Sunteți pe pagina 1din 5

Biomedicine & Pharmacotherapy 117 (2019) 109122

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Current mechanistic insights into the role of infection in systemic lupus T


erythematosus

Qingjun Pan ,1, Zejian Liu1, Shuzhen Liao1, Lin Ye, Xing Lu, Xiaoqun Chen, Zhihang Li, Xinxin Li,

Yong-Zhi Xu, Huafeng Liu
Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University,
Zhanjiang, Guangdong, 524001, China

A R T I C LE I N FO A B S T R A C T

Keywords: Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized by inflammation and ab-
Systemic lupus erythematosus normal production of autoantibody, but the mechanisms of the aberrant immune responses are currently un-
Infectious agents known. Recently, growing evidence has suggested that infection plays a pivotal role in SLE. Here, we investigate
Mechanism the role of infectious agents (e.g., Epstein-Barr virus, parvovirus B19, human T-lymphotropic virus type 1,
Causative association
human immunodeficiency virus type 1, and endogenous retroviruses) in the pathogenesis of SLE. More im-
Protective association
portantly, we explore the known mechanisms underlying the involvement of infectious agents in the patho-
genesis of SLE, including molecular mimicry, epitope spreading, superantigen production, bystander activation,
persistent viral infection, altered apoptosis, clearance deficiency, and epigenetic alterations (e.g., DNA methy-
lation and microRNAs). However, some infectious agents (e.g., malaria parasites, hepatitis B virus, Toxoplasma
gondii, and Helicobacter pylori) may exert protective effects on SLE. Therefore, the relationship between infection
and SLE is multifaceted and multidirectional, including causative and/or protective associations, which warrant
further investigation in the future.

1. Introduction development of autoimmune diseases, which have been widely studied


in human and experimental animal models [8–11], particularly with
Systemic lupus erythematosus (SLE), a chronic systemic auto- respect to the pathogenesis of SLE [6,12]. The predominant pathogens
immune disease, is characterized by inflammation and abnormal pro- known to be associated with SLE include Epstein-Barr virus (EBV),
duction of autoantibody affecting multiple organs throughout the body. parvovirus B19 virus, exogenous retroviruses (e.g., human T-lympho-
However, the mechanisms underlying the aberrant immune responses tropic virus type 1 [HTLV-1] and human immunodeficiency virus type 1
remain not well known [1]. In the past several decades, infection has [HIV-1]), and endogenous retroviruses (ERVs) [5–7,12–19]. Other in-
been increasingly recognized as a possible trigger of SLE. More recently, fectious agents that may trigger SLE but their involvement in SLE pa-
accumulating evidence has indicated that infection and SLE are tightly thogenesis is less understood, including viruses such as transfusion-
linked, with a multifaceted and multidirectional relationship [2–7]. transmitted virus (TTV) and cytomegalovirus (CMV), and bacteria such
In this review, we investigated the involvement of infectious agents as vibrio cholera and group A streptococcus (GAS) [3,6,20–25]. Only
in the pathogenesis of SLE. More importantly, the currently understood one or two cases of some pathogens, including hepatitis C virus (HCV),
mechanisms underlying the involvement of infectious agents in SLE are C-type oncornavirus, parainfluenza type 1, rubella virus, and measles,
summarized. has been reported in of the involvement of SLE pathogenesis
[3,6,26,27]. The definite, probable, and possible influences of these
2. Infectious agents and the mechanisms of their involvement in infectious agents on SLE are summarized in Table 1.
SLE pathogenesis Among SLE etiologies, infectious agents have been suggested to play
pivotal roles in individuals genetically predisposition; these pathogens
Infectious agents, such as viruses, bacteria, parasites, and fungi, are induce aberrant innate and adaptive immunity, leading to a loss of
associated with the abnormal production of autoantibodies and the tolerance toward autoantigens. The potential mechanisms of infectious


Corresponding authors at: No. 57, South of People's Avenue, Xiashan, Zhanjiang, Guangdong, 524001, China.
E-mail addresses: stillwater2004@126.com (Q. Pan), hf-liu@263.net (H. Liu).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.biopha.2019.109122
Received 23 April 2019; Received in revised form 5 June 2019; Accepted 12 June 2019
0753-3322/ © 2019 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/BY/4.0/).
Q. Pan, et al. Biomedicine & Pharmacotherapy 117 (2019) 109122

Table 1
Infectious agents involved in the pathogenesis of SLE.
Definite Probable Possible

Viruses: Viruses: Viruses:


Epstein-Barr virus (EBV) Cytomegalovirus (CMV) Hepatitis C virus (HCV)
Parvovirus B19 virus Transfusion-transmitted virus (TTV) C-type oncornavirus
Exogenous retroviruses:HIV-1 Bacteria Measles
HTLV-1 Group A streptococcus (GAS) Parainfluenza type 1
Endogenous retroviruses (ERVs) Vibrio cholerae Rubella virus

Note: HIV-1, Human immunodeficiency virus type 1; HTLV-1, Human T-lymphotropic viruses type 1.

Fig. 1. The potential mechanisms of infectious agents in the pathogenesis of SLE. (APC, antigen-presenting cells; TCR, T cell receptors; MHC, major histocompat-
ibility complex).

agents in the pathogenesis of SLE are discussed in detail below (Fig. 1). trigger or initiate SLE, epitope spreading, which can progress among
multiple epitopes on a single antigen or from one antigenic molecule to
another, facilitates the presentation of autoantigens through antigen-
2.1. Molecular mimicry
presenting cells (APCs) and induces the activation of autoreactive
lymphocytes and the production of antibodies [16,28,32–34].
Molecular mimicry by infectious agents can activate autoreactive T
cells via exogenous antigens with sequence or structural similarities to
autoantigens, consequently disordering the immune system and leading 2.3. Superantigen production
to autoimmune responses [2,10,16,17,28,29]. However, studies also
showed that molecular mimicry did not account for T cell activation in Superantigens, produced by a variety of viruses and bacteria, can
autoimmune diseases. Molecular mimicry may be mistakenly identified bind to the variable domains of the T cell receptor beta chain and many
when a single T cell has dual T cell receptors (TCR) that can react with major histocompatibility complex (MHC) class II molecules. Because
exogenous antigens and autoantigens. Nonetheless, this mechanism superantigens lack of the antigenic specificity, this process leads to the
may provide a better understanding of the cross-reactivity [7,30,31]. activation of many T lymphocytes with different antigenic specificities,
resulting in the development of autoimmune reactions [3,6,9,10].
2.2. Epitope spreading
2.4. Bystander activation
The adaptive immune system recognizes epitopes of the initial an-
tigens in the early immune response. However, over time, T and B cells Bystander activation occurs when APCs are significantly activated
may respond to other epitopes. This process, known as epitope by the main virulence factors of infectious agents, which can potentially
spreading, is pivotal to promote the efficiency and breadth of the im- activate pre-primed autoreactive T cells by increasing the production of
mune response to infectious agents and tumors. When infectious agents cytokines. The bystander activation of autoreactive immune T cells can

2
Q. Pan, et al. Biomedicine & Pharmacotherapy 117 (2019) 109122

2.7. Epigenetic factors

MicroRNAs are a novel class of endogenous, non-coding small RNAs


with a length of about 19–25 nucleotides. MicroRNAs negatively reg-
ulate gene expression through degrading or inhibiting post-transcrip-
tional mRNA translation. In the immune system, the biological func-
tions of microRNAs mainly include regulation of the differentiation of
immune cells and innate and adaptive responses. Dysregulation of
microRNAs may result in autoimmune diseases, including SLE. Indeed,
many pathogens can induce changes in the expression of particular
microRNAs [36,37]. Regulation of innate immune responses by mi-
croRNAs may prompt increased production of inflammatory cytokines;
for example, microRNAs abnormally activate type I IFN signaling in-
volved in SLE [38]. Further, abnormal microRNAs may impair adaptive
immunity, causing B and T cell dysfunction. Abnormal T cell function,
such as Treg instability, loss of Treg suppressor function, and lympho-
cyte accumulation, can induce loss of tolerance and the development of
autoimmunity. Abnormal B cell function involves impaired IgG class-
switch and an inadequate germinal center response, which provokes the
production of antibodies [39,40]. Moreover, altered microRNA levels
may indirectly or directly inhibit DNA methyltransferases (DNMTs),
leading to DNA hypomethylation and upregulation of autoimmunity-
associated genes (e.g., CD70, CD11a, and CD40 L), all of which are
involved in the pathogenesis of SLE [38]. Other recent studies de-
monstrated that bacterial and viral DNA are hypomethylated, which
may induce autoimmune responses by activation of Toll-like receptor 9
(TLR-9) signaling, overexpression of type 1 INF genes, and activation of
polyclonal B cells and autoantibodies [41,42].

3. The possible protective effects of infectious agents in SLE

Accumulating evidence suggests that infection can induce auto-


immune diseases. However, the relationships between infection and
autoimmune diseases are multifaceted and multidirectional, including
causative and/or protective associations [43]. Thus far, numerous
epidemiological studies and animal experimental studies have shown
Fig. 2. The potential protective mechanisms of infectious agents in SLE. (Treg that infectious agents, primarily protozoa, may prevent autoimmunity
cells, regulatory T-cells; TLR, Toll-like receptor). or even abrogate an ongoing autoimmune process, depending on the
unique interaction between the microorganism and host [2]. The hy-
subsequently induce autoimmune diseases. In addition to bystander giene hypothesis, which was developed based on the concomitant in-
activation of autoreactive T cells, APCs can activate virus-specific T crease in autoimmune diseases and decrease in infections due to the
cells, which might initiate bystander activation through identifying improvement of hygiene and sanitation in western countries, proposes
virus-infected cells and releasing cytotoxic granules and cytokines, such that infections may play protective roles in autoimmune diseases [44].
as TNF, lymphotoxin (LT), and nitric oxide (NO), possibly causing by- In general, autoimmune protection can be achieved by blocking antigen
stander T cells to kill uninfected neighboring cells and activate auto- presentation or antigen competition.
immune responses [2,3,6,9,10,31]. The potential protective mechanisms of infectious agents involve
the following: firstly, antigenic competition leads to a decrease in the
response to autoantigens; secondly, the suppression of host immune
2.5. Persistent viral infection
response to self- or non-self molecules by irritating subgroups of Treg
cells; thirdly, TLR signaling directly or indirectly regulates immune
Persistent viral infection may result in autoimmunity by constantly
suppression of Treg cells (Fig. 2) [3,4,6,9,10,45].
stimulating the immune system. At the same time, lymphotropic viruses
So far, some reports have confirmed that infectious agents may play
might stimulate the development of polyclonal lymphocytes, increase
a protective role in SLE. Continued infection with lactate dehy-
the production of antibodies and circulating immune complexes, and
drogenase virus significantly reduced antibodies against nuclear an-
eventually damage self-tissues [3,6,9,28].
tigen and DNA in lupus mice, delaying the progression of nephritis
[46,47]. Malaria infection attenuates lupus nephritis in lupus mice by
2.6. Altered apoptosis and clearance deficiency reducing kidney oxidative stress and increasing the antioxidant defense
system [48,49]. Data from lupus animal studies indicated that FcγRIIb
Cell apoptosis induced by infection can trigger autoreactive Th17 deficiency prevents animals from infection, but increases susceptibility
cells to promote autoinflammation and autoantibody generation. In to autoimmune diseases [50]. Some researchers have also used lupus
addition, insufficient clearance of infectious agents and apoptotic cells mice infected with live Plasmodium chabaudi (P. chabaudi) to improve
due to genetic defects of the immune system such as complement de- histopathological changes kidney tissue [51]. Toxoplasma infection can
ficiency may lead to accumulation of nuclear material [35]. These non- delay the development of autoimmune nephropathy in lupus-suscep-
ingested nuclear materials may provide survival signals for autoreactive tible mice by altering mouse autoantibodies [52,53]. Parasitic worms
B cells, resulting in the production of antibodies against nuclear may protect kidney pathology by secreting glycoproteins to reduce
structures and immunological dysfunction [3]. antinuclear antibody production and immune complex deposition or

3
Q. Pan, et al. Biomedicine & Pharmacotherapy 117 (2019) 109122

altering the cytokine microenvironment [54,55]. Studies have also [12] Z.X. Li, S. Zeng, H.X. Wu, Y. Zhou, The risk of systemic lupus erythematosus as-
found that the worm derivative phosphocholine (PC) acts as an im- sociated with Epstein-Barr virus infection: a systematic review and meta-analysis,
Clin. Exp. Med. (2018).
munomodulatory molecule to significantly decrease proteinuria levels, [13] H.W. Lehmann, P. von Landenberg, S. Modrow, Parvovirus B19 infection and au-
diminish proinflammatory cytokines, increase anti-inflammatory cyto- toimmune disease, Autoimmun. Rev. 2 (4) (2003) 218–223.
kines, and significantly amplify CD4+CD25+FOXP3+ Treg cell through [14] S. Aslanidis, A. Pyrpasopoulou, K. Kontotasios, S. Doumas, C. Zamboulis, Parvovirus
B19 infection and systemic lupus erythematosus: Activation of an aberrant
multiple mechanisms [56]. Epidemiological studies have revealed that pathway? Eur. J. Intern. Med. 19 (5) (2008) 314–318.
Helicobacter pylori (H. pylori) serum reactivity is associated with SLE [15] G. Valencia, Y.E. Pacheco, Ueji Nakazawa, E.A. Rodriguez Dzul, A.V. Angulo
development in African American women, but recent studies have Ramirez, R.F. Lopez Villanueva, I.G. Quintal Ortiz, E.P. Rosado Paredes, Serological
and molecular analysis of parvovirus B19 infection in Mayan women with systemic
shown that topical H. pylori gastritis can induce systemic inflammatory lupus erythematosus in Mexico, Colomb. Med. (Cali) 48 (3) (2017) 105–112.
response and promote lupus progression in some lupus patients [16] D.S. Pisetsky, Role of Epstein-Barr virus infection in SLE: gene-environment inter-
[57–59]. Therefore, the existing evidence still does not fully support the actions at the molecular level, Ann. Rheum. Dis. 77 (9) (2018) 1249–1250.
[17] N. Tugnet, P. Rylance, D. Roden, M. Trela, P. Nelson, Human Endogenous
role of H. pylori in the development of SLE. Epidemiological studies
Retroviruses (HERVs) and Autoimmune Rheumatic Disease: Is There a Link? Open
from different regions have also shown that hepatitis B virus (HBV) Rheumatol. J. 7 (2013) 13–21.
infection rate in patients with SLE is lower than that in non-SLE control [18] L. Baudino, K. Yoshinobu, N. Morito, M.L. Santiago-Raber, S. Izui, Role of en-
group, suggesting that HBV may have protective effects on SLE, but the dogenous retroviruses in murine SLE, Autoimmun. Rev. 10 (1) (2010) 27–34.
[19] M. Pavlovic, A. Kats, M. Cavallo, Y. Shoenfeld, Clinical and molecular evidence for
specific mechanism has not been studied in depth [60,61]. association of SLE with parvovirus B19, LUPUS 19 (7) (2010) 783–792.
In summary, since there are still many unknowns about the pro- [20] T.L. Skare, J.S. Dagostini, P.I. Zanardi, R.M. Nisihara, Infections and systemic lupus
tective pathogens, proving their protective effects remains challenging erythematosus, Einstein (Sao Paulo, Brazil) 14 (1) (2016) 47–51.
[21] F.B. Vincent, P. Bourke, E.F. Morand, F. Mackay, D. Bossingham, Focus on systemic
and need working hard on its in-depth exploration. As research of this lupus erythematosus in indigenous Australians: towards a better understanding of
nature continues, novel insights into the protective effects of infectious autoimmune diseases, Intern. Med. J. 43 (3) (2013) 227–234.
agents on SLE may be gained. [22] A.E. Mohamed, A.M. Hasen, G.F. Mohammed, N.N. Elmaraghy, Real-Time PCR of
cytomegalovirus and Epstein-Barr virus in adult Egyptian patients with systemic
lupus erythematosus, Int. J. Rheum. Dis. 18 (4) (2015) 452–458.
4. Conclusion and future perspective [23] P. Gergely Jr., R. Pullmann, C. Stancato, L. Otvos Jr., A. Koncz, A. Blazsek, G. Poor,
K.E. Brown, P.E. Phillips, A. Perl, Increased prevalence of transfusion-transmitted
virus and cross-reactivity with immunodominant epitopes of the HRES-1/p28 en-
Identifying the infections involved in the development of SLE could dogenous retroviral autoantigen in patients with systemic lupus erythematosus,
provide insight into practicable measures to reduce trigger exposure Clin. Immunol. 116 (2) (2005) 124–134.
and prevent flares as well as the design of treatment strategies for high- [24] G.M. Deng, G.C. Tsokos, Cholera toxin B accelerates disease progression in lupus-
prone mice by promoting lipid raft aggregation, J. Immunol. 181 (6) (2008)
risk groups, all of which will benefit patients with SLE. Recent dis-
4019–4026.
coveries related to the mechanisms of infection in SLE implicated mo- [25] M.N. Seyyed Mousavi, B. Mehramuz, J. Sadeghi, N. Alizadeh, M.A. Oskouee,
lecular mimicry, epitope spreading, superantigen production, bystander H.S. Kafil, The pathogenesis of Staphylococcus aureus in autoimmune diseases,
activation, persistent viral infection, altered apoptosis, clearance defi- Microb. Pathog. 111 (2017) 503–507.
[26] O. Laitinen, A. Vaheri, Very high measles and rubella virus antibody titres asso-
ciency, and epigenetic alterations (e.g., DNA methylation and ciated with hepatitis, systemic lupus erythematosus, and infectious mononucleosis,
microRNAs), etc. However, the relationship between infection and SLE Lancet 1 (7850) (1974) 194–197.
is multifaceted and multidirectional, including causative and/or pro- [27] N. Mahroum, A. Hejly, S. Tiosano, O. Gendelman, D. Comaneshter, A.D. Cohen,
H. Amital, Chronic hepatitis C viral infection among SLE patients: the significance
tective associations, which warrant further investigation in the future. of coexistence, Immunol. Res. 65 (2) (2017) 477–481.
[28] R. Root-Bernstein, D. Fairweather, Complexities in the relationship between in-
Funding acknowledgment fection and autoimmunity, Curr. Allergy Asthma Rep. 14 (1) (2014) 407.
[29] M.B. Oldstone, Molecular mimicry: its evolution from concept to mechanism as a
cause of autoimmune diseases, Monoclon. Antib. Immunodiagn. Immunother. 33
The work was supported by Natural Science Foundation of (3) (2014) 158–165.
Guangdong Province, China (grant number 2015A030313526), Medical [30] M.F. Cusick, J.E. Libbey, R.S. Fujinami, Molecular mimicry as a mechanism of
autoimmune disease, Clin. Rev. Allergy Immunol. 42 (1) (2012) 102–111.
Scientific Research Foundation of Guangdong Province, China (grant
[31] R.S. Fujinami, M.G. von Herrath, U. Christen, J.L. Whitton, Molecular mimicry,
number A2014481). bystander activation, or viral persistence: infections and autoimmune disease, Clin.
Microbiol. Rev. 19 (1) (2006) 80–94.
[32] C. Cornaby, L. Gibbons, V. Mayhew, C.S. Sloan, A. Welling, B.D. Poole, B cell epi-
References
tope spreading: mechanisms and contribution to autoimmune diseases, Immunol.
Lett. 163 (1) (2015) 56–68.
[1] A. Kaul, C. Gordon, M.K. Crow, Z. Touma, M.B. Urowitz, R. van Vollenhoven, [33] U.S. Deshmukh, H. Bagavant, J. Lewis, F. Gaskin, S.M. Fu, Epitope spreading within
G. Ruiz-Irastorza, G. Hughes, Systemic lupus erythematosus, Nat. Rev. Dis. Primers lupus-associated ribonucleoprotein antigens, Clin. Immunol. 117 (2) (2005)
2 (2016) 16039. 112–120.
[2] J.Y. Jung, C.H. Suh, Infection in systemic lupus erythematosus, similarities, and [34] F. Monneaux, S. Muller, Epitope spreading in systemic lupus erythematosus: iden-
differences with lupus flare, Korean J. Intern. Med. 32 (3) (2017) 429–438. tification of triggering peptide sequences, Arthritis Rheum. 46 (6) (2002)
[3] D. Rigante, M.B. Mazzoni, S. Esposito, The cryptic interplay between systemic lupus 1430–1438.
erythematosus and infections, Autoimmun. Rev. 13 (2) (2014) 96–102. [35] L. Campisi, G. Barbet, Y. Ding, E. Esplugues, R.A. Flavell, J.M. Blander, Apoptosis in
[4] S. Praprotnik, S. Sodin-Semrl, M. Tomsic, Y. Shoenfeld, The curiously suspicious: response to microbial infection induces autoreactive TH17 cells, Nat. Immunol. 17
infectious disease may ameliorate an ongoing autoimmune destruction in systemic (9) (2016) 1084–1092.
lupus erythematosus patients, J. Autoimmun. 30 (1–2) (2008) 37–41. [36] Y.E. Guo, J.A. Steitz, Virus meets host microRNA: the destroyer, the booster, the
[5] T. Hod, G. Zandman-Goddard, P. Langevitz, H. Rudnic, Z. Grossman, P. Rotman- hijacker, Mol. Cell. Biol. 34 (20) (2014) 3780–3787.
Pikielny, Y. Levy, Does parvovirus infection have a role in systemic lupus er- [37] H. Long, X. Wang, Y. Chen, L. Wang, M. Zhao, Q. Lu, Dysregulation of microRNAs in
ythematosus? Immunol. Res. 65 (2) (2017) 447–453. autoimmune diseases: pathogenesis, biomarkers and potential therapeutic targets,
[6] D. Rigante, S. Esposito, Infections and systemic lupus erythematosus: Binding or Cancer Lett. 428 (2018) 90–103.
sparring partners? Int. J. Mol. Sci. 16 (8) (2015) 17331–17343. [38] N. Shen, D. Liang, Y. Tang, N. de Vries, P.P. Tak, MicroRNAs–novel regulators of
[7] P. Nelson, P. Rylance, D. Roden, M. Trela, N. Tugnet, Viruses as potential patho- systemic lupus erythematosus pathogenesis, Nat. Rev. Rheumatol. 8 (12) (2012)
genic agents in systemic lupus erythematosus, LUPUS 23 (6) (2014) 596–605. 701–709.
[8] D. Germolec, D.H. Kono, J.C. Pfau, K.M. Pollard, Animal models used to examine [39] K.M. Pauley, S. Cha, E.K. Chan, MicroRNA in autoimmunity and autoimmune dis-
the role of the environment in the development of autoimmune disease: findings eases, J. Autoimmun. 32 (3–4) (2009) 189–194.
from an NIEHS Expert Panel Workshop, J. Autoimmun. 39 (4) (2012) 285–293. [40] G. Amarilyo, A. La Cava, miRNA in systemic lupus erythematosus, Clin. Immunol.
[9] S. Kivity, N. Agmon-Levin, M. Blank, Y. Shoenfeld, Infections and autoimmunity– 144 (1) (2012) 26–31.
friends or foes? Trends Immunol. 30 (8) (2009) 409–414. [41] R. Baccala, K. Hoebe, D.H. Kono, B. Beutler, A.N. Theofilopoulos, TLR-dependent
[10] P. Sfriso, A. Ghirardello, C. Botsios, M. Tonon, M. Zen, N. Bassi, F. Bassetto, and TLR-independent pathways of type I interferon induction in systemic auto-
A. Doria, Infections and autoimmunity: the multifaceted relationship, J. Leukoc. immunity, Nat. Med. 13 (5) (2007) 543–551.
Biol. 87 (3) (2010) 385–395. [42] N.S. Rasmussen, C.T. Nielsen, G. Houen, S. Jacobsen, Humoral markers of active
[11] P.R. Nielsen, T.W. Kragstrup, B.W. Deleuran, M.E. Benros, Infections as risk factor Epstein-Barr virus infection associate with anti-extractable nuclear antigen auto-
for autoimmune diseases - A nationwide study, J. Autoimmun. 74 (2016) 176–181. antibodies and plasma galectin-3 binding protein in systemic lupus erythematosus,

4
Q. Pan, et al. Biomedicine & Pharmacotherapy 117 (2019) 109122

LUPUS 25 (14) (2016) 1567–1576. A. Yano, Toxoplasma gondii infection inhibits the development of lupus-like syn-
[43] O. Shamriz, Y. Shoenfeld, Infections: a double-edge sword in autoimmunity, Curr. drome in autoimmune (New Zealand Black x New Zealand White) F1 mice, Int.
Opin. Rheumatol. 30 (4) (2018) 365–372. Immunol. 16 (7) (2004) 937–946.
[44] M. Versini, P.Y. Jeandel, T. Bashi, G. Bizzaro, M. Blank, Y. Shoenfeld, Unraveling [53] I. Noel, A.H. Balfour, M.H. Wilcox, Toxoplasma infection and systemic lupus er-
the Hygiene Hypothesis of helminthes and autoimmunity: origins, pathophysiology, ythematosus: analysis of the serological response by immunoblotting, J. Clin.
and clinical applications, BMC Med. 13 (2015) 81. Pathol. 46 (7) (1993) 628–632.
[45] J.F. Bach, Infections and autoimmune diseases, J. Autoimmun. 25 (Suppl) (2005) [54] D.T. Rodgers, M.A. McGrath, M.A. Pineda, L. Al-Riyami, J. Rzepecka, F. Lumb,
74–80. W. Harnett, M.M. Harnett, The parasitic worm product ES-62 targets myeloid dif-
[46] M.B. Oldstone, F.J. Dixon, Inhibition of antibodies to nuclear antigen and to DNA in ferentiation factor 88-dependent effector mechanisms to suppress antinuclear an-
New Zealand mice infected with lactate dehydrogenase virus, Science 175 (4023) tibody production and proteinuria in MRL/lpr mice, Arthritis Rheumatol. 67 (4)
(1972) 784–786. (2015) 1023–1035.
[47] T. Hayashi, I. Mori, H. Yamamoto, Lactic dehydrogenase virus infection prevents [55] K. Miyake, K. Adachi, M. Watanabe, Y. Sasatomi, S. Ogahara, Y. Abe, K. Ito,
development of anti-nuclear antibody in (NZB x NZW)F1 mice; role of pros- Y.K. Dan Justin, T. Saito, H. Nakashima, S. Hamano, Parasites alter the pathological
taglandin E2 and macrophage Ia antigen expression, Int. J. Exp. Pathol. 73 (5) phenotype of lupus nephritis, Autoimmunity 47 (8) (2014) 538–547.
(1992) 593–601. [56] H. Neuman, H. Mor, T. Bashi, O. Givol, A. Watad, A. Shemer, A. Volkov, I. Barshack,
[48] G. Badr, A. Sayed, M.A. Abdel-Maksoud, A.O. Mohamed, A. El-Amir, F.A. Abdel- M. Fridkin, M. Blank, Y. Shoenfeld, O. Koren, Helminth-based product and the
Ghaffar, S. Al-Quraishy, M.H. Mahmoud, Infection of female BWF1 lupus mice with microbiome of mice with lupus, mSystems 4 (1) (2019).
malaria parasite attenuates B cell autoreactivity by modulating the CXCL12/CXCR4 [57] A.H. Sawalha, W.R. Schmid, S.R. Binder, D.K. Bacino, J.B. Harley, Association be-
Axis and its downstream signals PI3K/AKT, NFkappaB and ERK, PLoS One 10 (4) tween systemic lupus erythematosus and Helicobacter pylori seronegativity, J.
(2015) e0125340. Rheumatol. 31 (8) (2004) 1546–1550.
[49] S. Al-Quraishy, M.A. Abdel-Maksoud, A. El-Amir, F.A. Abdel-Ghaffar, G. Badr, [58] D.S. Smyk, A.L. Koutsoumpas, M.G. Mytilinaiou, E.I. Rigopoulou, L.I. Sakkas,
Malarial infection of female BWF1 lupus mice alters the redox state in kidney and D.P. Bogdanos, Helicobacter pylori and autoimmune disease: cause or bystander,
liver tissues and confers protection against lupus nephritis, Oxid. Med. Cell. Longev. World J. Gastroenterol. 20 (3) (2014) 613–629.
2013 (2013) 156562. [59] S. Surawut, W. Panpetch, J. Makjaroen, P. Tangtanatakul, A. Thim-Uam,
[50] L.C. Willcocks, E.J. Carr, H.A. Niederer, T.F. Rayner, T.N. Williams, W. Yang, J. Wongphoom, S. Tumwasorn, A. Leelahavanichkul, Helicobacter pylori Infection
J.A. Scott, B.C. Urban, N. Peshu, T.J. Vyse, Y.L. Lau, P.A. Lyons, K.G. Smith, A Increased Anti-dsDNA and Enhanced Lupus Severity in Symptomatic FcgammaRIIb-
defunctioning polymorphism in FCGR2B is associated with protection against ma- Deficient Lupus Mice, Front. Microbiol. 9 (2018) 1488.
laria but susceptibility to systemic lupus erythematosus, Proc. Natl. Acad. Sci. U. S. [60] M. Ram, J.M. Anaya, O. Barzilai, D. Izhaky, B.S. Porat Katz, M. Blank, Y. Shoenfeld,
A. 107 (17) (2010) 7881–7885. The putative protective role of hepatitis B virus (HBV) infection from autoimmune
[51] M.A. Abdel-Maksoud, F.A. Abdel-Ghaffar, A. El-Amir, S. Al-Quraishy, G. Badr, disorders, Autoimmun. Rev. 7 (8) (2008) 621–625.
Infection with Plasmodium chabaudi diminishes plasma immune complexes and [61] J. Zhao, M. Qiu, M. Li, C. Lu, J. Gu, Low prevalence of hepatitis B virus infection in
ameliorates the histopathological alterations in different organs of female BWF1 patients with systemic lupus erythematosus in southern China, Rheumatol. Int. 30
lupus mice, Eur. Rev. Med. Pharmacol. Sci. 20 (4) (2016) 733–744. (12) (2010) 1565–1570.
[52] M. Chen, F. Aosai, K. Norose, H.S. Mun, H. Ishikura, S. Hirose, L.X. Piao, H. Fang,

S-ar putea să vă placă și