Вы находитесь на странице: 1из 6

The Journal of Infectious Diseases

BRIEF REPORT

Comprehensive Characterization of state-of-the-art laboratory resources available in EVD epicen-


ters. Consequently, T- and B-cell dynamics remain incomplete-
Cellular Immune Responses Following ly understood. Human data are so far limited to very few reports
Ebola Virus Infection describing longitudinal T- and B-cell dynamics in the context of
Christine Dahlke,1,3,a Sebastian Lunemann,2,a Rahel Kasonta,1,3 Benno Kreuels,1
EVD [2–5].
Stefan Schmiedel,1 My L. Ly,1,3 Sarah K. Fehling,4,5 Thomas Strecker,4,5 Reports about EBOV persistence in sanctuary sites have
Stephan Becker,4,5 Marcus Altfeld,2,3 Abdourahmane Sow,6 Ansgar W. Lohse,1,3
emerged recently [6], yielding questions about how and where
César Muñoz-Fontela,2 and Marylyn M. Addo1,3
1 EBOV can hide from the immune system and about the conse-
1st Department of Medicine, University Medical Center Hamburg-Eppendorf, 2Leibniz Institute
quences that persistence has on both human hosts and their

Downloaded from https://academic.oup.com/jid/article-abstract/215/2/287/2420589 by guest on 15 September 2018


for Experimental Virology, Heinrich-Pette-Institute, Hamburg, 3German Center for Infection
Research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems, 4German Center for Infection contacts. A case of EVD survival at the University Medical Cen-
Research (DZIF), partner site Giessen-Marburg-Langen, Braunschweig, and 5Institute for
Virology, Philipps University Marburg, Germany; and 6West African Health Organisation, Bobo- ter Hamburg-Eppendorf (UKE) provided the unique opportu-
Dioulasso, Burkina Faso nity to perform a comprehensive examination of lymphocyte
dynamics during the convalescent phase at days 37 and 46
The West African Ebola virus disease (EVD) outbreak was the after first appearance of EVD symptoms [7]. Importantly, our
largest EVD outbreak in history. However, data on lymphocyte results represent the first immunity data from an EVD survivor
dynamics and the antigen specificity of T cells in Ebola survi-
during the convalescent phase who cleared the infection
vors are scarce, and our understanding of EVD pathophysiology
without experimental drugs [7]. Beyond the assessment of neu-
is limited. A case of EVD survival in which the patient cleared
Ebola virus (EBOV) infection without experimental drugs al- tralizing antibodies, our approach involved a detailed analysis of
lowed for the detailed examination of lymphocyte dynamics. T- and B-cell dynamics and the investigation of antigen-specific
We demonstrate the persistence of T-cell activation well beyond T cells via intracellular staining (ICS) and enzyme-linked im-
viral clearance and detect EBOV-specific T cells. Our study pro- munospot (ELISPOT) analysis.
vides significant insights into lymphocyte specificity during the Despite the patient’s full clinical recovery from EVD symp-
recovery phase of EVD and may inform novel strategies to treat toms, a high fraction of cytotoxic T cells and persistent immune
EVD. activation were detectable well beyond clearance of plasma vire-
Keywords. EVD; EVD survivor; convalescent phase; cellu- mia. EBOV glycoprotein (GP)–specific T cells were detectable
lar immune response; lymphocyte dynamics; T-cell activation; but of low magnitude and were predominantly found in the
Ebola GP–specific T-cell responses; polyfunctional T-cell CD8+ T-cell population with the largest fraction of cells produc-
responses.
ing tumor necrosis factor α (TNF-α) but not interferon γ (IFN-
γ), interleukin 2 (IL-2), or macrophage inflammatory protein
The Ebola outbreak in West Africa was the most dramatic Ebola (MIP-1β; hereafter, “TNF-α single producers”), as well as
virus disease (EVD) outbreak in history. During this outbreak, among CD107a+ cells. Our data provide detailed insight into
27 patients with EVD were medically evacuated from Africa to the understanding of lymphocyte dynamics during the recovery
Europe and the United States [1], providing novel opportunities phase of EVD, which may contribute to future design of thera-
to fill critical gaps in our understanding of the mechanisms un- peutics and vaccines.
derlying the immunopathophysiology of EVD.
Data on immunity to Ebola virus (EBOV) infection in Ebola METHODS
survivors are scarce, owing to factors including unpredictability,
Peptides
limited numbers of cases in previous outbreaks, and limited Overlapping peptides (OLPs) cover the Zaire EBOV GP. Se-
quences and pools are shown in Supplementary Table 1.
Received 1 July 2016; accepted 17 October 2016; published online 31 October 2016.
Presented in part: 13th Kongress für Infektionskrankheiten und Tropenmedizin, Würzburg, Patient and Control Specimens
Germany, June 2016; 46th Annual Meeting of the German Society for Immunology, Hamburg,
Germany, September 2016. Blood specimens from the patient and the healthy controls
a
C. D. and S. L. contributed equally to this study. were collected at the UKE. Donors provided written informed
Correspondence: M. M. Addo, University Medical Center Hamburg Eppendorf, 1st Depart-
ment of Medicine, Division of Tropical Medicine, Martinistr. 52, 20246 Hamburg, Germany
consent. Peripheral blood mononuclear cells (PBMCs) were
(m.addo@uke.de). isolated via Ficoll density gradient centrifugation from
The Journal of Infectious Diseases® 2017;215:287–92 blood collected in ethylenediaminetetraacetic acid (EDTA)–
© The Author 2016. Published by Oxford University Press for the Infectious Diseases Society of
America. All rights reserved. For permissions, e-mail journals.permissions@oup.com.
lined tubes and cryopreserved using standard operating
DOI: 10.1093/infdis/jiw508 procedures.

BRIEF REPORT • JID 2017:215 (15 January) • 287


Phenotyping and ICS 81% (on day 37) and 80% (on day 46) of the EM subpopulation,
Phenotyping and ICS were performed using cryopreserved compared with a mean (±SD) of 14% ± 9%. Analysis of CD4+ T
PBMCs. Staining panels are depicted in Supplementary Table 2. cells and their corresponding EM subset revealed increased T-
For ICS, PBMCs were incubated for 6 hours at 37°C with EBOV cell activation too, but to a lesser extent.
GP peptide pools in the presence of CD28/CD49d, GolgiStop, Cytotoxicity was assessed via GrzB staining. The CD4+ and
and GolgiPlug. Negative controls were unstimulated but treated CD8+ T-cell populations showed a high proportion of GrzB+
with dimethyl sulfoxide. Positive controls were stimulated with cells (Figure 1C).
PMA and CEF (a pool consisting of peptides from cytomegalo- Regulatory lymphocytes play an important role in immune
virus, Epstein-Barr virus, and influenza virus). Cells were ana- homeostasis. Whereas the regulatory T-cell (Treg) population
lyzed on a BD LSRFortessa and evaluated with FlowJo 10. (CD4+CD25 hiCD127low) was quantitatively comparable to
ELISPOT that in healthy donors (SF2), the 2 B-cell subsets
EBOV GP–specific T-cell responses were assessed using over- (CD19+CD24hiCD38hi and CD24hiCD27+), in which regulatory

Downloaded from https://academic.oup.com/jid/article-abstract/215/2/287/2420589 by guest on 15 September 2018


lapping peptides by IFN-γ ELISPOT (MabtechELISpotPLUS) B-cells are enriched [10], showed slightly lower proportions
as described by Streeck et al [8]. than in controls (Figure 1D).
The slight expansion of plasmablasts (CD19+CD24−CD38high)
Neutralizing Antibody Assay
in the patient with EVD indicates activation of the humoral im-
The neutralizing antibody assay was performed with plasma
mune response and may mirror generation of antibodies. Indeed,
samples, as previously described [9].
we detected EBOV-specific neutralizing antibodies with geomet-
RESULTS ric mean titers of 91 (on day 37) and 76 (on day 46; SF3).

The interaction of EBOV with the human immune system re- EBOV GP–Specific T-Cell Responses
mains incompletely understood. We here report new insight To define EBOV-specific cellular immune responses, we investi-
into the long-lasting immune responses of an EVD survivor fol- gated the induction of 4 cytokines (IFN-γ, IL-2, TNF-α, and
lowing recovery. MIP-1β) in response to EBOV GP–specific OLP stimulation,
Ten days after first symptoms, the patient was transferred to via ICS (Figure 2). Antigen-specific T cells were predominantly
Hamburg, Germany, and treated in an isolation unit for indi- found in the CD8+ compartment and not in the CD4+ compart-
viduals with highly contagious diseases. The initial clinical ment. Whereas TNF-α/IFN-γ double producers were predomi-
course has been previously described [7]. He experienced a se- nantly observed upon SP and GP2 stimulation, GP1a
vere EBOV infection with the associated complications of sep- stimulation resulted in TNF-α induction (Figure 2A and 2B).
ticemia, encephalopathy, and respiratory failure [7]. After Only limited expression was observed for MIP-1β, and no induc-
recovery and EBOV-negative results of polymerase chain reac- tion was detectable for IL-2. Limited polyfunctionality (maximal-
tion analysis of plasma, he was transferred to the infectious dis- ly 3 functions) was observed in CD8+ T cells in response to GP2
ease unit (Supplementary Figure 1). Blood specimens were and SP peptide pools (Figure 2C), and none were observed in
collected in EDTA-lined tubes on days 37 and 46 after illness. CD4+ T cells (data not shown). Antigen-specific cytotoxic lym-
phocytes were characterized by staining with CD107a, with the
Lymphocyte Dynamics
Initial analysis of CD4+, CD8+, and CD19+ lymphocyte dynam- strongest induction observed in the GP1a pool (Figure 2B).
ics revealed minor differences in their respective frequencies as To increase sensitivity, EBOV-specific T-cell responses were
compared to findings for healthy controls (SF2). Subsequently, also assessed by IFN-γ ELISPOT (Figure 2D). Significant
the expression of CCR7 and CD45RA was evaluated (Figure 1A). responses were observed for peptide pools GP2.2 (376 spot-
While antigen-experienced effector memory (EM) T cells forming cells [SFCs] per million peripheral blood mononuclear
(CCR7−CD45RA−) were only slightly increased in the CD4+ cells [PBMCs] on day 37) and SP (202 SFCs per million PBMCs
compartment (21% on days 37 and 46, compared with a on day 37), confirming the ICS data described above. A higher
mean [±SD] of 13% ± 1.5% in controls), the proportion of response was observed for the master pool MP1.
EM CD8+ T cells expanded, compared with controls (39% on In summary, both ICS and ELISPOT revealed EBOV-specific
day 37 and 36% on day 46, compared with a mean [±SD] of antiviral T-cell activity, albeit with a low magnitude.
19% ± 7% in controls).
DISCUSSION
T-cell activation is a critical element of the antiviral immune
defense. We therefore assessed activation via HLA-DR and EBOV represents one of the most deadly human pathogens, and
CD38 staining (Figure 1B). Analysis of CD8+ T cells revealed to date little is understood about the human immunity to EBOV
a high frequency of activated cells, composing 68% (on day infection. Immunopathogenesis, factors that influence survival,
37) and 62% (on day 46) of the main CD8+ T-cell population, and correlates of protection have not been completely
compared with a mean (±SD) of 8% ± 4% among controls, and elucidated.

288 • JID 2017:215 (15 January) • BRIEF REPORT


Downloaded from https://academic.oup.com/jid/article-abstract/215/2/287/2420589 by guest on 15 September 2018

Figure 1. Lymphocyte dynamics during the convalescent phase of Ebola virus (EBOV) disease (EVD). A, CD4+ (left) and CD8+ (right) T-cell subsets were stained by
CCR7 and CD45RA to distinguish naive (CCR7+CD45RA+), effector memory (EM; CCR7−CD45RA−), central memory (CM; CCR7+CD45RA−), and effector memory RA
(CCR7−CD45RA+) T cells. In the upper row, data are represented in contour plots. The control plot depicts a representative experiment. The lower row shows the
distribution of T-cell subsets. The control pie chart represents the median value of measured results for 5 controls. B, T-cell activation was analyzed by HLA-DR and
CD38 staining to investigate CD4+, CD8+, and the respective EM T cells. At left, contour plots show the gating strategy. At right, the graph depicts the results (control
values are for 5 individuals). C, Cytotoxic T cells were assessed via GrzB staining. At left, contour plots show results for the patient with EVD on day 37 after symptom
onset for CD4+ (upper plot) and CD8+ (lower plot) T cells. Results are highlighted in the graph (red triangle, EVD survivor; circle, 2 healthy controls). D, Regulatory B
cells are enriched in transitional B cells (CD24+CD38high; blue gate; upper plot) and the CD19+CD24hiCD27+ population (lower plot). At left, contour plots depict rep-
resentative plots for the patient with EVD (on day 37) and 1 healthy control. At right, results are highlighted in the graph (data are for 5 controls). Values from controls
are shown as median values with SDs.

BRIEF REPORT • JID 2017:215 (15 January) • 289


Downloaded from https://academic.oup.com/jid/article-abstract/215/2/287/2420589 by guest on 15 September 2018

Figure 2. Ebola virus (EBOV) glycoprotein (GP)–specific T-cell responses found in CD8+ T cells. A, EBOV GP–specific interferon γ (IFN-γ)/tumor necrosis factor α
(TNF-α) T-cell responses. At left, graphs depict the observed IFN-γ/TNF-α responses in CD8+ (upper graph) and CD4+ (lower graph) compartments specific for the 4
EBOV GP peptide pools (control values are for 2 individuals). At right, representative dot plots show EBOV GP–specific IFN-γ/TNF-α T-cell responses for both CD8+
(upper panel) and CD4+ (lower panel) T cells (data are for patients with EVD on day 37 after first appearance of EVD symptoms). B, Cytokine and cytotoxic T-
lymphocyte responses upon stimulation with EBOV GP peptide pools (control values are for 2 individuals). C, The distribution of the functionality of CD8+ T-cell
responses to the 4 EBOV GP peptide pools on days 37 and 46 after first appearance of EVD symptoms. D, IFN-γ enzyme-linked immunospot assay of responses
against EBOV GP peptide pools. A CEF pool (consisting of peptides from cytomegalovirus, Epstein-Barr virus, and influenza virus) was used as positive control. The
assay was performed in duplicate. The graph shows median values and SDs (control values are for 2 individuals). Abbreviations: PBMC, peripheral blood mono-
nuclear cell; SFC, spot-forming cell.

290 • JID 2017:215 (15 January) • BRIEF REPORT


The medical management of the EVD survivor at the UKE ongoing T-cell activation reflects the previously experienced
provided the unique opportunity to examine lymphocyte dy- septicemia, nonspecific bystander activation, or the persistence
namics during the early convalescent phase of EVD in compre- of virus or viral antigens in sanctuary sites. Support for possible
hensive detail. This report is the first to show T- and B-cell virus persistence is provided by 2 recent publications describing
dynamics and antigen-specific T cells in an EVD survivor, a similar phenotype of long-lasting activated cytotoxic T cells
who received only supportive therapy and no experimental [2, 3] and the emerging reports on post-EVD syndrome
drugs. (PEVDS) and EBOV persistence in compartments other than
There is no consensus about the correlates of protection the bloodstream [6]. It is therefore conceivable that antigen
against EBOV, but vaccine-induced antigen-specific antibodies may have persisted at a low level, contributing to T-cell activa-
were reported in surviving nonhuman primates [11]. Plasma tion and cytotoxicity.
from the Hamburg patient effectively inhibited replication of In light of EBOV persistence and PEVDS, the convalescent
EBOV (isolate Mayinga) with titers similar to levels observed phase of EVD survivors represents an important state to an-

Downloaded from https://academic.oup.com/jid/article-abstract/215/2/287/2420589 by guest on 15 September 2018


in the EVD survivor treated in Frankfurt, who was analyzed alyze. However, thus far human data are scarce. To this date,
at similar time points [12]. Furthermore, neutralizing antibodies only a few recent publications specifically investigated T- and
were detected in 3 patients treated in Spain during their conva- B-cell dynamics, of which 2 focused on the convalescent
lescent phase [13]. These data reveal robust EBOV-specific phase. A comparison of these data with results associated
humoral response following infection, with generation of with the Hamburg patient may contribute to our understand-
neutralizing antibodies that persisted for at least 5 weeks ing of immune responses following EBOV infection. Long-
after illness. lasting lymphocyte dynamics and immunity following
Antigen-specific cellular immunity may contribute impor- EBOV infection were analyzed in 6 patients. Of note, all of
tantly to protection against EBOV infection. But so far, immu- these analyzed patients received experimental drugs, in con-
nodominant EBOV-specific T-cell responses in humans remain trast to the Hamburg patient. Further, it needs to be consid-
largely unknown. Our approach involved the analysis of anti- ered that analysis time points in all these patients were highly
gen-experienced and polyfunctional T cells following EBOV in- heterogeneous, with different rates of disease progression, dis-
fection. The increased number of EM and TEMRA cells in the similar therapies, distinct viral loads, and differences in pro-
CD8+ compartment imply antigen experience, but ELISPOT tocols and assays.
analysis of the reactivity of EBOV GP–specific T cells revealed A clear difference was observed in the type of cytokines.
an overall low IFN-γ response upon stimulation with GP Ahmed et al investigated EBOV-specific T-cell specificity in
peptide pools. To increase the breadth of cytokine analysis, 3 EVD survivors and observed consistent IFN-γ responses in
we performed ICS for 3 additional readouts and observed pre- single-producing cells to nucleoprotein and variable responses
dominantly TNF-α single-producing T cells and a low frequen- to GP. Only 1 survivor showed a strong IFN-γ response to GP
cy of IFN-γ/TNF-α double-producing T cells. But taking into [2]. We detected predominantly TNF-α (and CD107a) induc-
consideration that immune responses were analyzed at days tion. High T-cell activation was consistent in all 3 studies,
37 and 46 after illness onset, some antiviral functions may al- and in the context of the observed cytotoxic profile, these
ready have played their part. One recent study in nonhuman data indicate a robust immune response following EBOV infec-
primates revealed a peak in IFN-γ and IL-2 expression by tion and may suggest possible antigen persistency. In contrast to
CD8+ T cells early, at day 14 after infection [14]. However, previous reports, recent data from us and others do not show
the significance of virus-specific CD8+ T cells, monofunctional- immune suppression but prolonged immune activation with
ity or polyfunctionality, and the combination of cytokines fol- high cytotoxicity, elements that may have contributed to viral
lowing EBOV infection in humans remains to be identified. control. Further, these similarities were detected irrespective
More human data are required to identify cellular key factors of the patient treatment strategy, suggesting that these experi-
contributing to survival or protection. mental treatments had little impact on T-cell activation and fur-
Lymphocyte activation has been suggested as a marker for ther assuming delayed clearance of EBOV in humans, which
survival following EBOV infection. This was first described by needs to be further investigated.
Baize et al, based on their observation of increased messenger In sum, detailed analysis of immune responses and dynamics
RNA levels for several activation markers in PBMCs [15]. following EBOV infection may help to broaden our understand-
Viral infections are typically associated with massive expansion ing of possible mechanisms leading to EBOV persistence and
of activated T cells, which generally declines following pathogen the development of PEVDS. Identification of immune respons-
clearance. Although viable EBOV was not detected for around es and of antigens and epitopes targeted in patients with natural
20 days in analyzed body fluids, T cells remained highly activat- immunity to EBOV infection may also be critical for our under-
ed with a strong cytotoxic profile, specifically in the CD8+ T-cell standing of immunopathogenesis and may inform strategic
compartment. There is a question of whether the observed Ebola vaccine design and monitoring.

BRIEF REPORT • JID 2017:215 (15 January) • 291


Supplementary Data 2. McElroy AK, Akondy RS, Davis CW, et al. Human Ebola virus infection results in
substantial immune activation. Proc Natl Acad Sci U S A 2015; 112:4719–24.
Supplementary materials are available at http://jid.oxfordjournals.org.
3. Ruibal P, Oestereich L, Ludtke A, et al. Unique human immune signature of Ebola
Consisting of data provided by the author to benefit the reader, the posted virus disease in Guinea. Nature 2016; 533:100–4.
materials are not copyedited and are the sole responsibility of the author, so 4. Agrati C, Castilletti C, Casetti R, et al. Longitudinal characterization of dysfunc-
questions or comments should be addressed to the author. tional T cell-activation during human acute Ebola infection. Cell Death Dis 2016;
7:e2164.
Notes 5. Ellebedy AH, Jackson KJ, Kissick HT, et al. Defining antigen-specific plasmablast
and memory B cell subsets in human blood after viral infection or vaccination. Nat
Acknowledgments. We thank Claudia Beisel, Till Bornscheuer, Jakob Immunol 2016; 17:1226–34.
Cramer, Claudia Frey, Johannes Jochum, Stefan Lüth, Hanna Matthews, 6. Hayden EC. Maternal health: Ebola’s lasting legacy. Nature 2015; 519:24–6.
Claudia Röder, Thierry Rolling, Camilla Rothe, Helmut Salzer, Guido Schä- 7. Kreuels B, Wichmann D, Emmerich P, et al. A case of severe Ebola virus
fer, Christoph Schramm, Julian Schulze zur Wiesch, Christof Vinnemeier, infection complicated by gram-negative septicemia. N Engl J Med 2014;
Tobias Werner, Dominic Wichmann, Dorothea Wiemer, and all of the 371:2394–401.
other physicians and nurses involved in the clinical treatment of the patient; 8. Streeck H, Frahm N, Walker BD. The role of IFN-gamma Elispot assay in HIV
Florian Harder, Tom Hildebrandt, Hannes Kalb, and Kristina Scholz, for vaccine research. Nat Protoc 2009; 4:461–9.
intensive support in setting up, maintaining, and running the unit for the 9. Agnandji ST, Huttner A, Zinser ME, et al. Phase 1 Trials of rVSV Ebola Vaccine in

Downloaded from https://academic.oup.com/jid/article-abstract/215/2/287/2420589 by guest on 15 September 2018


Africa and Europe. N Engl J Med 2016; 374:1647–60.
treatment of highly contagious infections at the University Medical Center
10. Rosser EC, Mauri C. Regulatory B cells: origin, phenotype, and function. Immu-
Hamburg–Eppendorf (UKE); Nadine Biedenkopf, for organizing sample nity 2015; 42:607–12.
shipment; Joseph Poetsch and Madeleine Zinser, for critical review of the 11. Marzi A, Engelmann F, Feldmann F, et al. Antibodies are necessary for rVSV/
manuscript; and Gotthard Ludwig and Michael Schmidt (Philipps Univer- ZEBOV-GP-mediated protection against lethal Ebola virus challenge in nonhu-
sity Marburg), for their technical assistance with biosafety level 4 man primates. Proc Natl Acad Sci U S A 2013; 110:1893–8.
procedures. 12. Wolf T, Kann G, Becker S, et al. Severe Ebola virus disease with vascular leakage
Financial support. This work was supported by the German Center for and multiorgan failure: treatment of a patient in intensive care. Lancet 2015;
Infection Research (DZIF). 385:1428–35.
Potential conflicts of interest. All authors: No reported conflicts. All au- 13. Luczkowiak J, Arribas JR, Gomez S, et al. Specific neutralizing response in plasma
from convalescent patients of Ebola virus disease against the West Africa Makona
thors have submitted the ICMJE Form for Disclosure of Potential Conflicts
variant of Ebola virus. Virus Res 2016; 213:224–9.
of Interest. Conflicts that the editors consider relevant to the content of the 14. Qiu X, Audet J, Wong G, et al. Sustained protection against Ebola virus infection
manuscript have been disclosed. following treatment of infected nonhuman primates with ZMAb. Sci Rep 2013;
3:3365.
References 15. Baize S, Leroy EM, Georges-Courbot MC, et al. Defective humoral responses and
1. Uyeki TM, Mehta AK, Davey RT Jr, et al. Clinical management of Ebola virus dis- extensive intravascular apoptosis are associated with fatal outcome in Ebola virus-
ease in the United States and Europe. N Engl J Med 2016; 374:636–46. infected patients. Nat Med 1999; 5:423–6.

292 • JID 2017:215 (15 January) • BRIEF REPORT

Вам также может понравиться