ArticlePDF AvailableLiterature Review

Environment and systemic lupus erythematosus

Authors:

Abstract

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a wide range of clinical manifestations and a relapsing-remitting course. SLE pathogenesis is the result of complex interactions between ethnic, genetic, epigenetic, immunoregulatory, hormonal and environmental factors, and several aspects of these multifactorial connections are still unclear. Overall, for the disease development, an environmental trigger may induce immunological dysfunction in genetically predisposed individuals. This review aims to summarise the most relevant data on the impact of environmental factors on the incidence of SLE and on disease activity and damage in patients with an established diagnosis of SLE.
1104 Clinical and Experimental Rheumatology 2024
1Rheumatology Unit, Department of
Clinical and Experimental Medicine,
University of Pisa;
2Department of Medical Biotechnologies,
University of Siena, Italy.
Chiara Cardelli, MD*
Dina Zucchi, MD*
Elena Elefante, MD PhD
Viola Signorini, MD
Marina Menchini
Chiara Stagnaro, MD
Marta Mosca, MD, PhD
Chiara Tani, MD, PhD
*Contributed equally.
Please address correspondence to:
Chiara Tani
U.O. di Reumatologia,
Dipartimento di Medicina
Clinica e Sperimentale,
Università di Pisa,
via Roma 67,
56126, Pisa, Italy
E-mail: chiara.tani@unipi.it
Received on February 29, 2024; accepted
in revised form on April 22, 2024.
Clin Exp Rheumatol 2024; 42: 1104-1114.
© Copyright CliniCal and
ExpErimEntal rhEumatology 2024.
Key words: systemic lupus
erythematosus, environment,
smoking, infections
Competing interests: none declared.
ABSTRACT
Systemic lupus erythematosus (SLE) is
a chronic autoimmune disease with a
wide range of clinical manifestations
and a relapsing-remitting course. SLE
pathogenesis is the result of com plex in-
teractions between ethnic, genetic, epi-
genetic, immunoregulatory, hor monal
and environmental factors, and several
aspects of these multifactori al connec-
tions are still unclear. Overall, for the
disease development, an environmen-
tal trigger may induce immunological
dysfunction in genetically predisposed
individuals. This review aims to sum-
marise the most relevant data on the
impact of environmental factors on the
incidence of SLE and on disease activ-
ity and damage in patients with an es-
tablished diagnosis of SLE.
Introduction
Systemic lupus erythematosus (SLE)
is a complex, chronic autoimmune
disease with a broad spectrum of clini-
cal manifestations, that mainly affects
young women (1). The aetiology of
SLE is multifactorial, encompassing
genetic, epigenetic and environmental
factors. The role of genetics in SLE
is suggested by mouse models, famil-
ial linkage studies and concordance of
the disease in twins (2-4). However, a
recent metastudy on SLE concordance
in twins revealed high rates of discord-
ance (up to 75% in monozygotic and to
95% in dizygotic twins) (5), conrming
the key role of environmental factors in
SLE susceptibility. This narrative re-
view focuses on the main environmen-
tal factors involved in the development
and course of SLE.
Environmental factors
involved in SLE
Ultraviolet (UV) light
Among environmental factors, UV ra-
diation exposure is recognised as a risk
factor for the development and reacti-
vation of SLE. Whereas UVC, with its
short wavelength, does not penetrate
the atmospheric ozone layer, UVA
(320–400 nm) and UVB (280–320 nm)
are environmentally relevant.
UVA penetrates the deeper dermis and
is weakly absorbed by cells: its role in
SLE pathogenesis is still unclear. In-
deed, some studies have found that UVA
induces a reduction in disease activity
in SLE patients and in patients with
cutaneous lupus erythematosus (CLE)
(6); conversely, other studies have
shown that UVA exposure induces lu-
pus skin lesions (7, 8). In contrast, UVB
is unable to penetrate the epidermal lay-
er and is more absorbed by DNA and
cellular proteins; therefore, UVB is the
most effective inducer of keratinocytes
apoptosis. Several phototesting studies
demonstrated as UVB and/or UVA can
induce skin lesions in patients with dif-
ferent subtypes of LE (9), proving that
UV radiation exposure may induce or
exacerbate skin manifestations, while
their role as risk factors for the devel-
opment of SLE remains unclear. Only a
few case-control studies have been able
to examine UV radiation exposure and
risk of SLE (10, 11).
As we know, UV radiation causes DNA
damage and apoptosis of keratinocytes;
apoptotic keratinocytes, known as “sun-
burn cells”, are characterised by eosin-
ophilic cytoplasm and pyknotic nuclei
and can be found as early as eight hours
after UV exposure (12). UVA induces
keratinocytes apoptosis mainly through
mitochondrial oxidative damage lead-
ing to increased production of reac-
tive oxygen species (13). Conversely,
UVB induces apoptosis through DNA
damage with strand breaks and pyrimi-
dine dimers formation (14). Kuhn et
al. showed that, after a single UV light
exposure, apoptotic cells accumulate
in the skin of patients with CLE, com-
Review
Environment and systemic lupus erythematosus
C. Cardelli1,2, D. Zucchi1,2, E. Elefante1, V. Signorini1, M. Menchini1,
C. Stagnaro1, M. Mosca1, C. Tani1
1105
Clinical and Experimental Rheumatology 2024
SLE and environment / C. Cardelli et al.
pared to controls, suggesting impaired
or delayed clearance (15).
Another mechanism through which UV
induces skin injury is by increasing au-
toantigens production in the epidermis,
such as Ro52, IF116, Sm, RNP, Ku and
ribosomal-P (16). Moreover, UV light
increases the expression of chemokines
and induces keratinocytes and immune
cells to release pro-inammatory cy-
tokines such as interferon (IFN) and
tumour necrosis factor alpha (TNFα).
Furthermore, even in healthy skin, UV
induces the upregulation of proteins
that act as autoantigens in SLE patients,
suggesting that chronic or intense UV
exposure can increase the tendency to
autoantigen exposure (16).
Silica
Of the chemical agents that act as trig-
gers for SLE development, silica has
been the most studied. Silica is com-
monly found in nature as quartz; expo-
sure to respirable crystalline silica (<10
μm) occurs most often in occupational
settings.
Several epidemiological studies have
shown that silica exposure is related to
the development of autoimmune diseas-
es such as rheumatoid arthritis, ANCA-
associated vasculitis, systemic sclerosis
and SLE (17). Both Parks et al. (18) and
Morotti et al. (19) reported that occupa-
tional silica exposure may be associated
with SLE, particularly in patients with
silicosis. The dose-response has been
associated with increasing intensity or
duration of exposure (18, 20); these
ndings are conrmed by other popu-
lation-based studies (21, 22). Interest-
ingly, patients with SLE and silicosis
are predominantly middle-aged and
elderly men, in which the age of SLE
onset has been found to be between 40
and 63 years (23).
Smoking
Cigarette smoking has also been associ-
ated with the development and progres-
sion of several autoimmune diseases,
including SLE (24).
The pathogenetic link between smok-
ing and SLE development remains de-
bated; potential mechanisms include
oxidative stress, increased cytokine-
driven systemic inammation and im-
paired T- and B-cell function, epige-
netic modications. In particular, ciga-
rette smoke increases CD95 expression
on B and T lymphocyte cell surfaces
(25); CD95 plays a central role in im-
mune homeostasis, being an essential
membrane receptor for transmitting
apoptosis signals in lymphocytes; as a
consequence, its increased expression
could be responsible for increased ap-
optosis leading to an overburdening of
apoptotic debris clearance mechanisms,
a major driver of autoimmunity in SLE.
Other potential cellular mechanisms in-
clude reduction and functional impair-
ment of natural killer cells and impair-
ment of Th17 and Th22 cells functions.
The causal association between smok-
ing and disease onset risk, antibodies
prole, organ damage and treatment ef-
cacy in SLE patients has been the fo-
cus of extensive investigations in recent
years.
The relationship between smoking ex-
posure and the risk of developing the
disease is still debated; the presence
of possible confounders and the low
incidence of the disease in the general
population would require big cohorts
to be followed and analysed to draw
sound conclusions; to overcome these
difculties, several metanalysis have
been published in the last years.
In 2019, a metanalysis of 9 case-control
studies by Parisis et al. (26) found a sig-
nicantly increased risk of SLE in cur-
rent-smokers compared to never-smok-
ers and in ever- versus never-smokers,
while former-smokers were not at in-
creased risk of SLE. Data on passive
smoking remain scarce and controver-
sial. In the same study, no over-risk of
anti-dsDNA, anti-Sm or anti-SSA posi-
tivity was observed according to smok-
ing status.
More recently, Chua et al. (27) pub-
lished a metanalysis using a Bayesian
approach, including 12 studies for a total
of 3234 individuals who developed SLE
and 288336 control subjects; the study
conrmed the association between cur-
rent smoking and the risk to develop
SLE; interestingly, in this meta-regres-
sion analysis, publication time, age and
gender did not have a signicant effect
on the disease occurrence risk.
Moreover, in 2006, Simard et al. (28)
investigated whether early-life expo-
sure to cigarette smoke was associated
with subsequent SLE development; to
this end, they examined approximately
18000 adults free of SLE at baseline
who provided information on perinatal
exposures; in total, 236 incident SLE
cases were identied, but maternal cig-
arette smoking did not increase the risk
of SLE nor did paternal smoking during
participants’ childhood.
The association between tobacco smok-
ing and morbidity and organ damage
has been poorly studied.
In the 1990s, Ward and Studenski’s
study showed a signicant association
between smoking and the development
of end-stage renal disease (29).
More recently, Montes et al. (30) inves-
tigated the chronic damage accrual ex-
pressed by the SLICC/DI in a cohort of
SLE patients exposed or not to tobacco
smoke. They found that being “never
exposed” to smoking confers a 22%
relative risk reduction of progressing to
an SDI score >0 compared to an “ever
exposed” status.
In 2022, a systematic review was pub-
lished to summarise the available evi-
dence on the effects of tobacco smoking
on developing a cardiovascular disease
(CVD) in SLE patients (31). The authors
included a total of 10 studies on 6984
participants. Compared to never-smok-
ers, the risk of developing CVD in cur-
rent-smokers was signicantly higher.
In the last years, growing evidence has
suggested that tobacco smoking could
also have an impact in the effectiveness
of therapies in SLE patients.
The most recent data on this topic were
reviewed by Parisis et al. (26); in 11
observational studies of CLE or SLE
patients, tobacco smoking signicantly
reduced the therapeutic effectiveness
of hydroxychloroquine (HCQ) in cu-
taneous lesions. However, some stud-
ies evaluated the correlation between
plasmatic HCQ levels and tobacco
smoking, and no signicant differences
were found between smokers and non-
smokers (32-35).
One single study evaluated the effect
of tobacco smoking on the clinical re-
sponse to belimumab (36). In a pro-
spective follow-up study that included
active SLE patients starting belimum-
1106 Clinical and Experimental Rheumatology 2024
SLE and environment / C. Cardelli et al.
ab, the authors observed that current
smokers showed decreased probability
and prolonged time to attain clinical re-
sponse compared to non-smokers. The
same group also demonstrated that cur-
rent or former smokers showed a higher
probability of unchanged/worsened
mucocutaneous manifestations com-
pared to never-smokers, while no im-
pact of smoking on belimumab efcacy
in articular SLE was reported (37).
In conclusion, available evidence sug-
gests that tobacco smoking exposure
is associated with SLE disease risk,
its clinical progression/damage and re-
sponse to treatment. However, it is im-
portant to note that the available studies
refer to a wide range of geographical lo-
cations, vary widely in smoking expo-
sure collection methods and SLE phe-
notype; thus, large prospective studies
could be encouraged to better explore
the complex interaction between the
disease and tobacco smoke. Moreover,
there is a large and intriguing area relat-
ed to the possible impact of heated to-
bacco products and electronic cigarette
smoking, for which no studies exist yet.
Infections
Exogenous microbial agents, such as
bacteria or viruses, interact with and
sometimes overcome the human im-
mune system, potentially leading to
autoimmunity. Among microbiological
agents, viruses in particular have been
implicated as potential triggers of au-
toimmune conditions (38, 39). Overall,
viral infections could interact with in-
nate and acquired immune responses
and lead to an aberrant response or lack
of immune control, facilitating the de-
velopment of SLE and other autoim-
mune diseases. From an aetiopathoge-
netic point of view, the mechanism of
molecular mimicry by specic micro-
bial agents might play a role in the de-
velopment of SLE.
Several studies on the association with
Epstein-Barr virus (EBV) have been
conducted over the years, with con-
icting results (40). The main ndings
on the positive association come from
studies focusing on seroconversion
rates in SLE patients.
Some papers reported an association
between SLE and not only seroconver-
sion but also the presence of EBV ge-
nome in peripheral blood lymphocytes
in children and teenagers (41, 42). Con-
cerning adults, a study involving 196
SLE patients tested for previous infec-
tions demonstrated that all but one had
been exposed to EBV, whereas no dif-
ferences were observed between SLE
patients and controls in seroconversion
against CMV, HSV-2, or VZV, support-
ing the possible role of EBV in the de-
velopment of SLE (43). Similar results
emerged from another case-control
study, where a signicant difference
between SLE patients and controls was
also found in EBV viral load, with the
EBV-DNA positivity rate tending to de-
cline with age in the latter but not in the
former (44).
In 2014, Hanlon et al. published a
metanalysis of 25 case-control studies
to determine whether prior EBV in-
fection occurs more frequently in SLE
patients compared to matched controls:
although publication bias cannot be
excluded, the metanalysis supports the
hypothesis that EBV infection predis-
poses to SLE development (45). Indeed,
signicantly higher seroprevalence of
anti-viral capsid antigen (VCA) IgG,
anti-early antigen (EA)-D IgG and anti-
VCA IgA emerged in cases compared
with controls.
A seroprevalence of almost 100% sug-
gests a signicant, although not entire-
ly clear, role of EBV infection in SLE
pathogenesis. The likelihood that EBV
infection causes SLE in some patients
is supported by the possible molecular
mimicry of the EBV peptide PPPGRRP
by the human spliceosome peptide
SmB’/B’s PPPGMRPP (46). Moreover,
the presence of a dominant epitope in the
C-terminal region of SmD, which ex-
hibits a striking resemblance to a region
of the EBV nuclear antigen coding for
the EBNA1 protein (47), has previously
been demonstrated, thus raising hypoth-
eses on the possible role of the immune
response to EBV in the induction of
anti-SmD antibodies (48). Besides, the
cross-reactivity of spontaneously devel-
oped anti-ribosomal P protein antibod-
ies with the B/B´ and D constituents of
the Sm complex has been proven (49).
Recently, potential underlying molecu-
lar mechanisms have been investigated
by genetic and transcriptomic analy-
ses: lower latent EBV markers and
higher lytic EBV markers were found
in lymphoblastoid cell lines (LCLs) of
SLE patients compared to healthy indi-
viduals, suggesting an EBV nuclear an-
tigen 2 (EBNA2)-mediated molecular
pathway in which SLE risk loci may in-
crease the tendency of LCLs to switch
to the lytic phase (50).
A case-control study published a few
months ago, with data on serological,
molecular and sequence markers of
EBV infection in SLE patients, demon-
strated a 24-fold higher chance of hav-
ing SLE in the presence of anti-EBV-
EA-D IgG antibodies (51). Further-
more, higher titres of anti-EBV-EA-D
IgG were identied as independent
factor associated with lymphopenia and
SLE haematological manifestations,
while a higher titre of anti-VCA IgG as
an independent factor associated with
alopecia in SLE.
Regarding other infections, the Caro-
lina Lupus Study showed that the risk
of SLE increased with a history of shin-
gles and with frequent colds in the three
years prior to diagnosis (52). A recent
systematic review suggested a potential
association between COVID-19 and
new onset of systemic autoimmune dis-
eases, mostly myositis and SLE (53),
although given the few cases and the
short follow-up period no rm conclu-
sions can be drawn to date.
It should be noted that infections might
not only be associated with the devel-
opment of SLE, but also act as triggers
for disease ares over time. Among the
viral infections of most interest, inu-
enza was found to be a risk factor for a
SLE are according to a recent study,
with an incidence ratio for ares of
25.75 during the risk interval compared
to the control interval (54).
Even more, the possibility that the in-
fection itself acts as a mimicker for a
SLE are (55) should not be under-
estimated. SLE patients, in fact, are a
population at increased infection risk
due to both the aberrant immune system
and glucocorticoids and other immuno-
suppressants used to treat the disease.
Although prognosis has considerably
improved over the years, infections still
remain a major cause of hospitalisation
1107
Clinical and Experimental Rheumatology 2024
SLE and environment / C. Cardelli et al.
and mortality in patients with SLE, es-
pecially in the early phase of disease
(56-58).
Although controversial, the crucial in-
terplay between SLE and infections
must draw attention to the importance
of preventive measures such as vacci-
nation.
Exogenous hormones
Sex hormones may play a role in patho-
genesis, clinical features and manage-
ment of SLE.
A correlation between incident SLE and
use of combined oral contraceptives
was described by Bernier et al. (59) in
a large ten-year cohort. Data from this
study suggested a possible acute ef-
fect of sexual hormones in susceptible
women, as an increased risk of SLE
was observed particularly in women
who had recently started hormone ther-
apy and increased with the dose of ethi-
nylestradiol.
A unique population is represented by
transgender women receiving female
sex hormones before or after sexual
reassignment surgery. Few cases of
transgender women who have been
diagnosed with lupus following use of
exogenous female sex hormones were
reported (60-64). None of them had a
previous lupus diagnosis, and renal in-
volvement was described in three cases
(60, 63, 64). Despite the low number,
these data focused on the potential rela-
tionship between female sex hormones
and SLE in susceptible individuals.
It is known that SLE predominantly af-
fects women, and patients may require
hormone therapy during their life. Sev-
eral clinical trials have investigated the
effects of hormone therapies in SLE pa-
tients, with a particular attention to oral
contraceptives and menopause hormo-
nal therapy.
In a randomised controlled trial (RCT)
on 162 SLE women, patients were
randomised to combined oral contra-
ceptive, progestin-only pill or copper
intrauterine device (65). The results
demonstrated the absence of inuence
of these contraceptive methods on dis-
ease activity and adverse events in this
SLE subgroup. In another RTC on 183
SLE women, the authors observed that
combined hormonal oral contracep-
tives did not increase the risk of disease
are compared to placebo (66).
In both studies, all patients had stable
disease and those with medium/high
titre of antiphospholipid antibodies
(aPL) were excluded, as well as pa-
tients with history of thrombosis. Of
note, in the rst study (65) thrombosis
occurred in four patients (two for each
group receiving hormones), and all
four patients had low aPL titres.
Conversely, studies on effects of com-
bined contraceptives on aPL-positive
patients showed an increased risk of
thrombosis. A large multicentre popu-
lation-based case-control study, named
RATIO (Risk of Arterial Thrombo-
sis In relation to Oral contraceptives)
(67), showed an increased risk of arte-
rial events in combined users compared
with non-users. In particular, lupus an-
ticoagulant (LAC) resulted a major risk
factor for arterial thrombotic events,
and the risk of myocardial infarction
and ischemic stroke increased further in
women who used combined oral con-
traceptives.
As a matter of the fact, scientic soci-
eties (EULAR and ACR) support the
use of combined hormonal contracep-
tives in patients with stable/inactive
SLE and negative aPL, while in women
with positive aPL contraception with
combined hormones should be discour-
aged and progesterone only contracep-
tion carefully weighed against the risk
of thrombosis (68, 69). Moreover, ACR
conditionally recommends against the
use of transdermal oestrogen-progestin
patch, since it results in greater estro-
gen exposure than oral or transvaginal
methods (70).
In SLE patients with menopausal symp-
toms, hormone replacement therapy
(HRT) seems to improve vasomotor
symptoms (71), and several studies
have investigated HRT impact in SLE
women.
In RCTs no signicant increase in se-
vere disease ares (72, 73) or cardio-
vascular (CV) events (74) was reported
in SLE patients receiving HRT com-
pared to controls. However, Buyon et
al. observed a small increase in mild-
moderate ares in SLE HRT group with
respect to placebo (1.14 vs. 0.86 ares/
person-year for HRT and placebo, re-
spectively) (73). We must acknowledge
that women with high disease activity,
previous thrombosis or aPL-positivity
were excluded from most studies.
As stated in the previously mentioned
EULAR and ACR recommendations
(68, 69), HRT should be reserved for
the management of severe and disa-
bling menopausal vasomotor symp-
toms, preferably in SLE women with
stable/inactive disease and negative
aPL, while in patients with positive aPL
the use of HRT should be discouraged
or carefully weighed against thrombot-
ic and CV risks.
Diet
Some macronutrients and micronutrients
seem to have antioxidant, anti-inam-
matory and immunomodulatory effects,
and over the years several studies have
investigated their impact on rheumatic
and musculoskeletal diseases (RMDs)
and SLE (75). However, we must con-
sider that most of the studies are pilot or
small samples studies, and thus reliable
conclusions cannot be drawn.
Overall, a healthy, balanced diet is in-
tegral to lifestyle improvement for peo-
ple with RMDs (76). However, patients
should be informed that consuming spe-
cic food types is unlikely to have large
benets for RMD outcomes.
CV risk is known to be higher in SLE
patients, and mediterranean diet has
been reported to down-regulate inam-
matory biomarkers related to atherogen-
esis in subjects at high CV risk (77). A
cross-sectional study on 280 SLE pa-
tients showed that greater adherence to
the mediterranean diet correlated with
benecial effect on disease activity and
CV risk (78).
In overweight women with corticoster-
oid-dependent SLE, Davies et al. (79)
analysed the inuence of low glycae-
mic index diet and calorie restricted
diet in a 6-week controlled trial. Both
diets led to signicant weight loss, im-
proved waist and hip measurements and
reduced fatigue. In addition, no disease
ares were observed during the study
period, conrming the safety of this di-
etary regimen.
Omega-3 fatty acids are involved in
serotonin synthesis (80), and effects of
omega-3 supplementation in SLE have
1108 Clinical and Experimental Rheumatology 2024
SLE and environment / C. Cardelli et al.
been investigated in few studies, with
controversial results. Some papers re-
ported benets from omega-3 supple-
mentation in SLE patients, while other
studies did not conrm these results, as
shown in Table I.
Vitamin D is often considered an anti-
inammatory agent, and has effects on
proliferation, apoptosis and function of
immune system cells that are involved
in SLE pathophysiology (87). Vitamin
D deciency is common in SLE, since
the use of medications such as glucocor-
ticoids and renal failure can alter its me-
tabolism (87). In addition, all SLE pa-
tients are advised to avoid sunlight, and
vitamin D deciency is also reported as
a potential risk factor for SLE (88).
In clinical setting, adding vitamin D to
the traditional pharmacological regi-
men in SLE has been found benecial
in some studies, whereas other studies
failed to replicate these results, as de-
tailed in Table II.
Lastly, only a few studies have evalu-
ated the association between different
types of diet and the risk of SLE. Ac-
cording to a recent prospective study,
no association was found between
dietary patterns and risk of SLE oc-
currence (98). However, a subsequent
study suggested that a diet high in car-
bohydrates and low in fat may be asso-
ciated with an increased risk of SLE in
African-American women (99).
Gut microbiota
Increasing studies have shown that gut
microbiota dysbiosis, inducing inam-
mation and immune system sensitisa-
tion, represents a signicant risk factor
for the development of autoimmune
diseases, such as SLE (100).
In 2014, Hevia et al. (101) observed a
lower Firmicutes/Bacteroidetes (F/B)
ratio in SLE patients than in healthy
people. This nding was conrmed by
subsequent studies (102-105); further-
more, a French study observed that F/B
ratio was signicantly lower in active
SLE patients compared to patients in
remission (102). A signicant decrease
in Lactobacillus was also found in SLE
patients compared with healthy con-
trols (105).
Recently, Xiang et al. (106) realised a
metanalysis including 11 case-control
studies that examined 373 SLE pa-
tients and 1288 healthy controls. They
observed that SLE patients had fewer
Ruminococcaceae, but higher levels
of Enterobacteriaceae and Entero-
coccaceae. A two-sample Mendelian
randomisation study (107) found that
Bacilli, Eggertella and Lactobacilla-
les were positively correlated with the
risk of SLE, whereas Bacillales, Act-
inobacteria, Coprobacter and Lachno-
spira were negatively correlated with
SLE risk.
Another crucial factor involved in SLE
pathogenesis is the impairment of in-
testinal barrier: indeed, leaky gut has
been observed in patients with SLE.
This nding was demonstrated by two
studies that observed how calprotectin
levels in SLE stool samples were sig-
nicantly increased compared to con-
trols (108, 109). Moreover, Azzouz et
al. (108) observed that serum soluble
CD14, α1-acid glycoprotein and li-
popolysaccharides levels were higher
in SLE patients than in healthy sub-
jects, suggesting the presence of intesti-
nal bacterial displacement.
Molecular mimicry is another poten-
tial mechanism linking gut microbiota
with SLE, and several microorganisms
are involved: B. thetaiotaomicron, con-
taining Ro60, induces T- and B-cell re-
sponses against human Ro60 and glo-
merular immune complexes deposition;
E. gallinarum and R. intestinalis induce
anti-β2GP1 antibodies secretion; O.
splanchnicus, presenting a peptide sim-
ilar to human Sm antigen epitope, in-
creases IFN-γ and IL-17A production;
A. muciniphila has a peptide that, mim-
icking human Fas antigen, binds to IgG
produced by memory B cells; lastly, R.
gnavus cross-reacts with anti-dsDNA
antibodies.
Gut microbiota dysbiosis leads to both
cytokines and immune cells dysregula-
tion, contributing to the development
and progression of SLE. Briey, E. gal-
linarum and Ruminococcus are associ-
ated with an increase in Th17 cells and
a reduction of Treg, with a Th17/Treg
imbalance; E. gallinarum and L. reuteri
increase the number of plasmacytoid
dendritic cells and promote the produc-
tion of IFN-I, one of the most important
pathogenetic factors in SLE (110).
Drug-induced SLE
Drug-induced lupus (DIL) was rst
described in 1945 by Hoffman, who re-
ported lupus-like symptoms due to sul-
fadiazine treatment (111). Since then,
Table I. Principal interventional studies on omega3 supplementation in SLE.
Reference n Types of intervention Duration Conclusions
Westberg et al. 1990 (81) 17 Omega-3 vs. olive oil rened 6 months Short-term benecial effect on disease activity
(placebo group)
Duffy et al. 2004 (82) 52 Omega 3 with/without copper 24 weeks Improvement in disease activity (SLAM-R)
vs. copper vs. placebo
Wright et al. 2008 (83) 60 Omega-3 vs. olive oil 24 weeks Improvement in disease activity (SLAM-R, BILAG) and
(placebo group) endothelial function; reduction in oxidative stress
Bello et al. 2013 (84) 85 Omega-3 vs. starch 12 weeks No improvement in disease activity, endothelial function,
(placebo group) nor decrease in inammatory markers
Arriens et al. 2015 (85) 50 Omega-3 vs. olive oil rened 6 months Improvement in PGA, SF-36 and some circulating
(placebo group) inammatory markers; no impact on disease activity
Curado Borges et al. 2016 (86) 49 Omega-3; no placebo group 12 weeks No impact on IL-6, IL-10, leptin and adiponectin; signicant
decrease of CRP levels and impact on cholesterol levels
1109
Clinical and Experimental Rheumatology 2024
SLE and environment / C. Cardelli et al.
the list of drugs potentially involved in
the genesis of DIL has been expanding.
DIL is not a typical allergic drug reac-
tion, but is the result of a drug-induced
self-tolerance breakdown process (112).
According to studies of the early 2000s,
15000–30000 cases of DIL are esti-
mated to occur annually in the United
States, meaning that up to 10% of SLE
cases may be drug-induced.
Overall, compared to idiopathic SLE,
DIL tends to develop in elderly people
(partly because more exposed to drugs),
has a lower female predominance (with
a female-to-male ratio ranging from
4.3:1 to 1:1) and presents with a “lupus-
like syndrome” usually characterised
by fewer and milder clinical symptoms.
Arnaud et al. recently updated the
list of drugs associated with DIL. In
12166 DIL cases from the WHO phar-
macovigilance database, 118 putative
drugs were identied, and among these
42 had not been previously reported in
association with DIL. DIL was consid-
ered denite for nine drugs: procaina-
mide, hydralazine, minocycline, quini-
dine, isoniazid, terbinane, methyldo-
pa, dihydralazine and chlorpromazine
(113). Although less used nowadays,
procainamide and hydralazine are asso-
ciated with the highest risk of DIL, with
an estimated 20% incidence and 5–8%
risk per year of treatment (114).
In a recent matched case-control study
conducted on incident cases of CLE
and SLE in the Danish National Patient
Register, new plausible associations
were observed with some common
drugs: fexofenadine hydrochloride,
metoclopramide hydrochloride, metro-
nidazole hydrochloride and levothyrox-
ine sodium (115).
Moreover, a French pharmacoepidemi-
ological study suggested a link between
statin exposure and DIL, with a reported
OR >1 for each statin but uvastatin
(116).
Several anticonvulsants have been re-
ported to be associated to DIL, includ-
ing carbamazepine (117) and valproic
acid-induced lupus (118).
From a rheumatological perspective, it
is worth noting that, according to the
WHO pharmacovigilance database,
since 2007 onwards, anti-TNF agents
have been the drugs most commonly
associated with systemic DIL (113). We
can speculate if anti-TNF treatment may
unmask an underlying SLE in patients
with a baseline higher risk of overlap
syndrome, rather than cause de novo
drug-induced lupus (112).
Anti-TNF are known to induce autoan-
tibody production. Specically, in pro-
spective placebo-controlled trials, anti-
Table II. Principal interventional studies on vitamin D supplementation in SLE.
Reference n Types of intervention Duration Conclusions
Ruiz-Irastorza et al. 2010 (89) 60 Oral vitamin D3; no placebo group 24 months Benecial effect on fatigue; no effect on SLE
severity
Terrier et al. 2012 (90) 20 Vitamin D3 100000 IU/week for 4 weeks, 6 months Benecial effect on immunological and
followed by 100000 IU/month for 6 months; inammatory markers
no placebo group
Petri et al. 2013 (91) 1006 In patients with levels <40 ng/mL vitamin 128 weeks A 20 ng/mL increase in vitamin D level was
D2 50000 IU/week plus calcium/vitamin associated with decrease in the odds of having a
D3 200 IU twice daily high disease activity score and of having clinically
important proteinuria; no evidence of additional
benet of vitamin D beyond a level of 40 ng/ml
Aranow et al. 2015 (92) 54 Vitamin D3 4000 IU/day (high dose group) 12 weeks No effect on IFN signature in vitamin D-decient
or 2000 IU/day (low dose group) SLE patients
Andreoli et al. 2015 (93) 34 Vitamin D3 25000 IU/month (standard 24 months Neither regimen of supplementation affects disease
regimen) or 300000 IU initial bolus activity or SLE serology
followed by 50000 IU/month (intensive
regimen) for one year and then switched to
the other regimen in the second year
Piantoni et al. 2015 (94) 34 Vitamin D3 25000 IU/month (standard 24 months Enhancement of T-reg cells
regimen) or 300000 IU initial bolus followed
by 50000 IU/month (intensive regimen) for
one year and then switched to the other
regimen in the second year
Marinho et al. 2016 (95) 24 Vitamin D3 at variable dosage during the 6 months Reduction in SLEDAI and improvement in the
study period Treg/Th17 ratio
Karimzadeh et al. 2017 (96) 90 Vitamin D3 50000 IU/week for 12 weeks 6 months No effects on disease activity
and then 50000 IU/month for 3 months vs.
placebo
Al-Kushi et al. 2018 (97) 81 Corticosteroid treatment with vitamin D3 6 months Vitamin D and calcium supplementation
1400 IU/day and calcium carbonate 1250 signicantly improved the bone mineral density in
mg/day vs. corticosteroid treatment without vitamin D-decient patients; no effect on immune
supplementation vs. no corticosteroid markers or disease activity
treatment
1110 Clinical and Experimental Rheumatology 2024
SLE and environment / C. Cardelli et al.
dsDNA antibodies were induced in 20%
of iniximab, 15% of etanercept, 10-
12% of adalimumab and 4% of certoli-
zumab pegol patients. On the contrary,
in TNF-α antagonist-induced lupus-like
syndrome (TAILS), anti-histone an-
tibodies are less common (positive in
only 17–57% of patients). Moreover, in
a review of 72 patients with TAILS, the
presence of 12% anti-Ro/La, 10% anti-
Sm and 7% anti-RNP antibodies was
reported (119).
However, despite a quite high propor-
tion of patients developing autoanti-
bodies during anti-TNF treatment, the
development of a clinically relevant
lupus-like syndrome is much more in-
frequent and has been estimated to oc-
cur at a rate between 0.5 and 1%. Ac-
cording to a French study, iniximab
and adalimumab carry a higher risk of
TAILS than etanercept (120). From a
clinical point of view, TAILS, compared
to other DIL forms, is characterised by a
higher frequency of cutaneous manifes-
tations (up to 72% of patients).
Concurrent use of immunosuppres-
sants may reduce the risk of developing
autoantibodies and potentially TAILS.
Isolated induction of autoantibodies
is not an indication for discontinuing
therapy. Finally, a few studies investi-
gate if patients can switch to other anti-
TNF without recurrence of TAILS: 10
patients who tolerated long-term treat-
ment with similar-acting agents with-
out recurrence of TAILS have been
reported in the literature (121).
Anecdotal cases of DIL and ares of
preexisting SLE induced by chemo-
therapy (paclitaxel, capecitabine and
doxorubicin) have been described (122-
124).
A greater interest is currently being di-
rected towards immune checkpoint in-
hibitors (ICIs), known to be associated
with the risk of developing autoim-
mune rheumatic manifestations. How-
ever, DIL seems a rare event in patients
receiving ICIs. In the FDA Adverse
Event Reporting System, among 4870
rheumatic events reported with ICIs,
only 18 cases of SLE, 7 cases of CLE,
2 cases of lupus-like syndrome and 1
case each for lupus nephritis and cen-
tral nervous system lupus were identi-
ed (125). Only PD1/PDL1 were asso-
ciated with DIL (mainly nivolumab in
12 cases, followed by pembrolizumab
in 4 cases).
Such as idiopathic lupus, DIL can be
classied in systemic and cutaneous
DIL. Among skin-limited DIL, SCLE
has been mostly reported (126), while
discoid form of DIL seems to be very
rare (127).
In a population-based case-control study
of 234 SCLE incident cases in Sweden,
38% could be attributed to drug expo-
sure, with people <50 years at higher
risk compared to older ones (126). The
highest association was found for terbi-
nane, followed by anti-TNF. Among
anti-hypertensive drugs, ACE-inhibitors
resulted associated with SCLE. This
study also showed an association be-
tween proton pump inhibitors (PPIs)
exposure and SCLE, further conrmed
in a recent study on the French phar-
macovigilance database where, among
60 cases of DIL associated with PPIs,
79.6% were skin-limited (128).
The epidemiology and clinical spectrum
of DIL evolve with changes in the phar-
macopoeia. Rheumatologists should be
aware of this clinical entity, as prompt
discontinuation of the causative drug
can lead to rapid improvement of clini-
cal manifestations.
Conclusion
Although the exact patterns of SLE
onset and disease progression are not
fully understood, signicant progress
has been made over the years in under-
standing this complex disease. Besides
historically known environmental fac-
tors, recent advances have been made
in deciphering triggers and drivers that
contribute to the development of SLE.
In this review we summarised the most
recent literature investigating environ-
mental factors potentially involved in
the disease onset and clinical expres-
sion; despite the big amount of data
available, the causal relationship be-
tween certain exposures and the disease
onset or progression remains frequently
elusive and not fully demonstrated. In-
deed, epidemiological research on SLE
is difcult due to the low prevalence of
the disease, its scattered distribution,
and the inuence of several socio-eco-
nomic factors on disease expression and
evolution. For instance, environmental
studies may suffer from serious weak-
nesses, including the lack of adjustment
for individual confounding factors and
the inaccuracy in capturing the relevant
exposure at the time of disease induc-
tion. Thus, environmental-epidemiolo-
gy studies often lack sufcient power to
detect important effects.
Moreover, many environmental-epide-
miology studies are cross-sectional. In
such designs, the simultaneous assess-
ment of outcome and exposure can give
rise to difculties in determining the
temporal aspects of the causal associa-
tion between exposure and outcome.
Lastly, it is important to note that the
available studies refer to a wide range
of geographical locations and vary
widely in exposure collection methods
and SLE phenotypes.
In conclusion, high quality research
designs that could help identify the en-
vironmental components of the causal
pathways leading to SLE and its clinical
phenotype are still scarce and should be
strongly encouraged.
References
1. ZUCCHI D, SILVAGNI E, ELEFANTE E et al.:
Systemic lupus erythematosus: one year in
review 2023. Clin Exp Rheumatol 2023; 41:
997-1008. https://
doi.org/10.55563/clinexprheumatol/4uc7e8
2. COOPER GS, MILLER FW, PANDEY JP: The
role of genetic factors in autoimmune dis-
ease: implications for environmental re-
search. Environ Health Perspect 1999;107
Suppl 5: 693-700.
https://doi.org/10.1289/ehp.99107s5693
3. DEAFEN D, ESCALANTE A, WEINRIB L et
al.: A revised estimate of twin concordance
in systemic lupus erythematosus. Arthritis
Rheum 1992; 35: 311-18.
https://doi.org/10.1002/art.1780350310
4. SESTAK AL, FÜRNROHR BG, HARLEY JB
et al.: The genetics of systemic lupus ery-
thematosus and implications for targeted
therapy. Ann Rheum Dis 2011; 70 Suppl. 1.
https://doi.org/10.1136/ard.2010.138057
5. MARGERY-MUIR AA, BUNDELL C, NELSON
D et al.: Gender balance in patients with
systemic lupus erythematosus. Autoimmun
Rev 2017; 16: 258-68.
https://doi.org/10.1016/J.autrev.2017.01.007
6. McGRATH H: Ultraviolet-A1 irradiation
decreases clinical disease activity and au-
toantibodies in patients with systemic lupus
erythematosus. Clin Exp Rheumatol 1994;
12: 129-35.
7. HASAN T, NYBERG F, STEPHANSSON E et
al.: Photosensitivity in lupus erythematosus,
UV photoprovocation results compared with
history of photosensitivity and clinical nd-
1111
Clinical and Experimental Rheumatology 2024
SLE and environment / C. Cardelli et al.
ings. Br J Dermatol 1997; 136: 699-705.
8. van WEELDEN H, VELTHUIS PJ, BAART DE
LA FAILLE H: Light-induced skin lesions in
lupus erythematosus: photobiological stud-
ies. Arch Dermatol Res 1989; 281: 470-74.
https://doi.org/10.1007/bf00510082
9. KUHN A, WENZEL J, WEYD H: Photosensi-
tivity, apoptosis, and cytokines in the patho-
genesis of lupus erythematosus: a critical
review. Clin Rev Allergy Immunol 2014; 47:
148-62.
https://doi.org/10.1007/S12016-013-8403-x
10. BULLER DB, COKKINIDES V, HALL HI et al.:
Prevalence of sunburn, sun protection, and
indoor tanning behaviors among Ameri-
cans: review from national surveys and case
studies of 3 states. J Am Acad Dermatol
2011; 65: S114.e1-S114.e11.
https://doi.org/10.1016/j.jaad.2011.05.033
11. BENGTSSON AA, RYLANDER L, HAGMAR L
et al.: Risk factors for developing systemic
lupus erythematosus: a case-control study in
southern Sweden. Rheumatology (Oxford)
2002; 41: 563-71. https://
doi.org/10.1093/rheumatology/41.5.563
12. MATSUNAGA T, HIEDA K, NIKAIDO O:
Wavelength dependent formation of thy-
mine dimers and (6-4) photoproducts in
DNA by monochromatic ultraviolet light
ranging from 150 to 365 nm. Photochem
Photobiol 1991; 54: 403-10. https://
doi.org/10.1111/J.1751-1097.1991.tb02034.x
13. MITCHELL D: Revisiting the photochemis-
try of solar UVA in human skin. Proc Natl
Acad Sci USA 2006; 103: 13567-68.
https://doi.org/10.1073/pnas.0605833103
14. BARBHAIYA M, COSTENBADER KH: Ultra-
violet radiation and systemic lupus erythe-
matosus. Lupus 2014; 23: 588-95.
https://doi.org/10.1177/0961203314530488
15. KUHN A, HERRMANN M, KLEBER S et al.:
Accumulation of apoptotic cells in the
epidermis of patients with cutaneous lupus
erythematosus after ultraviolet irradiation.
Arthritis Rheum 2006; 54: 939-50.
https://doi.org/10.1002/art.21658
16. HILE GA, KAHLENBERG JM: Immunopatho-
genesis of skin injury in systemic lupus
erythematosus. Curr Opin Rheumatol 2021;
33: 173-80. https://
doi.org/10.1097/bor.0000000000000770
17. MILLER FW, ALFREDSSON L, COSTEN-
BADER KH et al.: Epidemiology of environ-
mental exposures and human autoimmune
diseases: ndings from a National Institute
of Environmental Health Sciences Expert
Panel Workshop. J Autoimmun 2012; 39:
259-71.
https://doi.org/10.1016/j.jaut.2012.05.002
18. PARKS CG, COOPER GS, NYLANDER-
FRENCH LA et al.: Occupational exposure to
crystalline silica and risk of systemic lupus
erythematosus: A population-based, case-
control study in the southeastern United
States. Arthritis Rheum 2002; 46: 1840-50.
https://doi.org/10.1002/art.10368
19. MOROTTI A, SOLLAKU I, CATALANI S et
al.: Systematic review and meta-analysis of
epidemiological studies on the association of
occupational exposure to free crystalline sil-
ica and systemic lupus erythematosus. Rheu-
matology (Oxford) 2021; 60: 81-91. https://
doi.org/10.1093/rheumatology/keaa444
20. COOPER GS, WITHER J, BERNATSKY S et
al.: Occupational and environmental expo-
sures and risk of systemic lupus erythemato-
sus: silica, sunlight, solvents. Rheumatology
(Oxford) 2010; 49: 2172-80. https://
doi.org/10.1093/rheumatology/keq214
21. LI X, SUNDQUIST J, SUNDQUIST K et al.:
Occupational risk factors for systemic lupus
erythematosus: a nationwide study based on
hospitalizations in Sweden. J Rheumatol.
2012; 39: 743-51.
https://doi.org/10.3899/jrheum.110789
22. GOLD LS, WARD MH, DOSEMECI M et al.:
Systemic autoimmune disease mortality and
occupational exposures. Arthritis Rheum
2007; 56: 3189-201.
https://doi.org/10.1002/art.22880
23. CONRAD K, MEHLHORN J, LÜTHKE K et al.:
Systemic lupus erythematosus after heavy
exposure to quartz dust in uranium mines:
clinical and serological characteristics. Lu-
pus 1996; 5: 62-29. https://
doi.org/10.1177/096120339600500112
24. SPEYER CB, COSTENBADER KH: Cigarette
smoking and the pathogenesis of systemic
lupus erythematosus. Expert Rev Clin Im-
munol 2018; 14: 481-87. https://
doi.org/10.1080/1744666x.2018.1473035
25. BIJL M, HORST G, LIMBURG PC et al.:
Effects of smoking on activation markers,
Fas expression and apoptosis of peripheral
blood lymphocytes. Eur J Clin Invest 2001;
31: 550-53. https://
doi.org/10.1046/j.1365-2362.2001.00842.x
26. PARISIS D, BERNIER C, CHASSET F et al.:
Impact of tobacco smoking upon disease
risk, activity and therapeutic response in
systemic lupus erythematosus: a systematic
review and meta-analysis. Autoimmun Rev
2019; 18: 102393. https://
doi.org/10.1016/j.autrev.2019.102393
27. YAN CHUA MH, TING NG IA, CHEUNG MWL
et al.: Association between cigarette smok-
ing and systemic lupus erythematosus: an
updated multivariate Bayesian metaanaly-
sis. J Rheumatol 2020; 47: 1514-21.
https://doi.org/10.3899/jrheum.190733
28. SIMARD JF, COSTENBADER KH, LIANG MH
et al.: Exposure to maternal smoking and
incident SLE in a prospective cohort study.
Lupus 2009; 18: 431-35.
https://doi.org/10.1177/0961203308098186
29. WARD MM, STUDENSKI S: Clinical prog-
nostic factors in lupus nephritis. The impor-
tance of hypertension and smoking. Arch
Intern Med 1992; 152: 2082-88.
30. MONTES RA, MOCARZEL LO, LANZIERI
PG et al.: Smoking and its association with
morbidity in systemic lupus erythematosus
evaluated by the Systemic Lupus Inter-
national Collaborating Clinics/American
College of Rheumatology Damage Index:
preliminary data and systematic review. Ar-
thritis Rheumatol 2016; 68: 441-48.
https://doi.org/10.1002/art.39427
31. ZHANG WT, LIU Z, ZHU BC et al.: Effects of
tobacco smoking on cardiovascular disease
in patients with systemic lupus erythemato-
sus: A systematic review and meta-analysis.
Front Immunol 2022; 13.
https://doi.org/10.3389/mmu.2022.967506
32. LEROUX G, COSTEDOAT-CHALUMEAU
N, HULOT J-S et al.: Relationship between
blood hydroxychloroquine and desethyl-
chloroquine concentrations and cigarette
smoking in treated patients with connective
tissue diseases. Ann Rheum Dis 2007; 66:
1547-48.
https://doi.org/10.1136/ard.2007.072587
33. FRANCÈS C, COSNES A, DUHAUT P et al.:
Low blood concentration of hydroxychloro-
quine in patients with refractory cutaneous
lupus erythematosus: a French multicenter
prospective study. Arch Dermatol 2012;
148: 479-84. https://
doi.org/10.1001/archdermatol.2011.2558
34. JALLOULI M, GALICIER L, ZAHR N et al.:
Determinants of hydroxychloroquine blood
concentration variations in systemic lupus
erythematosus. Arthritis Rheumatol 2015;
67: 2176-84.
https://doi.org/10.1002/art.39194
35. YEON LEE J, LEE J, KI KWOK S et al.: Fac-
tors related to blood hydroxychloroquine
concentration in patients with systemic lu-
pus erythematosus. Arthritis Care Res 2017;
69: 536-42.
https://doi.org/10.1002/acr.22962
36. PARODIS I, SJÖWALL C, JÖNSEN A et al.:
Smoking and pre-existing organ damage re-
duce the efcacy of belimumab in systemic
lupus erythematosus. Autoimmun Rev 2017;
16: 343-51.
https://doi.org/10.1016/j.autrev.2017.02.005
37. PARODIS I, GOMEZ A, FRODLUND M et al.:
Smoking reduces the efcacy of belimumab
in mucocutaneous lupus. Expert Opin Biol
Ther 2018; 18: 911-20. https://
doi.org/10.1080/14712598.2018.1494719
38. ILLESCAS-MONTES R, CORONA-CASTRO
CC, MELGUIZO-RODRÍGUEZ L et al.: Infec-
tious processes and systemic lupus erythe-
matosus. Immunology 2019; 158: 153-60.
https://doi.org/10.1111/imm.13103
39. NELSON P, RYLANCE P, RODEN D et al.:
Viruses as potential pathogenic agents in
systemic lupus erythematosus. Lupus 2014;
23: 596-605.
https://doi.org/10.1177/0961203314531637
40 McCLAIN MT, HARLEY JB, JAMES JA:
The role of Epstein-Barr virus in systemic
lupus erythematosus. Front Biosci 2001; 6:
E137-47. https://doi.org/10.2741/mcclain
41. JAMES JA, KAUFMAN KM, FARRIS AD et al.:
An increased prevalence of Epstein-Barr vi-
rus infection in young patients suggests a
possible etiology for systemic lupus erythe-
matosus. J Clin Invest 1997; 100: 3019-26.
https://doi.org/10.1172/jci119856
42. HARLEY JB, JAMES JA: Epstein-Barr vi-
rus infection may be an environmental
risk factor for systemic lupus erythema-
tosus in children and teenagers. Arthri-
tis Rheum 1999; 42: 1782-83. https://doi.
org/10.1002/1529-0131(199908)42:8<1782
::aid-anr36>3.0.co;2-x
43. JAMES JA, NEAS BR, MOSER KL et al.: Sys-
temic lupus erythematosus in adults is asso-
ciated with previous Epstein-Barr virus ex-
posure. Arthritis Rheum 2001; 44: 1122-26.
https://doi.org/10.1002/1529-0131(200105)
44:5<1122::aid-anr193>3.0.co;2-d
44. YU SF, WU HC, TSAI WC et al.: Detecting
1112 Clinical and Experimental Rheumatology 2024
SLE and environment / C. Cardelli et al.
Epstein-Barr virus DNA from peripheral
blood mononuclear cells in adult patients
with systemic lupus erythematosus in Tai-
wan. Med Microbiol Immunol 2005; 194:
115-20.
https://doi.org/10.1007/s00430-004-0230-5
45. HANLON P, AVENELL A, AUCOTT L et al.:
Systematic review and meta-analysis of the
sero-epidemiological association between
Epstein-Barr virus and systemic lupus ery-
thematosus. Arthritis Res Ther 2014; 16.
https://doi.org/10.1186/ar4429
46. ZANDMAN-GODDARD G, SHOENFELD Y:
SLE and infections. Clin Rev Allergy Immu-
nol 2003; 25: 29-39.
https://doi.org/10.1385/criai:25:1:29
47. ROKEACH LA, HASELBY JA, HOCH SO: Mo-
lecular cloning of a cDNA encoding the hu-
man Sm-D autoantigen. Proc Natl Acad Sci
USA 1988; 85: 4832-36.
https://doi.org/10.1073/pnas.85.13.4832
48. SABBATINI A, DOLCHER MP, MARCHINI
B et al.: Mapping of epitopes on the SmD
molecule: the use of multiple antigen pep-
tides to measure autoantibodies in systemic
lupus erythematosus. J Rheumatol 1993; 20:
1679-83.
49. CAPONI L, BOMBARDIERI S, MIGLIORINI P:
Anti-ribosomal antibodies bind the Sm pro-
teins D and B/B’. Clin Exp Immunol 1998;
112: 139-43. https://
doi.org/10.1046/j.1365-2249.1998.00545.x
50. AFRASIABI A, KEANE JT, ONG LTC et al.:
Genetic and transcriptomic analyses support
a switch to lytic phase in Epstein Barr virus
infection as an important driver in develop-
ing systemic lupus erythematosus. J Auto-
immun 2022; 127.
https://doi.org/10.1016/j.jaut.2021.102781
51. BANKO A, CIRKOVIC A, MISKOVIC R et al.:
Epstein-Barr virus infection as potential in-
dicator of the occurrence and clinical pres-
entation of systemic lupus erythematosus.
Front Immunol 2023; 14: 1307589. https://
doi.org/10.3389/mmu.2023.1307589
52. COOPER GS, DOOLEY MA, TREADWELL EL
et al.: Risk factors for development of sys-
temic lupus erythematosus: Allergies, infec-
tions, and family history. J Clin Epidemiol
2002; 55: 982-89. https://doi.org/10.1016/
s0895-4356(02)00429-8
53. KOURANLOO K, DEY M, ELWELL H et al.: A
systematic review of the incidence, manage-
ment and prognosis of new-onset autoim-
mune connective tissue diseases after COV-
ID-19. Rheumatol Int 2023; 43: 1221-43.
https://doi.org/10.1007/s00296-023-05283-9
54. JOO Y BIN, KIM KJ, PARK KS et al.: Inuen-
za infection as a trigger for systemic lupus
erythematosus ares resulting in hospitali-
zation. Sci Rep 2021; 11.
https://doi.org/10.1038/S41598-021-84153-5
55. OSPINA FE, ECHEVERRI A, ZAMBRANO D
et al.: Distinguishing infections vs ares in
patients with systemic lupus erythematosus.
Rheumatology (Oxford) 2017; 56: i46-54.
https://
doi.org/10.1093/rheumatology/kew340
56. UROWITZ MB, BOOKMAN AA, KOEHLER BE
et al.: The bimodal mortality pattern of sys-
temic lupus erythematosus. Am J Med 1976;
60: 221-25. https://doi.org/10.1016/0002-
9343(76)90431-9
57. FRODLUND M, JÖNSEN A, REMKUS L et
al.: Glucocorticoid treatment in SLE is as-
sociated with infections, comorbidities, and
mortality-a national cohort study. Rheuma-
tology (Oxford) 2024; 63: 1104-12. https://
doi.org/10.1093/rheumatology/kead348
58. TEKTONIDOU MG, DASGUPTA A, WARD
MM: Interhospital variation in mortality
among patients with systemic lupus erythe-
matosus and sepsis in the USA. Rheumatol-
ogy (Oxford) 2019; 58: 1794-801. https://
doi.org/10.1093/rheumatology/kez103
59. BERNIER MO, MIKAELOFF Y, HUDSON M
et al.: Combined oral contraceptive use and
the risk of systemic lupus erythematosus.
Arthritis Rheum 2009; 61: 476-81.
https://doi.org/10.1002/art.24398
60. SANTOS-OCAMPO AS: New onset systemic
lupus erythematosus in a transgender man:
possible role of feminizing sex hormones. J
Clin Rheumatol 2007; 13: 29-30. https://doi.
org/10.1097/01.rhu.0000256169.05087.ad
61. ZANDMAN-GODDARD G, SOLOMON M,
BARZILAI A et al.: Lupus erythematosus tu-
midus induced by sex reassignment surgery.
J Rheumatol 2007; 34: 1938-40.
62. CHAN KL, MOK CC: Development of sys-
temic lupus erythematosus in a male-to-fe-
male transsexual: the role of sex hormones
revisited. Lupus 2013; 22: 1399-402.
https://doi.org/10.1177/0961203313500550
63. PONTES LT, CAMILO DT, DE BORTOLI MR
et al.: New-onset lupus nephritis after male-
to-female sex reassignment surgery. Lupus
2018; 27: 2166-69.
https://doi.org/10.1177/0961203318800571
64. HILL BG, HODGE B, MISISCHIA R: Lupus
nephritis in a transgender woman on cross-
sex hormone therapy: a case for the role of
oestrogen in systemic lupus erythematosus.
Lupus 2020; 29: 1807-10.
https://doi.org/10.1177/0961203320946372
65. SÁNCHEZ-GUERRERO J, URIBE AG, JIMÉN-
EZ-SANTANA L et al.: A trial of contracep-
tive methods in women with systemic lupus
erythematosus. N Engl J Med 2005; 353:
2539-49.
https://doi.org/10.1056/nejmoa050817
66. PETRI M, KIM MY, KALUNIAN KC et al.:
Combined oral contraceptives in women
with systemic lupus erythematosus. N Engl
J Med 2005; 353: 2550-58.
https://doi.org/10.1056/nejmoa051135
67. URBANUS RT, SIEGERINK B, ROEST M et
al.: Antiphospholipid antibodies and risk of
myocardial infarction and ischaemic stroke
in young women in the RATIO study: a
case-control study. Lancet Neurol 2009; 8:
998-1005. https://
doi.org/10.1016/s1474-4422(09)70239-x
68. ANDREOLI L, BERTSIAS GK, AGMON-
LEVIN N et al.: EULAR recommendations
for women’s health and the management
of family planning, assisted reproduction,
pregnancy and menopause in patients with
systemic lupus erythematosus and/or an-
tiphospholipid syndrome. Ann Rheum Dis
2017; 76: 476-85. https://
doi.org/10.1136/annrheumdis-2016-209770
69. SAMMARITANO LR, BERMAS BL,
CHAKRAVARTY EE et al.: 2020 American
College of Rheumatology Guideline for
the Management of Reproductive Health in
Rheumatic and Musculoskeletal Diseases.
Arthritis Rheumatol 2020; 72: 529-56.
https://doi.org/10.1002/art.41191
70. GALZOTE RM, RAFIE S, TEAL R et al.:
Transdermal delivery of combined hormo-
nal contraception: a review of the current
literature. Int J Womens Health 2017; 9:
315-21.
https://doi.org/10.2147/ijwh.s102306
71. CRAVIOTO MDC, DURAND-CARBAJAL
M, JIMÉNEZ-SANTANA L et al.: Efcacy
of estrogen plus progestin on menopausal
symptoms in women with systemic lupus
erythematosus: a randomized, double-blind,
controlled trial. Arthritis Care Res (Hobo-
ken) 2011; 63: 1654-63.
https://doi.org/10.1002/acr.20608
72. SÁNCHEZ-GUERRERO J, GONZÁLEZ-PÉREZ
M, DURAND-CARBAJAL M et al.: Meno-
pause hormonal therapy in women with sys-
temic lupus erythematosus. Arthritis Rheum
2007; 56: 3070-79.
https://doi.org/10.1002/art.22855
73. BUYON JP, PETRI MA, KIM MY et al.:
The effect of combined estrogen and pro-
gesterone hormone replacement therapy on
disease activity in systemic lupus erythema-
tosus: a randomized trial. Ann Intern Med
2005; 142. https://doi.org/10.7326/0003-
4819-142-12_part_1-200506210-00004
74. HOCHMAN J, UROWITZ MB, IBAÑZ D et al.:
Hormone replacement therapy in women
with systemic lupus erythematosus and risk
of cardiovascular disease. Lupus 2009; 18:
313-37.
https://doi.org/10.1177/0961203308097475
75. JIAO H, ACAR G, ROBINSON GA et al.: Diet
and systemic lupus erythematosus (SLE):
from supplementation to intervention. Int J
Environ Res Public Health 2022; 19.
https://doi.org/10.3390/ijerph191911895
76. GWINNUTT JM, WIECZOREK M, BALA-
NESCU A et al.: 2021 EULAR recommen-
dations regarding lifestyle behaviours and
work participation to prevent progression
of rheumatic and musculoskeletal diseases.
Ann Rheum Dis 2023; 82: 48-56. https://
doi.org/10.1136/annrheumdis-2021-222020
77. ESTRUCH R: Anti-inammatory effects of
the Mediterranean diet: the experience of
the PREDIMED study. Proc Nutr Soc 2010;
69: 333-40.
https://doi.org/10.1017/s0029665110001539
78. POCOVI-GERARDINO G, CORREA-RODRÍ-
GUEZ M, CALLEJAS-RUBIO JL et al.: Bene-
cial effect of Mediterranean diet on disease
activity and cardiovascular risk in systemic
lupus erythematosus patients: a cross-sec-
tional study. Rheumatology (Oxford) 2021;
60: 160-69. https://
doi.org/10.1093/rheumatology/keaa210
79. DAVIES RJ, LOMER M, YEO SI et al.: Weight
loss and improvements in fatigue in system-
ic lupus erythematosus: a controlled trial of
a low glycaemic index diet versus a calorie
restricted diet in patients treated with corti-
costeroids. Lupus 2012; 21: 649-55.
https://doi.org/10.1177/0961203312436854
80. PATRICK RP, AMES BN: Vitamin D and
the omega-3 fatty acids control serotonin
1113
Clinical and Experimental Rheumatology 2024
SLE and environment / C. Cardelli et al.
synthesis and action, part 2: relevance for
ADHD, bipolar disorder, schizophrenia,
and impulsive behavior. FASEB J 2015; 29:
2207-22.
https://doi.org/10.1096/fj.14-268342
81. WESTBERG G, TARKOWSKI A: Effect of
MaxEPA in patients with SLE. A double-
blind, crossover study. Scand J Rheumatol
1990; 19: 137-43. https://
doi.org/10.3109/03009749009102117
82. DUFFY EM, MEENAGH GK, MCMILLAN SA
et al.: The clinical effect of dietary sup-
plementation with omega-3 sh oils and/or
copper in systemic lupus erythematosus. J
Rheumatol 2004; 31: 1551-56.
83. WRIGHT S, O’PREY FM, McHENRY MT et
al.: A randomised interventional trial of
omega-3-polyunsaturated fatty acids on
endothelial function and disease activity in
systemic lupus erythematosus. Ann Rheum
Dis 2008; 67: 841-48.
https://doi.org/10.1136/ard.2007.077156
84. BELLO KJ, FANG H, FAZELI P et al.: Ome-
ga-3 in SLE: a double-blind, placebo-con-
trolled randomized clinical trial of endothe-
lial dysfunction and disease activity in sys-
temic lupus erythematosus. Rheumatol Int
2013; 33: 2789-96.
https://doi.org/10.1007/S00296-013-2811-3
85. ARRIENS C, HYNAN LS, LERMAN RH et al.:
Placebo-controlled randomized clinical trial
of sh oil’s impact on fatigue, quality of
life, and disease activity in Systemic Lupus
Erythematosus. Nutr J 2015; 14.
https://doi.org/10.1186/s12937-015-0068-2
86. CURADO BORGES M, DE MIRANDA MOURA
DOS SANTOS F, WEISS TELLES R et al.:
Omega-3 fatty acids, inammatory status
and biochemical markers of patients with
systemic lupus erythematosus: a pilot study.
Rev Bras Reumatol 2017; 57: 526-34.
https://doi.org/10.1016/j.rbre.2016.09.014
87. MOK CC: Vitamin D and systemic lupus
erythematosus: an update. Expert Rev Clin
Immunol 2013; 9: 453-63.
https://doi.org/10.1586/eci.13.19
88. HASSANALILOU T, KHALILI L, GHAVAMZA-
DEH S et al.: Role of vitamin D deciency in
systemic lupus erythematosus incidence and
aggravation. Auto Immun Highlights 2017;
9: 1.
https://doi.org/10.1007/s13317-017-0101-x
89. RUIZ-IRASTORZA G, GORDO S, OLIVARES
N et al.: Changes in vitamin D levels in
patients with systemic lupus erythemato-
sus: Effects on fatigue, disease activity,
and damage. Arthritis Care Res (Hoboken)
2010; 62: 1160-65.
https://doi.org/10.1002/acr.20186
90. TERRIER B, DERIAN N, SCHOINDRE Y et al.:
Restoration of regulatory and effector T cell
balance and B cell homeostasis in systemic
lupus erythematosus patients through vita-
min D supplementation. Arthritis Res Ther
2012; 14. https://doi.org/10.1186/ar4060
91. PETRI M, BELLO KJ, FANG H et al.: Vitamin
D in systemic lupus erythematosus: mod-
est association with disease activity and the
urine protein-to-creatinine ratio. Arthritis
Rheum 2013; 65: 1865-71.
https://doi.org/10.1002/art.37953
92. ARANOW C, KAMEN DL, DALL’ERA M et
al.: Randomized, double-blind, placebo-
controlled trial of the effect of Vitamin D3
on the interferon signature in patients with
systemic lupus erythematosus. Arthritis
Rheumatol 2015; 67: 1848-57.
https://doi.org/10.1002/art.39108
93. ANDREOLI L, DALL’ARA F, PIANTONI S et
al.: A 24-month prospective study on the ef-
cacy and safety of two different monthly
regimens of vitamin D supplementation in
pre-menopausal women with systemic lupus
erythematosus. Lupus 2015; 24: 499-506.
https://doi.org/10.1177/0961203314559089
94. PIANTONI S, ANDREOLI L, SCARSI M et
al.: Phenotype modications of T-cells and
their shift toward a Th2 response in patients
with systemic lupus erythematosus supple-
mented with different monthly regimens of
vitamin D. Lupus 2015; 24: 490-98. https://
doi.org/10.1177/0961203314559090
95. MARINHO A, CARVALHO C, BOLEIXA D et
al.: Vitamin D supplementation effects on
FoxP3 expression in T cells and FoxP3+/
IL-17A ratio and clinical course in systemic
lupus erythematosus patients: a study in a
Portuguese cohort. Immunol Res 2017; 65:
197-206.
https://doi.org/10.1007/S12026-016-8829-3
96. KARIMZADEH H, SHIRZADI M, KARIMIFAR
M: The effect of Vitamin D supplementation
in disease activity of systemic lupus erythe-
matosus patients with Vitamin D deciency:
A randomized clinical trial. J Res Med Sci
2017; 22.
https://doi.org/10.4103/1735-1995.199089
97. AL-KUSHI A, AZZEH F, HEADER E et al.: Ef-
fect of Vitamin D and calcium supplementa-
tion in patients with systemic lupus erythe-
matosus. Saudi J Med Med Sci 2018; 6: 137.
https://
doi.org/10.4103/sjmms.sjmms_134_17
98. TEDESCHI SK, BARBHAIYA M, SPARKS JA
et al.: Dietary patterns and risk of system-
ic lupus erythematosus in women. Lupus
2020; 29: 67-73.
https://doi.org/10.1177/0961203319888791
99. CASTRO-WEBB N, COZIER YC, BARBHAIYA
M et al.: Association of macronutrients and
dietary patterns with risk of systemic lupus
erythematosus in the Black Women’s Health
Study. Am J Clin Nutr 2021; 114: 1486-94.
https://doi.org/10.1093/ajcn/nqab224
100. BROWN K, DECOFFE D, MOLCAN E et al.:
Diet-induced dysbiosis of the intestinal
microbiota and the effects on immunity
and disease. Nutrients 2012; 4: 1095-119.
https://doi.org/10.3390/nu4081095
101. HEVIA A, MILANI C, LÓPEZ P et al.:
Intestinal dysbiosis associated with system-
ic lupus erythematosus. MBio 2014; 5.
https://doi.org/10.1128/mbio.01548-14
102. TOUMI E, GOUTORBE B, PLAUZOLLES A et
al.: Gut microbiota in systemic lupus ery-
thematosus patients and lupus mouse mod-
el: a cross species comparative analysis for
biomarker discovery. Front Immunol 2022;
13: 943241.
https://doi.org/10.3389/mmu.2022.943241
103. HE Z, SHAO T, LI H et al.: Alterations of the
gut microbiome in Chinese patients with
systemic lupus erythematosus. Gut Pathog
2016; 8.
https://doi.org/10.1186/s13099-016-0146-9
104. van der MEULEN TA, HARMSEN HJM, VILA
AV et al.: Shared gut, but distinct oral mi-
crobiota composition in primary Sjögren’s
syndrome and systemic lupus erythemato-
sus. J Autoimmun 2019; 97: 77-87.
https://doi.org/10.1016/j.jaut.2018.10.009
105. GERGES MA, ESMAEEL NE, MAKRAM WK
et al.: Altered prole of fecal microbiota in
newly diagnosed systemic lupus erythemato-
sus Egyptian patients. Int J Microbiol 2021;
2021. https://doi.org/10.1155/2021/9934533
106. XIANG S, QU Y, QIAN S et al.: Association
between systemic lupus erythematosus
and disruption of gut microbiota: a meta-
analysis. Lupus Sci Med 2022; 9: e000599.
https://doi.org/10.1136/lupus-2021-000599
107. XIANG K, WANG P, XU Z et al.: Causal ef-
fects of gut microbiome on systemic lupus
erythematosus: a two-sample Mendelian
randomization study. Front Immunol 2021;
12: 667097.
https://doi.org/10.3389/mmu.2021.667097
108. AZZOUZ D, OMARBEKOVA A, HEGUY A et
al.: Lupus nephritis is linked to disease-ac-
tivity associated expansions and immunity
to a gut commensal. Ann Rheum Dis 2019;
78: 947-56. https:/
doi.org/10.1136/annrheumdis-2018-214856
109. MANFREDO VIEIRA S, HILTENSPERGER
M, KUMAR V et al.: Translocation of a gut
pathobiont drives autoimmunity in mice
and humans. Science 2018; 359: 1156-61.
https://doi.org/10.1126/science.aar7201
110. YAO K, XIE Y, WANG J et al.: Gut microbio-
ta: a newly identied environmental factor
in systemic lupus erythematosus. Front Im-
munol 2023; 14.
https://doi.org/10.3389/mmu.2023.1202850
111. HE Y, SAWALHA AH: Drug-induced lupus
erythematosus: an update on drugs and
mechanisms. Curr Opin Rheumatol 2018;
30: 490-97. https://
doi.org/10.1097/bor.0000000000000522
112. VAGLIO A, GRAYSON PC, FENAROLI P et
al.: Drug-induced lupus: Traditional and
new concepts. Autoimmun Rev 2018; 17:
912-18.
https://doi.org/10.1016/j.autrev.2018.03.016
113. ARNAUD L, MERTZ P, GAVAND PE et al.:
Drug-induced systemic lupus: revisiting the
ever-changing spectrum of the disease using
the WHO pharmacovigilance database. Ann
Rheum Dis 2019; 78: 504-8. https://
doi.org/10.1136/annrheumdis-2018-214598
114. ARAÚJO-FERNÁNDEZ S, AHIJÓN-LANA M,
ISENBERG DA: Drug-induced lupus: includ-
ing anti-tumour necrosis factor and inter-
feron induced. Lupus 2014; 23: 545-53.
https://doi.org/10.1177/0961203314523871
115. HAUGAARD JH, KOFOED K, GISLASON G et
al.: Association between drug use and sub-
sequent diagnosis of lupus erythematosus.
JAMA Dermatol 2020; 156: 1199-207. https://
doi.org/10.1001/jamadermatol.2020.2786
116. MOULIS G, BÉNÉ J, SOMMET A et al.:
Statin-induced lupus: a case/non-case study
in a nationwide pharmacovigilance data-
base. Lupus 2012; 21: 885-89.
https://doi.org/10.1177/0961203312436861
117. SCHOONEN WM, THOMAS SL, SOMERS EC
et al.: Do selected drugs increase the risk of
1114 Clinical and Experimental Rheumatology 2024
SLE and environment / C. Cardelli et al.
lupus? A matched case-control study. Br J
Clin Pharmacol 2010; 70: 588-96. https://
doi.org/10.1111/j.1365-2125.2010.03733.x
118. PAPADOPOULOU E, SAROGLOU M, ISMAI-
LOS G et al.: Pearls for the diagnosis and
possible pathophysiological mechanisms of
valproic acid-induced lupus erythematosus:
A literature review. Lupus 2022; 31: 650-58.
https://doi.org/10.1177/09612033221088445
119. RAMOS-CASALS M, BRITO-ZERÓN P, MU-
ÑOZ S et al.: Autoimmune diseases induced
by TNF-targeted therapies: analysis of 233
cases. Medicine (Baltimore) 2007; 86: 242-
51. https://
doi.org/10.1097/md.0b013e3181441A68
120. MOULIS G, SOMMET A, LAPEYRE-MESTRE
M et al.: Is the risk of tumour necrosis factor
inhibitor-induced lupus or lupus-like syn-
drome the same with monoclonal antibod-
ies and soluble receptor? A case/non-case
study in a nationwide pharmacovigilance
database. Rheumatology (Oxford) 2014; 53:
1864-71. https://
doi.org/10.1093/rheumatology/keu214
121. WILLIAMS VL, COHEN PR: TNF alpha an-
tagonist-induced lupus-like syndrome: re-
port and review of the literature with impli-
cations for treatment with alternative TNF
alpha antagonists. Int J Dermatol 2011;50:
619-25. https://
doi.org/10.1111/j.1365-4632.2011.04871.x
122. DASANU CA, ALEXANDRESCU DT: System-
ic lupus erythematosus associated with pa-
clitaxel use in the treatment of ovarian can-
cer. South Med J 2008; 101: 1161-62. https://
doi.org/10.1097/smj.0b013e31818956f6
123. WIESENTHAL A, MATHERNE R, GIBSON B:
Capecitabine-induced systemic lupus ery-
thematosus and palmoplantar erythrodyses-
thesia. J Drugs Dermatol 2012; 11: 769-71.
124. HUANG JY, WU CH, SHIH IH et al.: Compli-
cations mimicking lupus are-up in a ure-
mic patient undergoing pegylated liposomal
doxorubicin therapy for cervical cancer.
Anticancer Drugs 2004; 15: 239-41.
https://doi.org/10.1097/00001813-2004030
00-00008
125. RASCHI E, ANTONAZZO IC, POLUZZI E et
al.: Drug-induced systemic lupus erythema-
tosus: should immune checkpoint inhibitors
be added to the evolving list? Ann Rheum
Dis 2021; 80. https://
doi.org/10.1136/annrheumdis-2019-215819
126. GRÖNHAGEN CM, FORED CM, LINDER M
et al.: Subacute cutaneous lupus erythema-
tosus and its association with drugs: a pop-
ulation-based matched case-control study
of 234 patients in Sweden. Br J Dermatol
2012; 167: 296-305. https://
doi.org/10.1111/J.1365-2133.2012.10969.x
127. BREHON A, MOGUELET P, GUÉGAN S et al.:
Discoid drug-induced lupus erythematosus
induced by antitumor necrosis factor agents
is a very rare subtype of cutaneous lupus:
Three cases and literature review. Dermatol
Ther 2020; 33: e13364.
https://doi.org/10.1111/dth.13364
128. BATAILLE P, LEBRUN-VIGNES B, TUBACH
F et al.: Proton pump inhibitors associated
with drug-induced lupus erythematosus.
JAMA Dermatol 2022; 158: 1208-10. https://
doi.org/10.1001/jamadermatol.2022.2421
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Introduction The relationship between Systemic lupus erythematosus (SLE) and Epstein-Barr virus (EBV) infection has been suggested for decades, but the underlying mechanism of the EBV influence on SLE development remains to be elucidated. Methods The goals of this research, which included 103 SLE patients and 99 controls, were to investigate the association of the parameters of EBV infection and SLE, to explore whether pooled demographic, clinical and EBV markers achieve a more significant effect on SLE development than each of them individually, and to evaluate EBV nuclear antigen 1 (EBNA1) and latent membrane protein 1 (LMP1) gene polymorphisms in isolates from SLE patients. Results Comprehensive results related to serological, molecular and sequence markers of EBV infection in SLE patients demonstrated even 24 times higher possibility of having SLE if there is the presence of anti-EBV-EA(D) (early antigen) IgG antibodies (OR=24.086 95%CI OR=2.86-216.07, p=0.004). There was the same distribution of glucocorticoids (p=0.130), antimalarials (p=0.213), and immunosuppressives (p=0.712) in anti-EBV-EA(D) IgG positive and negative SLE patients. Further, higher anti-EBV-EA(D) IgG antibodies titers were identified as independent factors associated with lymphopenia, hematological SLE manifestation (OR=1.041, 95%CI OR=1.01-1.08, p=0.025, while a higher titer of anti-CA (viral capsid antigen) IgG antibodies (OR=1.015, 95%CI OR=1.01-1.03, p=0.019) and positive RF (rheumatoid factors) (OR=4.871, 95%CI OR=1.52-15.61, p=0.008) were identified as independent factors associated with alopecia within SLE. Finally, novel data on EBV EBNA1 and LMP1 gene polymorphisms in lupus are reported. Conclusion The results support further investigation targeting EBV as a prognostic marker and therapeutic goal for lupus.
Article
Full-text available
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that predominantly affects women of childbearing age and is characterized by the damage to multiple target organs. The pathogenesis of SLE is complex, and its etiology mainly involves genetic and environmental factors. At present, there is still a lack of effective means to cure SLE. In recent years, growing evidence has shown that gut microbiota, as an environmental factor, triggers autoimmunity through potential mechanisms including translocation and molecular mimicry, leads to immune dysregulation, and contributes to the development of SLE. Dietary intervention, drug therapy, probiotics supplement, fecal microbiome transplantation and other ways to modulate gut microbiota appear to be a potential treatment for SLE. In this review, the dysbiosis of gut microbiota in SLE, potential mechanisms linking gut microbiota and SLE, and immune dysregulation associated with gut microbiota in SLE are summarized.
Article
Full-text available
Objectives: Patients with SLE have an increased risk of comorbidities and impaired survival. We aimed to assess if various thresholds of oral corticosteroids (OCS) can predict development of infections, comorbidities, malignancies and survival in SLE using data from national health registries in Sweden. Methods: All incident SLE cases, age >18 years, in Sweden (n = 5309) between 2005 and 2020 and matched population controls (n = 26545) were included and followed until 2020, total 257 942 patient years. Data from national registers were retrieved including information from the National Prescribed Drug Register. Risk factors were analysed using time-dependent Cox regression models. Results: Compared with no OCS, >0 to < 5.0 mg/day, 5.0-7.5 mg/day as well as > 7.5 mg OCS/day predicted development of infections (pneumonia, influenza, herpes zoster and urinary tract infection), osteoporosis, osteonecrosis, gastroduodenal ulcers, cataracts, hypertension and mortality (all p< 0.05). OCS >0 to < 5.0 mg/day was associated with lower hazard ratios for these comorbidities than higher doses of OCS. Fifteen years after diagnosis, 48% of patients were taking OCS at a median dose of 5.7 mg/day. A small reduction of OCS treatment 5 years after diagnosis in patients diagnosed with SLE 2006-2010 compared with 2011-2015 was observed, 49% vs. 46% respectively (p= 0.039). Conclusion: Results highlight the potential harm associated with even low OCS dose treatment in SLE and the need to judiciously use OCS at the lowest possible dose to maximize efficacy and minimize harm.
Article
Full-text available
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a wide range of clinical manifestations and a relapsing-remitting course. New data regarding pathogenic pathways, biomarkers and clinical manifestations of SLE are emerging, and new drugs and therapeutic protocols have been proposed to improve the control of disease activity. Furthermore, new insights into comorbidities and reproductive health in SLE patients are constantly emerging.This annual review aims to summarise the most relevant data on SLE that was published in 2022.
Article
Full-text available
A literature review on new-onset autoimmune connective tissue diseases (ACTDs) following COVID-19 is lacking. We evaluated potential associations between COVID-19 and the development of new-onset ACTDs. The “population” was adults with disease terms for ACTDs, including systemic lupus erythematosus (SLE), Sjogren’s syndrome, systemic sclerosis (SSc), idiopathic inflammatory myositis (IIM), anti-synthetase syndrome, mixed CTD and undifferentiated CTD, and “intervention” as COVID-19 and related terms. Databases were searched for English-language articles published until September 2022. We identified 2236 articles with 28 ultimately included. Of the 28 included patients, 64.3% were female, with a mean age was 51.1 years. The USA reported the most cases (9/28). ACTD diagnoses comprised: 11 (39.3%) IIM (including four dermatomyositis); 7 (25%) SLE; four (14.3%) anti-synthetase syndrome; four (14.3%) SSc; two (7.1%) other ACTD (one lupus/MCTD overlap). Of eight, four (14.3%) patients (including that with lupus/MCTD) had lupus nephritis. The average time from COVID-19 to ACTD diagnosis was 23.7 days. A third of patients were admitted to critical care, one for treatment of haemophagocytic lymphohistiocytosis in SLE (14 sessions of plasmapheresis, rituximab and intravenous corticosteroids) and nine due to COVID-19. 80% of patients went into remission of ACTD following treatment, while three (10%) patients died—one due to macrophage activation syndrome with anti-synthetase syndrome and two from unreported causes. Our results suggest a potential association between COVID-19 and new-onset ACTDs, notably in young females, reflecting more comprehensive CTD epidemiology. The most common diagnosis in our cohort was IIM. The aetiology and mechanisms by which ACTDs emerge following COVID-19 remain unknown and require further research.
Article
Full-text available
Background: Systemic lupus erythematosus (SLE) is a chronic autoimmune inflammatory disease characterised by immune dysregulation affecting multiple organs. Current anti-inflammatory treatments used in SLE are associated with unwanted side-effects. Dietary supplementation has been suggested as a safe and effective addition to conventional treatment, but evidence of efficacy in SLE or preventing associated comorbidities is uncertain. Methods: We identified literature on clinical trials focused on nutritional interventions in SLE aiming to improve inflammation and comorbidities. A systematic-type search on Embase, Medline, and the Cochrane Library, was conducted to identify nutritional interventions among SLE patients in the past 15 years that met our inclusion criteria. Results: We identified 2754 articles, of which 14 were eligible for inclusion based on our set criteria and were subsequently quality assessed. Vitamin D or E supplementation was associated with respective improvement of inflammatory markers or antibody production, but not disease activity scores in most studies. Despite their expected synergistic actions, the addition of curcumin on vitamin D supplementation had no additional effects on disease activity or inflammatory markers. Trials of omega-3 fatty acid supplementation presented significant reductions in ESR, CRP, disease activity, inflammatory markers, and oxidative stress, and improved lipid levels and endothelial function, while a low glycaemic index (GI) diet showed evidence of reduced weight and improved fatigue in patients. Conclusions: Different dietary guidelines can therefore be implicated to target specific SLE symptoms or therapeutic side-effects. This systematic review highlights the scarcity of larger and longer in duration trials with homogenous methodologies and verifiable outcomes to assess disease progression.
Article
Full-text available
An increasing number of studies have provided strong evidence that gut microbiota interact with the immune system and stimulate various mechanisms involved in the pathogenesis of auto-immune diseases such as Systemic Lupus Erythematosus (SLE). Indeed, gut microbiota could be a source of diagnostic and prognostic biomarkers but also hold the promise to discover novel therapeutic strategies. Thus far, specific SLE microbial signatures have not yet been clearly identified with alteration patterns that may vary between human and animal studies. In this study, a comparative analysis of a clinically well-characterized cohort of adult patients with SLE showed reduced biodiversity, a lower Firmicutes/Bacteroidetes (F/B) ratio, and six differentially abundant taxa compared with healthy controls. An unsupervised clustering of patients with SLE patients identified a subgroup of patients with a stronger alteration of their gut microbiota. Interestingly, this clustering was strongly correlated with the disease activity assessed with the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) score (p = 0.03, odd ratio = 15) and the identification of specific alterations involving the F/B ratio and some different taxa. Then, the gut microbiota of pristane-induced lupus and control mice were analyzed for comparison with our human data. Among the six differentially abundant taxa of the human disease signature, five were common with our murine model. Finally, an exhaustive cross-species comparison between our data and previous human and murine SLE studies revealed a core-set of gut microbiome species that might constitute biomarker panels relevant for future validation studies.
Article
Full-text available
Background Patients with systemic lupus erythematosus (SLE) are at increased risk of cardiovascular disease (CVD) compared to the general population. However, little is known about the effects of tobacco smoking on CVD in patients with SLE. Objective To systematically review and summarize the available literature regarding the effects of tobacco smoking on developing CVD in patients with SLE. Methods We retrieved relevant studies from the following databases: PubMed, EMBASE, Web of Science and China National Knowledge Internet (CNKI) database. Two reviewers independently reviewed the eligible studies, assessed their validity, and extracted relevant data. Sensitivity and subgroup analyses were performed to distinguish sources of heterogeneity. Results A total of 10 studies, which comprised 6984 participants, were included in the analysis. The overall quality of evidence was rated as moderate to low. The smoking prevalence among CVD patients was 39.28% (271/690), which was higher than 31.36% (1974/6294) among non-CVD patients. Compared with never-smokers, the risk of developing CVD in current smokers was 1.42 (95% CI: 1.21–1.66). No significant publication bias was found in our meta-analysis. Conclusions In spite of the several negative results, this study found that current smokers with SLE have an increased risk of developing CVD, although most of the included studies were in low-to-moderate quality. Systematic Review Registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022338109.
Article
Full-text available
Objective Recent studies reported that SLE is characterised by altered interactions between the microbiome and immune system. We performed a meta-analysis of publications on this topic. Methods Case–control studies that compared patients with SLE and healthy controls (HCs) and determined the diversity of the gut microbiota and the abundance of different microbes were examined. Stata/MP V.16 was used for the meta-analysis. A Bonferroni correction for multiple tests was used to reduce the likelihood of false-positive results. Results We included 11 case–control studies that examined 373 patients with SLE and 1288 HCs. These studies were performed in five countries and nine cities. Compared with HCs, patients with SLE had gut microbiota with lower Shannon-Wiener diversity index (weighted mean difference=−0.22, 95% CI −0.32 to –0.13, p<0.001) and lower Chao1 richness (standardised mean difference (SMD)=−0.62, 95% CI −1.04 to –0.21, p=0.003). Patients with SLE had lower abundance of Ruminococcaceae (SMD = −0.49, 95% CI −0.84 to −0.15,p=0.005), but greater abundance of Enterobacteriaceae (SMD=0.45, 95% CI 0.01 to 0.89, p=0.045) and Enterococcaceae (SMD=0.53, 95% CI 0.05 to 1.01, p=0.03). However, only the results for Ruminococcaceae passed the Bonferroni correction (p=0.0071). The two groups had no significant differences in Lachnospiraceae and Bacteroides (both p>0.05). Patients with SLE who used high doses of glucocorticoids had altered gut microbiota based on the Chao1 species diversity estimator, and hydroxychloroquine use appeared to reduce the abundance of Enterobacteriaceae. Conclusions Patients with SLE have imbalanced gut microbiota, with a decrease in beneficial bacteria and an increase in harmful bacteria. Drugs used to treat SLE may also alter the gut microbiota of these patients.
Article
Full-text available
Objectives A European League Against Rheumatism taskforce was convened to review the literature and develop recommendations on lifestyle behaviours for rheumatic and musculoskeletal diseases (RMDs). Methods Six lifestyle exposures (exercise, diet, weight, alcohol, smoking, work participation) and seven RMDs (osteoarthritis, rheumatoid arthritis, axial spondyloarthritis, psoriatic arthritis, systemic lupus erythematosus, systemic sclerosis, gout) were considered. The taskforce included health professionals in rheumatology, geriatricians, epidemiologists, public health experts, people with RMDs and exposure domain experts. Systematic reviews were conducted to gather available evidence, from which recommendations were developed. Results Five overarching principles and 18 specific recommendations were defined based on available evidence. The overarching principles define the importance of a healthy lifestyle, how lifestyle modifications should be implemented, and their role in relation to medical treatments. Exercise recommendations highlight the safety and benefits of exercise on pain and disability, particularly among people with osteoarthritis and axial spondyloarthritis. The diet recommendations emphasise the importance of a healthy, balanced diet for people with RMDs. People with RMDs and health professionals should work together to achieve and maintain a healthy weight. Small amounts of alcohol are unlikely to negatively affect the outcomes of people with RMDs, although people with rheumatoid arthritis and gout may be at risk of flares after moderate alcohol consumption. Smokers should be supported to quit. Work participation may have benefits on RMD outcomes and should be discussed in consultations. Conclusions These recommendations cover a range of lifestyle behaviours and can guide shared decision making between people with RMDs and health professionals when developing and monitoring treatment plans.