Open access peer-reviewed chapter

Interaction of Ebola Virus with the Innate Immune System

Written By

Parastoo Yousefi and Alireza Tabibzadeh

Reviewed: 07 April 2022 Published: 06 May 2022

DOI: 10.5772/intechopen.104843

From the Edited Volume

Macrophages - Celebrating 140 Years of Discovery

Edited by Vijay Kumar

Chapter metrics overview

186 Chapter Downloads

View Full Metrics

Abstract

The Ebola viruses (EBOVs) are known as one the most lethal viruses. EBOV systemic infection can cause damage to vital organs and lead to death. The immune responses of the innate immune system and inflammatory cascade are critical elements in the EBOV pathogenesis and mortality. The primary innate immune system response can shape the adaptive immune responses. The innate immune response, due to the pattern-recognition receptors (PRRs), can induce interferons (IFN). IFN is a critical element in the antiviral response. The EBOV can evade the IFN and innate immunity using different mechanisms, whereas a well-controlled and sufficient innate immune response is vital for limiting the EBOV infection. In this regard, a hyperactive inflammation response may lead to cytokine storms and death. In this chapter, we have tried to provide a perspective on the pathogenesis and molecular mechanisms of the innate immune system and its interaction with EBOV infection.

Keywords

  • Ebola virus
  • immunity
  • innate immune responses
  • macrophages
  • VP35

1. Introduction

The Ebola virus (EBOV) genus and Marburg viruses are classified in the Filoviridae family and Mononegavirales order [1]. The EBOV contains a linear single-stranded RNA genome of 18.9-kilobase length and a membrane glycoprotein [2, 3]. Over the past 40 years, there have been 34 episodes of EBOV outbreaks in 11 different Sub-Saharan African countries. The first outbreaks occurred in 1971 in the Democratic Republic of the Congo (DRC) and Sudan [3]. These outbreaks amassed a total of more than 34000 cases and led to 14000 deaths [3]. EBOV Sudan, Bundibugyo, Zaire, and Reston are important species of the EBOV infection in humans [4]. The basic reproductive number (R0) of the EBOV is estimated to be in the range of 1.51–2.53 [4].

Some therapeutic and vaccination strategies have been introduced against the EBOV so far. The remdesivir, monoclonal antibodies [5], and rVSV-ZEBOV (vesicular stomatitis virus-based vaccine which is expressing the glycoprotein of a Zaire Ebola virus) [6] are considered ongoing advances in this area for EBOV treatment and transmission control.

Innate immunity and interferons (IFN) production are one of the most important immune responses to viral infections. Interaction between the Ebola virus and innate immunity with a well-regulated inflammation response can be life-saving in patients with the Ebola virus disease (EBD) [7]. The interaction of the EBOV with the innate immune system is a multifactorial condition, and it is largely associated with cytokines, chemokines, inflammation mediators, the NK cell receptors, and pathogen-associated molecular patterns (PAMPs), such as killer immunoglobulin-like receptor (KIR) and Toll-like receptors (TLRs) [8, 9]. The signaling pathways in the innate immune system are triggered after PAMPs detected by pattern-recognition receptors (PRRs) [10]. Based on protein domain homology, the PRRs are classified into six groups TLRs, a retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), nucleotide oligomerization domain (NOD)-like receptors (NLRs), C-type lectin receptors (CLRs), absent in melanoma-2 (AIM2)-like receptors (ALRs), and cyclic GMP-AMP synthase and stimulator of interferon genes (cGAS-STING pathway) [11, 12]. PRRs are also able to recognize molecules released by damaged cells (The damage-associated molecular pattern-DAMPs) and activate natural immunity [13, 14]. The detection of PAMPs or DAMPs by PRRs increases the transcription of genes encoding cytokine and chemokine, IFN type 1, and antimicrobial proteins (AMPs) [15]. Furthermore, most of the TLRs recognize double-stranded RNA (dsRNA) whereas TLR7 and TLR8 bind single-stranded RNA (ssRNA) in endosomes [16]. After recognition, TLRs recruit several adaptor proteins, including Myeloid Differentiation primary response 88 (MyD88), TIR-domain-containing adaptor protein (TIRAP), TIR-domain-containing adapter-inducing interferon-β (TRIF), and TRIF-related adapter molecule (TRAM) [17]. This initiates downstream signaling cascades that lead to the activation of transcription factors, such as transcription factors NF-κB, interferon regulatory factor 3/7 (IRF3/7), and activator protein-1 (AP-1). These factors stimulate the transcription of genes in the cell nucleus and increase the secretion of pro-inflammatory cytokines and IFN [15, 18]. RIG-1 and melanoma differentiation-associated gene 5 (MDA5), recognize viral ss/dsRNA molecule leads to the translocation of NF-kB, mitogen-activated protein kinases (MAPKs), as well as interferon regulatory factors IRF3, IRF7 [19]. RIG-1 signaling pathway plays a crucial role in the antiviral innate immune response. Activated RIG-1 can interact with signaling adaptor protein mitochondrial antiviral signaling protein (MAVS), also known as IFN-β promoter stimulator 1 (IPS-1), to induction of NF-κB signaling or interferon pathways. It was revealed that upon viral infection, MAVs form high-molecular-weight aggregates downstream of RIG-1 and MDA5 signaling [20]. Activated MAVS–RIG-I signaling ultimately results in the activation of the antiviral IFN-I pathway [21]. The binding of type I and III IFNs to their receptors activates the Jak–STAT pathway, which is responsible for enhancing IFN-stimulated gene expression, which includes antiviral genes, such as Viperin, MxA, MxB, IFITMs, OAS, and PKR [22, 23]. One of the most important complexes, which participate in host defense by sensing viral infection and promoting innate immune system response, is the inflammasome, first described by Martinon in 2002 [24]. The best-studied inflammasome sensor is NOD-like receptor pyrin domain-containing protein 3 (NLRP3), which consists of a sensor molecule (NLRP3), the adaptor protein ASC, also called PYCARD, and an effector pro-caspase-1 [25]. Activated caspase is required for the cleavage of the inactivated interleukin-1 family (IL-1), such as pro-IL-18 and pro-IL-1β to the mature forms to initiate inflammasome [26].

Dendritic cells (DCs) and macrophages are one of the main axes for filoviruses infections due to the activation of the adaptive immune [27, 28]. In addition, the macrophage’s infection and attachment by EBOV result in downstream signaling for alteration of the expression profile in these cells. This alteration leads to the pro-inflammatory cytokine release and virus spreading, which both are important elements in the disease progression and outcome [29, 30]. In this regard, in the current chapter, we tried to provide a perspective on the pathophysiology and molecular mechanisms of the innate immune system and its interaction with EBOV infection.

Advertisement

2. Ebola virus: history and epidemiology

Over the past 40 years, there are 34 episodes of the EBOV outbreak in more than 10 different Sub-Saharan African countries leading to numerous cases and deaths [31]. Different species of the EBOV have been reported thus far. Three important species are known as Sudan, Bundibugyo, and Reston [32]. The responsible species for the 2014 outbreak of EBOV was identified as the Zaire ebolavirus [32]. The EBOV disease is a zoonotic disease in nature and the major route for transmission is contact with the infected animal especially chimpanzees, fruit bats, and antelope [2, 33]. The virus genome could be detectable for 10 weeks postmortem [34]. Any contact with bodily fluids is a major route for transmission of EBOV [35]. Even after the survival from the disease the viral genome can be detected in semen for 179 days and leads to sexual transmission in some cases [36]. Fever, fatigue, diarrhea, and tachycardia are the most common symptoms and based on the infected strain and the patient’s age, 43% of mortality is reported over the 8 days following the infection [37]. In convalesce or a long time after the remission, some rare complications and long-term sequels, such as uveitis or blurred vision, sleeping problems, and meningoencephalitis are reported [38, 39, 40].

Advertisement

3. EBOVE virology and pathogenicity

3.1 Virology and classification

The Ebola virus (EBOV) genus in Filoviridae is a member of the great order of Mononegavirales [1]. There are seven important genes in EBOV. The nucleoprotein (NP) is curial for replication and virus formation. Also, viral protein 35 (VP35) is important in the IFN synthesis blocking and virus replication while VP40 acts as an element in virus formation and intracellular trafficking. The glycoprotein (GP) is a major element for viral entry and induces lymphocyte apoptosis by soluble GPs. EBOV VP30 has been assumed to function as a transcription activation factor, which is essential for viral replication. VP24 is necessary for the formation of nucleocapsids (NC) and nucleocapsid-like structures, and Ebola virus L proteins act as subunits of RNA-dependent RNA polymerase, which along with VP35, is necessary for viral replication and transcription [41, 42, 43]. A summary of the function of genes is provided in Table 1.

Viral gene nameImportant functions
VP24formation of nucleocapsids (NC) and nucleocapsid-like structures, inhibit IFN signaling
LRNA-dependent RNA polymerase
glycoprotein (GP)viral entry induces lymphocyte apoptosis and reduces neutralizing antibodies by soluble GPs
NPNucleoprotein formation
VP40Virus assembly
VP35inhibits the induction of IFN type I
VP30transcription activation factor

Table 1.

A summary of the important functions of the Ebola virus genes [41, 44].

The VP35 is a key viral protein in the EBOV virulence. This unique protein inhibits the induction of IFN type I in the infected cell. The IFN type I has long been declared a key element in the host’s innate antiviral response [45]. In this regard, the VP35 is considered the most important for virus immune evasion [46]. The IFN blocking strategy in EBOV is not limited to VP35. Other EBOV protein VP24 can actively bind to the karyopherin alpha (KPNA) and interfere with the IFN STAT1 downstream signaling [47]. Furthermore, VP24 seems to be essential for the EBOV replication in macrophages of the guinea pig as an animal model [48].

3.2 Pathogenicity and treatment

The EBOV represents an affinity to a wide range of the cellular receptors for virus attachment to the cells. Following the systemic virus infection, viral cytopathology and immune-mediated cell damage are two essential steps of the EBOV pathogenesis and tissue damage [49]. One of the suggested pathogenic mechanisms of the EBOV is the antibody-dependent augmentation of the infection through the attachment of the antibodies to the virus and the C1q-C1q receptor of the complement in the cell surfaces [50].

One of the most important therapeutic agents for EBOV is the remdesivir. The remdesivir acts as an inhibitor of the virus RNA polymerase and leads to chain termination [51]. Efforts for the treatment of the EBOV also lead to some monoclonal antibodies against this virus; for instance, old and newer versions of these monoclonal antibodies are ZMapp, MAb114, and REGN-EB3 [5].

Advertisement

4. Innate immune system and EBOV

4.1 Primary innate immune response and inflammation

Interaction between the Ebola virus and innate immunity suggested that a fast and well-regulated inflammation response could be life-saving in patients with the Ebola virus disease (EBD). While a massive monocyte/macrophage activation could be lethal [7]. The mononuclear phagocytes are import element in the EBD pathology [52].

Biomarkers are suggestive elements for disease prognosis and pathogenesis. Some markers such as apoptosis antigen-Fas, IFN-β, IL-29, IL-5, TNFR-II, and FAS ligand levels are associated with the moderate disease while the D-dimer, Granzyme B, IL-10, IL-6, IL-8, TNFR-I, vWF (von Willebrand factor), monocyte chemoattractant proteins and thrombomodulin are associate with severe disease [53]. Furthermore, up-regulation of the IL-1β and IL-6 can suggest a non-fatal infection while the increase in TNF-α, IFN-ɤ, IL-10, IL-1 receptor antagonist, neopterin, IL-8, IL-15, and IL-16 is associated with the lethal outcome [7, 54, 55, 56]. By considering all these markers, it has been suggested that in a general view an increase in cytokines and cytokine storm, which it is, represents a hyperactive of the innate immune responses and in the other way, suppression of the adaptive immune responses and lymphocyte apoptosis is the main pathogenesis feature of the EBD and lethal infections [55].

The lymphocyte apoptosis leads to lymphocyte depletion. This apoptotic feature is not due to the replication and infection of the lymphocytes but it mediates through viral and immune system stimulations [57]. sGP, a viral protein produced during EBOV infection and accumulates at high concentrations in the serum, serves as a decoy to prevent the immune system from fighting the infection by binding EBOV-neutralizing antibodies [58]. It is assumed that glycosylation of transmembrane GP may affect neutralizing antibody binding [59]. Virally infected cells, release inflammation mediators that induce Fas and TNF-associated apoptosis-inducing ligands (TRAIL) pathways that can result in lymphocyte apoptosis and lack of an effective adaptive immune response [60, 61, 62]. In EBOV infection, lymphocytes are not directly infected, but apoptosis of lymphocytes is a pathological feature of infection. It is hypothesized that the factors (such as TRAIL, TNF-a, and Fas ligand) secreted by macrophages and dendritic cells infected in EBOV, cause lymphocyte apoptosis [52, 60].

Investigation of the EBOV infection in asymptomatic people suggested strong activation of the innate immune system and inflammation, which leads to adaptive immune activation and cytotoxic T cell responses. The strong activation of the innate immune system, inflammation and adaptive immune activation are considered as the optimum immune response to EBOV [63]. The main concept of the current section is summarized in Figure 1. The figure represents the EBOV infection in lethal and non-lethal scenarios and the role of the innate immune and inflammation in this process. In addition, some important interactions of the EBOV proteins are noted, for instance, the role of the VP35 and VP24 in type I interferon blocking or the role of the soluble glycoproteins in lymphocyte apoptosis.

Figure 1.

A summary of the EBOV infection in lethal and non-lethal scenarios and the role of the innate immune and inflammation. After the EBOV, infection such as any other viral infection innate immunity induces inflammation and IFN. The VP35 and VP24 of the EBOV block the IFN production. A well-controlled and sufficient innate immune response, while it leads to the adaptive immune response, is assumed main cause of asymptomatic or non-lethal infections.

4.2 Innate immunity receptors and EBOV

The interaction of the EBOV and the innate immune system is not only limited to the cytokines, chemokines, or inflammation mediators. In this regard, the NK cell and T cell receptors, which are known as killer immunoglobulin-like receptors (KIR), are critical. The role of the NK cells in response to the EBOV viral-like particles highlighted the importance of these cells in the innate immune response to the EBOV [64]. The KIRs are important elements in the host response to infectious diseases. KIR2DS1 and KIR2DS3 of repertoire genotypes of the KIR represent more susceptibility to fatal EBOV [8].

In the EBOV infection, the interaction of the innate and adaptive immune responses is critical for the disease outcome. The Toll-like receptor 4 (TLR4) is a vital element in response to the EBOV glycoprotein and activation of the antigen-presenting cells and T cells. By considering this, TLR4 responses represent a vital role in the regulation of innate and adaptive immunity [9]. The interaction of the TLR4 and monocytes (as antigen-presenting cells) highlighted the importance of the monocytes in the regulation of the immune response. The evidence supports the alteration of the transcriptional patterns in monocytes in EBD [65]. Furthermore, one of the major elements in inflammation and IFN stimulation is NLRP3 [66]. The EBOV infection could increase the IL-1β and IL-18 by the NLRP3 inflammasome activation [67]. This factor highlights the importance of the NLRP3 in pro-inflammatory cytokine production and innate immune system responses.

Advertisement

5. Macrophages and other important innate immune cells in Ebola virus innate immune responses

The DCs and macrophages infection with the EBOV is critical for adaptive immune activation. The EBOV infection leads to macrophage activation (by increasing the CD163 marker) and decreases T cell activation (by reducing in CD25 marker) in severe cases [68]. The macrophage’s infection leads to reductions in the co-stimulatory molecules in these cells, which are known as the main adaptive immune activation and the axis of the antigen presentation [27, 28]. The macrophage infection also leads to alteration in pro-inflammatory cytokine releases, such as IL-1β, TNF-α, and IL-6, which could dysregulate the inflammation [69]. Furthermore, monocytes infection with EBOV is a toll on the virus spreading all around the body [69]. The EBOV uses a mimicking of the apoptosis process for attachment and entry macrophages due to the TAM receptor tyrosine kinases and integrin αV [70]. However, it seems that other cell surface receptors such as DC-SIGN and DC-SIGNR or macrophage galactose-type calcium-type lectin (MGL) are critical for the EBOV infection in DC and macrophages [71, 72]. The attachment of the virus to macrophages regardless of the virus entry or macrophage infection affects the macrophage’s expression profile. This alteration is led macrophages to produce high levels of pro-inflammatory and pro-apoptotic signals [30].

Macrophages and dendritic cells are important cell targets of EBOV and the main innate immune cells that secrete cytokines, and chemokines following infection [29, 69]. Although macrophages and DCs are still able to initiate coagulation and inflammation, they are not able to stop the spreading of the Ebola virus systemically due to their impaired ability [28, 73]. These dysfunctions have a major impact on the innate and adaptive immune systems [74]. Macrophages and DCs are the essential cells of innate immunity and provide a bridge between innate and adaptive immunity [75]. It will highlight the importance of these cells in the disease outcome (Figure 1). Furthermore, VP24 and VP35 block latent lymphocyte stimulation through the IFN response [73]. All these clues are critical to combine and work as chains for limiting the infection through sufficient and well-controlled innate immune response activation, which leads to adaptive immune responses.

Advertisement

6. Conclusions

In this chapter, we tried to provide a perspective on the EBOV infection and innate immune responses. In a glimpse, it is worth mentioning that innate immune responses are critical in the EBOV infection. Sufficient and well-controlled innate immune responses may lead to optimum cytokine release and adaptive immune activation. In contrast, an overreacted innate immunity could affect and hyper-inflammation response.

The macrophages and DCs are also key elements in EBOV infection due to pro-inflammatory response and virus spreading. The EBOV uses different strategies to dysregulate and evade innate immune responses.

VP35 and VP24 of the virus inhibit the IFN type I stimulation in infected cells. Furthermore, the soluble GPs of the EBOV can induce apoptosis in T cells. The interaction of the EBOV with innate immunity is the most fundamental feature of the infection and determines the disease outcome.

Advertisement

Acknowledgments

We sincerely acknowledge the IntechOpen publication editorial office and Jelena Vrdoljak the Author Service Manager in IntechOpen for her incredible efforts.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. ICTV. https://talk.ictvonline.org/taxonomy/. visit 2 March 2022
  2. 2. Pourrut X et al. The natural history of Ebola virus in Africa. Microbes and Infection. 2005;7(7-8):1005-1014
  3. 3. Li Y, Chen S. Evolutionary history of Ebola virus. Epidemiology & Infection. 2014;142(6):1138-1145
  4. 4. Althaus CL. Estimating the reproduction number of Ebola virus (EBOV) during the 2014 outbreak in West Africa. PLoS Currents. 2014;2014:6
  5. 5. Mulangu S et al. A randomized, controlled trial of Ebola virus disease therapeutics. New England Journal of Medicine. 2019;381(24):2293-2303
  6. 6. Lévy Y et al. Prevention of Ebola virus disease through vaccination: Where we are in 2018. The Lancet. 2018;392(10149):787-790
  7. 7. Baize S et al. Inflammatory responses in Ebola virus-infected patients. Clinical & Experimental Immunology. 2002;128(1):163-168
  8. 8. Wauquier N et al. Association of KIR2DS1 and KIR2DS3 with fatal outcome in Ebola virus infection. Immunogenetics. 2010;62(11):767-771
  9. 9. Lai C-Y et al. Ebola virus glycoprotein induces an innate immune response in vivo via TLR4. Frontiers in Microbiology. 2017;8:1571
  10. 10. Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nature Immunology. 2004;5(10):987-995
  11. 11. Abe T, Marutani Y, Shoji I. Cytosolic DNA-sensing immune response and viral infection. Microbiology and Immunology. 2019;63(2):51-64
  12. 12. He S et al. Research advancement of innate immunity and pattern recognition receptors. Chinese Journal of Animal Nutrition. 2017;29(11):3844-3851
  13. 13. Amarante-Mendes GP et al. Pattern recognition receptors and the host cell death molecular machinery. Frontiers in Immunology. 2018:2379
  14. 14. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124(4):783-801
  15. 15. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140(6):805-820
  16. 16. Dalpke AH, Helm M. RNA mediated Toll-like receptor stimulation in health and disease. RNA Biology. 2012;9(6):828-842
  17. 17. Jang T-h, Park HH. Crystal structure of TIR domain of TLR6 reveals novel dimeric interface of TIR–TIR interaction for toll-like receptor signaling pathway. Journal of Molecular Biology. 2014;426(19):3305-3313
  18. 18. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nature Immunology. 2010;11(5):373-384
  19. 19. Jensen S, Thomsen AR. Sensing of RNA viruses: A review of innate immune receptors involved in recognizing RNA virus invasion. Journal of Virology. 2012;86(6):2900-2910
  20. 20. Hou F et al. MAVS forms functional prion-like aggregates to activate and propagate antiviral innate immune response. Cell. 2011;146(3):448-461
  21. 21. Servant MJ, Grandvaux N, Hiscott J. Multiple signaling pathways leading to the activation of interferon regulatory factor 3. Biochemical Pharmacology. 2002;64(5-6):985-992
  22. 22. Schoggins JW. Interferon-stimulated genes: Roles in viral pathogenesis. Current Opinion in Virology. 2014;6:40-46
  23. 23. He F et al. Filovirus VP24 proteins differentially regulate RIG-I and MDA5-dependent Type I and III interferon promoter activation. Frontiers in Immunology. Jan 5, 2022;12:694105
  24. 24. Martinon F, Burns K, Tschopp J. The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-β. Molecular Cell. 2002;10(2):417-426
  25. 25. Menu P, Vince J. The NLRP3 inflammasome in health and disease: The good, the bad and the ugly. Clinical & Experimental Immunology. 2011;166(1):1-15
  26. 26. Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. 2014;157(5):1013-1022
  27. 27. Mahanty S et al. Cutting edge: Impairment of dendritic cells and adaptive immunity by Ebola and Lassa viruses. The Journal of Immunology. 2003;170(6):2797-2801
  28. 28. Bosio CM et al. Ebola and Marburg viruses replicate in monocyte-derived dendritic cells without inducing the production of cytokines and full maturation. The Journal of Infectious Diseases. 2003;188(11):1630-1638
  29. 29. Gupta M et al. Monocyte-derived human macrophages and peripheral blood mononuclear cells infected with Ebola virus secrete MIP-1α and TNF-α and inhibit poly-IC-induced IFN-α in vitro. Virology. 2001;284(1):20-25
  30. 30. Wahl-Jensen V et al. Ebola virion attachment and entry into human macrophages profoundly effects early cellular gene expression. PLoS Neglected Tropical Diseases. 2011;5(10):e1359
  31. 31. Rugarabamu S et al. Forty-two years of responding to Ebola virus outbreaks in Sub-Saharan Africa: A review. BMJ Global Health. 2020;5(3):e001955
  32. 32. Baize S et al. Emergence of Zaire Ebola virus disease in Guinea. New England Journal of Medicine. 2014;371(15):1418-1425
  33. 33. Rewar S, Mirdha D. Transmission of Ebola virus disease: An overview. Annals of Global Health. 2014;80(6):444-451
  34. 34. Prescott J et al. Postmortem stability of Ebola virus. Emerging Infectious Diseases. 2015;21(5):856
  35. 35. Judson S, Prescott J, Munster V. Understanding ebola virus transmission. Viruses. 2015;7(2):511-521
  36. 36. Mate SE et al. Molecular evidence of sexual transmission of Ebola virus. New England Journal of Medicine. 2015;373(25):2448-2454
  37. 37. Bah EI et al. Clinical presentation of patients with Ebola virus disease in Conakry, Guinea. New England Journal of Medicine. 2015;372(1):40-47
  38. 38. Varkey JB et al. Persistence of Ebola virus in ocular fluid during convalescence. New England Journal of Medicine. 2015;372(25):2423-2427
  39. 39. Jacobs M et al. Late Ebola virus relapse causing meningoencephalitis: A case report. The Lancet. 2016;388(10043):498-503
  40. 40. Clark DV et al. Long-term sequelae after Ebola virus disease in Bundibugyo, Uganda: A retrospective cohort study. The Lancet Infectious Diseases. 2015;15(8):905-912
  41. 41. Jain S et al. Structural and functional aspects of Ebola virus proteins. Pathogens. 2021;10(10):1330
  42. 42. Watt A et al. A novel life cycle modeling system for Ebola virus shows a genome length-dependent role of VP24 in virus infectivity. Journal of Virology. 2014;88(18):10511-10524
  43. 43. Boehmann Y et al. A reconstituted replication and transcription system for Ebola virus Reston and comparison with Ebola virus Zaire. Virology. 2005;332(1):406-417
  44. 44. He FB, Melén K, Kakkola L, Julkunen I. Interaction of ebola virus with the innate immune system. In: Okware SI, editor. Emerging Challenges in Filovirus Infections [Internet]. London: IntechOpen; 2019 [cited 2022 May 02]. Available from: https://www.intechopen.com/chapters/67614. doi: 10.5772/intechopen.86749
  45. 45. Basler CF et al. The Ebola virus VP35 protein functions as a type I IFN antagonist. Proceedings of the National Academy of Sciences. 2000;97(22):12289-12294
  46. 46. Woolsey C et al. A VP35 mutant Ebola virus lacks virulence but can elicit protective immunity to wild-type virus challenge. Cell Reports. 2019;28(12):3032-3046
  47. 47. Xu W et al. Ebola virus VP24 targets a unique NLS binding site on karyopherin alpha 5 to selectively compete with nuclear import of phosphorylated STAT1. Cell Host & Microbe. 2014;16(2):187-200
  48. 48. Mateo M et al. VP24 is a molecular determinant of Ebola virus virulence in guinea pigs. The Journal of Infectious Diseases. 2011;204(suppl_3):S1011-S1020
  49. 49. Baseler L et al. The pathogenesis of Ebola virus disease. Annual Review of Pathology: Mechanisms of Disease. 2017;12:387-418
  50. 50. Takada A et al. Antibody-dependent enhancement of Ebola virus infection. Journal of Virology. 2003;77(13):7539-7544
  51. 51. Tchesnokov EP et al. Mechanism of inhibition of Ebola virus RNA-dependent RNA polymerase by remdesivir. Viruses. 2019;11(4):326
  52. 52. Hensley LE et al. Proinflammatory response during Ebola virus infection of primate models: Possible involvement of the tumor necrosis factor receptor superfamily. Immunology Letters. 2002;80(3):169-179
  53. 53. McElroy AK et al. Kinetic analysis of biomarkers in a cohort of US patients with Ebola virus disease. Reviews of Infectious Diseases. 2016;63(4):460-467
  54. 54. Villinger F et al. Markedly elevated levels of interferon (IFN)-γ, IFN-α, interleukin (IL)-2, IL-10, and tumor necrosis factor-α associated with fatal Ebola virus infection. The Journal of Infectious Diseases. 1999;179(Suppl_1):S188-S191
  55. 55. Wauquier N et al. Human fatal zaire ebola virus infection is associated with an aberrant innate immunity and with massive lymphocyte apoptosis. PLoS Neglected Tropical Diseases. 2010;4(10):e837
  56. 56. Hutchinson KL, Rollin PE. Cytokine and chemokine expression in humans infected with Sudan Ebola virus. The Journal of Infectious Diseases. 2007;196(Suppl_2):S357-S363
  57. 57. Geisbert TW et al. Apoptosis induced in vitro and in vivo during infection by Ebola and Marburg viruses. Laboratory Investigation. 2000;80(2):171-186
  58. 58. Ito H et al. Ebola virus glycoprotein: Proteolytic processing, acylation, cell tropism, and detection of neutralizing antibodies. Journal of Virology. 2001;75(3):1576-1580
  59. 59. Francica JR et al. Steric shielding of surface epitopes and impaired immune recognition induced by the ebola virus glycoprotein. PLoS Pathogens. 2010;6(9):e1001098
  60. 60. Bradfute SB et al. Mechanisms and consequences of ebolavirus-induced lymphocyte apoptosis. The Journal of Immunology. 2010;184(1):327-335
  61. 61. Zaki S, Goldsmith C. Pathologic features of filovirus infections in humans. Current Topics in Microbiology and Immunology. 1999;235:97-116
  62. 62. Prescott JB et al. Immunobiology of Ebola and Lassa virus infections. Nature Reviews Immunology. 2017;17(3):195-207
  63. 63. Leroy E et al. Early immune responses accompanying human asymptomatic Ebola infections. Clinical & Experimental Immunology. 2001;124(3):453-460
  64. 64. Warfield KL et al. Role of natural killer cells in innate protection against lethal ebola virus infection. The Journal of Experimental Medicine. 2004;200(2):169-179
  65. 65. Menicucci AR et al. Transcriptome analysis of circulating immune cell subsets highlight the role of monocytes in Zaire Ebola virus Makona pathogenesis. Frontiers in Immunology. 2017;8:1372
  66. 66. Chong WC et al. The complex interplay between endoplasmic reticulum stress and the NLRP3 inflammasome: A potential therapeutic target for inflammatory disorders. Clinical & Translational Immunology. 2021;10(2):e1247
  67. 67. Halfmann P, Hill-Batorski L, Kawaoka Y. The induction of IL-1β secretion through the NLRP3 inflammasome during Ebola virus infection. The Journal of Infectious Diseases. 2018;218(suppl_5):S504-S507
  68. 68. McElroy AK et al. Macrophage activation marker soluble CD163 associated with fatal and severe Ebola virus disease in humans. Emerging Infectious Diseases. 2019;25(2):290
  69. 69. Ströher U et al. Infection and activation of monocytes by Marburg and Ebola viruses. Journal of Virology. 2001;75(22):11025-11033
  70. 70. Dahlmann F et al. Analysis of Ebola virus entry into macrophages. The Journal of Infectious Diseases. 2015;212(suppl_2):S247-S257
  71. 71. Simmons G et al. DC-SIGN and DC-SIGNR bind ebola glycoproteins and enhance infection of macrophages and endothelial cells. Virology. 2003;305(1):115-123
  72. 72. Usami K et al. Involvement of viral envelope GP2 in Ebola virus entry into cells expressing the macrophage galactose-type C-type lectin. Biochemical and Biophysical Research Communications. 2011;407(1):74-78
  73. 73. Bray M, Geisbert TW. Ebola virus: The role of macrophages and dendritic cells in the pathogenesis of Ebola hemorrhagic fever. The International Journal of Biochemistry & Cell Biology. 2005;37(8):1560-1566
  74. 74. Mohamadzadeh M, Chen L, Schmaljohn AL. How Ebola and Marburg viruses battle the immune system. Nature Reviews Immunology. 2007;7(7):556-567
  75. 75. Olukitibi TA et al. Dendritic cells/macrophages-targeting feature of Ebola glycoprotein and its potential as immunological facilitator for antiviral vaccine approach. Microorganisms. 2019;7(10):402

Written By

Parastoo Yousefi and Alireza Tabibzadeh

Reviewed: 07 April 2022 Published: 06 May 2022