• EBV infects lymphoid, myeloid, and stem cells in patients with CAEBV, so HSCT is currently the only curative treatment for CAEBV disease.

  • EBV-infected HSCs have a higher rate of differentiation, which speeds up the cellular infection and alters immune response in CAEBV.

Abstract

Chronic active Epstein-Barr virus (EBV) disease (CAEBV) is a lethal syndrome because of persistent EBV infection. When diagnosed as CAEBV, EBV infection was observed in multiple hematopoietic lineages, but the etiology of CAEBV is still elusive. Bone marrow and peripheral cells derived from 5 patients with CAEBV, 1 patient with EBV-associated hemophagocytic lymphohistiocytosis, and 2 healthy controls were analyzed. Multiple assays were applied to identify and characterize EBV-infected cells, including quantitative polymerase chain reaction, PrimeFlow, and single-cell RNA-sequencing (scRNA-seq). Based on scRNA-seq data, alterations in gene expression of particular cell types were analyzed between patients with CAEBV and controls, and between infected and uninfected cells. One patient with CAEBV was treated with allogeneic hematopoietic stem cell transplantation (HSCT), and the samples derived from this patient were analyzed again 6 months after HSCT. EBV infected the full spectrum of the hematopoietic system including both lymphoid and myeloid lineages, as well as the hematopoietic stem cells (HSCs) of the patients with CAEBV. EBV-infected HSCs exhibited a higher differentiation rate toward downstream lineages, and the EBV infection had an impact on both the innate and adaptive immunity, resulting in inflammatory symptoms. EBV-infected cells were thoroughly removed from the hematopoietic system after HSCT. Taken together, multiple lines of evidence presented in this study suggest that CAEBV disease originates from the infected HSCs, which might potentially lead to innovative therapy strategies for CAEBV.

1.
Damania
B
,
Kenney
SC
,
Raab-Traub
N
.
Epstein-Barr virus: biology and clinical disease
.
Cell
.
2022
;
185
(
20
):
3652
-
3670
.
2.
Rostgaard
K
,
Wohlfahrt
J
,
Hjalgrim
H
.
A genetic basis for infectious mononucleosis: evidence from a family study of hospitalized cases in Denmark
.
Clin Infect Dis
.
2014
;
58
(
12
):
1684
-
1689
.
3.
Okuno
Y
,
Murata
T
,
Sato
Y
, et al
.
Defective Epstein-Barr virus in chronic active infection and haematological malignancy
.
Nat Microbiol
.
2019
;
4
(
3
):
404
-
413
.
4.
Cohen
JI
,
Iwatsuki
K
,
Ko
YH
, et al
.
Epstein-Barr virus NK and T cell lymphoproliferative disease: report of a 2018 International Meeting
.
Leuk Lymphoma
.
2020
;
61
(
4
):
808
-
819
.
5.
Fujiwara
S
,
Nakamura
H
.
Chronic active Epstein-Barr virus infection: is it immunodeficiency, malignancy, or both?
.
Cancers
.
2020
;
12
(
11
):
3202
.
6.
Smith
NA
,
Coleman
CB
,
Gewurz
BE
,
Rochford
R
.
CD21 (complement receptor 2) is the receptor for Epstein-Barr virus entry into T cells
.
J Virol
.
2020
;
94
(
11
):
e00428-20
.
7.
Ichigi
Y
,
Naitoh
K
,
Tokushima
M
, et al
.
Generation of cells with morphological and antigenic properties of microglia from cloned EBV-transformed lymphoid progenitor cells derived from human fetal liver
.
Cell Immunol
.
1993
;
149
(
1
):
193
-
207
.
8.
Ohga
S
,
Ishimura
M
,
Yoshimoto
G
, et al
.
Clonal origin of Epstein-Barr virus (EBV)-infected T/NK-cell subpopulations in EBV-positive T/NK-cell lymphoproliferative disorders of childhood
.
J Clin Virol
.
2011
;
51
(
1
):
31
-
37
.
9.
Fernandez-Pol
S
,
Silva
O
,
Natkunam
Y
.
Defining the elusive boundaries of chronic active Epstein-Barr virus infection
.
Haematologica
.
2018
;
103
(
6
):
924
-
927
.
10.
Lai
W
,
Wang
Y
,
Wang
J
,
Wu
L
,
Jin
Z
,
Wang
Z
.
Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis in adults and adolescents-a life-threatening disease: analysis of 133 cases from a single center
.
Hematology
.
2018
;
23
(
10
):
810
-
816
.
11.
Imashuku
S
.
Clinical features and treatment strategies of Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis
.
Crit Rev Oncol Hematol
.
2002
;
44
(
3
):
259
-
272
.
12.
Henter
JI
,
Horne
A
,
Arico
M
, et al
.
HLH-2004: diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis
.
Pediatr Blood Cancer
.
2007
;
48
(
2
):
124
-
131
.
13.
Dura
B
,
Choi
JY
,
Zhang
K
, et al
.
scFTD-seq: freeze-thaw lysis based, portable approach toward highly distributed single-cell 3’ mRNA profiling
.
Nucleic Acids Res
.
2019
;
47
(
3
):
e16
.
14.
Su
M
,
Pan
T
,
Chen
QZ
, et al
.
Data analysis guidelines for single-cell RNA-seq in biomedical studies and clinical applications
.
Mil Med Res
.
2022
;
9
(
1
):
68
.
15.
Kang
MS
,
Kieff
E
.
Epstein-Barr virus latent genes
.
Exp Mol Med
.
2015
;
47
(
1
):
e131
.
16.
Belluschi
S
,
Calderbank
EF
,
Ciaurro
V
, et al
.
Myelo-lymphoid lineage restriction occurs in the human haematopoietic stem cell compartment before lymphoid-primed multipotent progenitors
.
Nat Commun
.
2018
;
9
(
1
):
4100
.
17.
Zhang
YW
,
Mess
J
,
Aizarani
N
, et al
.
Hyaluronic acid-GPRC5C signalling promotes dormancy in haematopoietic stem cells
.
Nat Cell Biol
.
2022
;
24
(
7
):
1038
-
1048
.
18.
Quintanilla-Martinez
L
,
Swerdlow
SH
,
Tousseyn
T
,
Barrionuevo
C
,
Nakamura
S
,
Jaffe
ES
.
New concepts in EBV-associated B, T, and NK cell lymphoproliferative disorders
.
Virchows Arch
.
2023
;
482
(
1
):
227
-
244
.
19.
Dunmire
SK
,
Odumade
OA
,
Porter
JL
, et al
.
Primary EBV infection induces an expression profile distinct from other viruses but similar to hemophagocytic syndromes
.
PloS One
.
2014
;
9
(
1
):
e85422
.
20.
Sawada
A
,
Inoue
M
,
Kawa
K
.
How we treat chronic active Epstein-Barr virus infection
.
Int J Hematol
.
2017
;
105
(
4
):
406
-
418
.
21.
Luo
Y
,
Wei
A
,
Wang
B
, et al
.
Allogeneic hematopoietic stem cell transplantation with the modified myeloablative conditioning regimen for children with chronic active Epstein-Barr virus infection
.
Pediatr Investig
.
2022
;
6
(
4
):
250
-
259
.
22.
Bollard
CM
,
Cohen
JI
.
How I treat T-cell chronic active Epstein-Barr virus disease
.
Blood
.
2018
;
131
(
26
):
2899
-
2905
.
23.
Kebschull
JM
,
Zador
AM
.
Cellular barcoding: lineage tracing, screening and beyond
.
Nat Methods
.
2018
;
15
(
11
):
871
-
879
.
24.
Miller
TE
,
Lareau
CA
,
Verga
JA
, et al
.
Mitochondrial variant enrichment from high-throughput single-cell RNA sequencing resolves clonal populations
.
Nat Biotechnol
.
2022
;
40
(
7
):
1030
-
1034
.
25.
Janjetovic
S
,
Hinke
J
,
Balachandran
S
, et al
.
Non-random pattern of integration for Epstein-Barr virus with preference for gene-poor genomic chromosomal regions into the genome of Burkitt lymphoma cell lines
.
Viruses
.
2022
;
14
(
1
):
86
.
26.
von Laer
D
,
Meyer-Koenig
U
,
Serr
A
, et al
.
Detection of cytomegalovirus DNA in CD34+ cells from blood and bone marrow
.
Blood
.
1995
;
86
(
11
):
4086
-
4090
.
27.
Mendelson
M
,
Monard
S
,
Sissons
P
,
Sinclair
J
.
Detection of endogenous human cytomegalovirus in CD34+ bone marrow progenitors
.
J Gen Virol
.
1996
;
77
(
pt 12
):
3099
-
3102
.
28.
Youngblood
B
,
Hale
JS
,
Kissick
HT
, et al
.
Effector CD8 T cells dedifferentiate into long-lived memory cells
.
Nature
.
2017
;
552
(
7685
):
404
-
409
.
29.
Cobaleda
C
,
Jochum
W
,
Busslinger
M
.
Conversion of mature B cells into T cells by dedifferentiation to uncommitted progenitors
.
Nature
.
2007
;
449
(
7161
):
473
-
477
.
30.
Dalecki
AG
,
Greer
BD
,
Duverger
A
, et al
.
Host T cell dedifferentiation effects drive HIV-1 latency stability
.
J Virol
.
2022
;
96
(
5
):
e0197421
.
31.
Gutzeit
C
,
Nagy
N
,
Gentile
M
, et al
.
Exosomes derived from Burkitt’s lymphoma cell lines induce proliferation, differentiation, and class-switch recombination in B cells
.
J Immunol
.
2014
;
192
(
12
):
5852
-
5862
.
32.
Canitano
A
,
Venturi
G
,
Borghi
M
,
Ammendolia
MG
,
Fais
S
.
Exosomes released in vitro from Epstein-Barr virus (EBV)-infected cells contain EBV-encoded latent phase mRNAs
.
Cancer Lett
.
2013
;
337
(
2
):
193
-
199
.
33.
Lee
JH
,
Choi
J
,
Ahn
YO
,
Kim
TM
,
Heo
DS
.
CD21-independent Epstein-Barr virus entry into NK cells
.
Cell Immunol
.
2018
;
327
:
21
-
25
.
You do not currently have access to this content.
Sign in via your Institution