Thus, EBOV GP infused with a specific viral antigenic protein can be incorporated into VSV as a vector-based vaccine (Figure 1C) to induce stronger and more robust immune responses against the specific virus [116].

6. Conclusions

Dendritic cell-targeted vaccines and EBOV GP-based vaccines are very potent, long-lasting, durable, and safe vaccines [31]. Based on the ability of EBOV GP to induce the stimulation of DCs and macrophages for the modulation of the cellular and humoral adaptive immune responses, EBOV GP can be used for the development of a DC-targeting vaccine approach and used as a natural adjuvant to elicit robust acquired immune responses. Since adjuvants are substances which can either be biological (microbial products, saponins, cytokines, and liposomes), chemical (mineral salts, polymers, and emulsions), or even particles (microparticles and nanoparticles) that can aid in the production of robust and stronger immune responses when combined with a specific antigen more than using the antigen alone [178], we propose that EBOV GP can also serve as natural adjuvant with no adverse effects. As mentioned above, since EBOV GP can target different peptides to the DCs, it can thus aid in the production of robust immune responses for specific antigens. Few pieces of research, as earlier mentioned, support this claim. Therefore, further investigations are recommended for the usage of EBOV GP as a substance to direct specific antigens to DCs for induction of stronger immune responses than what the peptide can produce alone.
Likewise, since EBOV GP1,2 also play an essential role in the stimulation of innate immune responses, this viral glycoprotein can be used to stimulate the induction of inflammatory cytokines. Using the technology of EBOV GP VLP immunological basis, it is possible to incorporate EBOV GP VLPs with other viral antigens to induce strong humoral and cellular adaptive immune responses (Figure 2). Since studies have established the immunogenic properties of EBOV VLPs and successful development of EBOV VLP vaccines and DCs-targeted vaccines, these immunological approaches can be further researched to develop a vaccine for other viruses, including HIV, influenza, Zika viruses, and other epidemic and pandemic viral infections. Also, EBOV GP can be fused with other viral proteins and inserted in the deleted G domain of VSV as a vector-based vaccine to induce stronger immune responses. Having shown the potentials of EBOV GP in inducing robust immune responses by directing specific antigens of peptides to DCs, further studies are recommended using EBOV GP to develop innate and adaptive immune responses to any desired pathogen. Furthermore, it is expedient to investigate possible limitations and variations to this technology.

Author Contributions

T.A.O., Z.A., and M.M. wrote and revised the manuscript draft; G.A.K. and X.Y. provided conceptual advice and input to the manuscript, and reviewed and revised the paper.

Funding

T.A.O. is a recipient of the Research Manitoba/Children’s Hospital Research Institute of Manitoba Ph.D. Studentship Award. X.Y. holds a Research Manitoba Chair Award from the Manitoba Health Research Council (MHRC). This work was supported by the Canadian Institute of Health Research (CIHR)/MHRC RPP grant (#RPA-132176) to X.Y., and CIHR-IAVI Grant (# OVV-152411) to G.A.K.

Acknowledgments

We thank Chidi Onyejiegbu and Lijun Wang for helpful discussions.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Graham, B.S. Advances in antiviral vaccine development. Immunol. Rev. 2013, 255, 230–242. [Google Scholar] [CrossRef] [PubMed]
  2. Ada, G.L. The ideal vaccine. World J. Microbiol. Biotechnol. 1991, 7, 105–109. [Google Scholar] [CrossRef] [PubMed]
  3. Norrby, E. Yellow fever and max theiler: The only nobel prize for a virus vaccine. J. Exp. Med. 2007, 204, 2779–2784. [Google Scholar] [CrossRef] [PubMed]
  4. He, H. Vaccines and antiviral agents. In Current Issues in Molecular Virology-Viral Genetics and Biotechnological Applications; InTech: London, UK, 2013. [Google Scholar] [CrossRef]
  5. World Health Organization. WHO Position paper on vaccines and vaccination against yellow fever. Weekly epidemiological record. Available online: https://www.who.int/wer/2013/wer8827.pdf?ua=1 (accessed on 5 July 2013).
  6. Grohskopf, L.A.; Sokolow, L.Z.; Broder, K.R.; Walter, E.B.; Fry, A.M.; Jernigan, D.B. Prevention and control of seasonal influenza with vaccines: Recommendations of the advisory committee on immunization practices-”united states, 2018–2019 influenza season. Mmwr Recomm. Rep. 2018, 67, 1–20. [Google Scholar] [CrossRef] [PubMed]
  7. Sequeira, D.P.; Correia, R.; Carrondo, M.J.T.; Roldao, A.; Teixeira, A.P.; Alves, P.M. Combining stable insect cell lines with baculovirus-mediated expression for multi-ha influenza vlp production. Vaccine 2018, 36, 3112–3123. [Google Scholar] [CrossRef] [PubMed]
  8. Karlsson, I.; Borggren, M.; Rosenstierne, M.W.; Trebbien, R.; Williams, J.A.; Vidal, E.; Vergara-Alert, J.; Foz, D.S.; Darji, A.; Sisteré-Oró, M.; et al. Protective effect of a polyvalent influenza DNA vaccine in pigs. Vet. Immunol. Immunopathol. 2018, 195, 25–32. [Google Scholar] [CrossRef]
  9. Jorba, J.; Diop, O.M.; Iber, J.; Henderson, E.; Sutter, R.W.; Wassilak, S.G.F.; Burns, C.C. Update on vaccine-derived polioviruses-worldwide, january 2016–june 2017. Mmwr. Morb. Mortal. Wkly. Rep. 2017, 66, 1185–1191. [Google Scholar] [CrossRef]
  10. Scott, H. James, Measles, mumps and rubella viruses. In Infectious Diseases; Elsevier: Amsterdam, The Netherlands, 2017; pp. 1399–1405.e1391. [Google Scholar]
  11. Van Damme, P.; Bonanni, P.; Bosch, F.X.; Joura, E.; Kjaer, S.K.G.; Meijer, C.J.L.M.; Petry, K.-U.; Soubeyrand, B.; Verstraeten, T.; Stanley, M. Use of the nonavalent hpv vaccine in individuals previously fully or partially vaccinated with bivalent or quadrivalent hpv vaccines. Vaccine 2016, 34, 757–761. [Google Scholar] [CrossRef]
  12. Uddin, M.N. Formulation of Bioactive Molecules: Antisense Oliginucleotide, Dexamethasone, and Human Papillomavirus (hpv) in a Novel Particulate Drug Delivery System. Ph.D. Thesis, Mercer University, Macon, GA, USA, September 2011. [Google Scholar]
  13. Zanetti, A.R.; Van Damme, P.; Shouval, D. The global impact of vaccination against hepatitis b: A historical overview. Vaccine 2008, 26, 6266–6273. [Google Scholar] [CrossRef]
  14. McAleer, W.J.; Buynak, E.B.; Maigetter, R.Z.; Wampler, D.E.; Miller, W.J.; Hilleman, M.R. Human hepatitis b vaccine from recombinant yeast. Nature 1984, 307, 178. [Google Scholar] [CrossRef]
  15. Mast, E.; Mahoney, F.; Kane, M.; Margolis, H. Hepatitis B vaccine. In Vaccines, 4th ed.; Plotkin, S.A., Orenstein, W.A., Eds.; W.B. Saunders Co: St. Louis, MO, USA, 2004; pp. 299–314. [Google Scholar]
  16. Gillet, Y.; Habermehl, P.; Thomas, S.; Eymin, C.; Fiquet, A. Immunogenicity and safety of concomitant administration of a measles, mumps and rubella vaccine (M-M-RvaxPro®) and a varicella vaccine (VARIVAX®) by intramuscular or subcutaneous routes at separate injection sites: A randomised clinical trial. BMC Med. 2009, 7, 16–27. [Google Scholar] [CrossRef] [PubMed]
  17. O‘Neal, C.M.; Crawford, S.E.; Estes, M.K.; Conner, M.E. Rotavirus virus-like particles administered mucosally induce protective immunity. J. Virol. 1997, 71, 8707–8717. [Google Scholar] [PubMed]
  18. Deen, J.; Lopez, A.L.; Kanungo, S.; Wang, X.-Y.; Anh, D.D.; Tapia, M.; Grais, R.F. Improving rotavirus vaccine coverage: Can newer-generation and locally produced vaccines help? Hum. Vaccines Immunother. 2018, 14, 495–499. [Google Scholar] [CrossRef] [PubMed]
  19. Tohidi, F.; Sadat, S.M.; Bolhassani, A.; Yaghoubi, R.; Larijani, M.S. Induction of a robust humoral response using HIV-1 VLPMPER-V3 as a novel candidate vaccine in BALB/C mice. Curr. HIV Res. 2019, 17, 33–41. [Google Scholar] [CrossRef] [PubMed]
  20. Huang, X.; Zhu, Q.; Huang, X.; Yang, L.; Song, Y.; Zhu, P.; Zhou, P. In vivo electroporation in DNA-VLP prime-boost preferentially enhances HIV-1 envelope-specific IgG2a, neutralizing antibody and CD8 T cell responses. Vaccine 2017, 35, 2042–2051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Jones, S.M.; Feldmann, H.; Ströher, U.; Geisbert, J.B.; Fernando, L.; Grolla, A.; Klenk, H.D.; Sullivan, N.J.; Volchkov, V.E.; Fritz, E.A.; et al. Live attenuated recombinant vaccine protects nonhuman primates against ebola and marburg viruses. Nat. Med. 2005, 11, 786–790. [Google Scholar] [CrossRef] [PubMed]
  22. Kennedy, S.B.; Bolay, F.; Kieh, M.; Grandits, G.; Badio, M.; Ballou, R.; Eckes, R.; Feinberg, M.; Follmann, D.; Grund, B.; et al. Phase 2 placebo-controlled trial of two vaccines to prevent ebola in liberia. N. Engl. J. Med. 2017, 377, 1438–1447. [Google Scholar] [CrossRef]
  23. Wang, M.; Jokinen, J.; Tretyakova, I.; Pushko, P.; Lukashevich, I.S. Alphavirus vector-based replicon particles expressing multivalent cross-protective lassa virus glycoproteins. Vaccine 2018, 36, 683–690. [Google Scholar] [CrossRef]
  24. McCormick, J.B.; Mitchell, S.W.; Kiley, M.P.; Ruo, S.; Fisher-Hoch, S.P. Inactivated lassa virus elicits a non protective immune response in rhesus monkeys. J. Med. Virol. 1992, 37, 1–7. [Google Scholar] [CrossRef]
  25. Cashman, K.A.; Wilkinson, E.R.; Wollen, S.E.; Shamblin, J.D.; Zelko, J.M.; Bearss, J.J.; Zeng, X.; Broderick, K.E.; Schmaljohn, C.S. DNA vaccines elicit durable protective immunity against individual or simultaneous infections with lassa and ebola viruses in guinea pigs. Hum. Vaccines Immunother. 2017, 13, 3010–3019. [Google Scholar] [CrossRef]
  26. Gans, H.; Yasukawa, L.; Rinki, M.; DeHovitz, R.; Forghani, B.; Beeler, J.; Audet, S.; Maldonado, Y.; Arvin, A.M. Immune responses to measles and mumps vaccination of infants at 6, 9, and 12 months. J. Infect. Dis. 2001, 184, 817–826. [Google Scholar] [CrossRef] [PubMed]
  27. Meixlsperger, S.; Leung, C.S.; Rämer, P.C.; Pack, M.; Vanoaica, L.D.; Breton, G.; Pascolo, S.; Salazar, A.M.; Dzionek, A.; Schmitz, J. Cd141+ dendritic cells produce prominent amounts of ifn-γ after dsrna recognition and can be targeted via dec-205 in humanized mice. Blood 2013, 121, 5034–5044. [Google Scholar] [CrossRef] [PubMed]
  28. Banchereau, J.; Palucka, A.K. Dendritic cells as therapeutic vaccines against cancer. Nat. Rev. Immunol. 2005, 5, 296–306. [Google Scholar] [CrossRef] [PubMed]
  29. Brandão, J.G.; Scheper, R.J.; Lougheed, S.A.M.; Curiel, D.T.; Tillman, B.W.; Gerritsen, W.R.; Van Den Eertwegh, A.J.M.; Pinedo, H.M.; Haisma, H.J.; De Gruijl, T.D. Cd40-targeted adenoviral gene transfer to dendritic cells through the use of a novel bispecific single-chain FV antibody enhances cytotoxic T cell activation. Vaccine 2003, 21, 2268–2272. [Google Scholar] [CrossRef]
  30. Querec, T.; Bennouna, S.; Alkan, S.; Laouar, Y.; Gorden, K.; Flavell, R.; Akira, S.; Ahmed, R.; Pulendran, B. Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity. J. Exp. Med. 2006, 203, 413–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Chen, P.; Liu, X.; Sun, Y.; Zhou, P.; Wang, Y.; Zhang, Y. Dendritic cell targeted vaccines: Recent progresses and challenges. Hum. Vaccines Immunother. 2016, 12, 612–622. [Google Scholar] [CrossRef]
  32. Hawiger, D.; Inaba, K.; Dorsett, Y.; Guo, M.; Mahnke, K.; Rivera, M.; Ravetch, J.V.; Steinman, R.M.; Nussenzweig, M.C. Dendritic cells induce peripheral t cell unresponsiveness under steady state conditions in vivo. J. Exp. Med. 2001, 194, 769–780. [Google Scholar] [CrossRef] [PubMed]
  33. Zaneti, A.B.; Yamamoto, M.M.; Sulczewski, F.B.; Almeida, B.d.S.; Souza, H.F.S.; Ferreira, N.l.S.; Maeda, D.L.N.F.; Sales, N.S.; Rosa, D.S.; Ferreira, S.; et al. Dendritic cell targeting using a DNA vaccine induces specific antibodies and CD4+ T cells to the dengue virus envelope protein domain III. Front. Immunol. 2019, 10, 1–15. [Google Scholar] [CrossRef]
  34. Cheng, C.; Gall, J.G.D.; Kong, W.-P.; Sheets, R.L.; Gomez, P.L.; King, C.R.; Nabel, G.J. Mechanism of AD5 vaccine immunity and toxicity: Fiber shaft targeting of dendritic cells. PLoS Pathog. 2007, 3, e25–e32. [Google Scholar] [CrossRef]
  35. Boudewijns, S.; Westdorp, H.; Koornstra, R.H.T.; Aarntzen, E.H.J.G.; Schreibelt, G.; Creemers, J.H.A.; Punt, C.J.A.; Figdor, C.G.; de Vries, I.J.M.; Gerritsen, W.R. Immune-related adverse events of dendritic cell vaccination correlate with immunologic and clinical outcome in stage iii and iv melanoma patients. J. Immunother. (Hagerstown Md. 1997) 2016, 39, 241–248. [Google Scholar] [CrossRef]
  36. Almand, B.; Clark, J.I.; Nikitina, E.; van Beynen, J.; English, N.R.; Knight, S.C.; Carbone, D.P.; Gabrilovich, D.I. Increased production of immature myeloid cells in cancer patients: A mechanism of immunosuppression in cancer. J. Immunol. 2001, 166, 678–689. [Google Scholar] [CrossRef] [PubMed]
  37. Apostólico, J.d.S.; Lunardelli, V.A.S.; Yamamoto, M.M.; Souza, H.F.S.; Cunha-Neto, E.; Boscardin, S.B.; Rosa, D.S. Dendritic cell targeting effectively boosts t cell responses elicited by an hiv multiepitope DNA vaccine. Front. Immunol. 2017, 8, 101–117. [Google Scholar] [CrossRef]
  38. Graham, J.P.; Authie, P.; Palucka, A.K.; Zurawski, G. Targeting interferon-alpha to dendritic cells enhances a CD8+ T cell response to a human CD40-targeted cancer vaccine. Vaccine 2017, 35, 4532–4539. [Google Scholar] [CrossRef] [PubMed]
  39. Garu, A.; Moku, G.; Gulla, S.K.; Chaudhuri, A. Genetic immunization with in vivo dendritic cell-targeting liposomal DNA vaccine carrier induces long-lasting antitumor immune response. Mol. Ther. 2016, 24, 385–397. [Google Scholar] [CrossRef] [PubMed]
  40. Nchinda, G.; Kuroiwa, J.; Oks, M.; Trumpfheller, C.; Park, C.G.; Huang, Y.; Hannaman, D.; Schlesinger, S.J.; Mizenina, O.; Nussenzweig, M.C. The efficacy of DNA vaccination is enhanced in mice by targeting the encoded protein to dendritic cells. J. Clin. Investig. 2008, 118, 1427–1436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Fossum, E.; Gradeland, G.; Terhorst, D.; Tveita, A.A.; Vikse, E.; Mjaaland, S.; Henri, S.; Malissen, B.; Bogen, B. Vaccine molecules targeting Xcr1 on cross-presenting dcs induce protective CD8+ T-cell responses against influenza virus. Eur. J. Immunol. 2014, 45, 624–635. [Google Scholar] [CrossRef]
  42. Gudjonsson, A.; Lysén, A.; Balan, S.; Sundvold-Gjerstad, V.; Arnold-Schrauf, C.; Richter, L.; Bækkevold, E.S.; Dalod, M.; Bogen, B.; Fossum, E. Targeting influenza virus hemagglutinin to Xcr1+ dendritic cells in the absence of receptor-mediated endocytosis enhances protective antibody responses. J. Immunol. 2017, 198, 2785–2795. [Google Scholar] [CrossRef] [PubMed]
  43. Ye, C.; Choi, J.G.; Abraham, S.; Shankar, P.; Manjunath, N. Targeting DNA vaccines to myeloid cells using a small peptide. Eur. J. Immunol. 2015, 45, 82–88. [Google Scholar] [CrossRef] [PubMed]
  44. Varol, C.; Mildner, A.; Jung, S. Macrophages: Development and tissue specialization. Annu. Rev. Immunol. 2015, 33, 643–675. [Google Scholar] [CrossRef]
  45. Steinman, R.M.; Hemmi, H. Dendritic cells: Translating innate to adaptive immunity. In From Innate Immunity to Immunological Memor; Springer: Berlin, Heidelberg, 2006; pp. 17–58. [Google Scholar]
  46. Jego, G.; Palucka, A.K.; Blanck, J.-P.; Chalouni, C.; Pascual, V.; Banchereau, J. Plasmacytoid dendritic cells induce plasma cell differentiation through type i interferon and interleukin 6. Immunity 2003, 19, 225–234. [Google Scholar] [CrossRef]
  47. Fernandez, N.C.; Lozier, A.; Flament, C.; Ricciardi-Castagnoli, P.; Bellet, D.; Suter, M.; Perricaudet, M.; Tursz, T.; Maraskovsky, E.; Zitvogel, L. Dendritic cells directly trigger nk cell functions: Cross-talk relevant in innate anti-tumor immune responses in vivo. Nat. Med. 1999, 5, 405–411. [Google Scholar] [CrossRef] [PubMed]
  48. Heink, S.; Yogev, N.; Garbers, C.; Herwerth, M.; Aly, L.; Gasperi, C.; Husterer, V.; Croxford, A.L.; Möller-Hackbarth, K.; Bartsch, H.S.; et al. Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells. Nat. Immunol. 2017, 18, 74–85. [Google Scholar] [CrossRef] [PubMed]
  49. Humeniuk, P.; Dubiela, P.; Hoffmann-Sommergruber, K. Dendritic cells and their role in allergy: Uptake, proteolytic processing and presentation of allergens. Int. J. Mol. Sci. 2017, 18, 1491. [Google Scholar] [CrossRef] [PubMed]
  50. Chun, I.Y.; Becker, C.; Metang, P.; Marches, F.; Wang, Y.; Toshiyuki, H.; Banchereau, J.; Merad, M.; Palucka, K. Human CD141+ dendritic cells induce CD4+ T cells to produce type 2 cytokines. J. Immunol. 2014, 193, 4335–4343. [Google Scholar]
  51. Figdor, C.G.; van Kooyk, Y.; Adema, G.J. C-type lectin receptors on dendritic cells and langerhans cells. Nat. Rev. Immunol. 2002, 2, 77–84. [Google Scholar] [CrossRef] [PubMed]
  52. Sehgal, K.; Dhodapkar, K.M.; Dhodapkar, M.V. Targeting human dendritic cells in situ to improve vaccines. Immunol. Lett. 2014, 162, 59–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Scott, C.L.; Aumeunier, A.M.; Mowat, A.M. Intestinal CD103+ dendritic cells: Master regulators of tolerance? Trends Immunol. 2011, 32, 412–419. [Google Scholar] [CrossRef]
  54. Christmas, P. Toll-like receptors: Sensors that detect infection. Nat. Educ. 2010, 3, 85. [Google Scholar]
  55. Arpaia, N.; Barton, G.M. The impact of toll-like receptors on bacterial virulence strategies. Curr. Opin. Microbiol. 2013, 16, 17–22. [Google Scholar] [CrossRef]
  56. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on toll-like receptors. Nat. Immunol. 2010, 11, 373. [Google Scholar] [CrossRef]
  57. Fukuda, D.; Nishimoto, S.; Aini, K.; Tanaka, A.; Nishiguchi, T.; Kim-Kaneyama, J.-R.; Lei, X.-F.; Masuda, K.; Naruto, T.; Tanaka, K. Toll-like receptor 9 plays a pivotal role in angiotensin II-induced atherosclerosis. J. Am. Heart Assoc. 2019, 8, e010860–e010884. [Google Scholar] [CrossRef] [PubMed]
  58. Koblansky, A.A.; Jankovic, D.; Oh, H.; Hieny, S.; Sungnak, W.; Mathur, R.; Hayden, M.S.; Akira, S.; Sher, A.; Ghosh, S. Recognition of profilin by toll-like receptor 12 is critical for host resistance to toxoplasma gondii. Immunity 2013, 38, 119–130. [Google Scholar] [CrossRef] [PubMed]
  59. Chaturvedi, A.; Pierce, S.K. How location governs toll-like receptor signaling. Traffic 2009, 10, 621–628. [Google Scholar] [CrossRef] [PubMed]
  60. Raetz, M.; Kibardin, A.; Sturge, C.R.; Pifer, R.; Li, H.; Burstein, E.; Ozato, K.; Larin, S.; Yarovinsky, F. Cooperation of TLR12 and TLR11 in the IRF8-dependent IL-12 response to toxoplasma gondii profilin. J. Immunol. 2013, 191, 4818–4827. [Google Scholar] [CrossRef]
  61. Majewska, M.; Szczepanik, M. The role of toll-like receptors (TLR) in innate and adaptive immune responses and their function in immune response regulation. Postepy Hig. I Med. Dosw. (Online) 2006, 60, 52–63. [Google Scholar] [PubMed]
  62. Okumura, A.; Pitha, P.M.; Yoshimura, A.; Harty, R.N. Interaction between ebola virus glycoprotein and host toll-like receptor 4 leads to induction of proinflammatory cytokines and socs1. J. Virol. 2010, 84, 27–33. [Google Scholar] [CrossRef] [PubMed]
  63. Lai, C.-Y.; Strange, D.P.; Wong, T.A.S.; Lehrer, A.T.; Verma, S. Ebola virus glycoprotein induces an innate immune response in vivo via TLR4. Front. Microbiol. 2017, 8, 1571–1586. [Google Scholar] [CrossRef] [PubMed]
  64. Goubau, D.; Schlee, M.; Deddouche, S.; Pruijssers, A.J.; Zillinger, T.; Goldeck, M.; Schuberth, C.; Van der Veen, A.G.; Fujimura, T.; Rehwinkel, J. Antiviral immunity via RIG-I-mediated recognition of RNA bearing 5’-diphosphates. Nature 2017, 514, 372–375. [Google Scholar] [CrossRef]
  65. Martinez, I.; Oliveros, J.C.; Cuesta, I.; de la Barrera, J.; Ausina, V.; Casals, C.; de Lorenzo, A.; Garcia, E.; Garcia-Fojeda; Garmendia, J. Apoptosis, toll-like, RIG-I-like and nod-like receptors are pathways jointly induced by diverse respiratory bacterial and viral pathogens. Front. Microbiol. 2017, 8, 276. [Google Scholar] [CrossRef]
  66. Weber, M.; Gawanbacht, A.; Habjan, M.; Rang, A.; Borner, C.; Schmidt, A.M.; Veitinger, S.; Jacob, R.; Devignot, S.P.; Kochs, G. Incoming RNA virus nucleocapsids containing a 5’-triphosphorylated genome activate RIG-I and antiviral signaling. Cell Host Microbe 2013, 13, 336–346. [Google Scholar] [CrossRef]
  67. He, F.; Melen, K.; Maljanen, S.; Lundberg, R.; Jiang, M.; Osterlund, P.; Kakkola, L.; Julkunen, I. Ebolavirus protein VP24 interferes with innate immune responses by inhibiting interferon-λ1 gene expression. Virology 2017, 509, 23–34. [Google Scholar] [CrossRef] [PubMed]
  68. Ting, J.P.Y.; Duncan, J.A.; Lei, Y. How the noninflammasome nlrs function in the innate immune system. Science 2010, 327, 286–290. [Google Scholar] [CrossRef] [PubMed]
  69. Inohara, N.; Ogura, Y.; Fontalba, A.; Gutierrez, O.; Pons, F.; Crespo, J.; Fukase, K.; Inamura, S.; Kusumoto, S.; Hashimoto, M. Host recognition of bacterial muramyl dipeptide mediated through NOD2 implications for crohn’s disease. J. Biol. Chem. 2003, 278, 5509–5512. [Google Scholar] [CrossRef] [PubMed]
  70. Heath, W.R.; Carbone, F.R. Dendritic cell subsets in primary and secondary T cell responses at body surfaces. Nat. Immunol. 2009, 10, 1237–1244. [Google Scholar] [CrossRef]
  71. Ueno, H.; Schmitt, N.; Palucka, A.K.; Banchereau, J. Dendritic cells and humoral immunity in humans. Immunol. Cell Biol. 2010, 88, 376–380. [Google Scholar] [CrossRef]
  72. Uematsu, S.; Fujimoto, K.; Jang, M.H.; Yang, B.-G.; Jung, Y.-J.; Nishiyama, M.; Sato, S.; Tsujimura, T.; Yamamoto, M.; Yokota, Y. Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing toll-like receptor 5. Nat. Immunol. 2008, 9, 769–776. [Google Scholar] [CrossRef]
  73. Steinman, R.M.; Banchereau, J. Taking dendritic cells into medicine. Nature 2007, 449, 419–426. [Google Scholar] [CrossRef] [PubMed]
  74. Dang, Y.; Wagner, W.M.; Gad, E.; Rastetter, L.; Berger, C.M.; Holt, G.E.; Disis, M.L. Dendritic cell-activating vaccine adjuvants differ in the ability to elicit antitumor immunity due to an adjuvant-specific induction of immunosuppressive cells. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2012, 18, 3122–3131. [Google Scholar] [CrossRef]
  75. Steinman, R.M.; Bona, C.; Inaba, K. Dendritic Cells: Important Adjuvants during DNA Vaccination; Landes Bioscience: Georgetown, TX, USA, 2002. [Google Scholar]
  76. Epelman, S.; Lavine, K.J.; Randolph, G.J. Origin and functions of tissue macrophages. Immunity 2014, 41, 21–35. [Google Scholar] [CrossRef]
  77. Sang, Y.; Miller, L.C.; Blecha, F. Macrophage polarization in virus-host interactions. J. Clin. Cell. Immunol. 2015, 6, 311–334. [Google Scholar]
  78. Delves, P.J.; Martin, S.J.; Burton, D.R.; Roitt, I.M. Essential Immunology; Chichester, West Sussex John Wiley & Sons: Hoboken, NJ, USA, 2017. [Google Scholar]
  79. Ferenbach, D.; Hughes, J. Macrophages and dendritic cells: What is the difference? Kidney Int. 2008, 74, 5–7. [Google Scholar] [CrossRef] [PubMed]
  80. Quattrocchi, V.; Langellotti, C.; Pappalardo, J.S.N.; Olivera, V.; Di Giacomo, S.N.; Van Rooijen, N.; Mongini, C.; Waldner, C.; Zamorano, P.I.S. Role of macrophages in early protective immune responses induced by two vaccines against foot and mouth disease. Antivir. Res. 2011, 92, 262–270. [Google Scholar] [CrossRef] [PubMed]
  81. Bermejo-Jambrina, M.; Eder, J.; Helgers, L.C.; Hertoghs, N.; Nijmeijer, B.M.; Stunnenberg, M.; Geijtenbeek, T.B.H. C-type lectin receptors in antiviral immunity and viral escape. Front. Immunol. 2018, 9, 590–602. [Google Scholar] [CrossRef] [PubMed]
  82. Kumar, A. Ebola virus altered innate and adaptive immune response signalling pathways: Implications for novel therapeutic approaches. Infect. Disord. Drug Targets (Former. Curr. Drug Targets-Infect. Disord.) 2016, 16, 79–94. [Google Scholar] [CrossRef]
  83. Bosio, C.M.; Aman, M.J.; Grogan, C.; Hogan, R.; Ruthel, G.; Negley, D.; Mohamadzadeh, M.; Bavari, S.; Schmaljohn, A. Ebola and marburg viruses replicate in monocyte-derived dendritic cells without inducing the production of cytokines and full maturation. J. Infect. Dis. 2003, 188, 1630–1638. [Google Scholar] [CrossRef] [PubMed]
  84. Marcinkiewicz, J.; Bryniarski, K.; Nazimek, K. Ebola haemorrhagic fever virus: Pathogenesis, immune responses, potential prevention. Folia Med. Cracov. 2014, 54, 39–48. [Google Scholar] [PubMed]
  85. Martinez, O.; Leung, L.W.; Basler, C.F. The role of antigen-presenting cells in filoviral hemorrhagic fever: Gaps in current knowledge. Antivir. Res. 2012, 93, 416–428. [Google Scholar] [CrossRef] [PubMed]
  86. Hensley, L.E.; Young, H.A.; Jahrling, P.B.; Geisbert, T.W. Proinflammatory response during ebola virus infection of primate models: Possible involvement of the tumor necrosis factor receptor superfamily. Immunol. Lett. 2002, 80, 169–179. [Google Scholar] [CrossRef]
  87. Marzi, A.; Muller, P.; Hanna, S.L.; Harrer, T.; Eisemann, J.; Steinkasserer, A.; Becker, S.; Baribaud, F.; Pohlmann, S. Analysis of the interaction of ebola virus glycoprotein with DC-SIGN (dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin) and its homologue dc-signr. J. Infect. Dis. 2007, 196, S237–S246. [Google Scholar] [CrossRef]
  88. Younan, P.; Iampietro, M.; Bukreyev, A. Disabling of lymphocyte immune response by Ebola virus. PLoS Pathog. 2018, 14, e1006932–e1006939. [Google Scholar] [CrossRef]
  89. Lubaki, N.M.; Ilinykh, P.; Pietzsch, C.; Tigabu, B.; Freiberg, A.N.; Koup, R.A.; Bukreyev, A. The lack of maturation of Ebola virus-infected dendritic cells results from the cooperative effect of at least two viral domains. J. Virol. 2013, 87, 7471–7485. [Google Scholar] [CrossRef] [PubMed]
  90. Lubaki, N.M.; Younan, P.; Santos, R.I.; Meyer, M.; Iampietro, M.; Koup, R.A.; Bukreyev, A. The ebola interferon inhibiting domains attenuate and dysregulate cell-mediated immune responses. PLoS Pathog. 2016, 12, e1006031–e1006065. [Google Scholar] [CrossRef] [PubMed]
  91. Groseth, A.; Marzi, A.; Hoenen, T.; Herwig, A.; Gardner, D.; Becker, S.; Ebihara, H.; Feldmann, H. The ebola virus glycoprotein contributes to but is not sufficient for virulence in vivo. PLoS Pathog. 2012, 8, e1002847–e1002859. [Google Scholar] [CrossRef] [PubMed]
  92. Baize, S.; Leroy, E.M.; Georges, A.J.; Georges-Courbot; Capron, M.; Bedjabaga, I.; Lansouda-Soukate, J.; Mavoungou, E. Inflammatory responses in ebola virus-infected patients. Clin. Exp. Immunol. 2002, 128, 163–168. [Google Scholar] [CrossRef] [PubMed]
  93. Leroy, E.M.; Gonzalez, J.-P.; Baize, S. Ebola and marburg haemorrhagic fever viruses: Major scientific advances, but a relatively minor public health threat for africa. Clin. Microbiol. Infect. 2011, 17, 964–976. [Google Scholar] [CrossRef] [PubMed]
  94. Ludtke, A.; Ruibal, P.; Becker-Ziaja, B.; Rottstegge, M.; Wozniak, D.M.; Cabeza-Cabrerizo, M.; Thorenz, A.; Weller, R.; Kerber, R.; Idoyaga, J. Ebola virus disease is characterized by poor activation and reduced levels of circulating CD16+ monocytes. J. Infect. Dis. 2016, 214, S275–S280. [Google Scholar] [CrossRef] [PubMed]
  95. Lüdtke, A.; Ruibal, P.; Wozniak, D.M.; Pallasch, E.; Wurr, S.; Bockholt, S.; Gómez-Medina, S.; Qiu, X.; Kobinger, G.P.; RodrÃguez, E. Ebola virus infection kinetics in chimeric mice reveal a key role of T cells as barriers for virus dissemination. Sci. Rep. 2017, 7, 43776–43786. [Google Scholar] [CrossRef] [PubMed]
  96. Silvin, A.; Chun, I.Y.; Lahaye, X.; Imperatore, F.; Brault, J.-B.; Cardinaud, S.; Becker, C.; Kwan, W.-H.; Conrad, C.; Maurin, M. Constitutive resistance to viral infection in human CD141+ dendritic cells. Sci. Immunol. 2017, 2, eaai8071–eaai8099. [Google Scholar] [CrossRef] [PubMed]
  97. Wahl-Jensen, V.; Kurz, S.; Feldmann, F.; Buehler, L.K.; Kindrachuk, J.; DeFilippis, V.; da Silva Correia, J.; Fruh, K.; Kuhn, J.H.; Burton, D.R. Ebola virion attachment and entry into human macrophages profoundly effects early cellular gene expression. PLoS Negl. Trop. Dis. 2011, 5, e1359–e1375. [Google Scholar] [PubMed]
  98. Rao, M.; Bray, M.; Alving, C.R.; Jahrling, P.; Matyas, G.R. Induction of immune responses in mice and monkeys to ebola virus after immunization with liposome-encapsulated irradiated ebola virus: Protection in mice requires cd4+ t cells. J. Virol. 2002, 76, 9176–9185. [Google Scholar] [CrossRef] [PubMed]
  99. Ayithan, N.; Bradfute, S.B.; Anthony, S.M.; Stuthman, K.S.; Dye, J.M.; Bavari, S.; Bray, M.; Ozato, K. Ebola virus-like particles stimulate type i interferons and proinflammatory cytokine expression through the toll-like receptor and interferon signaling pathways. J. Interferon Cytokine Res. 2014, 34, 79–89. [Google Scholar] [CrossRef] [PubMed]
  100. Regules, J.A.; Beigel, J.H.; Paolino, K.M.; Voell, J.; Castellano, A.R.; Hu, Z.; MuÃoz, P.; Moon, J.E.; Ruck, R.C.; Bennett, J.W. A recombinant vesicular stomatitis virus ebola vaccine. N. Engl. J. Med. 2017, 376, 330–341. [Google Scholar] [CrossRef] [PubMed]
  101. Lee, J.E.; Fusco, M.L.; Hessell, A.J.; Oswald, W.B.; Burton, D.R.; Saphire, E.O. Structure of the ebola virus glycoprotein bound to an antibody from a human survivor. Nature 2008, 454, 177. [Google Scholar] [CrossRef] [PubMed]
  102. Sanchez, A.; Trappier, S.G.; Mahy, B.W.; Peters, C.J.; Nichol, S.T. The virion glycoproteins of ebola viruses are encoded in two reading frames and are expressed through transcriptional editing. Proc. Natl. Acad. Sci. USA 1996, 93, 3602–3607. [Google Scholar] [CrossRef] [PubMed]
  103. Alazard-Dany, N.; Volchkova, V.; Reynard, O.; Carbonnelle, C.; Dolnik, O.; Ottmann, M.; Khromykh, A.; Volchkov, V.E. Ebola virus glycoprotein gp is not cytotoxic when expressed constitutively at a moderate level. J. Gen. Virol. 2006, 87, 1247–1257. [Google Scholar] [CrossRef] [PubMed]
  104. Volchkov, V.E.; Feldmann, H.; Volchkova, V.A.; Klenk, H.-D. Processing of the ebola virus glycoprotein by the proprotein convertase furin. Proc. Natl. Acad. Sci. USA 1998, 95, 5762–5767. [Google Scholar] [CrossRef]
  105. Lee, J.E.; Saphire, E.O. Ebolavirus glycoprotein structure and mechanism of entry. Future Virol. 2009, 4, 621–635. [Google Scholar] [CrossRef]
  106. Jeffers, S.A.; Sanders, D.A.; Sanchez, A. Covalent modifications of the ebola virus glycoprotein. J. Virol. 2002, 76, 12463–12472. [Google Scholar] [CrossRef]
  107. Martinez, O.; Tantral, L.; Mulherkar, N.; Chandran, K.; Basler, C.F. Impact of ebola mucin-like domain on antiglycoprotein antibody responses induced by Ebola virus-like particles. J. Infect. Dis. 2011, 204, S825–S832. [Google Scholar] [CrossRef]
  108. Lennemann, N.J.; Rhein, B.A.; Ndungo, E.; Chandran, K.; Qiu, X.; Maury, W. Comprehensive functional analysis of N-linked glycans on Ebola virus GP1. mBio 2014, 5, e00862-13. [Google Scholar] [CrossRef]
  109. Zhao, D.; Han, X.; Zheng, X.; Wang, H.; Yang, Z.; Liu, D.; Han, K.; Liu, J.; Wang, X.; Yang, W. The Myeloid LSECtin is a DAP12-coupled receptor that is crucial for inflammatory response induced by Ebola virus Glycoprotein. PLoS Pathog. 2016, 12, e1005487–e1005502. [Google Scholar] [CrossRef] [PubMed]
  110. Alvarez, C.P.; Lasala, F.; Carrillo, J.; Muiz, O.; Corbi, A.L.; Delgado, R. C-type lectins DC-SIGN and L-SIGN mediate cellular entry by Ebola virus in cis and in trans. J. Virol. 2002, 76, 6841–6844. [Google Scholar] [CrossRef] [PubMed]
  111. Domínguez-Soto, A.A.; Aragoneses-Fenoll, L.; Martin-Gayo, E.; Martinez-Prats, L.; Colmenares, M.; Naranjo-Gomez, M.; Borras, F.E.; Munoz, P.; Zubiaur, M.; Toribio, M.L. The DC-SIGN-“related lectin LSECtin mediates antigen capture and pathogen binding by human myeloid cells. Blood 2007, 109, 5337–5345. [Google Scholar] [CrossRef] [PubMed]
  112. Sato, K.; Imai, Y.; Higashi, N.; Kumamoto, Y.; Onami, T.M.; Hedrick, S.M.; Irimura, T. Lack of antigen-specific tissue remodeling in mice deficient in the macrophage galactose-type calcium-type LECtin 1/CD301A. Blood 2005, 106, 207–215. [Google Scholar] [CrossRef] [PubMed]
  113. Sato, K.; Imai, Y.; Higashi, N.; Kumamoto, Y.; Mukaida, N.; Irimura, T. Redistributions of macrophages expressing the macrophage galactose-type C-type lectin (MGL) during antigen-induced chronic granulation tissue formation. Int. Immunol. 2005, 17, 559–568. [Google Scholar] [CrossRef] [Green Version]
  114. Bornholdt, Z.A.; Ndungo, E.; Fusco, M.L.; Bale, S.; Flyak, A.I.; Crowe, J.E.; Chandran, K.; Saphire, E.O. Host-primed ebola virus gp exposes a hydrophobic npc1 receptor-binding pocket, revealing a target for broadly neutralizing antibodies. mBio 2016, 7, e02154-15. [Google Scholar] [CrossRef] [PubMed]
  115. Ao, Z.; Wang, L.; Mendoza, E.J.; Cheng, K.; Zhu, W.; Cohen, E.A.; Fowke, K.; Qiu, X.; Kobinger, G.; Yao, X. Incorporation of Ebola glycoprotein into hiv particles facilitates dendritic cell and macrophage targeting and enhances HIV-specific immune responses. PLoS ONE 2019, 14, e0216949–e0217067. [Google Scholar] [CrossRef]
  116. Wong, G.; Qiu, X.; Ebihara, H.; Feldmann, H.; Kobinger, G.P. Characterization of a bivalent vaccine capable of inducing protection against both Ebola and cross-clade H5N1 influenza in mice. J. Infect. Dis. 2015, 212, S435–S442. [Google Scholar] [CrossRef]
  117. Khataby, K.; Kasmi, Y.; Hammou, R.A.; Laasri, F.E.; Boughribi, S.; Ennaji, M.M. Ebola virus’s glycoproteins and entry mechanism. In Ebola; InTech: London, UK, 2016. [Google Scholar] [CrossRef]
  118. Majid, M.U.; Tahir, M.S.; Ali, Q.; Rao, A.Q.; Rashid, B.; Ali, A.; Nasir, I.A.; Husnain, T. Nature and history of Ebola virus: An overview. Arch. Neurosci. 2016, 3, e35027–e35038. [Google Scholar] [CrossRef]
  119. Ning, Y.-J.; Deng, F.; Hu, Z.; Wang, H. The roles of ebolavirus glycoproteins in viral pathogenesis. Virol. Sin. 2017, 32, 3–15. [Google Scholar] [CrossRef]
  120. Francica, J.R.; Varela-Rohena, A.; Medvec, A.; Plesa, G.; Riley, J.L.; Bates, P. Steric shielding of surface epitopes and impaired immune recognition induced by the Ebola virus glycoprotein. PLoS Pathog. 2010, 6, e1001098–e1001113. [Google Scholar] [CrossRef] [PubMed]
  121. Yang, Z.-Y.; Duckers, H.J.; Sullivan, N.J.; Sanchez, A.; Nabel, E.G.; Nabel, G.J. Identification of the Ebola virus glycoprotein as the main viral determinant of vascular cell cytotoxicity and injury. Nat. Med. 2000, 6, 886–889. [Google Scholar] [CrossRef] [PubMed]
  122. Hood, C.L.; Abraham, J.; Boyington, J.C.; Leung, K.; Kwong, P.D.; Nabel, G.J. Biochemical and structural characterization of cathepsin L-processed ebola virus glycoprotein: Implications for viral entry and immunogenicity. J. Virol. 2010, 84, 2972–2982. [Google Scholar] [CrossRef] [PubMed]
  123. Zhao, Y.; Ren, J.; Harlos, K.; Jones, D.M.; Zeltina, A.; Bowden, T.A.; Padilla-Parra, S.; Fry, E.E.; Stuart, D.I. Toremifene interacts with and destabilizes the Ebola virus glycoprotein. Nature 2016, 535, 169–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Lee, J.; Nyenhuis, D.A.; Nelson, E.A.; Cafiso, D.S.; White, J.M.; Tamm, L.K. Structure of the Ebola virus envelope protein MPER/TM domain and its interaction with the fusion loop explains their fusion activity. Proc. Natl. Acad. Sci. USA 2017, 114, 177–182. [Google Scholar] [CrossRef]
  125. Wang, B.; Wang, Y.; Frabutt, D.A.; Zhang, X.; Yao, X.; Hu, D.; Zhang, Z.; Liu, C.; Zheng, S.; Xiang, S.-H. Mechanistic understanding of N-glycosylation in Ebola virus glycoprotein maturation and function. J. Biol. Chem. 2017, 292, 5860–5870. [Google Scholar] [CrossRef] [Green Version]
  126. De La Vega, M.-A.; Wong, G.; Kobinger, G.P.; Qiu, X. The multiple roles of sGP in Ebola pathogenesis. Viral Immunol. 2015, 28, 3–9. [Google Scholar] [CrossRef] [PubMed]
  127. Radoshitzky, S.R.; Warfield, K.L.; Chi, X.; Dong, L.; Kota, K.; Bradfute, S.B.; Gearhart, J.D.; Retterer, C.; Kranzusch, P.J.; Misasi, J.N. Ebolavirus δ-peptide immunoadhesins inhibit marburgvirus and Ebolavirus cell entry. J. Virol. 2011, 85, 8502–8513. [Google Scholar] [CrossRef]
  128. Volchkova, V.A.; Feldmann, H.; Klenk, H.-D.; Volchkov, V.E. The nonstructural small glycoprotein sGP of Ebola virus is secreted as an antiparallel-orientated homodimer. Virology 1998, 250, 408–414. [Google Scholar] [CrossRef]
  129. Wahl-Jensen, V.; Kurz, S.K.; Hazelton, P.R.; Schnittler, H.-J.; StrÃher, U.; Burton, D.R.; Feldmann, H. Role of Ebola virus secreted glycoproteins and virus-like particles in activation of human macrophages. J. Virol. 2005, 79, 2413–2419. [Google Scholar] [CrossRef]
  130. Monath, T.P.; Fast, P.E.; Modjarrad, K.; Clarke, D.K.; Martin, B.K.; Fusco, J.; Nichols, R.; Heppner, D.G.; Simon, J.K.; Dubey, S. rVSV ”G-ZEBOV-GP (also designated V920) recombinant vesicular stomatitis virus pseudotyped with Ebola Zaire Glycoprotein: Standardized template with key considerations for a risk/benefit assessment. Vaccine X 2019, 1, 100009. [Google Scholar] [CrossRef] [PubMed]
  131. Ilinykh, P.A.; Shen, X.; Flyak, A.I.; Kuzmina, N.; Ksiazek, T.G.; Crowe, J.E.; Bukreyev, A. Chimeric filoviruses for identification and characterization of monoclonal antibodies. J. Virol. 2016, 90, 3890–3901. [Google Scholar] [CrossRef] [PubMed]
  132. Huttner, A.; Dayer, J.-A.; Yerly, S.; Combescure, C.; Auderset, F.; Desmeules, J.; Eickmann, M.; Finckh, A.; Goncalves, A.R.; Hooper, J.W. The effect of dose on the safety and immunogenicity of the VSV Ebola candidate vaccine: A randomised double-blind, placebo-controlled phase 1/2 trial. Lancet Infect. Dis. 2015, 15, 1156–1166. [Google Scholar] [CrossRef]
  133. Pallesen, J.; Murin, C.D.; de Val, N.; Cottrell, C.A.; Hastie, K.M.; Turner, H.L.; Fusco, M.L.; Flyak, A.I.; Zeitlin, L.; Crowe, J.E., Jr. Structures of Ebola virus GP and sGP in complex with therapeutic antibodies. Nat. Microbiol. 2017, 1, 16128–16151. [Google Scholar] [CrossRef]
  134. Davidson, E.; Bryan, C.; Fong, R.H.; Barnes, T.; Pfaff, J.; Mabila, M.; Rucker, J.B.; Doranz, B.J. Mechanism of binding to Ebola virus glycoprotein by the ZMapp, ZMAb, and MB-003 cocktail antibodies. J. Virol. 2015, 89, 10982–10992. [Google Scholar] [CrossRef]
  135. Hacke, M.; Björkholm, P.; Hellwig, A.; Himmels, P.; Ruiz de Almodóvar, C.; Brügger, B.; Wieland, F.; Ernst, A.M. Inhibition of Ebola virus glycoprotein-mediated cytotoxicity by targeting its transmembrane domain and cholesterol. Nat. Commun. 2015, 6, 7688–7697. [Google Scholar] [CrossRef]
  136. Mohan, G.S.; Li, W.; Ye, L.; Compans, R.W.; Yang, C. Antigenic subversion: A novel mechanism of host immune evasion by Ebola virus. PLoS Pathog. 2012, 8, e1003065–e1003079. [Google Scholar] [CrossRef]
  137. Speranza, E.; Connor, J.H. Host transcriptional response to Ebola virus infection. Vaccines 2017, 5, 30. [Google Scholar] [CrossRef]
  138. Agnandji, S.T.; Huttner, A.; Zinser, M.E.; Njuguna, P.; Dahlke, C.; Fernandes, J.F.; Yerly, S.; Dayer, J.-A.; Kraehling, V.; Kasonta, R. Phase 1 trials of rVSV Ebola vaccine in africa and europe. N. Engl. J. Med. 2016, 374, 1647–1660. [Google Scholar] [CrossRef]
  139. Marzi, A.; Engelmann, F.; Feldmann, F.; Haberthur, K.; Shupert, W.L.; Brining, D.; Scott, D.P.; Geisbert, T.W.; Kawaoka, Y.; Katze, M.G. Antibodies are necessary for rVSV/ZEBOV-GP-mediated protection against lethal Ebola virus challenge in nonhuman primates. Proc. Natl. Acad. Sci. USA 2012, 110, 1893–1898. [Google Scholar] [CrossRef]
  140. Venkatraman, N.; Silman, D.; Folegatti, P.M.; Hill, A.V.S. Vaccines against Ebola virus. Vaccine 2017, 36, 5454–5459. [Google Scholar] [CrossRef] [PubMed]
  141. Takada, A.; Kawaoka, Y. Antibody-dependent enhancement of viral infection: Molecular mechanisms and in vivo implications. Rev. Med Virol. 2003, 13, 387–398. [Google Scholar] [CrossRef] [PubMed]
  142. Alberts, B.; Johnson, A.; Lewis, J.; Raff, M.; Roberts, K.; Walter, P. Innate immunity. In Molecular Biology of the Cell, 4th ed.; Garland Science: New York, NY, USA, 2002; Chapter 25; p. 712. [Google Scholar]
  143. Henao-Restrepo, A.M.; Longini, I.M.; Egger, M.; Dean, N.E.; Edmunds, W.J.; Camacho, A.; Carroll, M.W.; Doumbia, M.; Draguez, B.; Duraffour, S. Efficacy and effectiveness of an rVSV-vectored vaccine expressing ebola surface glycoprotein: Interim results from the guinea ring vaccination cluster-randomised trial. Lancet 2015, 386, 857–866. [Google Scholar] [CrossRef]
  144. Sakabe, S.; Sullivan, B.M.; Hartnett, J.N.; Robles-Sikisaka, R.; Gangavarapu, K.; Cubitt, B.; Ware, B.C.; Kotliar, D.; Branco, L.M.; Goba, A. Analysis of CD8+ t cell response during the 2013–2016 Ebola epidemic in west africa. Proc. Natl. Acad. Sci. USA 2018, 115, E7578–E7586. [Google Scholar] [CrossRef] [PubMed]
  145. Dahlke, C.; Lunemann, S.; Kasonta, R.; Kreuels, B.; Schmiedel, S.; Ly, M.L.; Fehling, S.K.; Strecker, T.; Becker, S.; Altfeld, M. Comprehensive characterization of cellular immune responses following Ebola virus infection. J. Infect. Dis. 2016, 215, 287–292. [Google Scholar] [CrossRef] [PubMed]
  146. Warfield, K.L.; Olinger, G.; Deal, E.M.; Swenson, D.L.; Bailey, M.; Negley, D.L.; Hart, M.K.; Bavari, S. Induction of humoral and CD8+ T cell responses are required for protection against lethal Ebola virus infection. J. Immunol. 2005, 175, 1184–1191. [Google Scholar] [CrossRef] [PubMed]
  147. Olejnik, J.; Forero, A.; Deflubé, L.R.; Hume, A.J.; Manhart, W.A.; Nishida, A.; Marzi, A.; Katze, M.G.; Ebihara, H.; Rasmussen, A.L. Ebolaviruses associated with differential pathogenicity induce distinct host responses in human macrophages. J. Virol. 2017, 91, e00179-17. [Google Scholar] [CrossRef]
  148. Rhein, B.A.; Maury, W.J. Ebola virus entry into host cells: Identifying therapeutic strategies. Curr. Clin. Microbiol. Rep. 2015, 2, 115–124. [Google Scholar] [CrossRef] [PubMed]
  149. Baribaud, F.; Doms, R.W.; Pohlmann, S. The role of DC-sign and DC-signr in HIV and Ebola virus infection: Can potential therapeutics block virus transmission and dissemination? Expert Opin. Ther. Targets 2002, 6, 423–431. [Google Scholar] [CrossRef] [PubMed]
  150. Raska, M.; Czernekova, L.; Moldoveanu, Z.; Zachova, K.; Elliott, M.C.; Novak, Z.; Hall, S.; Hoelscher, M.; Maboko, L.; Brown, R. Differential glycosylation of envelope GP120 is associated with differential recognition of HIV-1 by virus-specific antibodies and cell infection. Aids Res. Ther. 2018, 11, 23–39. [Google Scholar] [CrossRef]
  151. Shimojima, M.; Takada, A.; Ebihara, H.; Neumann, G.; Fujioka, K.; Irimura, T.; Jones, S.; Feldmann, H.; Kawaoka, Y. Tyro3 family-mediated cell entry of EBOLA and marburg viruses. J. Virol. 2006, 80, 10109–10116. [Google Scholar] [CrossRef] [PubMed]
  152. Dahlmann, F.; Biedenkopf, N.; Babler, A.; Jahnen-Dechent, W.; Karsten, C.B.; Gnirß, K.K.; Schneider, H.; Wrensch, F.; O’Callaghan, C.A.; Bertram, S. Analysis of Ebola virus entry into macrophages. J. Infect. Dis. 2015, 212, S247–S257. [Google Scholar] [CrossRef] [PubMed]
  153. de Witte, L.; Abt, M.; Schneider-Schaulies, S.; van Kooyk, Y.; van Kooyk, Y.; Geijtenbeek, T.B. Measles virus targets DC-sign to enhance dendritic cell infection. J. Virol. 2006, 80, 3477–3486. [Google Scholar] [CrossRef] [PubMed]
  154. Geijtenbeek, T.B.; van Kooyk, Y. DC-sign: A novel HIV receptor on dcs that mediates HIV-1 transmission. Curr. Top. Microbiol. Immunol. 2003, 276, 31–54. [Google Scholar] [PubMed]
  155. Rinaldo, C.R. Dendritic cell-based human immunodeficiency virus vaccine. J. Intern. Med. 2009, 265, 138–158. [Google Scholar] [CrossRef] [PubMed]
  156. Hillaire, M.L.; Nieuwkoop, N.J.; Boon, A.C.; de Mutsert, G.; Vogelzang-van Trierum, S.E.; Fouchier, R.A.; Osterhaus, A.D.; Rimmelzwaan, G.F. Binding of DC-sign to the hemagglutinin of influenza a viruses supports virus replication in DC-sign expressing cells. PLoS ONE 2013, 8, e56164–e56174. [Google Scholar] [CrossRef] [PubMed]
  157. Garcia-Pineres, A.J.; Hildesheim, A.; Trivett, M.; Williams, M.; Wu, L.; Kewalramani, V.N.; Pinto, L.A.; Pinto, L.A. Role of DC-sign in the activation of dendritic cells by HPV-16 l1 virus-like particle vaccine. Eur. J. Immunol. 2006, 36, 437–445. [Google Scholar] [CrossRef]
  158. Fujihira, H.; Usami, K.; Matsuno, K.; Takeuchi, H.; Denda-Nagai, K.; Furukawa, J.-I.; Shinohara, Y.; Takada, A.; Kawaoka, Y.; Irimura, T. A critical domain of ebolavirus envelope glycoprotein determines glycoform and infectivity. Sci. Rep. 2018, 8, 5495–5508. [Google Scholar] [CrossRef]
  159. Lin, G.; Simmons, G.; Pohlmann, S.; Baribaud, F.; Ni, H.; Leslie, G.J.; Haggarty, B.S.; Bates, P.; Weissman, D.; Hoxie, J.A. Differential N-linked glycosylation of human immunodeficiency virus and Ebola virus envelope glycoproteins modulates interactions with DC-sign and DC-signr. J. Virol. 2003, 77, 1337–1346. [Google Scholar] [CrossRef]
  160. Varki, A.; Cummings, R.D.; Esko, J.D.; Freeze, H.H.; Stanley, P.; Bertozzi, C.R.; Hart, G.W.; Etzler, M.E. Nematoda-Essentials of Glycobiology; Cold Spring Harbor Laboratory Press: New York, NY, USA, 2009; pp. 1068–1072. [Google Scholar]
  161. Liu, W.; Tang, L.; Zhang, G.; Wei, H.; Cui, Y.; Guo, L.; Gou, Z.; Chen, X.; Jiang, D.; Zhu, Y. Characterization of a novel C-type lectin-like gene, lesctin: Demonstration of carbohydrate binding and expression in sinusoidal endothelial cells of liver and lymph node. J. Biol. Chem. 2004, 279, 18748–18758. [Google Scholar] [CrossRef]
  162. Domínguez-Soto, A.; Aragoneses-Fenoll, L.; Gómez-Aguado, F.; Corcuera, M.T.; Clária, J.; García-Monzón, C.; Bustos, M.; Corbí, A.L. The pathogen receptor liver and lymph node sinusoidal endotelial cell C-type Lectin is expressed in human kupffer cells and regulated by PU. 1. Hepatology 2009, 49, 287–296. [Google Scholar] [CrossRef] [PubMed]
  163. Gramberg, T.; Soilleux, E.; Fisch, T.; Lalor, P.F.; Hofmann, H.; Wheeldon, S.; Cotterill, A.; Wegele, A.; Winkler, T.; Adams, D.H. Interactions of lsectin and DC-sign/DC-signr with viral ligands: Differential ph dependence, internalization and virion binding. Virology 2008, 373, 189–201. [Google Scholar] [CrossRef] [PubMed]
  164. Sakakura, M.; Oo-Puthinan, S.; Moriyama, C.; Kimura, T.; Moriya, J.; Irimura, T.; Shimada, I. Carbohydrate binding mechanism of the macrophage galactose-type C-type LECtin 1 revealed by saturation transfer experiments. J. Biol. Chem. 2008, 283, 33665–33673. [Google Scholar] [CrossRef] [PubMed]
  165. Takada, A.; Fujioka, K.; Tsuiji, M.; Morikawa, A.; Higashi, N.; Ebihara, H.; Kobasa, D.; Feldmann, H.; Irimura, T.; Kawaoka, Y. Human macrophage c-type lectin specific for galactose and n-acetylgalactosamine promotes filovirus entry. J. Virol. 2004, 78, 2943–2947. [Google Scholar] [CrossRef]
  166. Usami, K.; Matsuno, K.; Matsuno, K.; Igarashi, M.; Irimura, T.; Irimura, T. Involvement of viral envelope GP2 in Ebola virus entry into cells expressing the macrophage galactose-type C-type lectin. Biochem. Biophys. Res. Commun. 2011, 407, 74–78. [Google Scholar] [CrossRef] [PubMed]
  167. Sharma, R.; Jangid, K.; Anuradha. Ebola vaccine: How far are we? J. Clin. Diagn. Res. 2017, 11, DE01–DE04. [Google Scholar] [CrossRef] [PubMed]
  168. Weik, M.; Modrof, J.; Klenk, H.D.; Becker, S.; Mühlberger, E. Ebola virus VP30-mediated transcription is regulated by RNA secondary structure formation. J. Virol. 2002, 76, 8532–8539. [Google Scholar] [CrossRef]
  169. Warfield, K.L.; Bosio, C.M.; Welcher, B.C.; Deal, E.M.; Mohamadzadeh, M.; Schmaljohn, A.; Aman, M.J.; Bavari, S. Ebola virus-like particles protect from lethal ebola virus infection. Proc. Natl. Acad. Sci. USA 2003, 100, 15889–15894. [Google Scholar] [CrossRef]
  170. Schweneker, M.; Laimbacher, A.S.; Zimmer, G.; Wagner, S.; Schraner, E.M.; Wolferstotter, M.; Klingenberg, M.; Dirmeier, U.; Steigerwald, R.; Lauterbach, H.; et al. Recombinant modified vaccinia virus ankara generating ebola virus-like particles. J. Virol. 2017, 91, e00343-17. [Google Scholar] [CrossRef]
  171. Wang, Y.; Li, J.; Hu, Y.; Liang, Q.; Wei, M.; Zhu, F. Ebola vaccines in clinical trial: The promising candidates. Hum. Vaccines Immunother. 2017, 13, 153–168. [Google Scholar] [CrossRef]
  172. Chahal, J.S.; Khan, O.F.; Cooper, C.L.; McPartlan, J.S.; Tsosie, J.K.; Tilley, L.D.; Sidik, S.M.; Lourido, S.; Langer, R.; Bavari, S. Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and toxoplasma gondii challenges with a single dose. Proc. Natl. Acad. Sci. USA 2016, 113, E4133–E4142. [Google Scholar] [CrossRef] [PubMed]
  173. Chepurnov, A.A.; Ternovoi, V.A.; Dadaeva, A.A.; Dmitriev, I.P.; Sizikova, L.P.; Volchkov, V.E.; Kudoiarova, N.M.; Rudzevich, T.N.; Netesov, S.V. Immunobiological properties of vp24 protein of ebola virus expressed by recombinant vaccinia virus. Vopr Virusol 1997, 42, 115–120. [Google Scholar] [PubMed]
  174. Suder, E.; Furuyama, W.; Feldmann, H.; Marzi, A.A.-O.; de Wit, E. The vesicular stomatitis virus-based ebola virus vaccine: From concept to clinical trials. Hum. Vaccines Immunother. 2018, 14, 2107–2113. [Google Scholar] [CrossRef] [PubMed]
  175. Ewer, K.; Rampling, T.; Venkatraman, N.; Bowyer, G.; Wright, D.; Lambe, T.; Imoukhuede, E.B.; Payne, R.; Fehling, S.K.; Strecker, T. A monovalent chimpanzee adenovirus Ebola vaccine boosted with mva. N. Engl. J. Med. 2016, 374, 1635–1646. [Google Scholar] [CrossRef] [PubMed]
  176. Tapia, M.D.; Sow, S.O.; Lyke, K.E.; Haidara, F.C.; Diallo, F.; Doumbia, M.; Traore, A.; Coulibaly, F.; Kodio, M.; Onwuchekwa, U. Use of ChAd3-EBO-Z ebola virus vaccine in malian and US adults, and boosting of Malian adults with MVA-BN-Filo: A phase 1, single-blind, randomised trial, a phase 1b, open-label and double-blind, dose-escalation trial, and a nested, randomised, double-blind, placebo-controlled trial. Lancet Infect. Dis. 2016, 16, 31–42. [Google Scholar] [PubMed]
  177. Zhu, F.-C.; Hou, L.-H.; Li, J.-X.; Wu, S.-P.; Liu, P.; Zhang, G.-R.; Hu, Y.-M.; Meng, F.-Y.; Xu, J.-J.; Tang, R. Safety and immunogenicity of a novel recombinant adenovirus type-5 vector-based ebola vaccine in healthy adults in china: Preliminary report of a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet 2015, 385, 2272–2279. [Google Scholar] [CrossRef]
  178. Awate, S.; Babiuk, L.A.B.; Mutwiri, G. Mechanisms of action of adjuvants. Front. Immunol. 2013, 4, 1–10. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A schematic structure of (A) EBOV GP, indicating GP1,2. GP1 comprised of RBD, glycan cap, and MLD, while GP2 contains the HR1 and HR2. Y denotes the N-glycosylation sites. Receptors on DCs have an affinity with the N-linked glycans on GP1, indicating that the binding sites of EBOV GP with DCs are on the RBD, while the glycan cap contributes to its binding because of the presence of N-glycosylation sites [109]. The receptors on DCs for GP1 include DC-SIGN [110], L-SIGN, LSECtin [111], hMGLs [112,113], and NPC-1 [114]. Although N-glycosylation sites are present on the MLD, MLD is dispensable, and its absence contributes to more efficient cell entry of EBOV GP [115]. (B) Schematic diagram showing the incorporation of EBOV GP with a different pathogen antigen into VLPs [115]. (C) Schematic structure of vesicular stomatitis virus (VSV) with deleted glycoprotein and having EBOV GP with different pathogen antigen in the deleted G domain of VSV [116]. (N, nucleoprotein; M, matrix protein; L, RNA polymerase; G, glycoprotein; P, phosphoprotein)
Figure 1. A schematic structure of (A) EBOV GP, indicating GP1,2. GP1 comprised of RBD, glycan cap, and MLD, while GP2 contains the HR1 and HR2. Y denotes the N-glycosylation sites. Receptors on DCs have an affinity with the N-linked glycans on GP1, indicating that the binding sites of EBOV GP with DCs are on the RBD, while the glycan cap contributes to its binding because of the presence of N-glycosylation sites [109]. The receptors on DCs for GP1 include DC-SIGN [110], L-SIGN, LSECtin [111], hMGLs [112,113], and NPC-1 [114]. Although N-glycosylation sites are present on the MLD, MLD is dispensable, and its absence contributes to more efficient cell entry of EBOV GP [115]. (B) Schematic diagram showing the incorporation of EBOV GP with a different pathogen antigen into VLPs [115]. (C) Schematic structure of vesicular stomatitis virus (VSV) with deleted glycoprotein and having EBOV GP with different pathogen antigen in the deleted G domain of VSV [116]. (N, nucleoprotein; M, matrix protein; L, RNA polymerase; G, glycoprotein; P, phosphoprotein)
Microorganisms 07 00402 g001
Figure 2. Schematic diagram of the incorporation or infusing pathogen antigen on EBOV GP to target DCs for the induction of immune responses. Using the technology of EBOV GP VLP immunological basis, it is possible to incorporate EBOV VLPs with other viral antigens to induce an efficient humoral and adaptive cellular immune responses.
Figure 2. Schematic diagram of the incorporation or infusing pathogen antigen on EBOV GP to target DCs for the induction of immune responses. Using the technology of EBOV GP VLP immunological basis, it is possible to incorporate EBOV VLPs with other viral antigens to induce an efficient humoral and adaptive cellular immune responses.
Microorganisms 07 00402 g002
Table 1. Vaccine development strategies for selected viral infections.
Table 1. Vaccine development strategies for selected viral infections.
S/NViral InfectionVaccine Development StrategyRoute of AdministrationVaccine StatusReferences
1Yellow fever Attenuated virusSubcutaneousAvailable in circulation[5]
2InfluenzaLive attenuated virus, inactivated virus, recombinant influenza vaccine, VLPIntramuscular, intranasal, intradermal, subcutaneousAvailable in circulation
VLP and DNA vaccine not in circulation (needs development of a novel universal vaccine)
[6,7,8]
3.PoliovirusLive attenuated vaccineOralAvailable in circulation[9]
4.Measles, mumps and rubella Live attenuated vaccineSubcutaneousAvailable in circulation[10]
5Human papilloma virusVLP, inactivated vaccineIntramuscular, oralMonovalent, bivalent, tetravalent, nonavalent vaccines available in circulation[11,12]
6Hepatitis B virusLive inactivated, recombinant DNAIntramuscularAvailable in circulation (it gives short-term protection; issue raised concerning its safety)[13,14,15]
7VaricellaWeakened live virus or attenuated virusSubcutaneous, intramuscularAvailable in circulation[16]
8RotavirusLive attenuated, VLPOral, intranasalAvailable in circulation; VLP not in circulation[17,18]
9HIVVLP, DNA vaccineSubcutaneous, intramuscularNot in circulation (development in progress)[19,20]
10EBOVLive attenuated, VLPIntramuscularAvailable but not yet in circulation (phase trial in progress)[21,22]
11Lassa virusVLP, live attenuated virus, DNA vaccineIntradermalNo available vaccine[23,24,25]
Note: EBOV, Ebola virus; VLP, virus-like particle; HIV, human immunodeficiency virus.
Table 2. Development of vaccine using a dendritic cell (DC)-targeted approach in the selected viruses.
Table 2. Development of vaccine using a dendritic cell (DC)-targeted approach in the selected viruses.
S/N.VaccineImmune Responses InducedVaccine Development StrategyRoute of AdministrationDC-Targeting SubstanceProtection PercentageRef
1Cancer vaccineCellular immune response and humoral responsesDNA vaccineNot applicableIFN-αNot applicable[38,39]
DNA vaccineSubcutaneousLiposome and melanoma80–100%[39]
2Yellow feverInnate immune responses (proinflammatory cytokines interleukin (IL)-12p40, IL-6, and interferon-α), adaptive immune responses (T helper cell (Th)1/Th2 cytokine profile and antigen-specific CD8+ T cell)Live attenuated vaccineSubcutaneous Not applicable[30]
3AdenovirusCytolytic T lymphocyte cellsRecombinant vaccineNot applicableRecombinant single-chain (sc) mAb Fv fragmentsNot applicable[29]
4HIVIFN-γ, CD4+, and CD8+ T cellRecombinant vaccinia virus (DNA vaccine)IntranasalRecombinant single-chain (sc) mAb Fv fragments (scFv)
HIV gagp41-scFv
100%[40]
5Influenza ACytotoxic CD8+T, cell CD4+ Th1, IgG2a antibodiesDNA vaccinesIntradermalXcl1-hemagglutinin (HA)100%[41]
DNA vaccine Intravenous tail injection and electroporationXcl1-HA or Xcl2-HA90%[42]
6West Nile Virus vaccineHumoral and T-cell responsesDNA vaccine (immunodominant vaccinia B8R gene)Intravenous injectionRabies glycoprotein (GP) fused to protamine residue (RVG-P)80%[43]

Share and Cite

MDPI and ACS Style

Olukitibi, T.A.; Ao, Z.; Mahmoudi, M.; Kobinger, G.A.; Yao, X. Dendritic Cells/Macrophages-Targeting Feature of Ebola Glycoprotein and its Potential as Immunological Facilitator for Antiviral Vaccine Approach. Microorganisms 2019, 7, 402. https://doi.org/10.3390/microorganisms7100402

AMA Style

Olukitibi TA, Ao Z, Mahmoudi M, Kobinger GA, Yao X. Dendritic Cells/Macrophages-Targeting Feature of Ebola Glycoprotein and its Potential as Immunological Facilitator for Antiviral Vaccine Approach. Microorganisms. 2019; 7(10):402. https://doi.org/10.3390/microorganisms7100402

Chicago/Turabian Style

Olukitibi, Titus Abiola, Zhujun Ao, Mona Mahmoudi, Gary A. Kobinger, and Xiaojian Yao. 2019. "Dendritic Cells/Macrophages-Targeting Feature of Ebola Glycoprotein and its Potential as Immunological Facilitator for Antiviral Vaccine Approach" Microorganisms 7, no. 10: 402. https://doi.org/10.3390/microorganisms7100402

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop