1
|
Mani V, Chu WT, Yang HJ, Morris CP, Laux J, Byrum R, Cooper K, Liu DX, Wang H, Johnson C, Hadley K, Bernbaum JG, Hart R, Anthony SM, Marketon AE, Bernbaum-Cutler R, Pahar B, Worwa G, Kuhn JH, Crozier I, Calcagno C, Gale E. Reactive oxygen species-related oxidative changes are associated with splenic lymphocyte depletion in Ebola virus infection. NPJ IMAGING 2025; 3:16. [PMID: 40291761 PMCID: PMC12021656 DOI: 10.1038/s44303-025-00079-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025]
Abstract
The dysregulated production of reactive oxygen species (ROS) during viral infections may lead to immune cell death and ineffective host responses. ROS dynamics have been under-investigated in severe Ebola virus disease (EVD), a condition in which hyperinflammation and excessive immune cell death are well described but poorly understood. Through ex vivo immunohistochemistry and in vivo ROS-sensitive magnetic resonance imaging (MRI) we demonstrate significant ROS-related oxidative changes in the spleens of domestic ferrets exposed to Ebola virus (EBOV). By immunohistochemistry or MRI, detection of splenic ROS was inversely correlated with the number of CD4+/CD8+ T lymphocytes and apoptotic CD8+ lymphocytes, but detection was positively correlated with the frequency of apoptotic CD4+ cells and the number and frequency of apoptotic B lymphocytes. These results suggest that ROS-induced apoptosis may contribute to the loss of splenic CD4+ T lymphocytes in EBOV-exposed ferrets and warrant further investigation of the role of ROS in severe EVD.
Collapse
Affiliation(s)
- Venkatesh Mani
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Winston T. Chu
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Hee-Jeong Yang
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - C. Paul Morris
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Joseph Laux
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Russell Byrum
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Kurt Cooper
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - David X. Liu
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Hui Wang
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Cristal Johnson
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Kyra Hadley
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - John G. Bernbaum
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Randy Hart
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Scott M. Anthony
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Anthony E. Marketon
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Rebecca Bernbaum-Cutler
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Bapi Pahar
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Gabriella Worwa
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Ian Crozier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD USA
| | - Claudia Calcagno
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD USA
| | - Eric Gale
- Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA USA
- Harvard Medical School, Boston, MA USA
| |
Collapse
|
2
|
Haase JA, Marzi A. Molecular virulence determinants of human-pathogenic filoviruses. Adv Virus Res 2025; 121:1-29. [PMID: 40379380 DOI: 10.1016/bs.aivir.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2025]
Abstract
The Filoviridae family encompasses Ebola virus (EBOV) and Marburg virus (MARV), some of the most lethal viruses known to cause sporadic, recurring outbreaks of severe hemorrhagic fever mainly throughout central Africa. However, other lesser-known viruses also belong to the filovirus family as they are closely related, such as Bundibugyo, Reston and Taï Forest virus. These viruses differ in their virulence in humans significantly: while EBOV and MARV show lethality in humans of up to 90 %, Reston virus appears to be avirulent in humans. Here, underlying molecular factors leading to differences in virulence via changes in filovirus entry, replication and immune evasion strategies are summarized and assessed. While the filovirus glycoprotein contributes towards virulence by facilitating entry into a wide variety of tissues, differences in virus-host interactions and replication efficacies lead to measurable variances of progeny virus production. Additionally, immune evasion strategies lead to alterations in replication efficacy thus changing who has the upper hand between the virus and the host. Understanding and unraveling the contributions of these molecular determinants on filovirus virulence provide insights into the processes causing the underlying pathogenesis. It will further help to assess the pathogenicity of newly discovered filoviruses. Finally, these molecular determinants and processes present attractive targets for therapeutic intervention and development of novel antiviral countermeasures.
Collapse
Affiliation(s)
- Jil A Haase
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States.
| |
Collapse
|
3
|
Zhang Y, Zhang M, Wu H, Wu X, Zheng H, Feng J, Wang M, Wang J, Luo L, Xiao H, Qiao C, Li X, Zheng Y, Huang W, Wang Y, Wang Y, Feng J, Chen G. Afucosylated anti-EBOV antibody MIL77-3 engages sGP to elicit NK cytotoxicity. J Virol 2024; 98:e0068524. [PMID: 39162435 PMCID: PMC11406966 DOI: 10.1128/jvi.00685-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 06/24/2024] [Indexed: 08/21/2024] Open
Abstract
MIL77-3 is one component of antibody cocktail that is produced in our lab and represents an effective regimen for animals suffering from Zaire Ebolavirus (EBOV) infection. MIL77-3 is engineered to increase its affinity for the FcγRIIIa (CD16a) by deleting the fucose in the framework region. The potential effects of this modification on host immune responses, however, remain largely unknown. Herein, we demonstrated that MIL77-3 recognized secreted glycoproptein (sGP), produced by EBOV, and formed the immunocomplex to potently augment antibody-dependent cytotoxicity of human peripheral blood-derived natural killer cells (pNKs), including CD56dim and CD56bright subpopulations, in contrast to the counterparts (Mab114, rEBOV548, fucosylated MIL77-3). Intriguingly, this effect was not observed when NK92-CD16a cell line was utilized and restored by the addition of beads-coupled or membrane-anchored sGP in combination with MIL77-3. Furthermore, sGP bound to unrecognized receptors on T cells contaminated in pNKs rather than NK92-CD16a cells. Administration of beads-coupled sGP/MIL77-3 complex in mice elicited NK activation. Overall, this work reveals an immune-stimulating function of sGP/MIL77-3 complex by triggering cytotoxic activity of NK cells, highlighting the necessity to evaluate the potential impact of MIL77-3 on host immune reaction in clinical trials. IMPORTANCE Zaire Ebolavirus (EBOV) is highly lethal and causes sporadic outbreaks. The passive administration of monoclonal antibodies (mAbs) represents a promising treatment regimen against EBOV. Mounting evidence has shown that the efficacy of a subset of therapeutic mAbs in vivo is intimately associated with its capacity to trigger NK activity, supporting glycomodification of Fc region of anti-EBOV mAbs as a putative strategy to enhance Fc-mediated immune effector function as well as protection in vivo. Our work here uncovers the potential harmful influence of this modification on host immune responses, especially for mAbs with cross-reactivity to secreted glycoproptein (sGP) (e.g., MIL77-3), and highlights it is necessary to evaluate the NK-stimulating activity of a fucosylated mAb engaged with sGP when a new candidate is developed.
Collapse
Affiliation(s)
- Yuting Zhang
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Min Zhang
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Haiyan Wu
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiaonan Wu
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Hang Zheng
- Institute of Pharmacology and Toxicology, Beijing, China
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Junjuan Feng
- Institute of Pharmacology and Toxicology, Beijing, China
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Mianjing Wang
- Institute of Pharmacology and Toxicology, Beijing, China
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Jing Wang
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Longlong Luo
- Institute of Pharmacology and Toxicology, Beijing, China
| | - He Xiao
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Chunxia Qiao
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Xinying Li
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Yuanqiang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Yi Wang
- Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jiannan Feng
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Guojiang Chen
- Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
4
|
Kasereka MC, Mukadi-Bamuleka D, Kitenge-Omasumbu R, Edidi-Atani F, Kuamfumu MM, Mulangu S, Tshiani-Mbaya O, Malengera Vicky K, Mbala-Kingebeni P, Ahuka-Mundeke S, Muyembe-Tamfum JJ, Lee BE, Houston S, Mumtaz Z, Hawkes MT. Rhabdomyolysis, Acute Kidney Injury, and Mortality in Ebola Virus Disease: Retrospective Analysis of Cases From the Eastern Democratic Republic of the Congo, 2019. J Infect Dis 2024; 230:e465-e473. [PMID: 38696335 PMCID: PMC11326845 DOI: 10.1093/infdis/jiae224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/13/2024] [Accepted: 04/28/2024] [Indexed: 05/04/2024] Open
Abstract
BACKGROUND Skeletal muscle injury in Ebola virus disease (EVD) has been reported, but its association with morbidity and mortality remains poorly defined. METHODS This retrospective study included patients admitted to 2 EVD treatment units over an 8-month period in 2019 during an EVD epidemic in the Democratic Republic of the Congo. RESULTS An overall 333 patients (median age, 30 years; 58% female) had at least 1 creatine kinase (CK) measurement (n = 2229; median, 5/patient [IQR, 1-11]). Among patients, 271 (81%) had an elevated CK level (>380 U/L); 202 (61%) had rhabdomyolysis (CK >1000 IU/L); and 45 (14%) had severe rhabdomyolysis (≥5000 U/L). Among survivors, the maximum CK level was a median 1600 (IQR, 550-3400), peaking 3.4 days after admission (IQR, 2.3-5.5) and decreasing thereafter. Among fatal cases, the CK rose monotonically until death, with a median maximum CK level of 2900 U/L (IQR, 1500-4900). Rhabdomyolysis at admission was an independent predictor of acute kidney injury (adjusted odds ratio, 2.2 [95% CI, 1.2-3.8]; P = .0065) and mortality (adjusted hazard ratio, 1.7 [95% CI, 1.03-2.9]; P = .037). CONCLUSIONS Rhabdomyolysis is associated with acute kidney injury and mortality in patients with EVD. These findings may inform clinical practice by identifying laboratory monitoring priorities and highlighting the importance of fluid management.
Collapse
Affiliation(s)
- Masumbuko Claude Kasereka
- Department of Medicine, Université Catholique du Graben, Butembo, Democratic Republic of the Congo
- School of Public Health, University of Alberta, Edmonton, Canada
| | - Daniel Mukadi-Bamuleka
- Department of Virology, Institut National de Recherche Biomédicale
- Service of Microbiology, Department of Medical Biology, University of Kinshasa
| | - Richard Kitenge-Omasumbu
- Programme National d'Urgences et Actions Humanitaires, Ministry of Health of the Democratic Republic of the Congo, Kinshasa
| | - François Edidi-Atani
- Department of Virology, Institut National de Recherche Biomédicale
- Service of Microbiology, Department of Medical Biology, University of Kinshasa
| | - Meris Matondo Kuamfumu
- Department of Virology, Institut National de Recherche Biomédicale
- Service of Microbiology, Department of Medical Biology, University of Kinshasa
| | - Sabue Mulangu
- Department of Virology, Institut National de Recherche Biomédicale
- Service of Microbiology, Department of Medical Biology, University of Kinshasa
| | - Olivier Tshiani-Mbaya
- Department of Virology, Institut National de Recherche Biomédicale
- Service of Microbiology, Department of Medical Biology, University of Kinshasa
| | - Kambale Malengera Vicky
- Department of Medicine, Université Catholique du Graben, Butembo, Democratic Republic of the Congo
| | - Placide Mbala-Kingebeni
- Department of Virology, Institut National de Recherche Biomédicale
- Service of Microbiology, Department of Medical Biology, University of Kinshasa
| | - Steve Ahuka-Mundeke
- Department of Virology, Institut National de Recherche Biomédicale
- Service of Microbiology, Department of Medical Biology, University of Kinshasa
| | - Jean-Jacques Muyembe-Tamfum
- Department of Virology, Institut National de Recherche Biomédicale
- Service of Microbiology, Department of Medical Biology, University of Kinshasa
| | - Bonita E Lee
- Department of Pediatrics, University of Alberta, Edmonton
| | - Stan Houston
- School of Public Health, University of Alberta, Edmonton, Canada
| | - Zubia Mumtaz
- School of Public Health, University of Alberta, Edmonton, Canada
| | - Michael T Hawkes
- School of Public Health, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
5
|
Zarate-Sanchez E, George SC, Moya ML, Robertson C. Vascular dysfunction in hemorrhagic viral fevers: opportunities for organotypic modeling. Biofabrication 2024; 16:032008. [PMID: 38749416 PMCID: PMC11151171 DOI: 10.1088/1758-5090/ad4c0b] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
The hemorrhagic fever viruses (HFVs) cause severe or fatal infections in humans. Named after their common symptom hemorrhage, these viruses induce significant vascular dysfunction by affecting endothelial cells, altering immunity, and disrupting the clotting system. Despite advances in treatments, such as cytokine blocking therapies, disease modifying treatment for this class of pathogen remains elusive. Improved understanding of the pathogenesis of these infections could provide new avenues to treatment. While animal models and traditional 2D cell cultures have contributed insight into the mechanisms by which these pathogens affect the vasculature, these models fall short in replicatingin vivohuman vascular dynamics. The emergence of microphysiological systems (MPSs) offers promising avenues for modeling these complex interactions. These MPS or 'organ-on-chip' models present opportunities to better mimic human vascular responses and thus aid in treatment development. In this review, we explore the impact of HFV on the vasculature by causing endothelial dysfunction, blood clotting irregularities, and immune dysregulation. We highlight how existing MPS have elucidated features of HFV pathogenesis as well as discuss existing knowledge gaps and the challenges in modeling these interactions using MPS. Understanding the intricate mechanisms of vascular dysfunction caused by HFV is crucial in developing therapies not only for these infections, but also for other vasculotropic conditions like sepsis.
Collapse
Affiliation(s)
- Evelyn Zarate-Sanchez
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Monica L Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Claire Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
- UC Davis Comprehensive Cancer Center, Davis, CA, United States of America
| |
Collapse
|
6
|
Guito JC, Arnold CE, Schuh AJ, Amman BR, Sealy TK, Spengler JR, Harmon JR, Coleman-McCray JD, Sanchez-Lockhart M, Palacios GF, Towner JS, Prescott JB. Peripheral immune responses to filoviruses in a reservoir versus spillover hosts reveal transcriptional correlates of disease. Front Immunol 2024; 14:1306501. [PMID: 38259437 PMCID: PMC10800976 DOI: 10.3389/fimmu.2023.1306501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/27/2023] [Indexed: 01/24/2024] Open
Abstract
Several filoviruses, including Marburg virus (MARV), cause severe disease in humans and nonhuman primates (NHPs). However, the Egyptian rousette bat (ERB, Rousettus aegyptiacus), the only known MARV reservoir, shows no overt illness upon natural or experimental infection, which, like other bat hosts of zoonoses, is due to well-adapted, likely species-specific immune features. Despite advances in understanding reservoir immune responses to filoviruses, ERB peripheral blood responses to MARV and how they compare to those of diseased filovirus-infected spillover hosts remain ill-defined. We thus conducted a longitudinal analysis of ERB blood gene responses during acute MARV infection. These data were then contrasted with a compilation of published primate blood response studies to elucidate gene correlates of filovirus protection versus disease. Our work expands on previous findings in MARV-infected ERBs by supporting both host resistance and disease tolerance mechanisms, offers insight into the peripheral immunocellular repertoire during infection, and provides the most direct known cross-examination between reservoir and spillover hosts of the most prevalently-regulated response genes, pathways and activities associated with differences in filovirus pathogenesis and pathogenicity.
Collapse
Affiliation(s)
- Jonathan C. Guito
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Catherine E. Arnold
- Biological Defense Research Directorate, Naval Medical Research Center, Frederick, MD, United States
- RD-CBR, Research and Development Directorate, Chemical and Biological Technologies Directorate, Research Center of Excellence, Defense Threat Reduction Agency, Fort Belvoir, VA, United States
| | - Amy J. Schuh
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Brian R. Amman
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Tara K. Sealy
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jessica R. Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jessica R. Harmon
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Joann D. Coleman-McCray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Mariano Sanchez-Lockhart
- Center for Genome Sciences, Molecular Biology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Gustavo F. Palacios
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jonathan S. Towner
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Joseph B. Prescott
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
- Center for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
7
|
Mwesigwa B, Houser KV, Hofstetter AR, Ortega-Villa AM, Naluyima P, Kiweewa F, Nakabuye I, Yamshchikov GV, Andrews C, O'Callahan M, Strom L, Schech S, Anne Eller L, Sondergaard EL, Scott PT, Amare MF, Modjarrad K, Wamala A, Tindikahwa A, Musingye E, Nanyondo J, Gaudinski MR, Gordon IJ, Holman LA, Saunders JG, Costner PJM, Mendoza FH, Happe M, Morgan P, Plummer SH, Hickman SP, Vazquez S, Murray T, Cordon J, Dulan CNM, Hunegnaw R, Basappa M, Padilla M, Gajjala SR, Swanson PA, Lin BC, Coates EE, Gall JG, McDermott AB, Koup RA, Mascola JR, Ploquin A, Sullivan NJ, Kibuuka H, Ake JA, Ledgerwood JE. Safety, tolerability, and immunogenicity of the Ebola Sudan chimpanzee adenovirus vector vaccine (cAd3-EBO S) in healthy Ugandan adults: a phase 1, open-label, dose-escalation clinical trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:1408-1417. [PMID: 37544326 PMCID: PMC10837320 DOI: 10.1016/s1473-3099(23)00344-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Sudan Ebola virus can cause severe viral disease, with an average case fatality rate of 54%. A recent outbreak of Sudan Ebola virus in Uganda caused 55 deaths among 164 confirmed cases in the second half of 2022. Although vaccines and therapeutics specific for Zaire Ebola virus have been approved for use during outbreak situations, Sudan Ebola virus is an antigenically distinct virus with no approved vaccines available. METHODS In this phase 1, open-label, dose-escalation trial we evaluated the safety, tolerability, and immunogenicity of a monovalent chimpanzee adenovirus 3 vaccine against Sudan Ebola virus (cAd3-EBO S) at Makerere University Walter Reed Project in Kampala, Uganda. Study participants were recruited from the Kampala metropolitan area using International Review Board-approved written and electronic media explaining the trial intervention. Healthy adults without previous receipt of Ebola, Marburg, or cAd3 vectored-vaccines were enrolled to receive cAd3-EBO S at either 1 × 1010 or 1 × 1011 particle units (PU) in a single intramuscular vaccination and were followed up for 48 weeks. Primary safety and tolerability endpoints were assessed in all vaccine recipients by reactogenicity for the first 7 days, adverse events for the first 28 days, and serious adverse events throughout the study. Secondary immunogenicity endpoints included evaluation of binding antibody and T-cell responses against the Sudan Ebola virus glycoprotein, and neutralising antibody responses against the cAd3 vector at 4 weeks after vaccination. This study is registered with ClinicalTrials.gov, NCT04041570, and is completed. FINDINGS 40 healthy adults were enrolled between July 22 and Oct 1, 2019, with 20 receiving 1 × 1010 PU and 20 receiving 1 × 1011 PU of cAd3-EBO S. 38 (95%) participants completed all follow-up visits. The cAd3-EBO S vaccine was well tolerated with no severe adverse events. The most common reactogenicity symptoms were pain or tenderness at the injection site (34 [85%] of 40), fatigue (29 [73%] of 40), and headache (26 [65%] of 40), and were mild to moderate in severity. Positive responses for glycoprotein-specific binding antibodies were induced by 2 weeks in 31 (78%) participants, increased to 34 (85%) participants by 4 weeks, and persisted to 48 weeks in 31 (82%) participants. Most participants developed glycoprotein-specific T-cell responses (20 [59%, 95% CI 41-75] of 34; six participants were removed from the T cell analysis after failing quality control parameters) by 4 weeks after vaccination, and neutralising titres against the cAd3 vector were also increased from baseline (90% inhibitory concentration of 47, 95% CI 30-73) to 4 weeks after vaccination (196, 125-308). INTERPRETATION The cAd3-EBO S vaccine was safe at both doses, rapidly inducing immune responses in most participants after a single injection. The rapid onset and durability of the vaccine-induced antibodies make this vaccine a strong candidate for emergency deployment in Sudan Ebola virus outbreaks. FUNDING National Institutes of Health via interagency agreement with Walter Reed Army Institute of Research.
Collapse
Affiliation(s)
- Betty Mwesigwa
- Makerere University Walter Reed Project, Kampala, Uganda
| | - Katherine V Houser
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Amelia R Hofstetter
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ana M Ortega-Villa
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Galina V Yamshchikov
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Charla Andrews
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mark O'Callahan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Larisa Strom
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Steven Schech
- Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Leigh Anne Eller
- Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Erica L Sondergaard
- Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Paul T Scott
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Mihret F Amare
- Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | | | - Amir Wamala
- Makerere University Walter Reed Project, Kampala, Uganda
| | | | - Ezra Musingye
- Makerere University Walter Reed Project, Kampala, Uganda
| | | | - Martin R Gaudinski
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ingelise J Gordon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - LaSonji A Holman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jamie G Saunders
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela J M Costner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Floreliz H Mendoza
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Myra Happe
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patricia Morgan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah H Plummer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Somia P Hickman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sandra Vazquez
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tamar Murray
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jamilet Cordon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Caitlyn N M Dulan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ruth Hunegnaw
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Manjula Basappa
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Marcelino Padilla
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Suprabhath R Gajjala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Phillip A Swanson
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bob C Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Emily E Coates
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jason G Gall
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aurélie Ploquin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hannah Kibuuka
- Makerere University Walter Reed Project, Kampala, Uganda
| | - Julie A Ake
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Julie E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Liu DX, Pahar B, Perry DL, Xu H, Cooper TK, Huzella LM, Hart RJ, Hischak AMW, Bernbaum J, St Claire M, Byrum R, Bennett RS, Warren T, Holbrook MR, Hensley LE, Crozier I, Schmaljohn CS. Depletion of Bone Marrow Hematopoietic Cells in Ebolavirus-Infected Rhesus Macaques: A Possible Cause of Hematologic Abnormalities in Ebolavirus Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:2031-2046. [PMID: 37689386 PMCID: PMC10699128 DOI: 10.1016/j.ajpath.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/18/2023] [Accepted: 08/25/2023] [Indexed: 09/11/2023]
Abstract
The pathophysiology of long-recognized hematologic abnormalities in Ebolavirus (EBOV) disease (EVD) is unknown. From limited human sampling (of peripheral blood), it has been postulated that emergency hematopoiesis plays a role in severe EVD, but the systematic characterization of the bone marrow (BM) has not occurred in human disease or in nonhuman primate models. In a lethal rhesus macaque model of EVD, 18 sternal BM samples exposed to the Kikwit strain of EBOV were compared to those from uninfected controls (n = 3). Immunohistochemistry, RNAscope in situ hybridization, transmission electron microscopy, and confocal microscopy showed that EBOV infects BM monocytes/macrophages and megakaryocytes. EBOV exposure was associated with severe BM hypocellularity, including depletion of myeloid, erythroid, and megakaryocyte hematopoietic cells. These depletions were negatively correlated with cell proliferation (Ki67 expression) and were not associated with BM apoptosis during disease progression. In EBOV-infected rhesus macaques with terminal disease, BM showed marked hemophagocytosis, megakaryocyte emperipolesis, and the release of immature hematopoietic cells into the sinusoids. Collectively, these data demonstrate not only direct EBOV infection of BM monocytes/macrophages and megakaryocytes but also that disease progression is associated with hematopoietic failure, notably in peripheral cytopenia. These findings inform current pathophysiologic unknowns and suggest a crucial role for BM dysfunction and/or failure, including emergency hematopoiesis, as part of the natural history of severe human disease.
Collapse
Affiliation(s)
- David X Liu
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland.
| | - Bapi Pahar
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Donna L Perry
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Huanbin Xu
- Department of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, Frederick, Maryland
| | - Timothy K Cooper
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Louis M Huzella
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Randy J Hart
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Amanda M W Hischak
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - John Bernbaum
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Marisa St Claire
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Russell Byrum
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Richard S Bennett
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Travis Warren
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Michael R Holbrook
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Lisa E Hensley
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| | - Ian Crozier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Connie S Schmaljohn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland
| |
Collapse
|
9
|
Djurkovic MA, Leavitt CG, Arnett E, Kriachun V, Martínez-Sobrido L, Titone R, Sherwood LJ, Hayhurst A, Schlesinger LS, Shtanko O. Ebola Virus Uses Tunneling Nanotubes as an Alternate Route of Dissemination. J Infect Dis 2023; 228:S522-S535. [PMID: 37723997 PMCID: PMC10651192 DOI: 10.1093/infdis/jiad400] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023] Open
Abstract
Ebola virus (EBOV) disease is marked by rapid virus replication and spread. EBOV enters the cell by macropinocytosis and replicates in the cytoplasm, and nascent virions egress from the cell surface to infect neighboring cells. Here, we show that EBOV uses an alternate route to disseminate: tunneling nanotubes (TNTs). TNTs, an actin-based long-range intercellular communication system, allows for direct exchange of cytosolic constituents between cells. Using live, scanning electron, and high-resolution quantitative 3-dimensional microscopy, we show that EBOV infection of primary human cells results in the enhanced formation of TNTs containing viral nucleocapsids. TNTs promote the intercellular transfer of nucleocapsids in the absence of live virus, and virus could replicate in cells devoid of entry factors after initial stall. Our studies suggest an alternate model of EBOV dissemination within the host, laying the groundwork for further investigations into the pathogenesis of filoviruses and, importantly, stimulating new areas of antiviral design.
Collapse
Affiliation(s)
- Marija A Djurkovic
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Carson G Leavitt
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Eusondia Arnett
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Valeriia Kriachun
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Luis Martínez-Sobrido
- Disease Prevention and Intervention, Texas Biomedical Research Institute, San Antonio
| | - Rossella Titone
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Laura J Sherwood
- Disease Prevention and Intervention, Texas Biomedical Research Institute, San Antonio
| | - Andrew Hayhurst
- Disease Prevention and Intervention, Texas Biomedical Research Institute, San Antonio
| | - Larry S Schlesinger
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Olena Shtanko
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
- Disease Prevention and Intervention, Texas Biomedical Research Institute, San Antonio
| |
Collapse
|
10
|
Escudero-Pérez B, Lawrence P, Castillo-Olivares J. Immune correlates of protection for SARS-CoV-2, Ebola and Nipah virus infection. Front Immunol 2023; 14:1156758. [PMID: 37153606 PMCID: PMC10158532 DOI: 10.3389/fimmu.2023.1156758] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/20/2023] [Indexed: 05/09/2023] Open
Abstract
Correlates of protection (CoP) are biological parameters that predict a certain level of protection against an infectious disease. Well-established correlates of protection facilitate the development and licensing of vaccines by assessing protective efficacy without the need to expose clinical trial participants to the infectious agent against which the vaccine aims to protect. Despite the fact that viruses have many features in common, correlates of protection can vary considerably amongst the same virus family and even amongst a same virus depending on the infection phase that is under consideration. Moreover, the complex interplay between the various immune cell populations that interact during infection and the high degree of genetic variation of certain pathogens, renders the identification of immune correlates of protection difficult. Some emerging and re-emerging viruses of high consequence for public health such as SARS-CoV-2, Nipah virus (NiV) and Ebola virus (EBOV) are especially challenging with regards to the identification of CoP since these pathogens have been shown to dysregulate the immune response during infection. Whereas, virus neutralising antibodies and polyfunctional T-cell responses have been shown to correlate with certain levels of protection against SARS-CoV-2, EBOV and NiV, other effector mechanisms of immunity play important roles in shaping the immune response against these pathogens, which in turn might serve as alternative correlates of protection. This review describes the different components of the adaptive and innate immune system that are activated during SARS-CoV-2, EBOV and NiV infections and that may contribute to protection and virus clearance. Overall, we highlight the immune signatures that are associated with protection against these pathogens in humans and could be used as CoP.
Collapse
Affiliation(s)
- Beatriz Escudero-Pérez
- WHO Collaborating Centre for Arbovirus and Haemorrhagic Fever Reference and Research, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, Braunschweig, Germany
- *Correspondence: Beatriz Escudero-Pérez, ; Javier Castillo-Olivares,
| | - Philip Lawrence
- CONFLUENCE: Sciences et Humanités (EA 1598), Université Catholique de Lyon (UCLy), Lyon, France
| | - Javier Castillo-Olivares
- Laboratory of Viral Zoonotics, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Beatriz Escudero-Pérez, ; Javier Castillo-Olivares,
| |
Collapse
|
11
|
Gourronc FA, Rebagliati M, Kramer-Riesberg B, Fleck AM, Patten JJ, Geohegan-Barek K, Messingham KN, Davey RA, Maury W, Klingelhutz AJ. Adipocytes are susceptible to Ebola Virus infection. Virology 2022; 573:12-22. [DOI: 10.1016/j.virol.2022.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 12/23/2022]
|
12
|
Bhattacharyya S. Mechanisms of Immune Evasion by Ebola Virus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:15-22. [PMID: 34661889 DOI: 10.1007/978-3-030-67452-6_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
The 2013-2016 Ebola virus epidemic in West Africa, which also spread to the USA, UK and Europe, was the largest reported outbreak till date (World Health Organization. 2016. https://apps.who.int/iris/bitstream/handle/10665/208883/ebolasitrep_10Jun2016_eng.pdf;jsessionid=8B7D74BC9D82D2BE1B110BAFFAD3A6E6?sequence=1 ). The recent Ebola outbreak in the Democratic Republic of the Congo has raised immense global concern on this severe and often fatal infection. Although sporadic, the severity and lethality of Ebola virus disease outbreaks has led to extensive research worldwide on this virus. Vaccine (World Health Organization. 2016. https://www.who.int/en/news-room/detail/23-12-2016-final-trial-results-confirm-ebola-vaccine-provides-high-protection-against-disease ; Henao-Restrepo et al. Lancet 389:505-518, 2017) and drug (Hayden. Nature, 557, 475-476, 2018; Dyall et al. J Infect Dis 218(suppl_5), S672-S678, 2018) development efforts against Ebola virus are research hotspots, and a few approved therapeutics are currently available (Centers for Disease Control and Prevention. 2021. https://www.cdc.gov/vhf/ebola/clinicians/vaccine/index.html; Centers for Disease Control and Prevention. 2021. https://www.cdc.gov/vhf/ebola/treatment/index.html). Ebola virus has evolved several mechanisms of host immune evasion, which facilitate its replication and pathogenesis. This chapter describes the Ebola virus morphology, genome, entry, replication, pathogenesis and viral proteins involved in host immune evasion. Further understanding of the underlying molecular mechanisms of immune evasion may facilitate development of additional novel and sustainable strategies against this deadly virus.
Collapse
Affiliation(s)
- Suchita Bhattacharyya
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK.
| |
Collapse
|
13
|
Wolfe DN, Sabourin CL, Merchlinsky MJ, Florence WC, Wolfraim LA, Taylor KL, Ward LA. Selection of Filovirus Isolates for Vaccine Development Programs. Vaccines (Basel) 2021; 9:vaccines9091045. [PMID: 34579282 PMCID: PMC8471873 DOI: 10.3390/vaccines9091045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 01/25/2023] Open
Abstract
The continuing outbreaks of ebola virus disease highlight the ongoing threat posed by filoviruses. Fortunately, licensed vaccines and therapeutics are now available for Zaire ebolavirus. However, effective medical countermeasures, such as vaccines for other filoviruses such as Sudan ebolavirus and the Marburg virus, are presently in early stages of development and, in the absence of a large outbreak, would require regulatory approval via the U.S. Food and Drug Administration (FDA) Animal Rule. The selection of an appropriate animal model and virus challenge isolates for nonclinical studies are critical aspects of the development program. Here, we have focused on the recommendation of challenge isolates for Sudan ebolavirus and Marburg virus. Based on analyses led by the Filovirus Animal and Nonclinical Group (FANG) and considerations for strain selection under the FDA Guidance for the Animal Rule, we propose prototype virus isolates for use in nonclinical challenge studies.
Collapse
Affiliation(s)
- Daniel N. Wolfe
- U.S. Department of Health and Human Services (DHHS), Assistant Secretary for Preparedness and Response (ASPR), Biomedical Advanced Research and Development Authority (BARDA), Washington, DC 20201, USA;
- Correspondence: ; Tel.: +1-(202)-205-8968
| | - Carol L. Sabourin
- Tunnell Government Services, Inc., Supporting Biomedical Advanced Research & Development Authority (BARDA), Assistant Secretary for Preparedness and Response (ASPR), U.S. Department of Health and Human Services (DHHS), Washington, DC 20201, USA;
| | - Michael J. Merchlinsky
- U.S. Department of Health and Human Services (DHHS), Assistant Secretary for Preparedness and Response (ASPR), Biomedical Advanced Research and Development Authority (BARDA), Washington, DC 20201, USA;
| | - William C. Florence
- U.S. Department of Health and Human Services (DHHS), National Institutes of Health (NIH), National Institute of Allergy and Infectious Diseases (NIAID), Rockville, MD 20852, USA; (W.C.F.); (L.A.W.); (K.L.T.)
| | - Larry A. Wolfraim
- U.S. Department of Health and Human Services (DHHS), National Institutes of Health (NIH), National Institute of Allergy and Infectious Diseases (NIAID), Rockville, MD 20852, USA; (W.C.F.); (L.A.W.); (K.L.T.)
| | - Kimberly L. Taylor
- U.S. Department of Health and Human Services (DHHS), National Institutes of Health (NIH), National Institute of Allergy and Infectious Diseases (NIAID), Rockville, MD 20852, USA; (W.C.F.); (L.A.W.); (K.L.T.)
| | - Lucy A. Ward
- U.S. Department of Defense (DOD), Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND), Joint Project Manager for Chemical, Biological, Radiological, and Nuclear Medical (JPM CBRN Medical), Fort Detrick, MD 21702, USA;
| |
Collapse
|
14
|
Borisevich GV, Kirillova SL, Shatokhina IV, Lebedev VN, Shagarova NV, Syromyatnikova SI, Andrus AF, Koval'chuk EA, Kirillov VB, Bespalov ML, Petrov AA, Koval'chuk AV, Pantyukhov VB, Kutayev DA, Borisevich SV, Kuznetsov SL. [Flow cytometry evaluation of the rhesus monkey cellular immunity following the Zaire ebolavirus (Filoviridae; Ebolavirus: Zaire ebolavirus) experimental infection]. Vopr Virusol 2021; 66:289-298. [PMID: 34545721 DOI: 10.36233/0507-4088-64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 09/18/2021] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The outbreaks of the Zaire ebolavirus (ZE) disease (ZED) that have arisen in the last decade determine the need to study the infection pathogenesis, the formation of specific immunity forming as well as the development of effective preventive and therapeutic means. All stages of fight against the ZED spread require the experimental infection in sensitive laboratory animals, which are rhesus monkeys in case of this disease .The aim of the study is to evaluate the rhesus monkey cellular immunity following the ZE experimental infection by the means of flow cytometry (cytofluorimetry). MATERIAL AND METHODS Male rhesus monkeys were intramuscularly infected by the dose of 15 LD50 (dose of the pathogen that causes 50% mortality of infected animals) of the ZE, the Zaire strain (ZEBOV). Levels of 18 peripheral blood lymphocyte populations of the animals before the ZE experimental infection and at the terminal stage of the disease were assessed using flow cytometry. RESULTS AND DISCUSSION The certain changes in the levels of the lymphocyte populations were observed following infection, indicating simultaneous activation and suppression of the immune system during ZED. The increase in content was observed for T-lymphocytes, T-helper and cytotoxic T-lymphocytes expressing the corresponding markers of early activation. The decrease was recorded for T-lymphocytes and double-positive T-lymphocytes expressing corresponding markers of late activation, as well as natural killer cells expressing CD8 (p < 0.05). CONCLUSION For the first time in the Russian Federation, the rhesus monkey cellular immunity before and after the ZE experimental infection was assessed using flow cytometry.
Collapse
Affiliation(s)
- G V Borisevich
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - S L Kirillova
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - I V Shatokhina
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - V N Lebedev
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - N V Shagarova
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - S I Syromyatnikova
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - A F Andrus
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - E A Koval'chuk
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - V B Kirillov
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | | | - A A Petrov
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - A V Koval'chuk
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - V B Pantyukhov
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - D A Kutayev
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - S V Borisevich
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - S L Kuznetsov
- Directorate of the Chief of the Radiation, Chemical, and Biological Defence Troops of the Armed Forces of the Ministry of Defence of the Russian Federation
| |
Collapse
|
15
|
Pinski AN, Maroney KJ, Marzi A, Messaoudi I. Distinct transcriptional responses to fatal Ebola virus infection in cynomolgus and rhesus macaques suggest species-specific immune responses. Emerg Microbes Infect 2021; 10:1320-1330. [PMID: 34112056 PMCID: PMC8253202 DOI: 10.1080/22221751.2021.1942229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Ebola virus (EBOV) is a negative single-stranded RNA virus within the Filoviridae family and the causative agent of Ebola virus disease (EVD). Nonhuman primates (NHPs), including cynomolgus and rhesus macaques, are considered the gold standard animal model to interrogate mechanisms of EBOV pathogenesis. However, despite significant genetic similarity (>90%), NHP species display different clinical presentation following EBOV infection, notably a ∼1-2 days delay in disease progression. Consequently, evaluation of therapeutics is generally conducted in rhesus macaques, whereas cynomolgus macaques are utilized to determine efficacy of preventative treatments, notably vaccines. This observation is in line with reported differences in disease severity and host responses between these two NHP following infection with simian varicella virus, influenza A and SARS-CoV-2. However, the molecular underpinnings of these differential outcomes following viral infections remain poorly defined. In this study, we compared published transcriptional profiles obtained from cynomolgus and rhesus macaques infected with the EBOV-Makona Guinea C07 using bivariate and regression analyses to elucidate differences in host responses. We report the presence of a shared core of differentially expressed genes (DEGs) reflecting EVD pathology, including aberrant inflammation, lymphopenia, and coagulopathy. However, the magnitudes of change differed between the two macaque species. These findings suggest that the differential clinical presentation of EVD in these two species is mediated by altered transcriptional responses.
Collapse
Affiliation(s)
- Amanda N Pinski
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine CA, USA
| | - Kevin J Maroney
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine CA, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine CA, USA.,Center for Virus Research, University of California Irvine, Irvine, CA, USA.,Institute for Immunology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
16
|
Antibody responses to filovirus infections in humans: protective or not? THE LANCET. INFECTIOUS DISEASES 2021; 21:e348-e355. [PMID: 34175003 DOI: 10.1016/s1473-3099(21)00006-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/17/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022]
Abstract
Disease outbreaks caused by Ebola virus and other filoviruses highlight the urgent need for an in-depth understanding of the role of antibody responses in recovery. In this Personal View we aim to discuss the controversial biological role of antibodies during natural filovirus infections in humans. Survival during natural human filovirus infections correlates with the magnitude of the process of antibodies binding to the filovirus glycoprotein and neutralising the virus. Despite the severity of the disease, highly potent monoclonal antibodies have been isolated from survivors of natural filovirus infections, suggesting that the magnitude of the antibody response is insufficient for prevention of severe disease. Unlike natural infections, filovirus vaccines, which express the viral glycoprotein, do induce protective concentrations of antibodies, albeit only when administered at very high doses. Multiple mechanisms by which filoviruses can delay and reduce the antibody response have been identified in the past decade. Furthermore, subneutralising antibody concentrations have been shown to enhance filovirus infections of immune cells bearing Fc receptors. Understanding the role of antibody responses during natural filovirus infections is important for the development of safe and potent vaccines and antibody-based treatments.
Collapse
|
17
|
Wawina-Bokalanga T, Vanmechelen B, Lhermitte V, Martí-Carreras J, Vergote V, Koundouno FR, Akoi-Boré J, Thom R, Tipton T, Steeds K, Moussa KB, Amento A, Laenen L, Duraffour S, Gabriel M, Ruibal P, Hall Y, Kader-Kondé M, Günther S, Baele G, Muñoz-Fontela C, Van Weyenbergh J, Carroll MW, Maes P. Human Diversity of Killer Cell Immunoglobulin-Like Receptors and Human Leukocyte Antigen Class I Alleles and Ebola Virus Disease Outcomes. Emerg Infect Dis 2021; 27:76-84. [PMID: 33350932 PMCID: PMC7774578 DOI: 10.3201/eid2701.202177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We investigated the genetic profiles of killer cell immunoglobulin-like receptors (KIRs) in Ebola virus–infected patients. We studied the relationship between KIR–human leukocyte antigen (HLA) combinations and the clinical outcomes of patients with Ebola virus disease (EVD). We genotyped KIRs and HLA class I alleles using DNA from uninfected controls, EVD survivors, and persons who died of EVD. The activating 2DS4–003 and inhibitory 2DL5 genes were significantly more common among persons who died of EVD; 2DL2 was more common among survivors. We used logistic regression analysis and Bayesian modeling to identify 2DL2, 2DL5, 2DS4–003, HLA-B-Bw4-Thr, and HLA-B-Bw4-Ile as probably having a significant relationship with disease outcome. Our findings highlight the importance of innate immune response against Ebola virus and show the association between KIRs and the clinical outcome of EVD.
Collapse
|
18
|
Schreiber-Stainthorp W, Solomon J, Lee JH, Castro M, Shah S, Martinez-Orengo N, Reeder R, Maric D, Gross R, Qin J, Hagen KR, Johnson RF, Hammoud DA. Longitudinal in vivo imaging of acute neuropathology in a monkey model of Ebola virus infection. Nat Commun 2021; 12:2855. [PMID: 34001896 PMCID: PMC8129091 DOI: 10.1038/s41467-021-23088-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 04/13/2021] [Indexed: 02/03/2023] Open
Abstract
Ebola virus (EBOV) causes neurological symptoms yet its effects on the central nervous system (CNS) are not well-described. Here, we longitudinally assess the acute effects of EBOV on the brain, using quantitative MR-relaxometry, 18F-Fluorodeoxyglucose PET and immunohistochemistry in a monkey model. We report blood-brain barrier disruption, likely related to high cytokine levels and endothelial viral infection, with extravasation of fluid, Gadolinium-based contrast material and albumin into the extracellular space. Increased glucose metabolism is also present compared to the baseline, especially in the deep gray matter and brainstem. This regional hypermetabolism corresponds with mild neuroinflammation, sporadic neuronal infection and apoptosis, as well as increased GLUT3 expression, consistent with increased neuronal metabolic demands. Neuroimaging changes are associated with markers of disease progression including viral load and cytokine/chemokine levels. Our results provide insight into the pathophysiology of CNS involvement with EBOV and may help assess vaccine/treatment efficacy in real time.
Collapse
Affiliation(s)
- William Schreiber-Stainthorp
- Hammoud Laboratory, Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jeffrey Solomon
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ji Hyun Lee
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Marcelo Castro
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Swati Shah
- Hammoud Laboratory, Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Neysha Martinez-Orengo
- Hammoud Laboratory, Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Rebecca Reeder
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA
| | - Robin Gross
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Jing Qin
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Katie R Hagen
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Reed F Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Dima A Hammoud
- Hammoud Laboratory, Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
19
|
Misasi J, Sullivan NJ. Immunotherapeutic strategies to target vulnerabilities in the Ebolavirus glycoprotein. Immunity 2021; 54:412-436. [PMID: 33691133 DOI: 10.1016/j.immuni.2021.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 12/18/2022]
Abstract
The 2014 Ebola virus disease (EVD) outbreak in West Africa and the subsequent outbreaks of 2018-2020 in Equator and North Kivu provinces of the Democratic Republic of the Congo illustrate the public health challenges of emerging and reemerging viruses. EVD has a high case fatality rate with a rapidly progressing syndrome of fever, rash, vomiting, diarrhea, and bleeding diathesis. Recently, two monoclonal-antibody-based therapies received United States Food and Drug Administration (FDA) approval, and there are several other passive immunotherapies that hold promise as therapeutics against other species of Ebolavirus. Here, we review concepts needed to understand mechanisms of action, present an expanded schema to define additional sites of vulnerability on the viral glycoprotein, and review current antibody-based therapeutics. The concepts described are used to gain insights into the key characteristics that represent functional targets for immunotherapies against Zaire Ebolavirus and other emerging viruses within the Ebolavirus genus.
Collapse
Affiliation(s)
- John Misasi
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, Vaccine Research Center, 40 Convent Drive, Bethesda, MD 20892, USA
| | - Nancy J Sullivan
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, Vaccine Research Center, 40 Convent Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
20
|
Development of a Well-Characterized Rhesus Macaque Model of Ebola Virus Disease for Support of Product Development. Microorganisms 2021; 9:microorganisms9030489. [PMID: 33652589 PMCID: PMC7996724 DOI: 10.3390/microorganisms9030489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 11/17/2022] Open
Abstract
Ebola virus (EBOV) is a negative-sense RNA virus that can infect humans and nonhuman primates with severe health consequences. Development of countermeasures requires a thorough understanding of the interaction between host and pathogen, and the course of disease. The goal of this study was to further characterize EBOV disease in a uniformly lethal rhesus macaque model, in order to support development of a well-characterized model following rigorous quality standards. Rhesus macaques were intramuscularly exposed to EBOV and one group was euthanized at predetermined time points to characterize progression of disease. A second group was not scheduled for euthanasia in order to analyze survival, changes in physiology, clinical pathology, terminal pathology, and telemetry kinetics. On day 3, sporadic viremia was observed and pathological evidence was noted in lymph nodes. By day 5, viremia was detected in all EBOV exposed animals and pathological evidence was noted in the liver, spleen, and gastrointestinal tissues. These data support the notion that EBOV infection in rhesus macaques is a rapid systemic disease similar to infection in humans, under a compressed time scale. Biomarkers that correlated with disease progression at the earliest stages of infection were observed thereby identifying potential "trigger-to-treat" for use in therapeutic studies.
Collapse
|
21
|
Transcriptional Analysis of Lymphoid Tissues from Infected Nonhuman Primates Reveals the Basis for Attenuation and Immunogenicity of an Ebola Virus Encoding a Mutant VP35 Protein. J Virol 2021; 95:JVI.01995-20. [PMID: 33408171 DOI: 10.1128/jvi.01995-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022] Open
Abstract
Infection with Zaire ebolavirus (EBOV), a member of the Filoviridae family, causes a disease characterized by high levels of viremia, aberrant inflammation, coagulopathy, and lymphopenia. EBOV initially replicates in lymphoid tissues and disseminates via dendritic cells (DCs) and monocytes to liver, spleen, adrenal gland, and other secondary organs. EBOV protein VP35 is a critical immune evasion factor that inhibits type I interferon signaling and DC maturation. Nonhuman primates (NHPs) immunized with a high dose (5 × 105 PFU) of recombinant EBOV containing a mutated VP35 (VP35m) are protected from challenge with wild-type EBOV (wtEBOV). This protection is accompanied by a transcriptional response in the peripheral blood reflecting a regulated innate immune response and a robust induction of adaptive immune genes. However, the host transcriptional response to VP35m in lymphoid tissues has not been evaluated. Therefore, we conducted a transcriptional analysis of axillary and inguinal lymph nodes and spleen tissues of NHPs infected with a low dose (2 × 104 PFU) of VP35m and then back-challenged with a lethal dose of wtEBOV. VP35m induced early transcriptional responses in lymphoid tissues that are distinct from those observed in wtEBOV challenge. Specifically, we detected robust antiviral innate and adaptive responses and fewer transcriptional changes in genes with roles in angiogenesis, apoptosis, and inflammation. Two of three macaques survived wtEBOV back-challenge, with only the nonsurvivor displaying a transcriptional response reflecting Ebola virus disease. These data suggest that VP35 is a key modulator of early host responses in lymphoid tissues, thereby regulating disease progression and severity following EBOV challenge.IMPORTANCE Zaire Ebola virus (EBOV) infection causes a severe and often fatal disease characterized by inflammation, coagulation defects, and organ failure driven by a defective host immune response. Lymphoid tissues are key sites of EBOV pathogenesis and the generation of an effective immune response to infection. A recent study demonstrated that infection with an EBOV encoding a mutant VP35, a viral protein that antagonizes host immunity, can protect nonhuman primates (NHPs) against lethal EBOV challenge. However, no studies have examined the response to this mutant EBOV in lymphoid tissues. Here, we characterize gene expression in lymphoid tissues from NHPs challenged with the mutant EBOV and subsequently with wild-type EBOV to identify signatures of a protective host response. Our findings are critical for elucidating viral pathogenesis, mechanisms of host antagonism, and the role of lymphoid organs in protective responses to EBOV to improve the development of antivirals and vaccines against EBOV.
Collapse
|
22
|
Jain S, Khaiboullina SF, Baranwal M. Immunological Perspective for Ebola Virus Infection and Various Treatment Measures Taken to Fight the Disease. Pathogens 2020; 9:E850. [PMID: 33080902 PMCID: PMC7603231 DOI: 10.3390/pathogens9100850] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/07/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
Ebolaviruses, discovered in 1976, belongs to the Filoviridae family, which also includes Marburg and Lloviu viruses. They are negative-stranded RNA viruses with six known species identified to date. Ebola virus (EBOV) is a member of Zaire ebolavirus species and can cause the Ebola virus disease (EVD), an emerging zoonotic disease that results in homeostatic imbalance and multi-organ failure. There are three EBOV outbreaks documented in the last six years resulting in significant morbidity (> 32,000 cases) and mortality (> 13,500 deaths). The potential factors contributing to the high infectivity of this virus include multiple entry mechanisms, susceptibility of the host cells, employment of multiple immune evasion mechanisms and rapid person-to-person transmission. EBOV infection leads to cytokine storm, disseminated intravascular coagulation, host T cell apoptosis as well as cell mediated and humoral immune response. In this review, a concise recap of cell types targeted by EBOV and EVD symptoms followed by detailed run-through of host innate and adaptive immune responses, virus-driven regulation and their combined effects contributing to the disease pathogenesis has been presented. At last, the vaccine and drug development initiatives as well as challenges related to the management of infection have been discussed.
Collapse
Affiliation(s)
- Sahil Jain
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala 147004, Punjab, India;
| | - Svetlana F. Khaiboullina
- Department of Microbiology and Immunology, University of Nevada, Reno, NV 89557, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Tatarstan, Russia
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala 147004, Punjab, India;
| |
Collapse
|
23
|
McElroy AK, Akondy RS, Mcllwain DR, Chen H, Bjornson-Hooper Z, Mukherjee N, Mehta AK, Nolan G, Nichol ST, Spiropoulou CF. Immunologic timeline of Ebola virus disease and recovery in humans. JCI Insight 2020; 5:137260. [PMID: 32434986 PMCID: PMC7259516 DOI: 10.1172/jci.insight.137260] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
A complete understanding of human immune responses to Ebola virus infection is limited by the availability of specimens and the requirement for biosafety level 4 (BSL-4) containment. In an effort to bridge this gap, we evaluated cryopreserved PBMCs from 4 patients who survived Ebola virus disease (EVD) using an established mass cytometry antibody panel to characterize various cell populations during both the acute and convalescent phases. Acute loss of nonclassical monocytes and myeloid DCs, especially CD1c+ DCs, was noted. Classical monocyte proliferation and CD38 upregulation on plasmacytoid DCs coincided with declining viral load. Unsupervised analysis of cell abundance demonstrated acute declines in monocytic, NK, and T cell populations, but some populations, many of myeloid origin, increased in abundance during the acute phase, suggesting emergency hematopoiesis. Despite cell losses during the acute phase, upregulation of Ki-67 correlated with recovery of cell populations over time. These data provide insights into the human immune response during EVD.
Collapse
Affiliation(s)
- Anita K McElroy
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Division of Pediatric Infectious Diseases and Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rama S Akondy
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David R Mcllwain
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Han Chen
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Zach Bjornson-Hooper
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Nilanjan Mukherjee
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Aneesh K Mehta
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Garry Nolan
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
24
|
Early Transcriptional Changes within Liver, Adrenal Gland, and Lymphoid Tissues Significantly Contribute to Ebola Virus Pathogenesis in Cynomolgus Macaques. J Virol 2020; 94:JVI.00250-20. [PMID: 32213610 DOI: 10.1128/jvi.00250-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/11/2020] [Indexed: 01/05/2023] Open
Abstract
Ebola virus (EBOV) continues to pose a significant threat to human health, as evidenced by the 2013-2016 epidemic in West Africa and the ongoing outbreak in the Democratic Republic of the Congo. EBOV causes hemorrhagic fever, organ damage, and shock culminating in death, with case fatality rates as high as 90%. This high lethality combined with the paucity of licensed medical countermeasures makes EBOV a critical human pathogen. Although EBOV infection results in significant damage to the liver and the adrenal glands, little is known about the molecular signatures of injury in these organs. Moreover, while changes in peripheral blood cells are becoming increasingly understood, the host responses within organs and lymphoid tissues remain poorly characterized. To address this knowledge gap, we tracked longitudinal transcriptional changes in tissues collected from EBOV-Makona-infected cynomolgus macaques. Following infection, both liver and adrenal glands exhibited significant and early downregulation of genes involved in metabolism, coagulation, hormone synthesis, and angiogenesis; upregulated genes were associated with inflammation. Analysis of lymphoid tissues showed early upregulation of genes that play a role in innate immunity and inflammation and downregulation of genes associated with cell cycle and adaptive immunity. Moreover, transient activation of innate immune responses and downregulation of humoral immune responses in lymphoid tissues were confirmed with flow cytometry. Together, these data suggest that the liver, adrenal gland, and lymphatic organs are important sites of EBOV infection and that dysregulating the function of these vital organs contributes to the development of Ebola virus disease.IMPORTANCE Ebola virus (EBOV) remains a high-priority pathogen since it continues to cause outbreaks with high case fatality rates. Although it is well established that EBOV results in severe organ damage, our understanding of tissue injury in the liver, adrenal glands, and lymphoid tissues remains limited. We begin to address this knowledge gap by conducting longitudinal gene expression studies in these tissues, which were collected from EBOV-infected cynomolgus macaques. We report robust and early gene expression changes within these tissues, indicating they are primary sites of EBOV infection. Furthermore, genes involved in metabolism, coagulation, and adaptive immunity were downregulated, while inflammation-related genes were upregulated. These results indicate significant tissue damage consistent with the development of hemorrhagic fever and lymphopenia. Our study provides novel insight into EBOV-host interactions and elucidates how host responses within the liver, adrenal glands, and lymphoid tissues contribute to EBOV pathogenesis.
Collapse
|
25
|
Pathogenesis of Uveitis in Ebola Virus Disease Survivors: Evolving Understanding from Outbreaks to Animal Models. Microorganisms 2020; 8:microorganisms8040594. [PMID: 32325950 PMCID: PMC7232169 DOI: 10.3390/microorganisms8040594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/05/2020] [Accepted: 04/13/2020] [Indexed: 01/24/2023] Open
Abstract
Ebola virus disease (EVD) and emerging infectious disease threats continue to threaten life, prosperity and global health security. To properly counteract EVD, an improved understanding of the long-term impact of recent EVD outbreaks in West Africa and the Democratic Republic of Congo are needed. In the wake of recent outbreaks, numerous health sequelae were identified in EVD survivors. These findings include joint pains, headaches, myalgias, and uveitis, a vision-threatening inflammatory condition of the eye. Retrospective and more recent prospective studies of EVD survivors from West Africa have demonstrated that uveitis may occur in 13–34% of patients with an increase in prevalence from baseline to 12-month follow-up. The clinical spectrum of disease ranges from mild, anterior uveitis to severe, sight-threatening panuveitis. Untreated inflammation may ultimately lead to secondary complications of cataract and posterior synechiae, with resultant vision impairment. The identification of Ebola virus persistence in immune privileged organs, such as the eye, with subsequent tissue inflammation and edema may lead to vision loss. Non-human primate models of EVD have demonstrated tissue localization to the eye including macrophage reservoirs within the vitreous matter. Moreover, in vitro models of Ebola virus have shown permissiveness in retinal pigment epithelial cells, potentially contributing to viral persistence. Broad perspectives from epidemiologic studies of the outbreak, animal modeling, and immunologic studies of EVD survivors have demonstrated the spectrum of the eye disease, tissue specificity of Ebola virus infection, and antigen-specific immunologic response. Further studies in these areas will elucidate the mechanisms of this highly prevalent disease with the potential for improved therapeutics for Ebola virus in immune-privileged sites.
Collapse
|
26
|
Iraqi M, Edri A, Greenshpan Y, Kundu K, Bolel P, Cahana A, Ottolenghi A, Gazit R, Lobel L, Braiman A, Porgador A. N-Glycans Mediate the Ebola Virus-GP1 Shielding of Ligands to Immune Receptors and Immune Evasion. Front Cell Infect Microbiol 2020; 10:48. [PMID: 32211339 PMCID: PMC7068452 DOI: 10.3389/fcimb.2020.00048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/24/2020] [Indexed: 12/14/2022] Open
Abstract
The Ebola Virus (EBOV) glycoprotein (GP) sterically shields cell-membrane ligands to immune receptors such as human leukocyte antigen class-1 (HLA-I) and MHC class I polypeptide-related sequence A (MICA), thus mediating immunity evasion. It was suggested that the abundant N-glycosylation of the EBOV-GP is involved in this steric shielding. We aimed to characterize (i) the GP N-glycosylation sites contributing to the shielding, and (ii) the effect of mutating these sites on immune subversion by the EBOV-GP. The two highly glycosylated domains of GP are the mucin-like domain (MLD) and the glycan cap domain (GCD) with three and six N-glycosylation sites, respectively. We mutated the N-glycosylation sites either in MLD or in GCD or in both domains. We showed that the glycosylation sites in both the MLD and GCD domains contribute to the steric shielding. This was shown for the steric shielding of either HLA-I or MICA. We then employed the fluorescence resonance energy transfer (FRET) method to measure the effect of N-glycosylation site removal on the distance in the cell membrane between the EBOV-GP and HLA-I (HLA.A*0201 allele). We recorded high FRET values for the interaction of CFP-fused HLA.A*0201 and YFP-fused EBOV-GP, demonstrating the very close distance (<10 nm) between these two proteins on the cell membrane of GP-expressing cells. The co-localization of HLA-I and Ebola GP was unaffected by the disruption of steric shielding, as the removal of N-glycosylation sites on Ebola GP revealed similar FRET values with HLA-I. However, these mutations directed to N-glycosylation sites had restored immune cell function otherwise impaired due to steric shielding over immune cell ligands by WT Ebola GP. Overall, we showed that the GP-mediated steric shielding aimed to impair immune function is facilitated by the N-glycans protruding from its MLD and GCD domains, but these N-glycans are not controlling the close distance between GP and its shielded proteins.
Collapse
Affiliation(s)
- Muhammed Iraqi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Avishay Edri
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Yariv Greenshpan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Kiran Kundu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Priyanka Bolel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Avishag Cahana
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Aner Ottolenghi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Roi Gazit
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Leslie Lobel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| |
Collapse
|
27
|
Hendricks K, Parrado MG, Bradley J. Opinion: An Existing Drug to Assess In Vivo for Potential Adjunctive Therapy of Ebola Virus Disease and Post-Ebola Syndrome. Front Pharmacol 2020; 10:1691. [PMID: 32082173 PMCID: PMC7002323 DOI: 10.3389/fphar.2019.01691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/24/2019] [Indexed: 01/02/2023] Open
Affiliation(s)
| | | | - John Bradley
- Division of Infectious Diseases, Department of Pediatrics, UCSD School of Medicine, San Diego, CA, United States
| |
Collapse
|
28
|
Rojek AM, Salam A, Ragotte RJ, Liddiard E, Elhussain A, Carlqvist A, Butler M, Kayem N, Castle L, Odondi L', Stepniewska K, Horby PW. A systematic review and meta-analysis of patient data from the West Africa (2013-16) Ebola virus disease epidemic. Clin Microbiol Infect 2019; 25:1307-1314. [PMID: 31284032 PMCID: PMC7116468 DOI: 10.1016/j.cmi.2019.06.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/25/2019] [Accepted: 06/28/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Over 28 000 individuals were infected with Ebola virus during the West Africa (2013-2016) epidemic, yet there has been criticism of the lack of robust clinical descriptions of Ebola virus disease (EVD) illness from that outbreak. OBJECTIVES To perform a meta-analysis of published data from the epidemic to describe the clinical presentation, evolution of disease, and predictors of mortality in individuals with EVD. To assess the quality and utility of published data for clinical and public health decision-making. DATA SOURCES Primary articles available in PubMed and published between January 2014 and May 2017. ELIGIBILITY Studies that sequentially enrolled individuals hospitalized for EVD and that reported acute clinical outcomes. METHODS We performed meta-analyses using random-effect models and assessed heterogeneity using the I2 method. We assessed data representativeness by comparing meta-analysis estimates with WHO aggregate data. We examined data utility by examining the availability and compatibility of data sets. RESULTS In all, 3653 articles were screened and 34 articles were included, representing 16 independent cohorts of patients (18 overlapping cohorts) and at least 6168 individuals. The pooled estimate for case fatality rate was 51% (95% CI 46%-56%). However, pooling of estimates for clinical presentation, progression, and predictors of mortality in individuals with EVD were hampered by significant heterogeneity, and inadequate data on clinical progression. Our assessment of data quality found that heterogeneity was largely unexplained, and data availability and compatibility were poor. CONCLUSIONS We have quantified a missed opportunity to generate reliable estimates of the clinical manifestations of EVD during the West Africa epidemic. Clinical data standards and data capture platforms are urgently needed.
Collapse
Affiliation(s)
- A M Rojek
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK.
| | - A Salam
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK; United Kingdom Public Health Rapid Support Team, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - R J Ragotte
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - E Liddiard
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - A Elhussain
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - A Carlqvist
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - M Butler
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - N Kayem
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - L Castle
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - L 'o Odondi
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - K Stepniewska
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK; WorldWide Antimalarial Resistance Network, Oxford, UK
| | - P W Horby
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| |
Collapse
|
29
|
Younan P, Santos RI, Ramanathan P, Iampietro M, Nishida A, Dutta M, Ammosova T, Meyer M, Katze MG, Popov VL, Nekhai S, Bukreyev A. Ebola virus-mediated T-lymphocyte depletion is the result of an abortive infection. PLoS Pathog 2019; 15:e1008068. [PMID: 31648236 PMCID: PMC6812753 DOI: 10.1371/journal.ppat.1008068] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 09/03/2019] [Indexed: 12/21/2022] Open
Abstract
Ebola virus (EBOV) infections are characterized by a pronounced lymphopenia that is highly correlative with fatalities. However, the mechanisms leading to T-cell depletion remain largely unknown. Here, we demonstrate that both viral mRNAs and antigens are detectable in CD4+ T cells despite the absence of productive infection. A protein phosphatase 1 inhibitor, 1E7-03, and siRNA-mediated suppression of viral antigens were used to demonstrate de novo synthesis of viral RNAs and antigens in CD4+ T cells, respectively. Cell-to-cell fusion of permissive Huh7 cells with non-permissive Jurkat T cells impaired productive EBOV infection suggesting the presence of a cellular restriction factor. We determined that viral transcription is partially impaired in the fusion T cells. Lastly, we demonstrate that exposure of T cells to EBOV resulted in autophagy through activation of ER-stress related pathways. These data indicate that exposure of T cells to EBOV results in an abortive infection, which likely contributes to the lymphopenia observed during EBOV infections.
Collapse
Affiliation(s)
- Patrick Younan
- Department of Pathology, the University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Rodrigo I. Santos
- Department of Pathology, the University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Palaniappan Ramanathan
- Department of Pathology, the University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Mathieu Iampietro
- Department of Pathology, the University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Mukta Dutta
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Tatiana Ammosova
- Department of Medicine, Howard University, Washington, D.C., United States of America
| | - Michelle Meyer
- Department of Pathology, the University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Michael G. Katze
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- National Primate Research Center, Seattle, Washington, United States of America
| | - Vsevolod L. Popov
- Department of Pathology, the University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, the University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Sergei Nekhai
- Department of Medicine, Howard University, Washington, D.C., United States of America
| | - Alexander Bukreyev
- Department of Pathology, the University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, the University of Texas Medical Branch, Galveston, Texas, United States of America
- Department Microbiology & Immunology, the University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
30
|
Sobarzo A, Stonier SW, Radinsky O, Gelkop S, Kuehne AI, Edri A, Herbert AS, Fedida-Metula S, Lutwama JJ, Yavelsky V, Davis C, Porgador A, Dye JM, Lobel L. Multiple viral proteins and immune response pathways act to generate robust long-term immunity in Sudan virus survivors. EBioMedicine 2019; 46:215-226. [PMID: 31326432 PMCID: PMC6710910 DOI: 10.1016/j.ebiom.2019.07.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 12/05/2022] Open
Abstract
Background Profiles of immunity developed in filovirus patients and survivors have begun to shed light on antigen-specific cellular immune responses that had been previously under-studied. However, our knowledge of the breadth and length of those responses and the viral targets which mediate long-term memory immunity still lags significantly behind. Methods We characterized antigen-specific immune responses in whole blood samples of fifteen years post-infected survivors of the Sudan virus (SUDV) outbreak in Gulu, Uganda (2000−2001). We examined T cell and IgG responses against SUDV complete antigen and four SUDV proteins; glycoprotein (GP), nucleoprotein (NP), and viral protein 30 (VP30), and 40 (VP40). Findings We found survivors-maintained antigen-specific CD4+ T cell memory immune responses mediated mainly by the viral protein NP. In contrast, activated CD8+ T cell responses were nearly absent in SUDV survivors, regardless of the stimulating antigen used. Analysis of anti-viral humoral immunity revealed antigen-specific IgG antibodies against SUDV and SUDV proteins. Survivor IgGs mediated live SUDV neutralization in vitro and FcγRI and FcγRIII antibody Fc-dependent responses, mainly via antibodies to the viral proteins GP and VP40. Interpretation We highlight the key role of several proteins, i.e., GP, NP, and VP40, to act as mediators of distinctive and sustained cellular memory immune responses in long-term SUDV survivors. We suggest that the inclusion of these viral proteins in vaccine development may best mimic survivor native memory immune responses with the potential of protecting against viral infection. Funds This research was funded by the Defense Threat Reduction Agency (CB4088) and by the National Institute Of Allergy And Infectious Diseases of the National Institutes of Health under Award Number R01AI111516. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Collapse
Affiliation(s)
- Ariel Sobarzo
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| | - Spencer W Stonier
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St, Fort Detrick, MD 21702-5011, USA
| | - Olga Radinsky
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Sigal Gelkop
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Ana I Kuehne
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St, Fort Detrick, MD 21702-5011, USA
| | - Avishay Edri
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Andrew S Herbert
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St, Fort Detrick, MD 21702-5011, USA
| | - Shlomit Fedida-Metula
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Julius Julian Lutwama
- Department of Arbovirology, Emerging and Re-Emerging Infection Uganda Virus Research Institute, Plot No: 51 -59, Nakiwogo Road, P.O.Box 49, Entebbe, Uganda
| | - Victoria Yavelsky
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Department of Arbovirology, Emerging and Re-Emerging Infection Uganda Virus Research Institute, Plot No: 51 -59, Nakiwogo Road, P.O.Box 49, Entebbe, Uganda
| | - Claytus Davis
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Angel Porgador
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - John M Dye
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter St, Fort Detrick, MD 21702-5011, USA.
| | - Leslie Lobel
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Department of Arbovirology, Emerging and Re-Emerging Infection Uganda Virus Research Institute, Plot No: 51 -59, Nakiwogo Road, P.O.Box 49, Entebbe, Uganda
| |
Collapse
|
31
|
Brunton B, Rogers K, Phillips EK, Brouillette RB, Bouls R, Butler NS, Maury W. TIM-1 serves as a receptor for Ebola virus in vivo, enhancing viremia and pathogenesis. PLoS Negl Trop Dis 2019; 13:e0006983. [PMID: 31242184 PMCID: PMC6615641 DOI: 10.1371/journal.pntd.0006983] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 07/09/2019] [Accepted: 05/17/2019] [Indexed: 01/11/2023] Open
Abstract
Background T cell immunoglobulin mucin domain-1 (TIM-1) is a phosphatidylserine (PS) receptor, mediating filovirus entry into cells through interactions with PS on virions. TIM-1 expression has been implicated in Ebola virus (EBOV) pathogenesis; however, it remains unclear whether this is due to TIM-1 serving as a filovirus receptor in vivo or, as others have suggested, TIM-1 induces a cytokine storm elicited by T cell/virion interactions. Here, we use a BSL2 model virus that expresses EBOV glycoprotein to demonstrate the importance of TIM-1 as a virus receptor late during in vivo infection. Methodology/Principal findings Infectious, GFP-expressing recombinant vesicular stomatitis virus encoding either full length EBOV glycoprotein (EBOV GP/rVSV) or mucin domain deleted EBOV glycoprotein (EBOV GPΔO/rVSV) was used to assess the role of TIM-1 during in vivo infection. GFP-expressing rVSV encoding its native glycoprotein G (G/rVSV) served as a control. TIM-1-sufficient or TIM-1-deficient BALB/c interferon α/β receptor-/- mice were challenged with these viruses. While G/rVSV caused profound morbidity and mortality in both mouse strains, TIM-1-deficient mice had significantly better survival than TIM-1-expressing mice following EBOV GP/rVSV or EBOV GPΔO/rVSV challenge. EBOV GP/rVSV or EBOV GPΔO/rVSV in spleen of infected animals was high and unaffected by expression of TIM-1. However, infectious virus in serum, liver, kidney and adrenal gland was reduced late in infection in the TIM-1-deficient mice, suggesting that virus entry via this receptor contributes to virus load. Consistent with higher virus loads, proinflammatory chemokines trended higher in organs from infected TIM-1-sufficient mice compared to the TIM-1-deficient mice, but proinflammatory cytokines were more modestly affected. To assess the role of T cells in EBOV GP/rVSV pathogenesis, T cells were depleted in TIM-1-sufficient and -deficient mice and the mice were challenged with virus. Depletion of T cells did not alter the pathogenic consequences of virus infection. Conclusions Our studies provide evidence that at late times during EBOV GP/rVSV infection, TIM-1 increased virus load and associated mortality, consistent with an important role of this receptor in virus entry. This work suggests that inhibitors which block TIM-1/virus interaction may serve as effective antivirals, reducing virus load at late times during EBOV infection. T cell immunoglobulin mucin domain-1 (TIM-1) is one of a number of phosphatidylserine (PS) receptors that mediate clearance of apoptotic bodies by binding PS on the surface of dead or dying cells. Enveloped viruses mimic apoptotic bodies by exposing PS on the outer leaflet of the viral membrane. While TIM-1 has been shown to serve as an adherence factor/receptor for filoviruses in tissue culture, limited studies have investigated the role of TIM-1 as a receptor in vivo. Here, we sought to determine if TIM-1 was critical for Ebola virus glycoprotein-mediated infection using a BSL2 model virus. We demonstrate that loss of TIM-1 expression results in decreased virus load late during infection and significantly reduced virus-elicited mortality. These findings provide evidence that TIM-1 serves as an important receptor for Ebola virus in vivo. Blocking TIM-1/EBOV interactions may be effective antiviral strategy to reduce viral load and pathogenicity at late times of EBOV infection.
Collapse
Affiliation(s)
- Bethany Brunton
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Kai Rogers
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Elisabeth K. Phillips
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Rachel B. Brouillette
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Ruayda Bouls
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Noah S. Butler
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
32
|
Ploquin A, Zhou Y, Sullivan NJ. Ebola Immunity: Gaining a Winning Position in Lightning Chess. THE JOURNAL OF IMMUNOLOGY 2019; 201:833-842. [PMID: 30038036 DOI: 10.4049/jimmunol.1700827] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 05/05/2018] [Indexed: 12/13/2022]
Abstract
Zaire ebolavirus (EBOV), one of five species in the genus Ebolavirus, is the causative agent of the hemorrhagic fever disease epidemic that claimed more than 11,000 lives from 2014 to 2016 in West Africa. The combination of EBOV's ability to disseminate broadly and rapidly within the host and its high pathogenicity pose unique challenges to the human immune system postinfection. Potential transmission from apparently healthy EBOV survivors reported in the recent epidemic raises questions about EBOV persistence and immune surveillance mechanisms. Clinical, virological, and immunological data collected since the West Africa epidemic have greatly enhanced our knowledge of host-virus interactions. However, critical knowledge gaps remain in our understanding of what is necessary for an effective host immune response for protection against, or for clearance of, EBOV infection. This review provides an overview of immune responses against EBOV and discusses those associated with the success or failure to control EBOV infection.
Collapse
Affiliation(s)
- Aurélie Ploquin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Yan Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
33
|
Olejnik J, Hume AJ, Leung DW, Amarasinghe GK, Basler CF, Mühlberger E. Filovirus Strategies to Escape Antiviral Responses. Curr Top Microbiol Immunol 2019; 411:293-322. [PMID: 28685291 PMCID: PMC5973841 DOI: 10.1007/82_2017_13] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This chapter describes the various strategies filoviruses use to escape host immune responses with a focus on innate immune and cell death pathways. Since filovirus replication can be efficiently blocked by interferon (IFN), filoviruses have evolved mechanisms to counteract both type I IFN induction and IFN response signaling pathways. Intriguingly, marburg- and ebolaviruses use different strategies to inhibit IFN signaling. This chapter also summarizes what is known about the role of IFN-stimulated genes (ISGs) in filovirus infection. These fall into three categories: those that restrict filovirus replication, those whose activation is inhibited by filoviruses, and those that have no measurable effect on viral replication. In addition to innate immunity, mammalian cells have evolved strategies to counter viral infections, including the induction of cell death and stress response pathways, and we summarize our current knowledge of how filoviruses interact with these pathways. Finally, this chapter delves into the interaction of EBOV with myeloid dendritic cells and macrophages and the associated inflammatory response, which differs dramatically between these cell types when they are infected with EBOV. In summary, we highlight the multifaceted nature of the host-viral interactions during filoviral infections.
Collapse
Affiliation(s)
- Judith Olejnik
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA
| | - Adam J Hume
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Christopher F Basler
- Microbial Pathogenesis, Georgia State University, Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Elke Mühlberger
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA.
| |
Collapse
|
34
|
Muñoz-Fontela C, McElroy AK. Ebola Virus Disease in Humans: Pathophysiology and Immunity. Curr Top Microbiol Immunol 2019; 411:141-169. [PMID: 28653186 PMCID: PMC7122202 DOI: 10.1007/82_2017_11] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Viruses of the Ebolavirus genus cause sporadic epidemics of severe and systemic febrile disease that are fueled by human-to-human transmission. Despite the notoriety of ebolaviruses, particularly Ebola virus (EBOV), as prominent viral hemorrhagic fever agents, and the international concern regarding Ebola virus disease (EVD) outbreaks, very little is known about the pathophysiology of EVD in humans and, in particular, about the human immune correlates of survival and immune memory. This lack of basic knowledge about physiological characteristics of EVD is probably attributable to the dearth of clinical and laboratory data gathered from past outbreaks. The unprecedented magnitude of the EVD epidemic that occurred in West Africa from 2013 to 2016 has allowed, for the first time, evaluation of clinical, epidemiological, and immunological parameters in a significant number of patients using state-of-the-art laboratory equipment. This review will summarize the data from the literature regarding human pathophysiologic and immunologic responses to filoviral infection.
Collapse
Affiliation(s)
- César Muñoz-Fontela
- Laboratory of Emerging Viruses, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistraße 52, 20251, Hamburg, Germany.
| | - Anita K McElroy
- Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Drive NE, Atlanta, GA, 30322, USA.
| |
Collapse
|
35
|
Jain S, Baranwal M. Conserved peptide vaccine candidates containing multiple Ebola nucleoprotein epitopes display interactions with diverse HLA molecules. Med Microbiol Immunol 2019; 208:227-238. [PMID: 30790057 DOI: 10.1007/s00430-019-00584-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/11/2019] [Indexed: 10/27/2022]
Abstract
Immunoinformatics has come by leaps and bounds to finding potent vaccine candidates against various pathogens. In the current study, a combination of different T (CD4+ and CD8+) and B cell epitope prediction tools was applied to find peptides containing multiple epitopes against Ebola nucleoprotein (NP) and the presentation of peptides to human leukocyte antigen (HLA) molecules was analyzed by prediction, docking and population coverage tools. Further, potential peptides were analyzed by ELISA for peptide induced IFN-γ secretion in peripheral blood mononuclear cells isolated from healthy volunteers. Six peptides were obtained after merging the overlapping multiple HLA I (CD8+) and II (CD4+) restricted T cell epitopes as well as B cell epitopes and eliminating the peptides liable to generate autoimmune and allergic response. All peptides displayed 100% conservancy in Zaire ebolavirus. In other Ebola virus species (Sudan, Bundibugyo and Taï forest) and Filoviridae members (Lloviuvirus and Margburgvirus), some peptides were found to be conserved with minor variations. Prediction tools confirmed the ability of predicted peptides to bind with diverse HLA (HLA-A, HLA-B, HLA-DP, HLA-DQ and HLA-DR) alleles. CABS-dock results displayed that the average root mean square deviation (RMSD) value was less than three in majority of cases representing strong binding affinity with HLA alleles. Population coverage analysis predicted high coverage (> 85%) for expected immune response in four continents (Africa, America, Asia and Europe). Nine out of ten blood samples exhibited enhanced IFN-γ secretion for two peptides (P2 and P3). Thus, the identified NP peptides can be considered as potential synthetic vaccine candidates against Ebola virus.
Collapse
Affiliation(s)
- Sahil Jain
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
36
|
Reynard S, Journeaux A, Gloaguen E, Schaeffer J, Varet H, Pietrosemoli N, Mateo M, Baillet N, Laouenan C, Raoul H, Mullaert J, Baize S. Immune parameters and outcomes during Ebola virus disease. JCI Insight 2019; 4:125106. [PMID: 30626757 DOI: 10.1172/jci.insight.125106] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/29/2018] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The West African Ebola virus epidemic from 2014-2016 highlighted the lack of knowledge about the pathogenicity of the virus and the factors responsible for outcome. A performant and rapid diagnosis is of crucial importance, as is overcoming the difficulty of providing high-quality patient management during such an extensive outbreak. Here, we propose to study the role of the immune mediators during Ebola virus disease and to define some molecules of importance in the outcome. METHODS Plasma from Guinean patients sampled during the outbreak were analyzed using RT-qPCR, magnetic bead assay, ELISA, and high-quality statistical analyses. We also performed a transcriptomic analysis in leukocytes samples. Therefore, we deeply characterized the immune responses involved in Ebola virus disease. RESULTS We evaluated the immune patterns depending on the outcome of the disease. Survivors presented an efficient and well-balanced immune response, whereas fatalities were characterized by an intense inflammatory response, overexpression of multiple cytokines, and a "chemokine storm." The plasma concentration of most of the parameters tested increased until death. Statistical analyses also allowed us to define a panel of markers highly predictive of outcome. CONCLUSION The immune response observed in fatalities was highly similar to that characterizing septic shock syndrome. Our results suggest that immune responses can play a major pathogenic role during severe Ebola virus infection and argue in favor of therapeutic approaches that act on both viral replication and the induction of shock syndrome. FUNDING French Ministry of Foreign Affairs, the Agence Française de Développement, and the Institut Pasteur.
Collapse
Affiliation(s)
- Stéphanie Reynard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| | - Alexandra Journeaux
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| | - Emilie Gloaguen
- Infection Antimicrobials Modelling Evolution, UMR 1137, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Justine Schaeffer
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| | - Hugo Varet
- Institut Pasteur, Hub Bioinformatique et Biostatistique, Centre de Bioinformatique, Biostatistique et Biologie Intégrative, C3BI, USR 3756 IP CNRS, Paris, France
| | - Natalia Pietrosemoli
- Institut Pasteur, Hub Bioinformatique et Biostatistique, Centre de Bioinformatique, Biostatistique et Biologie Intégrative, C3BI, USR 3756 IP CNRS, Paris, France
| | - Mathieu Mateo
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| | - Nicolas Baillet
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| | - Cédric Laouenan
- Infection Antimicrobials Modelling Evolution, UMR 1137, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Bichat Claude Bernard, Paris, France
| | - Hervé Raoul
- Laboratoire P4 Jean Mérieux-INSERM, INSERM, Lyon, France
| | - Jimmy Mullaert
- Infection Antimicrobials Modelling Evolution, UMR 1137, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Sylvain Baize
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| |
Collapse
|
37
|
Vaughan K, Xu X, Peters B, Sette A. Investigation of Outbreak-Specific Nonsynonymous Mutations on Ebolavirus GP in the Context of Known Immune Reactivity. J Immunol Res 2018; 2018:1846207. [PMID: 30581874 PMCID: PMC6276448 DOI: 10.1155/2018/1846207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/01/2018] [Accepted: 10/22/2018] [Indexed: 11/17/2022] Open
Abstract
The global response to the most recent EBOV outbreak has led to increased generation and availability of data, which can be globally analyzed to increase our understanding of immune responses to EBOV. We analyzed the published antibody epitope data to identify regions immunogenic for humans on the main GP antigenic target and determine sequence variance/nonsynonymous mutations between historical isolates and variants from the 2013-2016 outbreak. Approximately half of the GP sequence has been reported as targeted by antibody responses. Our results show an enrichment of nonsynonymous mutations (NSMs) within epitopic regions on GP (70%, p = 0.0133). Mapping NSMs to human epitope reactivity may be useful for future therapeutic and prophylaxis development as well as for our general understanding of immunity against EBOV.
Collapse
Affiliation(s)
- Kerrie Vaughan
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Xiaojun Xu
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- University of California San Diego, Department of Medicine, La Jolla, CA 92093, USA
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- University of California San Diego, Department of Medicine, La Jolla, CA 92093, USA
| |
Collapse
|
38
|
Cross RW, Fenton KA, Geisbert TW. Small animal models of filovirus disease: recent advances and future directions. Expert Opin Drug Discov 2018; 13:1027-1040. [DOI: 10.1080/17460441.2018.1527827] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Robert W. Cross
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Karla A. Fenton
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Thomas W. Geisbert
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
39
|
McElroy AK, Mühlberger E, Muñoz-Fontela C. Immune barriers of Ebola virus infection. Curr Opin Virol 2018; 28:152-160. [PMID: 29452995 PMCID: PMC5886007 DOI: 10.1016/j.coviro.2018.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/23/2018] [Accepted: 01/26/2018] [Indexed: 01/10/2023]
Abstract
Since its initial emergence in 1976 in northern Democratic Republic of Congo (DRC), Ebola virus (EBOV) has been a global health concern due to its virulence in humans, the mystery surrounding the identity of its host reservoir and the unpredictable nature of Ebola virus disease (EVD) outbreaks. Early after the first clinical descriptions of a disease resembling a 'septic-shock-like syndrome', with coagulation abnormalities and multi-system organ failure, researchers began to evaluate the role of the host immune response in EVD pathophysiology. In this review, we summarize how data gathered during the last 40 years in the laboratory as well as in the field have provided insight into EBOV immunity. From molecular mechanisms involved in EBOV recognition in infected cells, to antigen processing and adaptive immune responses, we discuss current knowledge on the main immune barriers of infection as well as outstanding research questions.
Collapse
Affiliation(s)
- Anita K McElroy
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, 3501 Fifth Ave, Pittsburgh, PA 15261, USA
| | - Elke Mühlberger
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, 02118 Boston, MA, USA
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Strasse 74, 20359 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg, Germany.
| |
Collapse
|
40
|
|
41
|
Menicucci AR, Versteeg K, Woolsey C, Mire CE, Geisbert JB, Cross RW, Agans KN, Jankeel A, Geisbert TW, Messaoudi I. Transcriptome Analysis of Circulating Immune Cell Subsets Highlight the Role of Monocytes in Zaire Ebola Virus Makona Pathogenesis. Front Immunol 2017; 8:1372. [PMID: 29123522 PMCID: PMC5662559 DOI: 10.3389/fimmu.2017.01372] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 10/05/2017] [Indexed: 12/27/2022] Open
Abstract
Existing models of Ebola virus disease (EVD) suggest antigen-presenting cells are initial targets of Zaire ebolavirus (ZEBOV). In vitro studies have shown that ZEBOV infection of monocytes and macrophages results in the production of inflammatory mediators, which may cause lymphocyte apoptosis. However, these findings have not been corroborated by in vivo studies. In this study, we report the first longitudinal analysis of transcriptional changes in purified monocytes, T-cells, and B-cells isolated from cynomolgus macaques following infection with ZEBOV-Makona. Our data reveal monocytes as one of the major immune cell subsets that supports ZEBOV replication in vivo. In addition, we report a marked increase in the transcription of genes involved in inflammation, coagulation, and vascular disease within monocytes, suggesting that monocytes contribute to EVD manifestations. Further, genes important for antigen presentation and regulation of immunity were downregulated, potentially subverting development of adaptive immunity. In contrast, lymphocytes, which do not support ZEBOV replication, showed transcriptional changes limited to a small number of interferon-stimulated genes (ISGs) and a failure to upregulate genes associated with an antiviral effector immune response. Collectively, these data suggest that ZEBOV-infected monocytes play a significant role in ZEBOV-Makona pathogenesis and strategies to suppress virus replication or modify innate responses to infection in these cells should be a priority for therapeutic intervention.
Collapse
Affiliation(s)
- Andrea R Menicucci
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA, United States
| | - Krista Versteeg
- Galveston National Laboratory, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Courtney Woolsey
- Galveston National Laboratory, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Chad E Mire
- Galveston National Laboratory, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Joan B Geisbert
- Galveston National Laboratory, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Robert W Cross
- Galveston National Laboratory, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Krystle N Agans
- Galveston National Laboratory, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Allen Jankeel
- Department of Molecular Biology and Biochemistry, College of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| | - Thomas W Geisbert
- Galveston National Laboratory, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, College of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
42
|
Seckler JM, Meyer NM, Burton ST, Bates JN, Gaston B, Lewis SJ. Detection of trace concentrations of S-nitrosothiols by means of a capacitive sensor. PLoS One 2017; 12:e0187149. [PMID: 29073241 PMCID: PMC5658150 DOI: 10.1371/journal.pone.0187149] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/14/2017] [Indexed: 01/25/2023] Open
Abstract
Small molecule S-nitrosothiols are a class of endogenous chemicals in the body, which have been implicated in a variety of biological functions. However, the labile nature of NO and the limits of current detection assays have made studying these molecules difficult. Here we present a method for detecting trace concentrations of S-nitrosothiols in biological fluids. Capacitive sensors when coupled to a semiconducting material represent a method for detecting trace quantities of a chemical in complex solutions. We have taken advantage of the semiconducting and chemical properties of polydopamine to construct a capacitive sensor and associated method of use, which specifically senses S-nitrosothiols in complex biological solutions.
Collapse
Affiliation(s)
- James M. Seckler
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Nikki M. Meyer
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Spencer T. Burton
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - James N. Bates
- Department of Anesthesia, University of Iowa, Iowa City, Iowa, United States of America
| | - Benjamin Gaston
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
- Rainbow Babies and Children’s Hospital, Cleveland, Ohio, United States of America
| | - Stephen J. Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
43
|
Younan P, Iampietro M, Nishida A, Ramanathan P, Santos RI, Dutta M, Lubaki NM, Koup RA, Katze MG, Bukreyev A. Ebola Virus Binding to Tim-1 on T Lymphocytes Induces a Cytokine Storm. mBio 2017; 8:e00845-17. [PMID: 28951472 PMCID: PMC5615193 DOI: 10.1128/mbio.00845-17] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022] Open
Abstract
Ebola virus (EBOV) disease (EVD) results from an exacerbated immunological response that is highlighted by a burst in the production of inflammatory mediators known as a "cytokine storm." Previous reports have suggested that nonspecific activation of T lymphocytes may play a central role in this phenomenon. T-cell immunoglobulin and mucin domain-containing protein 1 (Tim-1) has recently been shown to interact with virion-associated phosphatidylserine to promote infection. Here, we demonstrate the central role of Tim-1 in EBOV pathogenesis, as Tim-1-/- mice exhibited increased survival rates and reduced disease severity; surprisingly, only a limited decrease in viremia was detected. Tim-1-/- mice exhibited a modified inflammatory response as evidenced by changes in serum cytokines and activation of T helper subsets. A series of in vitro assays based on the Tim-1 expression profile on T cells demonstrated that despite the apparent absence of detectable viral replication in T lymphocytes, EBOV directly binds to isolated T lymphocytes in a phosphatidylserine-Tim-1-dependent manner. Exposure to EBOV resulted in the rapid development of a CD4Hi CD3Low population, non-antigen-specific activation, and cytokine production. Transcriptome and Western blot analysis of EBOV-stimulated CD4+ T cells confirmed the induction of the Tim-1 signaling pathway. Furthermore, comparative analysis of transcriptome data and cytokine/chemokine analysis of supernatants highlight the similarities associated with EBOV-stimulated T cells and the onset of a cytokine storm. Flow cytometry revealed virtually exclusive binding and activation of central memory CD4+ T cells. These findings provide evidence for the role of Tim-1 in the induction of a cytokine storm phenomenon and the pathogenesis of EVD.IMPORTANCE Ebola virus infection is characterized by a massive release of inflammatory mediators, which has come to be known as a cytokine storm. The severity of the cytokine storm is consistently linked with fatal disease outcome. Previous findings have demonstrated that specific T-cell subsets are key contributors to the onset of a cytokine storm. In this study, we investigated the role of Tim-1, a T-cell-receptor-independent trigger of T-cell activation. We first demonstrated that Tim-1-knockout (KO) mice survive lethal Ebola virus challenge. We then used a series of in vitro assays to demonstrate that Ebola virus directly binds primary T cells in a Tim-1-phosphatidylserine-dependent manner. We noted that binding induces a cytokine storm-like phenomenon and that blocking Tim-1-phosphatidylserine interactions reduces viral binding, T-cell activation, and cytokine production. These findings highlight a previously unknown role of Tim-1 in the development of a cytokine storm and "immune paralysis."
Collapse
Affiliation(s)
- Patrick Younan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mathieu Iampietro
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Palaniappan Ramanathan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Rodrigo I Santos
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mukta Dutta
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Ndongala Michel Lubaki
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael G Katze
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Washington National Primate Research Center, Seattle, Washington, USA
| | - Alexander Bukreyev
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
44
|
Younan P, Iampietro M, Nishida A, Ramanathan P, Santos RI, Dutta M, Lubaki NM, Koup RA, Katze MG, Bukreyev A. Ebola Virus Binding to Tim-1 on T Lymphocytes Induces a Cytokine Storm. mBio 2017; 8:e00845-17. [PMID: 28951472 DOI: 10.1128/mbio.00845-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/17/2017] [Indexed: 12/12/2022] Open
Abstract
Ebola virus (EBOV) disease (EVD) results from an exacerbated immunological response that is highlighted by a burst in the production of inflammatory mediators known as a "cytokine storm." Previous reports have suggested that nonspecific activation of T lymphocytes may play a central role in this phenomenon. T-cell immunoglobulin and mucin domain-containing protein 1 (Tim-1) has recently been shown to interact with virion-associated phosphatidylserine to promote infection. Here, we demonstrate the central role of Tim-1 in EBOV pathogenesis, as Tim-1-/- mice exhibited increased survival rates and reduced disease severity; surprisingly, only a limited decrease in viremia was detected. Tim-1-/- mice exhibited a modified inflammatory response as evidenced by changes in serum cytokines and activation of T helper subsets. A series of in vitro assays based on the Tim-1 expression profile on T cells demonstrated that despite the apparent absence of detectable viral replication in T lymphocytes, EBOV directly binds to isolated T lymphocytes in a phosphatidylserine-Tim-1-dependent manner. Exposure to EBOV resulted in the rapid development of a CD4Hi CD3Low population, non-antigen-specific activation, and cytokine production. Transcriptome and Western blot analysis of EBOV-stimulated CD4+ T cells confirmed the induction of the Tim-1 signaling pathway. Furthermore, comparative analysis of transcriptome data and cytokine/chemokine analysis of supernatants highlight the similarities associated with EBOV-stimulated T cells and the onset of a cytokine storm. Flow cytometry revealed virtually exclusive binding and activation of central memory CD4+ T cells. These findings provide evidence for the role of Tim-1 in the induction of a cytokine storm phenomenon and the pathogenesis of EVD.IMPORTANCE Ebola virus infection is characterized by a massive release of inflammatory mediators, which has come to be known as a cytokine storm. The severity of the cytokine storm is consistently linked with fatal disease outcome. Previous findings have demonstrated that specific T-cell subsets are key contributors to the onset of a cytokine storm. In this study, we investigated the role of Tim-1, a T-cell-receptor-independent trigger of T-cell activation. We first demonstrated that Tim-1-knockout (KO) mice survive lethal Ebola virus challenge. We then used a series of in vitro assays to demonstrate that Ebola virus directly binds primary T cells in a Tim-1-phosphatidylserine-dependent manner. We noted that binding induces a cytokine storm-like phenomenon and that blocking Tim-1-phosphatidylserine interactions reduces viral binding, T-cell activation, and cytokine production. These findings highlight a previously unknown role of Tim-1 in the development of a cytokine storm and "immune paralysis."
Collapse
Affiliation(s)
- Patrick Younan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mathieu Iampietro
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Palaniappan Ramanathan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Rodrigo I Santos
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mukta Dutta
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Ndongala Michel Lubaki
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael G Katze
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Washington National Primate Research Center, Seattle, Washington, USA
| | - Alexander Bukreyev
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
45
|
Speranza E, Connor JH. Host Transcriptional Response to Ebola Virus Infection. Vaccines (Basel) 2017; 5:E30. [PMID: 28930167 PMCID: PMC5620561 DOI: 10.3390/vaccines5030030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/12/2017] [Accepted: 09/12/2017] [Indexed: 01/09/2023] Open
Abstract
Ebola virus disease (EVD) is a serious illness that causes severe disease in humans and non-human primates (NHPs) and has mortality rates up to 90%. EVD is caused by the Ebolavirus and currently there are no licensed therapeutics or vaccines to treat EVD. Due to its high mortality rates and potential as a bioterrorist weapon, a better understanding of the disease is of high priority. Multiparametric analysis techniques allow for a more complete understanding of a disease and the host response. Analysis of RNA species present in a sample can lead to a greater understanding of activation or suppression of different states of the immune response. Transcriptomic analyses such as microarrays and RNA-Sequencing (RNA-Seq) have been important tools to better understand the global gene expression response to EVD. In this review, we outline the current knowledge gained by transcriptomic analysis of EVD.
Collapse
Affiliation(s)
- Emily Speranza
- Department of Microbiology, Bioinformatics Program, National Emerging Infectious Disease Laboratories, Boston University, Boston, MA 02118, USA.
| | - John H Connor
- Department of Microbiology, Bioinformatics Program, National Emerging Infectious Disease Laboratories, Boston University, Boston, MA 02118, USA.
| |
Collapse
|
46
|
Interferon-γ-Driven iNOS: A Molecular Pathway to Terminal Shock in Arenavirus Hemorrhagic Fever. Cell Host Microbe 2017; 22:354-365.e5. [DOI: 10.1016/j.chom.2017.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/11/2017] [Accepted: 07/11/2017] [Indexed: 01/21/2023]
|
47
|
Stonier SW, Herbert AS, Kuehne AI, Sobarzo A, Habibulin P, Dahan CVA, James RM, Egesa M, Cose S, Lutwama JJ, Lobel L, Dye JM. Marburg virus survivor immune responses are Th1 skewed with limited neutralizing antibody responses. J Exp Med 2017; 214:2563-2572. [PMID: 28724616 PMCID: PMC5584125 DOI: 10.1084/jem.20170161] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/16/2017] [Accepted: 06/28/2017] [Indexed: 11/21/2022] Open
Abstract
Immune responses that develop in survivors of filovirus infection may indicate critical parameters that could inform rational vaccine development. Stonier et al. characterize immune responses in Marburg virus survivors and demonstrate robust CD4+ T cell responses but limited CD8+ T cell and neutralizing antibody responses. Until recently, immune responses in filovirus survivors remained poorly understood. Early studies revealed IgM and IgG responses to infection with various filoviruses, but recent outbreaks have greatly expanded our understanding of filovirus immune responses. Immune responses in survivors of Ebola virus (EBOV) and Sudan virus (SUDV) infections have provided the most insight, with T cell responses as well as detailed antibody responses having been characterized. Immune responses to Marburg virus (MARV), however, remain almost entirely uncharacterized. We report that immune responses in MARV survivors share characteristics with EBOV and SUDV infections but have some distinct differences. MARV survivors developed multivariate CD4+ T cell responses but limited CD8+ T cell responses, more in keeping with SUDV survivors than EBOV survivors. In stark contrast to SUDV survivors, rare neutralizing antibody responses in MARV survivors diminished rapidly after the outbreak. These results warrant serious consideration for any vaccine or therapeutic that seeks to be broadly protective, as different filoviruses may require different immune responses to achieve immunity.
Collapse
Affiliation(s)
- Spencer W Stonier
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD
| | - Andrew S Herbert
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD
| | - Ana I Kuehne
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD
| | - Ariel Sobarzo
- Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Polina Habibulin
- Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Chen V Abramovitch Dahan
- Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Rebekah M James
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD
| | - Moses Egesa
- Uganda Research Unit on AIDS, Medical Research Council/Uganda Virus Research Institute, Entebbe, Uganda.,Department of Medical Microbiology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Stephen Cose
- Uganda Research Unit on AIDS, Medical Research Council/Uganda Virus Research Institute, Entebbe, Uganda.,Department of Medical Microbiology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda.,London School of Hygiene & Tropical Medicine, London, England, UK
| | - Julius Julian Lutwama
- Department of Arbovirology, Emerging, and Re-emerging Infection, Uganda Virus Research Institute, Entebbe, Uganda
| | - Leslie Lobel
- Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Arbovirology, Emerging, and Re-emerging Infection, Uganda Virus Research Institute, Entebbe, Uganda
| | - John M Dye
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD
| |
Collapse
|
48
|
|
49
|
Smith JR, Todd S, Ashander LM, Charitou T, Ma Y, Yeh S, Crozier I, Michael MZ, Appukuttan B, Williams KA, Lynn DJ, Marsh GA. Retinal Pigment Epithelial Cells are a Potential Reservoir for Ebola Virus in the Human Eye. Transl Vis Sci Technol 2017; 6:12. [PMID: 28721309 PMCID: PMC5512973 DOI: 10.1167/tvst.6.4.12] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/19/2017] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Success of Ebola virus (EBOV) as a human pathogen relates at the molecular level primarily to blockade the host cell type I interferon (IFN) antiviral response. Most individuals who survive Ebola virus disease (EVD) develop a chronic disease syndrome: approximately one-quarter of survivors suffer from uveitis, which has been associated with presence of EBOV within the eye. Clinical observations of post-Ebola uveitis indicate involvement of retinal pigment epithelial cells. METHODS We inoculated ARPE-19 human retinal pigment epithelial cells with EBOV, and followed course of infection by immunocytochemistry and measurement of titer in culture supernatant. To interrogate transcriptional responses of infected cells, we combined RNA sequencing with in silico pathway, gene ontology, transcription factor binding site, and network analyses. We measured infection-induced changes of selected transcripts by reverse transcription-quantitative polymerase chain reaction. RESULTS Human retinal pigment epithelial cells were permissive to infection with EBOV, and supported viral replication and release of virus in high titer. Unexpectedly, 28% of 560 upregulated transcripts in EBOV-infected cells were type I IFN responsive, indicating a robust type I IFN response. Following EBOV infection, cells continued to express multiple immunomodulatory molecules linked to ocular immune privilege. CONCLUSIONS Human retinal pigment epithelial cells may serve as an intraocular reservoir for EBOV, and the molecular response of infected cells may contribute to the persistence of live EBOV within the human eye. TRANSLATIONAL RELEVANCE This bedside-to-bench research links ophthalmic findings in survivors of EVD who suffer from uveitis with interactions between retinal pigment epithelial cells and EBOV.
Collapse
Affiliation(s)
- Justine R Smith
- Flinders University School of Medicine, Eye & Vision Health, Flinders Medical Centre, Adelaide, Australia.,Flinders University School of Medicine, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Adelaide, Australia.,South Australian Health & Medical Research Institute, SAHMRI: Mind & Brain Theme, Adelaide, Australia
| | - Shawn Todd
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Geelong, Australia
| | - Liam M Ashander
- Flinders University School of Medicine, Eye & Vision Health, Flinders Medical Centre, Adelaide, Australia
| | - Theodosia Charitou
- South Australian Health & Medical Research Institute, SAHMRI: EMBL Australia group, Infection & Immunity Theme, Adelaide, Australia.,Systems Biology Ireland, University College Dublin, UCD Conway Institute, Dublin, Ireland
| | - Yuefang Ma
- Flinders University School of Medicine, Eye & Vision Health, Flinders Medical Centre, Adelaide, Australia
| | - Steven Yeh
- Departments of Ophthalmology and Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ian Crozier
- Infectious Diseases Institute, Mulago Hospital Complex, Kampala, Uganda
| | - Michael Z Michael
- Flinders University School of Medicine, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Adelaide, Australia
| | - Binoy Appukuttan
- Flinders University School of Medicine, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Adelaide, Australia.,Flinders University School of Medicine, Eye & Vision Health, Flinders Medical Centre, Adelaide, Australia
| | - Keryn A Williams
- South Australian Health & Medical Research Institute, SAHMRI: Mind & Brain Theme, Adelaide, Australia.,Flinders University School of Medicine, Ophthalmology, Flinders Medical Centre, Adelaide, Australia
| | - David J Lynn
- Flinders University School of Medicine, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Adelaide, Australia.,South Australian Health & Medical Research Institute, SAHMRI: EMBL Australia group, Infection & Immunity Theme, Adelaide, Australia
| | - Glenn A Marsh
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Geelong, Australia
| |
Collapse
|
50
|
Kash JC, Walters KA, Kindrachuk J, Baxter D, Scherler K, Janosko KB, Adams RD, Herbert AS, James RM, Stonier SW, Memoli MJ, Dye JM, Davey RT, Chertow DS, Taubenberger JK. Longitudinal peripheral blood transcriptional analysis of a patient with severe Ebola virus disease. Sci Transl Med 2017; 9:9/385/eaai9321. [PMID: 28404864 DOI: 10.1126/scitranslmed.aai9321] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/21/2016] [Accepted: 02/03/2017] [Indexed: 12/14/2022]
Abstract
The 2013-2015 outbreak of Ebola virus disease in Guinea, Liberia, and Sierra Leone was unprecedented in the number of documented cases, but there have been few published reports on immune responses in clinical cases and their relationships with the course of illness and severity of Ebola virus disease. Symptoms of Ebola virus disease can include severe headache, myalgia, asthenia, fever, fatigue, diarrhea, vomiting, abdominal pain, and hemorrhage. Although experimental treatments are in development, there are no current U.S. Food and Drug Administration-approved vaccines or therapies. We report a detailed study of host gene expression as measured by microarray in daily peripheral blood samples collected from a patient with severe Ebola virus disease. This individual was provided with supportive care without experimental therapies at the National Institutes of Health Clinical Center from before onset of critical illness to recovery. Pearson analysis of daily gene expression signatures revealed marked gene expression changes in peripheral blood leukocytes that correlated with changes in serum and peripheral blood leukocytes, viral load, antibody responses, coagulopathy, multiple organ dysfunction, and then recovery. This study revealed marked shifts in immune and antiviral responses that preceded changes in medical condition, indicating that clearance of replicating Ebola virus from peripheral blood leukocytes is likely important for systemic viral clearance.
Collapse
Affiliation(s)
- John C Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Jason Kindrachuk
- Critical Care Medicine Department, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Baxter
- Institute for Systems Biology, Seattle, WA 98109, USA
| | | | - Krisztina B Janosko
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Rick D Adams
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Andrew S Herbert
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Rebekah M James
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Spencer W Stonier
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Matthew J Memoli
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John M Dye
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Richard T Davey
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel S Chertow
- Critical Care Medicine Department, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|