The Influence and Therapeutic Effect of Microbiota On LES

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Microbiological Research 281 (2024) 127613

Contents lists available at ScienceDirect

Microbiological Research
journal homepage: www.elsevier.com/locate/micres

The influence and therapeutic effect of microbiota in systemic


lupus erythematosus
Chuzi Mo a, 1, Jiaming Bi a, 1, Siwei Li a, Yunhe Lin a, Peiyan Yuan a, Zhongjun Liu a, *, Bo Jia b, *,
Shuaimei Xu a, *
a
Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
b
Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China

A R T I C L E I N F O A B S T R A C T

Keywords: Systemic erythematosus lupus (SLE) is an autoimmune disease involving multiple organs that poses a serious risk
Lupus erythematosus to the health and life of patients. A growing number of studies have shown that commensals from different parts
Microbiota of the body and exogenous pathogens are involved in SLE progression, causing barrier disruption and immune
Therapeutics
dysregulation through multiple mechanisms. However, they sometimes alleviate the symptoms of SLE. Many
factors, such as genetic susceptibility, metabolism, impaired barriers, food, and sex hormones, are involved in
SLE, and the microbiota drives the development of SLE either by depending on or interacting with these factors.
Among these, the crosstalk between genetic susceptibility, metabolism, and microbiota is a hot topic of research
and is expected to lay the groundwork for the amelioration of the mechanism, diagnosis, and treatment of SLE.
Furthermore, the microbiota has great potential for the treatment of SLE. Ideally, personalised therapeutic ap­
proaches should be developed in combination with more specific diagnostic methods. Herein, we provide a
comprehensive overview of the role and mechanism of microbiota in lupus of the intestine, oral cavity, skin, and
kidney, as well as the therapeutic potential of the microbiota.

1. Introduction localisation can lead to a loss of tolerance and development of autoim­


mune diseases (Belkaid YHarrison, 2017; Choi et al., 2020; Ruff et al.,
Systemic lupus erythematosus (SLE), a chronic autoimmune disease 2020). The disturbed commensals and infections play a role in autoim­
with a striking female predominance (sex ratio: 9:1) (Krasselt MBaer­ mune diseases through several mechanisms, including hidden/cryptic
wald, 2019), is characterised by an abnormal immune response that antigens, epitope spread, anti-idiotypes, molecular mimicry, adjuvant or
results in inflammatory reactions in multiple organs or systems such as bystander effects, and antigenic complementarity (Christen, 2014;
the kidney, the skin and the intestine (Rasaratnam IRyan, 1997). The Root-Bernstein and Fairweather, 2014; Ruff et al., 2020). Additionally, a
onset and progression of SLE are influenced by genetic, environmental, growing number of metabolomic studies are looking for potential bio­
and stochastic factors (Nandakumar KSNündel, 2022). Genome-wide markers for the diagnosis and disease activity surveillance of SLE (Li
association studies (GWASs) have substantially enhanced our under­ et al., 2020a; Zhang et al., 2021).
standing of the genetic basis of SLE. Using this method, more than 40 SLE has distinct and independent clinical manifestations in different
susceptibility loci have been identified and confirmed to be associated organs; patients experience low-activity SLE in certain organs but not in
with SLE. Most of these related genes participate in crucial processes, the entire body (Oku KAtsumi, 2018; Aringer and Johnson, 2020).
including the processing of immune complexes, Toll-like (TLR) receptor However, the treatment options for SLE primarily target immune regu­
signalling, and the production of type I interferon (IFN I) (Cui et al., lation and immunosuppression to maintain long-term remission or
2013). In recent years, increasing evidence has shown that the micro­ reduce disease activity (Pan et al., 2021). This approach lacks specificity
biota plays an important role in the pathogenesis of autoimmune dis­ and adequate attention to the affected organs in cases of heterogeneous
eases. Dysregulation of microbiota abundance, function, or anatomical SLE (Narváez, 2020). Given this situation and the role of microbiota on

* Corresponding authors.
E-mail addresses: liuzhongjundr@smu.edu.cn (Z. Liu), dentist-jia@163.com (B. Jia), xushuaimei@smu.edu.cn (S. Xu).
1
These authors contributed equally to the article.

https://doi.org/10.1016/j.micres.2024.127613
Received 14 September 2023; Received in revised form 18 December 2023; Accepted 9 January 2024
Available online 14 January 2024
0944-5013/© 2024 Elsevier GmbH. All rights reserved.
C. Mo et al. Microbiological Research 281 (2024) 127613

SLE, it is important to focus on the effects of disturbed commensals and et al., 2010; Tian and Zhang, 2010).
infections in different lupus-affected sites, and therapeutic approaches The role of microbiota in the development of this disease remains
that can regulate or target microbial disturbance and infections are ur­ inadequate and unclear. The proposed microbial trigger factors of LMV
gently required. include infections changing the intestinal microbiota, cytomegalovirus
This review discusses the disturbance of commensal microbiota and infections, and animal viruses (Tian and Zhang, 2010). However, the
their roles on various organs and sites to shed light on the bidirectional specific mechanisms and their interactions with the gut microbiota have
relationship between SLE and microbiota (He et al., 2020; Huang et al., not been reported. Pneumatosis cystoides intestinalis is an uncommon
2020), and the effects of infections in SLE are also summarised (James type of SLE-related gastrointestinal involvement with only 14 reported
Jog and James, 2020). Certain serological markers and changes in mi­ cases, and bacteria that produce gas in the mucosa may be involved in its
crobial abundance are potential markers for the diagnosis (Jog et al., pathogenesis (Tian and Zhang, 2010). In addition, SLE-related PLGE has
2019; Huang et al., 2020). In addition, therapeutic modalities based on been reported to be potentially associated with intestinal infections
microbiota are discussed (Manfredo Vieira et al., 2018; Esmaeili et al., (Chng et al., 2010).
2021). Although most of these methods are still in the experimental The lack of adequate experimental studies on the gastrointestinal
stage and serve as supplementary treatment, their specificity and effi­ manifestations of SLE could be due to the diversity of its manifestations,
cacy show promise for the development of individualised SLE unclear mechanisms, and complicated diagnostic processes resulting
treatments. from a broader differential diagnosis (Brewer and Kamen, 2018). In
addition, effective and reliable animal and cellular models of
2. Microbiota in lupus affected sites SLE-related gastrointestinal involvement have not been established,
leading to the inability to conduct specific studies. Current treatments
2.1. Gastrointestinal tract for gastrointestinal manifestations include corticosteroids and immu­
nosuppressants (Li et al., 2017); however, these can lead to a loss of the
The intestinal epithelial cells limit the penetration of exogenous overall diversity of the gastrointestinal microbiota, thus aggravating
antigens and leakage of endogenous substances by forming tight junc­ autoimmune diseases (Flannigan et al., 2018; Wei et al., 2020). Simul­
tions (TJs), and gut microbiota plays an important role in maintaining taneously, abnormal microbiota and a leaky gut are common in auto­
the gut permeability and the immunity of an organism (Fasano, 2012; immune diseases (Christovich and Luo, 2022). Studies on the
Miyamoto et al., 2015; Erttmann et al., 2022). In contrast, mucosal interrelationship between the microbiota and autoimmune diseases, as
barrier dysfunction that increases intestinal permeability (known as well as microbiota-related therapies, are ongoing. If the microbiological
leaky gut syndrome), and microbial disturbance can be observed in basis of SLE can be explored by establishing reproducible models of
various diseases, including autoimmune diseases (Hu et al., 2016; Kin­ gastrointestinal manifestations and experimental protocols, effective
ashi YHase, 2021). For example, lupus-derived Ruminococcus gnavus and specific treatments for SLE-related gastrointestinal involvement can
strains can induce high levels of intestinal permeability, which is asso­ be developed.
ciated with R. gnavus translocation to the mesenteric lymph nodes and
increased serum levels of zonulin, a regulator of TJ formation in the gut 2.1.2. The crosstalk between SLE and intestinal microbiota
barrier (Silverman et al., 2022). Additionally, alterations in the intesti­ Both intestinal leakage and SLE-specific intestinal microbiota exac­
nal microbiota change the metabolic environment and activate specific erbate SLE (Thim-Uam et al., 2020). Current studies on SLE microbial
pattern recognition receptors in epithelial cells, thereby inducing a disturbance and translocation have mostly focused on abnormal IFN I
pro-inflammatory state. Cytokines, such as tumour necrosis factor production and autoimmune reactions (Fig. 1).
secreted in response, result in the destruction of the intestinal barrier (Le The crosstalk between genetic susceptibility, alterations in the
et al., 2020). Epithelial barrier leakage caused by other stimuli (e.g. microbiota, and metabolism has also been the emphasis of current
chronic stress, dietary imbalance, or antibiotic administration) leads to studies. A metabolomic profiling study of serum and faecal samples of
tissue microinflammation by triggering microbial disturbance and bac­ patients with SLE showed a deficiency of gut microbiome mediated
terial translocation to the interepithelial and subepithelial areas. transformation of bile acids and its potential effects on host lipid
Although open epithelial TJs drain immune cells and pro-inflammatory metabolism (He et al., 2020). Lipid metabolism is important for the
molecules from subepithelial inflammation, they allow the entry of regulation of inflammation in acute and chronic diseases, and various
foreign substances, such as allergens and microbial product, deeper into lipids have immunomodulatory and proinflammatory/anti-
the tissues (Sugita et al., 2018; Akdis, 2021). inflammatory properties (Andersen, 2022). Moreover, patients with
Therefore, the intestinal barrier function and gut microbial SLE have elevated low-density lipoprotein cholesterol and
homoeostasis are critical for the health of an organism. Current studies medium-chain/free fatty acids as well as reduced high-density lipopro­
also suggest that intestinal homoeostasis and leakage are potential tein and long-chain fatty acids (Robinson et al., 2022). Interestingly,
contributors to SLE-associated gastrointestinal involvement (Brewer and dysfunctional high-density lipoprotein, which becomes
Kamen, 2018) and play a non-negligible role in LN (see Microbiota and pro-inflammatory during inflammation, was described in 48.2% of fe­
LN). Simultaneously, microbial disturbance and translocation interact male patients with SLE, and greatly increased the risk of developing
with the immune system and constitute the mechanism underlying subclinical atherosclerosis (McMahon et al., 2009). Based on these
abnormal IFN I production and autoimmune responses in SLE (Manfredo studies so far, it is urgent to fill the gap that whether intestinal micro­
Vieira et al., 2018). biota plays a direct or significant role to lipid metabolism in SLE.
Additionally, microbiota under the SLE condition significantly im­
2.1.1. The role of microbiota in SLE-related gastrointestinal involvement pacts amino acid metabolism. A recent combined plasma metabolomic
Gastrointestinal involvement is a common complaint in 40–60% of and transcriptomic analysis revealed that plasma histidine levels are
patients with SLE (Frittoli et al., 2021), with three main manifestations: potential biomarkers for patients with SLE, and that this decrease is
intestinal pseudo-obstruction (IPO), lupus mesenteric vasculitis (LMV), associated with damage accrual (Iwasaki et al., 2023). Germ-free mice
and protein-losing gastroenteropathy (PLGE). SLE-related gastrointes­ that underwent faecal microbiota transplantation from patients with
tinal lesions, such as mesenteric vasculitis and thrombosis, are associ­ SLE showed markedly altered histidine metabolism and developed
ated with lupus anticoagulants and cause severe colonic ischaemia, several lupus-like phenotypic characteristics (Ma et al., 2021).
leading to intestinal infarction, perforation, and SLE-related pancrea­ Furthermore, patients with SLE exhibit defects in macrophage-
titis, which are life-threatening when left untreated. Clinically, these mediated phagocytosis of apoptotic cells (efferocytosis), whereas the
conditions require urgent surgery (Takeno MIshigatsubo, 2006; Chng male murine lupus model exhibits enhanced splenic macrophage

2
C. Mo et al. Microbiological Research 281 (2024) 127613

Fig. 1. The effect of gut microbiota on SLE. In SLE, disturbance occurs in the intestinal microbiota, leading to abnormal IFN I production or autoimmune responses in
the local immune or tissue cells. Some microbiota and their products can induce the above-mentioned responses by translocating to other organs or tissues via
intestinal leakage. IFN I dysregulation can cause autoantibody production, aggravating SLE progression. Created using BioRender.com. Abbreviations: AhR, aryl
hydrocarbon receptor; B. theta, Bacteroides thetaiotaomicron; ERV gp70, endogenous retrovirus glycoprotein 70; ERV gp70-ICs, ERV gp70 immune complexes; GC B,
germinal centre B cells; GLB, gut-lymph barrier; GVB, gut-vascular barrier; JAM-A, junctional adhesion molecule-A; MLN, mesenteric lymph node; pDCs, plasma­
cytoid dendritic cells; RS, resistant starch; SCFAs, short-chain fatty acids; SFB, segmented filamentous bacteria; SLOs, secondary lymphoid organs; TFH, Follicular
helper T cell; Th17, T helper 17; TLR7, Toll-like receptor 7; TRP, tryptophan GALT, gut-associated lymphoid tissue; β2GPI, β2-glycoprotein I. (Tesser et al., 2021)
(Manfredo Vieira et al., 2018) (Zegarra-Ruiz et al., 2019) (Choi et al., 2020) (Brown et al., 2022) (Shirakashi et al., 2022) (Greiling et al., 2018a) (Steinberg
et al., 1969).

efferocytosis in an androgen-dependent manner. Macrophage effer­ composition of gut virome has altered in patients with SLE (Chen et al.,
ocytosis was deficient in female mice, but was restored when fed with 2022), and crAss-like phages, one of the main components of a healthy
male microbiota (caecal contents). One of the intestinal microbial me­ gut virome, have been significantly reduced (Tomofuji et al., 2022). In
tabolites, phytanic acid, was reported to restore macrophage effer­ addition, an in vitro study showed that virus-like particles extracted
ocytosis activity of the splenic macrophages of female mice by activating from faecal samples of patients with SLE increased IFN-α production in
the PPARγ/LXR signalling pathway, which may have an impact on the immune cells, suggesting a potential effect of SLE-related virus-like
development of SLE (Harder et al., 2023) and also illustrated that an­ particles on SLE pathogenesis (Chen et al., 2023); nevertheless, this
drogens affect both immunity and microbial composition, shedding light conclusion needs to be further verified by more complete research.
on the disproportionate prevalence of SLE in males and females. However, there is insufficient evidence to support the idea that mi­
The gut microbiota in the context of SLE disease not only holds the crobial disturbance initiates SLE. A research (Choi et al., 2020) sug­
potential to foster the progression of SLE itself but also may contribute to gested that gut microbiota abnormality did not trigger but amplified
the advancement of cardiovascular disease among individuals with SLE. autoimmune phenotypes and that a leaky gut does not necessarily imply
It was reported that transplantation of intestinal microbiota from hy­ lupus development. In their study, lupus autoimmune response was
pertensive SLE mice to recipient normotensive mice led to elevated triggered in healthy control mice after transplanting faeces from aged
blood pressure, with Bacteroides and Parasutterella being the major lupus-prone mice (which had exhibited lupus-associated phenotypes),
harmful bacteria. However, vancomycin or broad-spectrum antibiotic whereas faeces from young lupus-prone mice (which did not develop
treatment can inhibit hypertension, ameliorate endothelial dysfunction lupus-associated phenotypes) did not elicit a similar effect. Furthermore,
and vascular oxidative stress and reduce aortic Th17 infiltration (de la bacterial translocation was not observed prior to the production of au­
Visitación et al., 2021). Another study showed that endothelial toantibodies in lupus-prone mice, and a small number of translocated
dysfunction in SLE was associated with increased superoxide production Staphylococcus did not significantly correlate with autoimmune mani­
initiated by NADPH oxidase and eNOS phosphorylation at the inhibitory festations. In contrast, it has been shown that the faecal microbiome of
Thr495 (Toral et al., 2019). The activity of these factors returned to SLE mice induced the production of anti-dsDNA antibodies, stimulated
normal levels after oral gavage of Lactobacillus fermentum in lupus-prone inflammatory responses, and altered the expression of SLE susceptibility
mice, thus reducing endothelium-dependent vasodilator responses to genes in germ-free mice (Ma et al., 2019). A recent study offered a more
acetylcholine of aortas (de la Visitación et al., 2020). detailed demonstration of the roles of host genetics and the gut micro­
The virus is also a component of gut microbiota (Shanahan et al., biota in autoimmunity. This shows that defective TCR signalling disrupts
2021), but the effect of gut viruses on SLE remains insufficiently studied. the gut microbiota by inhibiting the positive selection of certain
Based on results of current studies, the diversity and taxonomic microbe-reactive T cells. Altered gut microbiota promotes the

3
C. Mo et al. Microbiological Research 281 (2024) 127613

differentiation of Th17 cells and the development of systemic autoim­ 2.3. Skin
mune diseases (Shirakashi et al., 2022). In individuals susceptible to
autoimmune diseases, the genetic factor-mediated changes in immunity Cutaneous lupus erythematosus (CLE) affects the skin and is divided
and metabolism, and the consequential responses of the microbiota into two subgroups based on the lesion type: subacute cutaneous LE
remain an area to be further explored. (SCLE) and discoid lupus erythematosus (DLE). Acute CLE (ACLE) is a
common manifestation of SLE (Li et al., 2020c). Discoid lesions and
2.2. Oral cavity butterfly rashes are typical SLE skin lesions (Zhao et al., 2020) that occur
in 5–25% of patients with CLE. These lesions can progress to SLE.
Oral lesions are common lupus symptom (AlSaleh et al., 2008; However, clear predictive guidelines are still lacking (Zhou et al., 2020).
Khatibi et al., 2012; Kudsi et al., 2021). As described for the gastroin­ Skin commensals play an important role in the maturation and
testinal manifestations (Brewer and Kamen, 2018), they are interpreted homoeostasis of skin immunity (Chen et al., 2018) and a disturbance in
as mucosal counterparts of cutaneous LE because they represent mucosal the skin microbiota causes inflammatory skin diseases such as atopic
lesions (Nico et al., 2008). dermatitis (Condrò et al., 2022). The results of 16 S rRNA sequencing of
Epstein-Barr virus (EBV) DNA was present in 71.4% of the patients SLE skin lesions revealed a lower alpha diversity of the microbiota and
with SLE-associated oral lesions, and most patients with Epstein-Barr different bacterial communities compared to adjacent healthy skin cells.
virus (EBV) DNA-positive SLE present with oral lesions (Buonavoglia At the species level, Staphylococcus aureus and S. epidermidis remained
et al., 2021). However, the relationship between systemic or local EBV two strong discriminative signatures for SLE lesions. Therefore, S. aureus
infection and oral lupus lesions requires further research. It is currently and S. epidermidis may serve as markers for the diagnosis of SLE.
reported that different regions of the EBV EBNA-1 protein cause auto­ Notably, the ratio of Firmicutes/Bacteroidetes of the skin microbiota of
immunity through molecular mimicry, epitope expansion, and other SLE patients is higher, contrary to previous observations from the in­
mechanisms, which in turn lead to the clinical characteristics of SLE testinal microbiota of SLE patients. This is possibly due to the different
(James Jog and James, 2020). A cohort study found a strong association bacterial profiles of the skin and intestines (Huang et al., 2020).
between the serological markers of EBV and SLE, with elevated numbers Furthermore, based on the gut-skin axis, the gut affects skin health
of EBV-infected cells, increased viral loads, and higher early antigen IgG owing to its immune and metabolic properties, and a highly diverse gut
levels in patients with SLE. Moreover, serological reactivation of EBV microbiota can affect skin microbiota; however, the exact mechanisms
increases the likelihood of transitioning to SLE (Jog et al., 2019). Sub­ involved in this relationship are not fully understood, and factors such as
sequently, EBV-specific CD8+ cytotoxic T cell damage and irregular altered gut permeability and immune and endocrine factors may also be
cytokine production in plasmacytoid dendritic cells (DCs) and involved (Sinha et al., 2021). However, probiotic therapy reduces dis­
CD69+CD4+ T cells occur in patients with SLE (James and Robertson, ease activity and decreases corticosteroid dose (Navarro-López et al.,
2012). 2018). IL-37b considerably increases the diversity of the skin microbiota
In addition to EBV, disturbance in the oral microbiota of patients by modulating autophagy in atopic dermatitis and reducing intestinal
with SLE is not negligible. The plaque index was twice as high in patients bacterial diversity and metabolite secretion, substantially alleviating
with SLE than in healthy controls despite brushing with fluoride at the eosinophil-mediated allergic inflammation (Hou et al., 2020).
same rate. Moreover, the abundance and diversity of the oral microbiota Although there are reports describing changes in the microbiota of
in patients with SLE have decreased, creating a favourable environment LE skin lesions, few have demonstrated the direct impact of the micro­
for pathogenic strains to flourish. A microbial disturbance between acid- biota on disease progression. Therefore, further studies on the rela­
producing uropathogenic bacteria and alkaline-producing commensal tionship between the microbiota and the development of LE skin lesions
bacteria present in the oral cavity increases the risk of caries in patients are needed to deepen our knowledge of how the microbiota interacts
with SLE (Yang et al., 2018). With respect to periodontitis, patients with with the immune system. Therefore, microbial markers for the accurate
SLE exhibit a higher prevalence of periodontitis at a younger age and diagnosis of LE skin lesions and new therapeutic strategies are urgently
have more severe forms of periodontitis with higher bacterial loads and required.
decreased microbial diversity (Corrêa et al., 2017). In addition, there is a
strong positive correlation between disease activity and oral health 3. Microbiota and LN
status in patients with SLE. The prevalence of dental caries, periodon­
titis, and oral mucosal lesions was higher in patients with SLE than in LN is a form of glomerulonephritis and is one of the most severe
those without SLE (Aurlene et al., 2020). A survey of 91 female patients manifestations of SLE. Twenty-five to fifty percent of unselected patients
with SLE showed that low-grade systemic inflammation influencing SLE with SLE develop kidney disease symptoms at the time of disease onset
disease activity and severity was correlated with oral microbiota ab­ and up to 60% of adult patients with SLE experience these signs or
normality in periodontal diseases (Pessoa et al., 2019). A two-sample symptoms over the course of the disease (Anders et al., 2020). The
Mendelian randomisation (MR) analysis suggested that periodontitis mechanisms underlying LN include genetic polymorphisms, environ­
was likely causally associated with an increased risk of SLE, although mental triggers, complement and neutrophil extracellular traps, intrinsic
one of its analyses (MR–Egger regression) failed to find a significant and adaptive immunity, and intrarenal inflammation (Lintner et al.,
causal association (p = 0.394) (Bae SCLee, 2020). Bouchra et al. found a 2016; Yin et al., 2021; Yu et al., 2022). Studies have been conducted on
possible bidirectional association between periodontitis and SLE the use of drugs aimed at specific targets in combination with immu­
through recent clinical and translational research that should be nosuppressants for the treatment of SLE, such as the combination of
considered when managing patients with SLE (Sojod et al., 2021). prednisolone and tacrolimus (a calcineurin inhibitor). The rate of renal
In summary, the mechanism by which SLE impairs the homoeostasis response was superior to that of a combination of prednisolone and
of the oral microbiota and how this disturbance causes oral LE lesions mycophenolate mofetil; however, the long-term results showed a higher
remains unclear. Similarly, although there are a growing number of proportion of proteinuric and nephritic renal flares (although with no
surveys on the relationship between periodontitis and the risk of significance) (Mok et al., 2016). Patients who received belimumab plus
developing SLE, there is a lack of direct evidence that periodontitis plays standard therapy had a higher primary efficacy renal response than
a role in the development of oral LE lesions. Therefore, more in-depth those who received standard therapy alone; however, the percentages
research should be conducted to understand this relationship and were both < 50% (Furie et al., 2020). Therefore, there is an urgent need
develop strategies for managing oral LE in patients with SLE. Pathogens to develop novel therapeutic methods for exploring the pathogenesis of
that can cause infections and symptomatic management to alleviate oral LN.
LE in SLE patients require special attention. Inflammation and damage to the kidneys are significantly influenced

4
C. Mo et al. Microbiological Research 281 (2024) 127613

by microbiota. Intestinal microbiota disturbance and barrier disruption 2016; Khoryati et al., 2016; Monteith et al., 2016; Shen et al., 2022).
result in microbiota producing toxins responsible for renal damage Peripheral blood Mx1 is a potential marker for the diagnosis of SLE and
(Mosterd et al., 2021), aberrant immune cell activation, antibody LN. It is highly expressed in the kidneys of patients with LN and its
overproduction, immune complexes, inflammatory agents, and inflam­ expression decreases following immunosuppressive treatment (Shimizu
matory cell infiltration, all of which can directly or indirectly harm the et al., 2018). In addition, Mx1 inhibits various viral families, including
renal parenchyma (Chi et al., 2021; Haniuda et al., 2021). Some viruses negative-stranded RNA viruses (Verhelst et al., 2013). Shojiro et al.
can cause renal damage through cytopathic actions or indirectly through demonstrated that Mx1 was expressed primarily in the mesangial region
immune-mediated glomerulopathy, tubulointerstitial disease, and other in the kidney and also found that the knockdown of IFN-α in human
conditions (Prasad et al., 2019). Similar processes also occurred in the thylakoid cells (MCs) decreased Mx1 expression induced by
LNs (Fig. 2 and below). Current approaches to LN management rely on polyinosinic-polycytidylic acid (poly IC; an authentic viral
high-dose corticosteroids combined with broad-spectrum immunosup­ double-stranded RNA). This suggests that the innate antiviral immunity
pressive therapy. This approach has limitations in LN treatment because is involved in the MCs of patients and induces the expression of Mx1 in
of its many side effects, including life-threatening infections (Parikh and MCs (Watanabe et al., 2013). However, there is a paucity of definitive
Rovin, 2016; Fontana et al., 2018). Disturbance of the commensal evidence linking Mx1 to viral infection in patients with LN, and further
microbiota is strongly associated with the development and progression research is needed to determine the link between and the consequences
of SLE and LN (Valiente et al., 2022). of the potential upstream virus and LN infection.
Secondary viral infections after LN treatment noticeably influence
the disease progression. A retrospective study showed that cytomega­
3.1. Infection and LN lovirus (CMV) was the most common pathogen among infected hospi­
talised LN patients (Liu et al., 2018), and serum anti-CMV antibodies
3.1.1. Viral infection were more prevalent in patients with SLE and a history of LN than in
As early as 1968, Fresco (Grausz et al., 1970) observed clustered those without nephritis (Brunekreef et al., 2021). CMV infection may
filamentous structures and dense deposits in the glomerular basement lead to failure of antibiotic therapy and make immunosuppressive
membrane of a patient with LN, and named them mucoviruses based on therapy detrimental. Despite receiving high-dose steroid therapy, the
their morphology. Since then, other researchers have published com­ patient with LN and CMV infections experienced persistent proteinuria
parable findings in patients with SLE. It has been hypothesised that SLE and active sedimentation, indicative of active renal lupus. Additionally,
could result from an immune reaction to cells carrying a foreign antigen notable ethnic differences have been reported between CMV-infected
and that this virus is the antigen that causes the initial insult that triggers and uninfected patients. Furthermore, baseline urine protein creati­
autoimmune mechanisms (Györkey et al., 1969). nine ratio levels were higher in the CMV-infected cohort than in unin­
The presence of myxovirus resistance protein 1 (Mx1) also indirectly fected individuals (Liu et al., 2018; Sebastiani et al., 2019).
suggests potential antiviral immunity in the kidneys of LN patients. Mx1
is an important link in the pathogenesis of SLE and is induced by IFN I, 3.1.2. Other infections
which is abnormally produced through several mechanisms such as the Cryptococcosis is a common pathogen that causes fungal meningitis.
activation of TLR7 and TLR9, defects in clearing apoptotic debris, and Cryptococcal meningitis (CM) is rare in patients with LN. Patients with
oxidised mitochondrial nucleoids (Haller OKochs, 2011; Caielli et al.,

Fig. 2. Positive and negative effects of identified microbiota on LN. Various commensal and exogenous microbiota are involved in the progression and amelioration
of LN. They cause changes in multiple kidney factors, and lead to different outcomes in terms of renal function and pathology. Created using BioRender.com.
Abbreviations: BUN, blood urine nitrogen; Cre, creatinine; IgG 2a: immunoglobulin G 2a; IL-10/IL-6, interleukin-10/interleukin-6; MCP-1: Monocyte chemo­
attractant protein-1; RBP, renin-binding protein; RS, resistant starch; Th 17, T helper cell 17; Treg, Regulatory T cell; UPCR, urine protein-to-creatinine ratio.
(Al-Quraishy et al., 2013) (Johnson et al., 2015) (Mu et al., 2017) (Li et al., 2020b) (Zegarra-Ruiz et al., 2019) (Valiente et al., 2022) (Chen and Chen, 2020) (Liu
et al., 2018).

5
C. Mo et al. Microbiological Research 281 (2024) 127613

LN and CM had considerably higher prednisone doses and SLE Disease adverse effects under certain genetic and environmental conditions
Activity Index scores than those without CM. The mortality rate of pa­ (Zegarra-Ruiz et al., 2019). In fact, the gut microbiota is known to be
tients with LN and CM is 25% and is associated with low serum albumin influenced by both genetic factors and the environment, exerting a
levels and high cumulative doses of prednisolone (Chen and Chen, significant impact on the development of diseases (Yoo et al., 2021).
2020). Furthermore, once the homoeostasis is disrupted, it can potentially
By contrast, tropical infections, particularly malaria, may protect cause harm to the body (Zheng et al., 2020). In addition, potential dif­
against SLE. Epidemiological studies have shown that people in the rural ferences in the strains used in the study may also serve as a significant
areas of tropical Africa and Asia, where malaria is common, rarely factor in experiment results.
develop SLE (Adebajo, 1997). It has been reported that young Diet can also affect LN by altering the intestinal microbiota. The pH
lupus-prone mice infected with Plasmodium have a higher survival rate of drinking water affected LN by alterations the microbial composition
and lower oxidative stress in renal tissues (Al-Quraishy et al., 2013). (Johnson et al., 2015). The intestinal microbiota of lupus-prone mice
However, malarial infection is also associated with the production of was characterised by a notable reduction in Lactobacillaceae, which was
large amounts of autoantibodies, and DNA-reactive antibodies in ma­ negatively correlated with signs of lupus (lymphadenopathy and
laria are comparable to those in SLE and may play a role in the devel­ glomerulonephritis) and was restored by vitamin A (Zhang et al., 2014),
opment of immune deposits in nephritic malarial kidneys (Lloyd et al., consistent with the findings of Mu et al. (2017). Furthermore, predicted
1994). metagenomes showed that retinoic acid (a vitamin A metabolite)
Therefore, primary and secondary infections caused by immuno­ reversed numerous lupus-associated changes in microbial activation,
suppressive agents should be considered. Doses of immunosuppressive deviating from the control (Zhang et al., 2014). Additionally, resistant
agents and possible infection-related symptoms should be evaluated starch (fermented to short-chain fatty acids) improved LN by lowering
scientifically. However, in vitro and animal model studies are necessary intestinal epithelial permeability and IFN-I expression (Zegarra-Ruiz
to elucidate the precise mechanisms of the pathogens in LN, as most et al., 2019), and prevented the development of hypertension in murine
studies assessing the effects of infection on LN have mainly focused on lupus model apart from renal injury (Moleón et al., 2023).
clinical indicators.
4. Progress in microbiota-associated treatment of SLE
3.2. Intestinal microbial homoeostasis and LN
Despite advances in SLE treatment, SLE remains associated with high
Not only is the intestinal microbiota important for gut homoeostasis, morbidity and premature death. Furthermore, an overreliance on
but several studies have shown that gut microbiota diversity is strongly corticosteroid therapy leads to long-term organ damage (Durcan et al.,
correlated with LN. The SLE subgroup with LN was markedly associated 2019). Supplementary therapeutic approaches using microbiota com­
with a decline in the Shannon index (Chen et al., 2021). Moreover, ponents are a viable and highly valuable choice in conjunction with
patients with LN have an increased R. gnavus abundance, which is analysis of the predominant pathogenic microbiota (Fig. 3).
directly proportional to the LN and overall LN activity (Azzouz et al.,
2019; Azzouz et al., 2023). R. gnavus can cross-react with native DNA, 4.1. Probiotic therapy
eliciting an immune response against double-stranded DNA (Azzouz
et al., 2019; Kim et al., 2019). Teri et al. used an inducible lupus mouse Probiotic therapy has been extensively studied for the treatment of
model for Bacteroides thetaiotaommicron (B. theta) mono-colonisation in autoimmune diseases (Jakubczyk et al., 2020). It is effective in allevi­
a germ-free setting and detected LN-like immune complex deposition in ating lupus-associated syndromes (de la Visitación et al., 2020), and may
88% of the glomeruli of the mice model (Greiling et al., 2018b). Gian­ also improve gut integrity (Ahmadi et al., 2020; de la Visitación et al.,
carlo et al. found that the transplantation of segmented filamentous 2020; Robles-Vera et al., 2020). However, probiotics may not be bene­
bacteria (SFB) can lead to intestinal microbiota disturbance and LN ficial for immunocompromised individuals, further exacerbating the
progression (Valiente et al., 2022). In contrast, chronic oral adminis­ condition (Kothari et al., 2019). Additionally, studies related to Heli­
tration of the probiotic L. fermentum in lupus-prone mice inhibited the cobacter pylori eradication have shown that probiotics alone cannot
infiltration of Th17 and Th1 cells, the increased pro-inflammatory cy­ provide sufficient treatment effects and thus should be combined with
tokines levels, NADPH oxidase activity in kidney and also significantly conventional therapy for better results (Feng et al., 2017; Losurdo et al.,
improved renal damage (de la Visitación et al., 2020). Interestingly, SFB 2018).
are substantially increased in the ileum of immunosuppressed mice, and Most studies of probiotics have focused only on their immunomod­
transplantation of L. plantarum restores SFB numbers (Fuentes et al., ulatory effects. L. delbrueckii and L. rhamnosus alone or in combination,
2008). It is valuable to further investigate the mechanism of can reduce the expression of chemokine receptors on the surface of DCs
L. plantarum, possibly lies at the antagonistic of probiotics (Peng et al., in SLE patients and prevent DCs migration, thereby alleviating inflam­
2022). Similarly, B. fragilis restored the Th17/Treg balance and allevi­ mation (Esmaeili et al., 2021). The combination of tacrolimus and
ated disease activity in a murine lupus model by lowering autoantibody L. acidophilus restored the dysregulated intestinal microbiota in an SLE
levels and alleviating LN symptoms (Li et al., 2020b). However, the mouse model, considerably inhibited Th17 cell differentiation, and
beneficial effects of Lactobacillus on LN can only be detected in female induced Treg cell differentiation (Kim et al., 2021). In addition, Akker­
and castrated male mice and not in normal male mice, suggesting that mansia muciniphila and L. plantarum were shown to not only promote an
the gut microbiota also control LN in a sex hormone-dependent manner anti-inflammatory environment by regulating cytokine levels in the
(Mu et al., 2017). The gut microbiome is involved in excretion and circulation and remodelling the gut microbiota but also to exert pro­
recycling of sex hormones. Sex steroids also influence the gut micro­ tective effects on intestinal barrier integrity by restoring the TJ structure
biome composition. The term “microgenderome” has been proposed to of colonic epithelial cells (Guo et al., 2023), indicating that the benefi­
reflect how sex hormones affect the gut microbiota (Thackray, 2019; cial effects of probiotics are multifaceted.
Yoon and Kim, 2021). Therefore, probiotics can be used as a complementary therapy to
Notably, one study indicated that L. reuteri worsens lupus by immunosuppressive therapy for SLE to reduce adverse effects and
increasing glomerulonephritis, plasmacytoid dendritic cell activation, maintain intestinal microbial homoeostasis and integrity in patients in
and IFN I expression, which is inconsistent with other studies showing an evidence-based manner.
the positive effects of Lactobacillus. This study used a TLR7-dependent
lupus mouse model in which the effects of L. reuteri on LN were
observed under certain conditions, suggesting that microbiota can show

6
C. Mo et al. Microbiological Research 281 (2024) 127613

Fig. 3. Envisioned microbiota-related therapeutic approaches to SLE lesions. The dominant pathogenic microbiota in the SLE lesions was identified using indi­
vidualised microbiome analysis and/or anti-microbial antibody testing. It can subsequently be treated with antimicrobial vaccines, probiotic therapies or FMT to
alleviate or cure lupus in the affected organs. Created using BioRender.com.

4.2. Faecal microbiota transplantation further investigation, this study has shown new evidence for FMT
application in individuals with SLE (Zhang et al., 2023).
Faecal microbiota transplantation (FMT) is a method to transfer stool In conclusion, FMT is a novel supplementary treatment modality for
from a healthy donor to a patient, directly changing the recipient’s gut patients with SLE, and it is worth investing in more clinical studies to
microbiota to normalise the composition and gain a therapeutic benefit clarify the efficacy and safety. Furthermore, the mechanism of FMT in
(Vindigni and Surawicz, 2017; Wang et al., 2019). FMT has proven to be SLE should also be further studied.
effective and relatively safe in various diseases including autoimmune
diseases (Matsuoka, 2021; Zeng et al., 2022; Zhang et al., 2022).
A study showed that early and short-term antibiotic exposure led to a 4.3. Vaccination
significant increase in serum anti-dsDNA autoantibodies in lupus-prone
mice, and FMT reduced serum anti-dsDNA autoantibodies to pre- Unlike antibiotics, which are associated with the risks of bacterial
antibiotic levels, inhibiting the progression of lupus (Zhang et al., disturbance and resistance, anti-microbial vaccines are more specific,
2020). Notably, the intestinal microbiota of prednisone-treated lupu­ and vaccine technology has a strong foundation and is dependable.
s-prone mice transplanted to the blank lupus-prone mice also had a Enterococcus gallinarum exacerbates SLE progression during bacterial
similar therapeutic effect as prednisone, but did not exhibit side effects disturbance. Some studies developed a vaccine from heat-killed
as prednisone, suggesting that FMT is a effective treatment with various E. gallinarum and intramuscularly administered it to a murine lupus
ways for improvement and can avoid the side effects of glucocorticoids model to prevent the translocation of E. gallinarum, reduce serum
(Wang et al., 2021). Recently, Huang et al. conducted the first clinical autoantibody levels, and prolong survival (Manfredo Vieira et al., 2018).
trial of FMT in patients with active SLE. After 12 weeks of oral encap­ In addition to microbial antigens, some studies have focused on the
sulated faecal microbiome from healthy donors, 20 patients with active effectiveness of vaccine adjuvants on microbiota. Flagellin is an excel­
SLE experienced significant decrease in SLEDAI-2 K scores and serum lent vaccine adjuvant, and flagellin from non-pathogenic bacteria is safe
anti-dsDNA antibody level, and bacterial taxa that produced short-chain (Cui et al., 2018). A previous study has shown that repeated injections of
fatty acids and a reduction in inflammation-associated bacterial taxa purified flagellin triggered a robust anti-flagellin faecal IgA response in
were also observed (Huang et al., 2022). mice. Flagellin-specific IgA encapsulates intestinal bacteria, prevents
FMT is usually reported to work by gut microbiota remodelling, microbiota encroachment, remodels microbiota composition, and re­
bowel permeability reduction and immunoregualtion (Shen et al., 2018; duces flagellin expression, thereby protecting against colitis induced by
Belvoncikova et al., 2022). The latest study suggested that FMT treat­ IL-10 deficiency. However, systemic rather than mucosal responses to
ment showed potential curative effect on SLE by reducing the major flagellin may be detrimental. Therefore, the development of flagellin
FMT-responder lymphocyte populations (Zheng et al., 2023). Epige­ injections into the intestinal mucosal immune system, perhaps using
netics is also a crucial part of research in autoimmune diseases. It is nanoparticles targeting the mucosa, may provide safer and more effec­
reported that global DNA methylation is decreased in lymphocytes in tive preferential induction of mucosal antibodies for the treatment of
patients with SLE and correlates with disease activity (Hedrich et al., various chronic inflammatory diseases (Tran et al., 2019). However, the
2017). FMT was showed to upregulated global methylation in some availability of mucosal vaccines is limited because of the lack of delivery
patients with SLE, and its possible mechanism was that FMT induced the systems that maintain antigen integrity and the strong adjuvanticity of
gut microbiota to interact with DNA methylation. Although the exact vaccines (Miquel-Clopés et al., 2019). Recently, researchers developed a
relationship between the methylation and altered gut mircobiota needs parenteral vaccine with emulsified curdlan and CpG oligodeox­
ynucleotides as adjuvants that could induce antigen-specific mucosal

7
C. Mo et al. Microbiological Research 281 (2024) 127613

and systemic immune responses with high titres of IgA after stimulation (www.editage.cn) for providing English language editing. Biorender.
with a small amount of antigen on the target mucosal surface, even if the com (Toronto, Ontario) tool was used to create the figures.
antigen-specific IgA on the mucosa is lost (Fujimoto KUematsu, 2020).
This optimised delivery system could be applied for the development of References
various mucosal vaccines in the future.
Although vaccine development against microbiota specifically Adebajo, A.O., 1997. Low frequency of autoimmune disease in tropical Africa. Lancet
349 (9048), 361–362. https://doi.org/10.1016/s0140-6736(05)62867-x.
altered in patients with SLE and the induction of a strong immune Ahmadi, S., Wang, S., Nagpal, R., Wang, B., Jain, S., Razazan, A., Mishra, S.P., Zhu, X.,
clearance response through mucosal immunity are important, these still Wang, Z., Kavanagh, K., Yadav, H., 2020. A human-origin probiotic cocktail
remains challenging. Moreover, there is a causal relationship between ameliorates aging-related leaky gut and inflammation via modulating the
microbiota/taurine/tight junction axis. JCI Insight 5 (9). https://doi.org/10.1172/
vaccine and adverse events such as anaphylaxis, arthralgia or arthritis, jci.insight.132055.
disseminated varicella infection and so on (Dudley et al., 2020). Current Akdis, C.A., 2021. Does the epithelial barrier hypothesis explain the increase in allergy,
data also suggests an increased risk of some auto-immune illnesses autoimmunity and other chronic conditions? Nat. Rev. Immunol. 21 (11), 739–751.
https://doi.org/10.1038/s41577-021-00538-7.
following vaccination, which is mostly based on the concept of molec­ Al-Quraishy, S., Abdel-Maksoud, M.A., El-Amir, A., Abdel-Ghaffar, F.A., Badr, G., 2013.
ular mimicry (Conklin et al., 2021). As a result, new vaccines need to go Malarial infection of female BWF1 lupus mice alters the redox state in kidney and
through more stringent testing before clinical translation. liver tissues and confers protection against lupus nephritis. Oxid. Med. Cell Longev.
2013, 156562 https://doi.org/10.1155/2013/156562.
AlSaleh, J., Jassim, V., ElSayed, M., Saleh, N., Harb, D., 2008. Clinical and
5. Conclusion immunological manifestations in 151 SLE patients living in Dubai. Lupus 17 (1),
62–66. https://doi.org/10.1177/0961203307084297.
Both exogenous microbiota and disturbance of commensal micro­ Anders, H.-J., Saxena, R., Zhao, M.-H., Parodis, I., Salmon, J.E., Mohan, C., 2020. Lupus
nephritis. Nat. Rev. Dis. Prim. 6 (1), 7 https://doi.org/10.1038/s41572-019-0141-9.
biota can not only promote SLE development but also can act as targets Andersen, C.J., 2022. Lipid metabolism in inflammation and immune function. Nutrients
for individualised therapy. Furthermore, some microbiota plays a posi­ 14 (7). https://doi.org/10.3390/nu14071414.
tive role in SLE, but can be hazardous to other individuals or under Aringer, S.R., Johnson, M., 2020. Classifying and diagnosing systemic lupus
erythematosus in the 21st century. Rheumatology 59 (Suppl5). https://doi.org/
specific genetic, environmental, and metabolic circumstances, as well as 10.1093/rheumatology/keaa379.
in the presence of certain sex hormones. Therefore, individualised care Aurlene, N., Manipal, S., Prabu, D., Rajmohan, 2020. Prevalence of oral mucosal lesions,
for SLE is urgently required. In addition to clinical indicator detection dental caries, and periodontal disease among patients with systemic lupus
erythematosus in a teaching hospital in Chennai, Tamil Nadu. J. Fam. Med. Prim.
and microbiomics, experimental studies should comprehensively assess Care 9 (7), 3374–3380. https://doi.org/10.4103/jfmpc.jfmpc_1263_19.
the immunoreactions and signalling pathways. Azzouz, D., Omarbekova, A., Heguy, A., Schwudke, D., Gisch, N., Rovin, B.H.,
Various microbiota-related therapeutic methods have been proposed Caricchio, R., Buyon, J.P., Alekseyenko, A.V., Silverman, G.J., 2019. Lupus nephritis
is linked to disease-activity associated expansions and immunity to a gut commensal.
to enhance the treatment of SLE and other autoimmune diseases using Ann. Rheum. Dis. 78 (7), 947–956. https://doi.org/10.1136/annrheumdis-2018-
nonspecific treatments with unfavourable side effects. However, lengthy 214856.
clinical studies are required to improve these processes, conduct thor­ Azzouz, D.F., Chen, Z., Izmirly, P.M., Chen, L.A., Li, Z., Zhang, C., Mieles, D., Trujillo, K.,
Heguy, A., Pironti, A., Putzel, G.G., Schwudke, D., Fenyo, D., Buyon, J.P.,
ough safety checks, and review ethical issues. In addition, anti-microbial
Alekseyenko, A.V., Gisch, N., Silverman, G.J., 2023. Longitudinal gut microbiome
antibody testing and personalised microbiome testing are crucial for analyses and blooms of pathogenic strains during lupus disease flares. Ann. Rheum.
precise microbiota targeting and rely mainly on 16 S rRNA testing Dis. https://doi.org/10.1136/ard-2023-223929.
(Azzouz et al., 2019) and shotgun sequencing (Chen et al., 2021). It Bae SCLee, Y.H., 2020. Causal association between periodontitis and risk of rheumatoid
arthritis and systemic lupus erythematosus: a Mendelian randomization. Z. Rheuma
would be beneficial and influential to develop more practical and 79 (9), 929–936. https://doi.org/10.1007/s00393-019-00742-w.
affordable microbiome testing methods. Belkaid, O.J., Harrison, Y., 2017. Homeostatic immunity and the microbiota. Immunity
46 (4), 562–576. https://doi.org/10.1016/j.immuni.2017.04.008.
Belvoncikova, P., Maronek, M., Gardlik, R., 2022. Gut dysbiosis and fecal microbiota
Funding transplantation in autoimmune diseases. Int. J. Mol. Sci. 23 (18) https://doi.org/
10.3390/ijms231810729.
This work was supported by Scientific Research Cultivation Project Brewer, D.L., Kamen, B.N., 2018. Gastrointestinal and hepatic disease in systemic lupus
erythematosus. Rheum. Dis. Clin. North Am. 44 (1), 165–175. https://doi.org/
of Stomatological Hospital, School of Stomatology, Southern Medical 10.1016/j.rdc.2017.09.011.
University (grant number PY2021018) and Hebei Natural Science Brown, J., Abboud, G., Ma, L., Choi, S.-C., Kanda, N., Zeumer-Spataro, L., Lee, J.,
Foundation (grant number H2023104011). Peng, W., Cagmat, J., Faludi, T., Mohamadzadeh, M., Garrett, T., Mandik-Nayak, L.,
Chervonsky, A., Perl, A., Morel, L., 2022. Microbiota-mediated skewing of
tryptophan catabolism modulates CD4+ T cells in lupus-prone mice. iScience 25 (5),
CRediT authorship contribution statement 104241. https://doi.org/10.1016/j.isci.2022.104241.
Brunekreef, T., Limper, M., Melchers, R., Mathsson-Alm, L., Dias, J., Hoefer, I.,
Haitjema, S., van Laar, J.M., Otten, H., 2021. Microarray testing in patients with
Lin Yunhe: Writing – review & editing. Yuan Peiyan: Writing –
systemic lupus erythematosus identifies a high prevalence of CpG DNA-binding
review & editing. Bi Jiaming: Writing – review & editing, Writing – antibodies. Lupus Sci. Med. 8 (1) https://doi.org/10.1136/lupus-2021-000531.
original draft, Visualization, Supervision. Li Siwei: Writing – review & Buonavoglia, A., Leone, P., Prete, M., Solimando, A.G., Guastadisegno, C., Lanave, G.,
Camero, M., Martella, V., Lo Muzio, L., Racanelli, V., 2021. Epstein-Barr virus in
editing. Mo Chuzi: Writing – review & editing, Writing – original draft,
salivary samples from systemic lupus erythematosus patients with oral lesions.
Visualization, Supervision, Conceptualization. Xu Shuaimei: Writing – J. Clin. Med. 10 (21) https://doi.org/10.3390/jcm10214995.
review & editing, Supervision, Funding acquisition. Jia Bo: Supervision, Caielli, S., Athale, S., Domic, B., Murat, E., Chandra, M., Banchereau, R., Baisch, J.,
Funding acquisition. Liu Zhongjun: Supervision, Funding acquisition. Phelps, K., Clayton, S., Gong, M., Wright, T., Punaro, M., Palucka, K., Guiducci, C.,
Banchereau, J., Pascual, V., 2016. Oxidized mitochondrial nucleoids released by
neutrophils drive type I interferon production in human lupus. J. Exp. Med. 213 (5),
Declaration of Competing Interest 697–713. https://doi.org/10.1084/jem.20151876.
Chen, B., Cao, J., Liu, W., Zhang, Y., Liu, Y., Wang, M., Xiao, F., Ma, J., Wang, J.,
Zhang, X., 2023. Disturbed gut virome with potent interferonogenic property in
None. systemic lupus erythematosus. Sci. Bull. 68 (3), 295–304. https://doi.org/10.1016/j.
scib.2023.01.021.
Data availability Chen, B.-d, Jia, X.-M., Xu, J.-Y., Zhao, L.-D., Ji, J.-Y., Wu, B.-X., Ma, Y., Li, H., Zuo, X.-X.,
Pan, W.-Y., Wang, X.-H., Ye, S., Tsokos, G.C., Wang, J., Zhang, X., 2021. An
autoimmunogenic and proinflammatory profile defined by the gut microbiota of
No data was used for the research described in the article. patients with untreated systemic lupus erythematosus. Arthritis Rheumatol. 73 (2),
232–243. https://doi.org/10.1002/art.41511.
Chen, C., Yan, Q., Yao, X., Li, S., Lv, Q., Wang, G., Zhong, Q., Tang, F., Liu, Z., Huang, Y.,
Acknowledgements An, Y., Zhou, J., Zhang, Q., Zhang, A., Ullah, H., Zhang, Y., Liu, C., Zhu, D., Li, H.,
Sun, W., Ma, W., 2022. Alterations of the gut virome in patients with systemic lupus
We thank all the reviewers for reviewing this manuscript and Editage

8
C. Mo et al. Microbiological Research 281 (2024) 127613

erythematosus. Front. Immunol. 13, 1050895 https://doi.org/10.3389/ erythematosus. N. Engl. J. Med. 283 (10), 506–511. https://doi.org/10.1056/
fimmu.2022.1050895. NEJM197009032831003.
Chen, P., Chen, J., 2020. Cryptococcal meningitis in patients with lupus nephritis. Clin. Greiling, T.M., Dehner, C., Chen, X., Hughes, K., I?Iguez, A.J., Boccitto, M., Ruiz, D.Z.,
Rheumatol. 39 (2), 407–412. https://doi.org/10.1007/s10067-019-04844-3. Renfroe, S.C., Vieira, S.M., Ruff, W.E., 2018a. Commensal orthologs of the human
Chen, Y.E., Fischbach, M.A., Belkaid, Y., 2018. Skin microbiota-host interactions. Nature autoantigen Ro60 as triggers of autoimmunity in lupus. Sci. Transl. Med. 10 (434),
553 (7689), 427–436. https://doi.org/10.1038/nature25177. eaan2306 https://doi.org/10.1126/scitranslmed.aan2306.
Chi, M., Ma, K., Wang, J., Ding, Z., Li, Y., Zhu, S., Liang, X., Zhang, Q., Song, L., Liu, C., Greiling, T.M., Dehner, C., Chen, X., Hughes, K., Iñiguez, A.J., Boccitto, M., Ruiz, D.Z.,
2021. The immunomodulatory effect of the gut microbiota in kidney disease. Renfroe, S.C., Vieira, S.M., Ruff, W.E., Sim, S., Kriegel, C., Glanternik, J., Chen, X.,
J. Immunol. Res. 2021, 5516035 https://doi.org/10.1155/2021/5516035. Girardi, M., Degnan, P., Costenbader, K.H., Goodman, A.L., Wolin, S.L., Kriegel, M.
Chng, H.H., Tan, B.E., Teh, C.L., Lian, T.Y., 2010. Major gastrointestinal manifestations A., 2018b. Commensal orthologs of the human autoantigen Ro60 as triggers of
in lupus patients in Asia: lupus enteritis, intestinal pseudo-obstruction, and protein- autoimmunity in lupus. Sci. Transl. Med. 10 (434) https://doi.org/10.1126/
losing gastroenteropathy. Lupus 19 (12), 1404–1413. https://doi.org/10.1177/ scitranslmed.aan2306.
0961203310374337. Guo, M., Lu, M., Chen, K., Xu, R., Xia, Y., Liu, X., Liu, Z., Liu, Q., 2023. Akkermansia
Choi, S.-C., Brown, J., Gong, M., Ge, Y., Zadeh, M., Li, W., Croker, B.P., Michailidis, G., muciniphila and Lactobacillus plantarum ameliorate systemic lupus erythematosus
Garrett, T.J., Mohamadzadeh, M., Morel, L., 2020. Gut microbiota dysbiosis and by possibly regulating immune response and remodeling gut microbiota. mSphere,
altered tryptophan catabolism contribute to autoimmunity in lupus-susceptible mice. e0007023. https://doi.org/10.1128/msphere.00070-23.
Sci. Transl. Med. 12 (551) https://doi.org/10.1126/scitranslmed.aax2220. Györkey, F., Min, K.W., Sincovics, J.G., Györkey, P., 1969. Systemic lupus erythematosus
Christen, U., 2014. Editorial: pathogen infection and autoimmunity. Int. Rev. Immunol. and myxovirus. N. Engl. J. Med. 280 (6), 333. https://doi.org/10.1056/
33 (4), 261–265. https://doi.org/10.3109/08830185.2014.897345. nejm196902062800620.
Christovich, X.M., Luo, A., 2022. Gut microbiota, leaky gut, and autoimmune diseases. Haller OKochs, G., 2011. Human MxA protein: an interferon-induced dynamin-like
Front. Immunol. 13, 946248 https://doi.org/10.3389/fimmu.2022.946248. GTPase with broad antiviral activity. J. Interferon Cytokine Res. 31 (1), 79–87.
Condrò, G., Guerini, M., Castello, M., Perugini, P., 2022. Acne vulgaris, atopic dermatitis https://doi.org/10.1089/jir.2010.0076.
and rosacea: the role of the skin microbiota-a review. Biomedicines 10 (10). https:// Haniuda, K., Gommerman, J.L., Reich, H.N., 2021. The microbiome and IgA
doi.org/10.3390/biomedicines10102523. nephropathy. Semin. Immunopathol. 43 (5), 649–656. https://doi.org/10.1007/
Conklin, L., Hviid, A., Orenstein, W.A., Pollard, A.J., Wharton, M., Zuber, P., 2021. s00281-021-00893-6.
Vaccine safety issues at the turn of the 21st century. BMJ Glob. Health 6 (Suppl 2). Harder, J.W., Ma, J., Alard, P., Sokoloski, K.J., Mathiowitz, E., Furtado, S., Egilmez, N.K.,
https://doi.org/10.1136/bmjgh-2020-004898. Kosiewicz, M.M., 2023. Male microbiota-associated metabolite restores macrophage
Corrêa, J.D., Calderaro, D.C., Ferreira, G.A., Mendonça, S.M.S., Fernandes, G.R., Xiao, E., efferocytosis in female lupus-prone mice via activation of PPARγ/LXR signaling
Teixeira, A.L., Leys, E.J., Graves, D.T., Silva, T.A., 2017. Subgingival microbiota pathways. J. Leukoc. Biol. 113 (1), 41–57. https://doi.org/10.1093/jleuko/qiac002.
dysbiosis in systemic lupus erythematosus: association with periodontal status. He, J., Chan, T., Hong, X., Zheng, F., Zhu, C., Yin, L., Dai, W., Tang, D., Liu, D., Dai, Y.,
Microbiome 5 (1), 34. https://doi.org/10.1186/s40168-017-0252-z. 2020. Microbiome and metabolome analyses reveal the disruption of lipid
Cui, B., Liu, X., Fang, Y., Zhou, P., Zhang, Y., Wang, Y., 2018. Flagellin as a vaccine metabolism in systemic lupus erythematosus. Front. Immunol. 11, 1703 https://doi.
adjuvant. Expert Rev. Vaccin. 17 (4), 335–349. https://doi.org/10.1080/ org/10.3389/fimmu.2020.01703.
14760584.2018.1457443. Hedrich, C.M., Mäbert, K., Rauen, T., Tsokos, G.C., 2017. DNA methylation in systemic
Cui, Y., Sheng, Y., Zhang, X., 2013. Genetic susceptibility to SLE: recent progress from lupus erythematosus. Epigenomics 9 (4), 505–525. https://doi.org/10.2217/epi-
GWAS. J. Autoimmun. 41, 25–33. https://doi.org/10.1016/j.jaut.2013.01.008. 2016-0096.
Dudley, M.Z., Halsey, N.A., Omer, S.B., Orenstein, W.A., O’Leary, S.T., Limaye, R.J., Hou, T., Sun, X., Zhu, J., Hon, K.-L., Jiang, P., Chu, I.M.-T., Tsang, M.S.-M., Lam, C.W.-K.,
Salmon, D.A., 2020. The state of vaccine safety science: systematic reviews of the Zeng, H., Wong, C.-K., 2020. IL-37 ameliorating allergic inflammation in atopic
evidence. Lancet Infect. Dis. 20 (5), e80–e89. https://doi.org/10.1016/S1473-3099 dermatitis through regulating microbiota and AMPK-mTOR signaling pathway-
(20)30130-4. modulated autophagy mechanism. Front. Immunol. 11, 752 https://doi.org/
Durcan, L., O’Dwyer, T., Petri, M., 2019. Management strategies and future directions for 10.3389/fimmu.2020.00752.
systemic lupus erythematosus in adults. Lancet 393 (10188), 2332–2343. https:// Hu, X., Wang, T., Jin, F., 2016. Alzheimer’s disease and gut microbiota. Sci. China Life
doi.org/10.1016/S0140-6736(19)30237-5. Sci. 59 (10), 1006–1023. https://doi.org/10.1007/s11427-016-5083-9.
Erttmann, S.F., Swacha, P., Aung, K.M., Brindefalk, B., Jiang, H., Härtlova, A., Uhlin, B. Huang, C., Yi, X., Long, H., Zhang, G., Wu, H., Zhao, M., Lu, Q., 2020. Disordered
E., Wai, S.N., Gekara, N.O., 2022. The gut microbiota prime systemic antiviral cutaneous microbiota in systemic lupus erythematosus. J. Autoimmun. 108, 102391
immunity via the cGAS-STING-IFN-I axis. Immunity 55 (5). https://doi.org/ https://doi.org/10.1016/j.jaut.2019.102391.
10.1016/j.immuni.2022.04.006. Huang, C., Yi, P., Zhu, M., Zhou, W., Zhang, B., Yi, X., Long, H., Zhang, G., Wu, H.,
Esmaeili, S.-A., Taheri, R.A., Mahmoudi, M., Momtazi-Borojeni, A.A., Morshedi, M., Tsokos, G.C., Zhao, M., Lu, Q., 2022. Safety and efficacy of fecal microbiota
Bahramifar, A., Fasihi-Ramandi, M., 2021. Inhibitory effects of tolerogenic transplantation for treatment of systemic lupus erythematosus: an EXPLORER trial.
probiotics on migratory potential of lupus patient-derived DCs. Iran. J. Basic Med. J. Autoimmun. 130, 102844 https://doi.org/10.1016/j.jaut.2022.102844.
Sci. 24 (11), 1509–1514. https://doi.org/10.22038/IJBMS.2021.58438.12982. Iwasaki, Y., Takeshima, Y., Nakano, M., Okubo, M., Ota, M., Suzuki, A., Kochi, Y.,
Fasano, A., 2012. Leaky gut and autoimmune diseases. Clin. Rev. Allergy Immunol. 42 Okamura, T., Endo, T., Miki, I., Sakurada, K., Yamamoto, K., Fujio, K., 2023.
(1), 71–78. https://doi.org/10.1007/s12016-011-8291-x. Combined plasma metabolomic and transcriptomic analysis identify histidine as a
Feng, J.-R., Wang, F., Qiu, X., McFarland, L.V., Chen, P.-F., Zhou, R., Liu, J., Zhao, Q., biomarker and potential contributor in SLE pathogenesis. Rheumatology 62 (2),
Li, J., 2017. Efficacy and safety of probiotic-supplemented triple therapy for 905–913. https://doi.org/10.1093/rheumatology/keac338.
eradication of Helicobacter pylori in children: a systematic review and network Jakubczyk, D., Leszczyńska, K., Górska, S., 2020. The effectiveness of probiotics in the
meta-analysis. Eur. J. Clin. Pharmacol. 73 (10), 1199–1208. https://doi.org/ treatment of inflammatory bowel disease (IBD)-a critical review. Nutrients 12 (7).
10.1007/s00228-017-2291-6. https://doi.org/10.3390/nu12071973.
Flannigan, K.L., Taylor, M.R., Pereira, S.K., Rodriguez-Arguello, J., Moffat, A.W., James, J.A., Robertson, J.M., 2012. Lupus and Epstein-Barr. Curr. Opin. Rheumatol. 24
Alston, L., Wang, X., Poon, K.K., Beck, P.L., Rioux, K.P., Jonnalagadda, M., (4), 383–388. https://doi.org/10.1097/BOR.0b013e3283535801.
Chelikani, P.K., Galipeau, H.J., Lewis, I.A., Workentine, M.L., Greenway, S.C., Jog, J.A., James, N.R., 2020. Epstein Barr Virus and autoimmune responses in systemic
Hirota, S.A., 2018. An intact microbiota is required for the gastrointestinal toxicity of lupus erythematosus. Front. Immunol. 11, 623944 https://doi.org/10.3389/
the immunosuppressant mycophenolate mofetil. J. Heart Lung Transpl. 37 (9), fimmu.2020.623944.
1047–1059. https://doi.org/10.1016/j.healun.2018.05.002. Jog, N.R., Young, K.A., Munroe, M.E., Harmon, M.T., Guthridge, J.M., Kelly, J.A.,
Fontana, F., Alfano, G., Leonelli, M., Cerami, C., Ligabue, G., Spinella, A., Citriniti, G., Kamen, D.L., Gilkeson, G.S., Weisman, M.H., Karp, D.R., Gaffney, P.M., Harley, J.B.,
Manzini, C.U., Ferri, C., Cappelli, G., 2018. Efficacy of Belimumab for active lupus Wallace, D.J., Norris, J.M., James, J.A., 2019. Association of Epstein-Barr virus
nephritis in a young Hispanic woman intolerant to standard treatment: a case report. serological reactivation with transitioning to systemic lupus erythematosus in at-risk
BMC Nephrol. 19 (1), 276 https://doi.org/10.1186/s12882-018-1066-3. individuals. Ann. Rheum. Dis. 78 (9), 1235–1241. https://doi.org/10.1136/
Frittoli, R.B., Vivaldo, J.F., Costallat, L.T.L., Appenzeller, S., 2021. Gastrointestinal annrheumdis-2019-215361.
involvement in systemic lupus erythematosus: a systematic review. J. Transl. Johnson, B.M., Gaudreau, M.C., Al-Gadban, M.M., Gudi, R., Vasu, C., 2015. Impact of
Autoimmun. 4, 100106 https://doi.org/10.1016/j.jtauto.2021.100106. dietary deviation on disease progression and gut microbiome composition in lupus-
Fuentes, S., Egert, M., Jimenez-Valera, M., Monteoliva-Sanchez, M., Ruiz-Bravo, A., prone SNF1 mice. Clin. Exp. Immunol. 181 (2), 323–337. https://doi.org/10.1111/
Smidt, H., 2008. A strain of Lactobacillus plantarum affects segmented filamentous cei.12609.
bacteria in the intestine of immunosuppressed mice. FEMS Microbiol. Ecol. 63 (1), Khatibi, M., Shakoorpour, A.H., Jahromi, Z.M., Ahmadzadeh, A., 2012. The prevalence
65–72. https://doi.org/10.1111/j.1574-6941.2007.00411.x. of oral mucosal lesions and related factors in 188 patients with systemic lupus
Fujimoto KUematsu, S., 2020. Development of prime-boost-type next-generation erythematosus. Lupus 21 (12), 1312–1315. https://doi.org/10.1177/
mucosal vaccines. Int. Immunol. 32 (9), 597–603. https://doi.org/10.1093/intimm/ 0961203312454589.
dxz085. Khoryati, L., Augusto, J.-F., Shipley, E., Contin-Bordes, C., Douchet, I., Mitrovic, S.,
Furie, R., Rovin, B.H., Houssiau, F., Malvar, A., Teng, Y.K.O., Contreras, G., Amoura, Z., Truchetet, M.-E., Lazaro, E., Duffau, P., Couzi, L., Jacquemin, C., Barnetche, T.,
Yu, X., Mok, C.-C., Santiago, M.B., Saxena, A., Green, Y., Ji, B., Kleoudis, C., Vacher, P., Schaeverbeke, T., Blanco, P., Richez, C., 2016. IgE inhibits toll-like
Burriss, S.W., Barnett, C., Roth, D.A., 2020. Two-year, randomized, controlled trial receptor 7- and toll-like receptor 9-mediated expression of interferon-α by
of belimumab in lupus nephritis. N. Engl. J. Med. 383 (12), 1117–1128. https://doi. plasmacytoid dendritic cells in patients with systemic lupus erythematosus. Arthritis
org/10.1056/NEJMoa2001180. Rheumatol 68 (9), 2221–2231. https://doi.org/10.1002/art.39679.
Grausz, H., Earley, L.E., Stephens, B.G., Lee, J.C., Hopper, J., 1970. Diagnostic import of Kim, D.S., Park, Y., Choi, J.-W., Park, S.-H., Cho, M.-L., Kwok, S.-K., 2021.
virus-like particles in the glomerular endothelium of patients with systemic lupus Supplementation exerts a synergistic effect on tacrolimus efficacy by modulating

9
C. Mo et al. Microbiological Research 281 (2024) 127613

Th17/Treg balance in lupus-prone mice the SIGNR3 pathway. Front. Immunol. 12, complications linked to systemic lupus erythematosus in a preclinical study. Gut
696074 https://doi.org/10.3389/fimmu.2021.696074. Microbes 15 (2), 2247053. https://doi.org/10.1080/19490976.2023.2247053.
Kim, J.-W., Kwok, S.-K., Choe, J.-Y., Park, S.-H., 2019. Recent advances in our Monteith, A.J., Kang, S., Scott, E., Hillman, K., Rajfur, Z., Jacobson, K., Costello, M.J.,
understanding of the link between the intestinal microbiota and systemic lupus Vilen, B.J., 2016. Defects in lysosomal maturation facilitate the activation of innate
erythematosus. Int. J. Mol. Sci. 20 (19) https://doi.org/10.3390/ijms20194871. sensors in systemic lupus erythematosus. Proc. Natl. Acad. Sci. USA 113 (15),
Kinashi YHase, K., 2021. Partners in leaky gut syndrome: intestinal dysbiosis and E2142–E2151. https://doi.org/10.1073/pnas.1513943113.
autoimmunity. Front. Immunol. 12, 673708 https://doi.org/10.3389/ Mosterd, C.M., Kanbay, M., van den Born, B.J.H., van Raalte, D.H., Rampanelli, E., 2021.
fimmu.2021.673708. Intestinal microbiota and diabetic kidney diseases: the Role of microbiota and
Kothari, D., Patel, S., Kim, S.-K., 2019. Probiotic supplements might not be universally- derived metabolites inmodulation of renal inflammation and disease progression.
effective and safe: a review. Biomed. Pharmacother. = Biomedecine Pharmacother. Best Pract. Res. Clin. Endocrinol. Metab. 35 (3), 101484 https://doi.org/10.1016/j.
111, 537–547. https://doi.org/10.1016/j.biopha.2018.12.104. beem.2021.101484.
Krasselt MBaerwald, C., 2019. Sex, symptom severity, and quality of life in Mu, Q., Zhang, H., Liao, X., Lin, K., Liu, H., Edwards, M.R., Ahmed, S.A., Yuan, R., Li, L.,
rheumatology. Clin. Rev. Allergy Immunol. 56 (3), 346–361. https://doi.org/ Cecere, T.E., Branson, D.B., Kirby, J.L., Goswami, P., Leeth, C.M., Read, K.A.,
10.1007/s12016-017-8631-6. Oestreich, K.J., Vieson, M.D., Reilly, C.M., Luo, X.M., 2017. Control of lupus
Kudsi, M., Nahas, L.D., Alsawah, R., Hamsho, A., Omar, A., 2021. The prevalence of oral nephritis by changes of gut microbiota. Microbiome 5 (1), 73. https://doi.org/
mucosal lesions and related factors in systemic lupus erythematosus patients. 10.1186/s40168-017-0300-8.
Arthritis Res. Ther. 23 (1), 229 https://doi.org/10.1186/s13075-021-02614-8. Nandakumar KSNündel, K., 2022. Editorial: Systemic lupus erythematosus -
Le, S.T., Toussi, A., Maverakis, N., Marusina, A.I., Barton, V.R., Merleev, A.A., predisposition factors, pathogenesis, diagnosis, treatment and disease models. Front.
Luxardi, G., Raychaudhuri, S.P., Maverakis, E., 2020. The cutaneous and intestinal Immunol. 13, 1118180 https://doi.org/10.3389/fimmu.2022.1118180.
microbiome in psoriatic disease. Clin. Immunol. 218, 108537 https://doi.org/ Narváez, J., 2020. Systemic lupus erythematosus. Med Clin. 155 (11), 494–501. https://
10.1016/j.clim.2020.108537. doi.org/10.1016/j.medcli.2020.05.009.
Li, B.-Z., Zhou, H.-Y., Guo, B., Chen, W.-J., Tao, J.-H., Cao, N.-W., Chu, X.-J., Meng, X., Navarro-López, V., Ramírez-Boscá, A., Ramón-Vidal, D., Ruzafa-Costas, B., Genovés-
2020a. Dysbiosis of oral microbiota is associated with systemic lupus erythematosus. Martínez, S., Chenoll-Cuadros, E., Carrión-Gutiérrez, M., Horga de la Parte, J.,
Arch. Oral. Biol. 113, 104708 https://doi.org/10.1016/j.archoralbio.2020.104708. Prieto-Merino, D., Codoñer-Cortés, F.M., 2018. Effect of oral administration of a
Li, D., Pan, Y., Xia, X., Liang, J., Liu, F., Dou, H., Hou, Y., 2020b. Bacteroides fragilis mixture of probiotic strains on SCORAD index and use of topical steroids in young
alleviates the symptoms of lupus nephritis via regulating CD1d and CD86 patients with moderate atopic dermatitis: a randomized clinical trial. JAMA
expressions in B cells. Eur. J. Pharmacol. 884, 173421 https://doi.org/10.1016/j. Dermatol. 154 (1), 37–43. https://doi.org/10.1001/jamadermatol.2017.3647.
ejphar.2020.173421. Nico, M.M.S., Vilela, M.A.C., Rivitti, E.A., Lourenço, S.V., 2008. Oral lesions in lupus
Li, Q., Wu, H., Zhou, S., Zhao, M., Lu, Q., 2020c. An update on the pathogenesis of skin erythematosus: correlation with cutaneous lesions. Eur. J. Dermatol. 18 (4),
damage in lupus. Curr. Rheumatol. Rep. 22 (5), 16 https://doi.org/10.1007/s11926- 376–381. https://doi.org/10.1684/ejd.2008.0388.
020-00893-9. Oku KAtsumi, T., 2018. Systemic lupus erythematosus: nothing stale her infinite variety.
Li, Z., Xu, D., Wang, Z., Wang, Y., Zhang, S., Li, M., Zeng, X., 2017. Gastrointestinal Mod. Rheumatol. 28 (5), 758–765. https://doi.org/10.1080/
system involvement in systemic lupus erythematosus. Lupus 26 (11), 1127–1138. 14397595.2018.1494239.
https://doi.org/10.1177/0961203317707825. Pan, Q., Guo, F., Huang, Y., Li, A., Chen, S., Chen, J., Liu, H.-F., Pan, Q., 2021. Gut
Lintner, K.E., Wu, Y.L., Yang, Y., Spencer, C.H., Hauptmann, G., Hebert, L.A., Atkinson, J. microbiota dysbiosis in systemic lupus erythematosus: novel insights into
P., Yu, C.Y., 2016. Early components of the complement classical activation pathway mechanisms and promising therapeutic strategies. Front. Immunol. 12, 799788
in human systemic autoimmune diseases. Front. Immunol. 7, 36. https://doi.org/ https://doi.org/10.3389/fimmu.2021.799788.
10.3389/fimmu.2016.00036. Parikh, B.H., Rovin, S.V., 2016. Current and emerging therapies for lupus nephritis.
Liu, P., Tan, H.Z., Li, H., Lim, C.C., Choo, J.C.J., 2018. Infections in hospitalized lupus J. Am. Soc. Nephrol. 27 (10), 2929–2939. https://doi.org/10.1681/
nephritis patients: characteristics, risk factors, and outcomes. Lupus 27 (7), ASN.2016040415.
1150–1158. https://doi.org/10.1177/0961203318768881. Peng, X., Ed-Dra, A., Song, Y., Elbediwi, M., Nambiar, R.B., Zhou, X., Yue, M., 2022.
Lloyd, C.M., Collins, I., Belcher, A.J., Manuelpillai, N., Wozencraft, A.O., Staines, N.A., Lacticaseibacillus rhamnosus alleviates intestinal inflammation and promotes
1994. Characterization and pathological significance of monoclonal DNA-binding microbiota-mediated protection against Salmonella fatal infections. Front. Immunol.
antibodies from mice with experimental malaria infection. Infect. Immun. 62 (5), 13, 973224 https://doi.org/10.3389/fimmu.2022.973224.
1982–1988. https://doi.org/10.1128/iai.62.5.1982-1988.1994. Pessoa, L., Aleti, G., Choudhury, S., Nguyen, D., Yaskell, T., Zhang, Y., Li, W., Nelson, K.
Losurdo, G., Cubisino, R., Barone, M., Principi, M., Leandro, G., Ierardi, E., Di Leo, A., E., Neto, L.L.S., Sant’Ana, A.C.P., Freire, M., 2019. Host-microbial interactions in
2018. Probiotic monotherapy and eradication: a systematic review with pooled-data systemic lupus erythematosus and periodontitis. Front. Immunol. 10, 2602 https://
analysis. World J. Gastroenterol. 24 (1), 139–149. https://doi.org/10.3748/wjg.v24. doi.org/10.3389/fimmu.2019.02602.
i1.139. Prasad, N., Novak, J.E., Patel, M.R., 2019. Kidney diseases associated with parvovirus
Ma, Y., Xu, X., Li, M., Cai, J., Wei, Q., Niu, H., 2019. Gut microbiota promote the B19, hanta, ebola, and dengue virus infection: a brief review. Adv. Chronic Kidney
inflammatory response in the pathogenesis of systemic lupus erythematosus. Mol. Dis. 26 (3), 207–219. https://doi.org/10.1053/j.ackd.2019.01.006.
Med. 25 (1), 35 https://doi.org/10.1186/s10020-019-0102-5. Rasaratnam IRyan, P.F., 1997. Systemic lupus erythematosus. Med. J. Aust. 166 (5),
Ma, Y., Guo, R., Sun, Y., Li, X., He, L., Li, Z., Silverman, G.J., Chen, G., Gao, F., Yuan, J., 266–270. https://doi.org/10.5694/j.1326-5377.1997.tb140110.x.
Wei, Q., Li, M., Lu, L., Niu, H., 2021. Lupus gut microbiota transplants cause Robinson, G., Pineda-Torra, I., Ciurtin, C., Jury, E.C., 2022. Lipid metabolism in
autoimmunity and inflammation. Clin. Immunol. 233, 108892 https://doi.org/ autoimmune rheumatic disease: implications for modern and conventional therapies.
10.1016/j.clim.2021.108892. J. Clin. Investig. 132 (2) https://doi.org/10.1172/JCI148552.
Manfredo Vieira, S., Hiltensperger, M., Kumar, V., Zegarra-Ruiz, D., Dehner, C., Khan, N., Robles-Vera, I., Toral, M., de la Visitación, N., Sánchez, M., Gómez-Guzmán, M.,
Costa, F.R.C., Tiniakou, E., Greiling, T., Ruff, W., Barbieri, A., Kriegel, C., Mehta, S. Romero, M., Yang, T., Izquierdo-Garcia, J.L., Jiménez, R., Ruiz-Cabello, J., Guerra-
S., Knight, J.R., Jain, D., Goodman, A.L., Kriegel, M.A., 2018. Translocation of a gut Hernández, E., Raizada, M.K., Pérez-Vizcaíno, F., Duarte, J., 2020. Probiotics
pathobiont drives autoimmunity in mice and humans. Science 359 (6380), prevent dysbiosis and the rise in blood pressure in genetic hypertension: role of
1156–1161. https://doi.org/10.1126/science.aar7201. short-chain fatty acids. Mol. Nutr. Food Res 64 (6), e1900616. https://doi.org/
Matsuoka, K., 2021. Fecal microbiota transplantation for ulcerative colitis. Immunol. 10.1002/mnfr.201900616.
Med. 44 (1), 30–34. https://doi.org/10.1080/25785826.2020.1792040. Root-Bernstein, D., Fairweather, R., 2014. Complexities in the relationship between
McMahon, M., Grossman, J., Skaggs, B., Fitzgerald, J., Sahakian, L., Ragavendra, N., infection and autoimmunity. Curr. Allergy Asthma Rep. 14 (1), 407 https://doi.org/
Charles-Schoeman, C., Watson, K., Wong, W.K., Volkmann, E., Chen, W., Gorn, A., 10.1007/s11882-013-0407-3.
Karpouzas, G., Weisman, M., Wallace, D.J., Hahn, B.H., 2009. Dysfunctional Ruff, W.E., Greiling, T.M., Kriegel, M.A., 2020. Host-microbiota interactions in immune-
proinflammatory high-density lipoproteins confer increased risk of atherosclerosis in mediated diseases. Nat. Rev. Microbiol 18 (9), 521–538. https://doi.org/10.1038/
women with systemic lupus erythematosus. Arthritis Rheumtol. 60 (8), 2428–2437. s41579-020-0367-2.
https://doi.org/10.1002/art.24677. Sebastiani, G.D., Iuliano, A., Canofari, C., Bracci, M., 2019. Cytomegalovirus infection in
Miquel-Clopés, A., Bentley, E.G., Stewart, J.P., Carding, S.R., 2019. Mucosal vaccines and systemic lupus erythematosus: report of four cases challenging the management of
technology. Clin. Exp. Immunol. 196 (2), 205–214. https://doi.org/10.1111/ the disease, and literature review. Lupus 28 (3), 432–437. https://doi.org/10.1177/
cei.13285. 0961203319825570.
Miyamoto, J., Mizukure, T., Park, S.-B., Kishino, S., Kimura, I., Hirano, K., Bergamo, P., Shanahan, F., Ghosh, T.S., O’Toole, P.W., 2021. The healthy microbiome-what is the
Rossi, M., Suzuki, T., Arita, M., Ogawa, J., Tanabe, S., 2015. A gut microbial definition of a healthy gut microbiome? Gastroenterology 160 (2), 483–494. https://
metabolite of linoleic acid, 10-hydroxy-cis-12-octadecenoic acid, ameliorates doi.org/10.1053/j.gastro.2020.09.057.
intestinal epithelial barrier impairment partially via GPR40-MEK-ERK pathway. Shen, M., Duan, C., Xie, C., Wang, H., Li, Z., Li, B., Wang, T., 2022. Identification of key
J. Biol. Chem. 290 (5), 2902–2918. https://doi.org/10.1074/jbc.M114.610733. interferon-stimulated genes for indicating the condition of patients with systemic
Mok, C.C., Ying, K.Y., Yim, C.W., Siu, Y.P., Tong, K.H., To, C.H., Ng, W.L., 2016. lupus erythematosus. Front. Immunol. 13, 962393 https://doi.org/10.3389/
Tacrolimus versus mycophenolate mofetil for induction therapy of lupus nephritis: a fimmu.2022.962393.
randomised controlled trial and long-term follow-up. Ann. Rheum. Dis. 75 (1), Shen, Z.-H., Zhu, C.-X., Quan, Y.-S., Yang, Z.-Y., Wu, S., Luo, W.-W., Tan, B., Wang, X.-Y.,
30–36. https://doi.org/10.1136/annrheumdis-2014-206456. 2018. Relationship between intestinal microbiota and ulcerative colitis: Mechanisms
Moleón, J., González-Correa, C., Robles-Vera, I., Miñano, S., de la Visitación, N., and clinical application of probiotics and fecal microbiota transplantation. World J.
Barranco, A.M., Martín-Morales, N., O’Valle, F., Mayo-Martínez, L., García, A., Gastroenterol. 24 (1) https://doi.org/10.3748/wjg.v24.i1.5.
Toral, M., Jiménez, R., Romero, M., Duarte, J., 2023. Targeting the gut microbiota Shimizu, Y., Yasuda, S., Kimura, T., Nishio, S., Kono, M., Ohmura, K., Shimamura, S.,
with dietary fibers: a novel approach to prevent the development cardiovascular Kono, M., Fujieda, Y., Kato, M., Oku, K., Bohgaki, T., Fukasawa, Y., Tanaka, S.,
Atsumi, T., 2018. Interferon-inducible Mx1 protein is highly expressed in renal

10
C. Mo et al. Microbiological Research 281 (2024) 127613

tissues from treatment-naïve lupus nephritis, but not in those under lpr mice. Arthritis Res. Ther. 23 (1), 240 https://doi.org/10.1186/s13075-021-
immunosuppressive treatment. Mod. Rheumatol. 28 (4), 661–669. https://doi.org/ 02620-w.
10.1080/14397595.2017.1404711. Watanabe, S., Imaizumi, T., Tsuruga, K., Aizawa, T., Ito, T., Matsumiya, T., Yoshida, H.,
Shirakashi, M., Maruya, M., Hirota, K., Tsuruyama, T., Matsuo, T., Watanabe, R., Joh, K., Ito, E., Tanaka, H., 2013. Glomerular expression of myxovirus resistance
Murata, K., Tanaka, M., Ito, H., Yoshifuji, H., Ohmura, K., Elewaut, D., Sakaguchi, S., protein 1 in human mesangial cells: possible activation of innate immunity in the
Fagarasan, S., Mimori, T., Hashimoto, M., 2022. Effect of impaired T cell receptor pathogenesis of lupus nephritis. Nephrology 18 (12), 833–837. https://doi.org/
signaling on the gut microbiota in a mouse model of systemic autoimmunity. 10.1111/nep.12155.
Arthritis Rheumatol. 74 (4), 641–653. https://doi.org/10.1002/art.42016. Wei, Y., Li, Y., Yan, L., Sun, C., Miao, Q., Wang, Q., Xiao, X., Lian, M., Li, B., Chen, Y.,
Silverman, G.J., Deng, J., Azzouz, D.F., 2022. Sex-dependent Lupus Blautia Zhang, J., Li, Y., Huang, B., Li, Y., Cao, Q., Fan, Z., Chen, X., Fang, J.-Y.,
(Ruminococcus) gnavus strain induction of zonulin-mediated intestinal permeability Gershwin, M.E., Tang, R., Ma, X., 2020. Alterations of gut microbiome in
and autoimmunity. Front. Immunol. 13, 897971 https://doi.org/10.3389/ autoimmune hepatitis. Gut 69 (3), 569–577. https://doi.org/10.1136/gutjnl-2018-
fimmu.2022.897971. 317836.
Sinha, S., Lin, G., Ferenczi, K., 2021. The skin microbiome and the gut-skin axis. Clin. Yang, L., Wang, J., Xiao, Y., Wang, X., Sun, Q., Shang, J., Zhao, Y., 2018. Saliva
Dermatol. 39 (5), 829–839. https://doi.org/10.1016/j.clindermatol.2021.08.021. dysfunction and oral microbial changes among systemic lupus erythematosus
Sojod, B., Pidorodeski Nagano, C., Garcia Lopez, G.M., Zalcberg, A., Dridi, S.M., patients with dental caries. Biomed. Res. Int. 2018, 8364042 https://doi.org/
Anagnostou, F., 2021. Systemic lupus erythematosus and periodontal disease: a 10.1155/2018/8364042.
complex clinical and biological interplay. J. Clin. Med. 10 (9) https://doi.org/ Yin, X., Kim, K., Suetsugu, H., Bang, S.-Y., Wen, L., Koido, M., Ha, E., Liu, L.,
10.3390/jcm10091957. Sakamoto, Y., Jo, S., Leng, R.-X., Otomo, N., Laurynenka, V., Kwon, Y.-C., Sheng, Y.,
Steinberg, A.D., Baron, S., Talal, N., 1969. The pathogenesis of autoimmunity in New Sugano, N., Hwang, M.Y., Li, W., Mukai, M., Yoon, K., Cai, M., Ishigaki, K.,
Zealand mice, I. Induction of antinucleic acid antibodies by polyinosinic- Chung, W.T., Huang, H., Takahashi, D., Lee, S.-S., Wang, M., Karino, K., Shim, S.-C.,
polycytidylic acid. Proc. Natl. Acad. Sci. USA 63 (4), 1102–1107. https://doi.org/ Zheng, X., Miyamura, T., Kang, Y.M., Ye, D., Nakamura, J., Suh, C.-H., Tang, Y.,
10.1073/pnas.63.4.1102. Motomura, G., Park, Y.-B., Ding, H., Kuroda, T., Choe, J.-Y., Li, C., Niiro, H., Park, Y.,
Sugita, K., Steer, C.A., Martinez-Gonzalez, I., Altunbulakli, C., Morita, H., Castro- Shen, C., Miyamoto, T., Ahn, G.-Y., Fei, W., Takeuchi, T., Shin, J.-M., Li, K.,
Giner, F., Kubo, T., Wawrzyniak, P., Rückert, B., Sudo, K., Nakae, S., Matsumoto, K., Kawaguchi, Y., Lee, Y.-K., Wang, Y., Amano, K., Park, D.J., Yang, W., Tada, Y.,
O’Mahony, L., Akdis, M., Takei, F., Akdis, C.A., 2018. Type 2 innate lymphoid cells Yamaji, K., Shimizu, M., Atsumi, T., Suzuki, A., Sumida, T., Okada, Y., Matsuda, K.,
disrupt bronchial epithelial barrier integrity by targeting tight junctions through IL- Matsuo, K., Kochi, Y., Kottyan, L.C., Weirauch, M.T., Parameswaran, S., Eswar, S.,
13 in asthmatic patients. J. Allergy Clin. Immunol. 141 (1) https://doi.org/10.1016/ Salim, H., Chen, X., Yamamoto, K., Harley, J.B., Ohmura, K., Kim, T.-H., Yang, S.,
j.jaci.2017.02.038. Yamamoto, T., Kim, B.-J., Shen, N., Ikegawa, S., Lee, H.-S., Zhang, X., Terao, C.,
Takeno MIshigatsubo, Y., 2006. Intestinal manifestations in systemic lupus Cui, Y., Bae, S.-C., 2021. Meta-analysis of 208370 East Asians identifies 113
erythematosus. Intern Med. 45 (2), 41–42. https://doi.org/10.2169/ susceptibility loci for systemic lupus erythematosus. Ann. Rheum. Dis. 80 (5),
internalmedicine.45.0136. 632–640. https://doi.org/10.1136/annrheumdis-2020-219209.
Tesser, A., Piperno, G.M., Pin, A., Piscianz, E., Boz, V., Benvenuti, F., Tommasini, A., Yoo, W., Zieba, J.K., Foegeding, N.J., Torres, T.P., Shelton, C.D., Shealy, N.G.,
2021. Priming of the cGAS-STING-TBK1 pathway enhances LPS-induced release of Byndloss, A.J., Cevallos, S.A., Gertz, E., Tiffany, C.R., Thomas, J.D., Litvak, Y.,
type I interferons. Cells 10 (4). https://doi.org/10.3390/cells10040785. Nguyen, H., Olsan, E.E., Bennett, B.J., Rathmell, J.C., Major, A.S., Bäumler, A.J.,
Thackray, V.G., 2019. Sex, microbes, and polycystic ovary syndrome. Trends Endocrinol. Byndloss, M.X., 2021. High-fat diet-induced colonocyte dysfunction escalates
Metab. 30 (1), 54–65. https://doi.org/10.1016/j.tem.2018.11.001. microbiota-derived trimethylamine N-oxide. Science 373 (6556), 813–818. https://
Thim-Uam, A., Surawut, S., Issara-Amphorn, J., Jaroonwitchawan, T., Hiengrach, P., doi.org/10.1126/science.aba3683.
Chatthanathon, P., Wilantho, A., Somboonna, N., Palaga, T., Pisitkun, P., Yoon, N., Kim, K., 2021. Roles of sex hormones and gender in the gut microbiota.
Leelahavanichkul, A., 2020. Leaky-gut enhanced lupus progression in the Fc gamma J. Neurogastroenterol. Motil. 27 (3), 314–325. https://doi.org/10.5056/jnm20208.
receptor-IIb deficient and pristane-induced mouse models of lupus. Sci. Rep. 10 (1), Yu, C., Li, P., Dang, X., Zhang, X., Mao, Y., Chen, X., 2022. Lupus nephritis: new progress
777 https://doi.org/10.1038/s41598-019-57275-0. in diagnosis and treatment. J. Autoimmun. 132, 102871 https://doi.org/10.1016/j.
Tian, X., Zhang, X.-P., 2010. Gastrointestinal involvement in systemic lupus jaut.2022.102871.
erythematosus: insight into pathogenesis, diagnosis and treatment. World J. Zegarra-Ruiz, D.F., El Beidaq, A., Iñiguez, A.J., Lubrano Di Ricco, M., Manfredo
Gastroenterol. 16 (24), 2971–2977. https://doi.org/10.3748/wjg.v16.i24.2971. Vieira, S., Ruff, W.E., Mubiru, D., Fine, R.L., Sterpka, J., Greiling, T.M., Dehner, C.,
Tomofuji, Y., Kishikawa, T., Maeda, Y., Ogawa, K., Nii, T., Okuno, T., Oguro-Igashira, E., Kriegel, M.A., 2019. A diet-sensitive commensal lactobacillus strain mediates TLR7-
Kinoshita, M., Yamamoto, K., Sonehara, K., Yagita, M., Hosokawa, A., Motooka, D., dependent systemic autoimmunity. Cell Host Microbe 25 (1). https://doi.org/
Matsumoto, Y., Matsuoka, H., Yoshimura, M., Ohshima, S., Nakamura, S., 10.1016/j.chom.2018.11.009.
Inohara, H., Mochizuki, H., Takeda, K., Kumanogoh, A., Okada, Y., 2022. Whole gut Zeng, L., Deng, Y., Yang, K., Chen, J., He, Q., Chen, H., 2022. Safety and efficacy of fecal
virome analysis of 476 Japanese revealed a link between phage and autoimmune microbiota transplantation for autoimmune diseases and autoinflammatory diseases:
disease. Ann. Rheum. Dis. 81 (2), 278–288. https://doi.org/10.1136/annrheumdis- a systematic review and meta-analysis. Front. Immunol. 13, 944387 https://doi.org/
2021-221267. 10.3389/fimmu.2022.944387.
Toral, M., Robles-Vera, I., Romero, M., de la Visitación, N., Sánchez, M., O’Valle, F., Zhang, B., Zhou, W., Liu, Q., Huang, C., Hu, Z., Zheng, M., Xin, Y., Zhao, M., Lu, Q., 2023.
Rodriguez-Nogales, A., Gálvez, J., Duarte, J., Jiménez, R., 2019. CECT5716: a novel Effects of fecal microbiota transplant on DNA methylation in patients with systemic
alternative for the prevention of vascular disorders in a mouse model of systemic lupus erythematosus. J. Autoimmun., 103047 https://doi.org/10.1016/j.
lupus erythematosus. FASEB J. 33 (9), 10005–10018. https://doi.org/10.1096/ jaut.2023.103047.
fj.201900545RR. Zhang, H., Liao, X., Sparks, J.B., Luo, X.M., 2014. Dynamics of gut microbiota in
Tran, H.Q., Ley, R.E., Gewirtz, A.T., Chassaing, B., 2019. Flagellin-elicited adaptive autoimmune lupus. Appl. Environ. Microbiol. 80 (24), 7551–7560. https://doi.org/
immunity suppresses flagellated microbiota and vaccinates against chronic 10.1128/AEM.02676-14.
inflammatory diseases. Nat. Commun. 10 (1), 5650 https://doi.org/10.1038/ Zhang, L., Qing, P., Yang, H., Wu, Y., Liu, Y., Luo, Y., 2021. Gut microbiome and
s41467-019-13538-y. metabolites in systemic lupus erythematosus: link, mechanisms and intervention.
Valiente, G.R., Munir, A., Hart, M.L., Blough, P., Wada, T.T., Dalan, E.E., Willis, W.L., Front. Immunol. 12, 686501 https://doi.org/10.3389/fimmu.2021.686501.
Wu, L.-C., Freud, A.G., Jarjour, W.N., 2022. Gut dysbiosis is associated with Zhang, S., Deng, F., Chen, J., Chen, F., Wu, Z., Li, L., Hou, K., 2022. Fecal microbiota
acceleration of lupus nephritis. Sci. Rep. 12 (1), 152 https://doi.org/10.1038/ transplantation treatment of autoimmune-mediated type 1 diabetes: a systematic
s41598-021-03886-5. review. Front. Cell. Infect. Microbiol. 12, 1075201 https://doi.org/10.3389/
Verhelst, J., Hulpiau, P., Saelens, X., 2013. Mx proteins: antiviral gatekeepers that fcimb.2022.1075201.
restrain the uninvited. Microbiol. Mol. Biol. Rev. 77 (4), 551–566. https://doi.org/ Zhang, Y., Liu, Q., Yu, Y., Wang, M., Wen, C., He, Z., 2020. Early and short-term
10.1128/MMBR.00024-13. interventions in the gut microbiota affects lupus severity, progression, and treatment
Vindigni, C.M., Surawicz, S.M., 2017. Fecal microbiota transplantation. Gastroenterol. in MRL/lpr mice. Front. Microbiol. 11, 628 https://doi.org/10.3389/
Clin. North Am. 46 (1), 171–185. https://doi.org/10.1016/j.gtc.2016.09.012. fmicb.2020.00628.
de la Visitación, N., Robles-Vera, I., Toral, M., O’Valle, F., Moleon, J., Gómez- Zhao, Y., Zhang, B., Liu, C., Ren, G., 2020. Method for accurate diagnose of lupus
Guzmán, M., Romero, M., Duarte, M., Sánchez, M., Jiménez, R., Duarte, J., 2020. erythematosus skin lesions based on microbial rDNA sequencing. Saudi J. Biol. Sci.
Lactobacillus fermentum CECT5716 prevents renal damage in the NZBWF1 mouse 27 (8), 2111–2115. https://doi.org/10.1016/j.sjbs.2020.05.035.
model of systemic lupus erythematosus. Food Funct. 11 (6), 5266–5274. https://doi. Zheng, M., Zhou, W., Huang, C., Hu, Z., Zhang, B., Lu, Q., Zhao, M., 2023. A single-cell
org/10.1039/d0fo00578a. map of peripheral alterations after FMT treatment in patients with systemic lupus
de la Visitación, N., Robles-Vera, I., Toral, M., Gómez-Guzmán, M., Sánchez, M., erythematosus. J. Autoimmun. 135, 102989 https://doi.org/10.1016/j.
Moleón, J., González-Correa, C., Martín-Morales, N., O’Valle, F., Jiménez, R., jaut.2022.102989.
Romero, M., Duarte, J., 2021. Gut microbiota contributes to the development of Zheng, Y., Gou, X., Zhang, L., Gao, H., Wei, Y., Yu, X., Pang, B., Tian, J., Tong, X., Li, M.,
hypertension in a genetic mouse model of systemic lupus erythematosus. Br. J. 2020. Interactions between gut microbiota, host, and herbal medicines: a review of
Pharmacol. 178 (18), 3708–3729. https://doi.org/10.1111/bph.15512. new insights into the pathogenesis and treatment of type 2 diabetes. Front. Cell.
Wang, J.-W., Kuo, C.-H., Kuo, F.-C., Wang, Y.-K., Hsu, W.-H., Yu, F.-J., Hu, H.-M., Hsu, P.- Infect. Microbiol. 10, 360 https://doi.org/10.3389/fcimb.2020.00360.
I., Wang, J.-Y., Wu, D.-C., 2019. Fecal microbiota transplantation: review and Zhou, W., Wu, H., Zhao, M., Lu, Q., 2020. New insights into the progression from
update. J. Formos. Med. Assoc. 118 (Suppl 1), S23–S31. https://doi.org/10.1016/j. cutaneous lupus to systemic lupus erythematosus. Expert Rev. Clin. Immunol. 16 (8),
jfma.2018.08.011. 829–837. https://doi.org/10.1080/1744666X.2020.1805316.
Wang, M., Zhu, Z., Lin, X., Li, H., Wen, C., Bao, J., He, Z., 2021. Gut microbiota mediated
the therapeutic efficacies and the side effects of prednisone in the treatment of MRL/

11

You might also like