Open access peer-reviewed chapter - ONLINE FIRST

Role of Metabolomics in Precision Medicine in the Context of Systemic Lupus Erythematosus and Lupus Nephritis

Written By

Alejandra Rojo-Sánchez, Agustín Abuchaibe, Ada Carmona, Valentina Arrieta-Bravo, David Chica-Valle, Yirys Díaz-Olmos and Lisandro Pacheco-Lugo

Submitted: 27 November 2023 Reviewed: 27 January 2024 Published: 24 June 2024

DOI: 10.5772/intechopen.1005132

Personalized Medicine - New Perspectives IntechOpen
Personalized Medicine - New Perspectives Edited by Xianquan Zhan

From the Edited Volume

Personalized Medicine - New Perspectives [Working Title]

scope

Chapter metrics overview

9 Chapter Downloads

View Full Metrics

Abstract

Systemic lupus erythematosus (SLE) is an autoimmune, multisystemic disease, the underlying causes of which are not fully understood. Clinically, SLE is a heterogeneous disease, and its clinical spectrum overlaps with other rheumatic diseases. Even though multiple organs can be involved in the progression of the disease, the kidney is the main indicator of morbidity and mortality in SLE, a condition known as lupus nephritis (LN). The diagnosis of LN still requires an invasive procedure that becomes impractical when monitoring patients with a confirmed diagnosis, and the described biomarkers do not meet the characteristics of a good biomarker. Recently, “omics” technologies have revolutionized the field of the molecular diagnosis, including autoimmune diseases. Metabolomics has been an extremely useful tool for identifying non-invasive biomarkers in the clinical context and is proposed as a powerful and promising tool to differentiate between individuals with SLE without renal damage and patients with SLE with renal involvement. In addition, this methodology will allow the categorization of patients according to renal damage, promising personalized management of LN in the context of SLE. Here, we review the potential of untargeted metabolomic approaches using LC/GC-MS and MNR to identify potential biomarkers for renal damage in patients with SLE.

Keywords

  • systemic lupus erythematosus
  • lupus nephritis
  • metabolomics
  • LC-MS
  • GC-MS
  • biomarkers

1. Introduction

Systemic lupus erythematosus (SLE) presents a high clinical complexity due to its wide variety of expression patterns. It can affect any organ and follows a relapsing-remitting course, with periods of activity and inactivity. Its multisystemic nature and the wide range of symptoms it can manifest require a careful diagnostic study and the use of differentiated therapy based on the severity of the condition [1]. The kidney is affected to varying degrees; in the majority of SLE patients, immune complex deposits are observed, only detectable at present by electron microscopy or immunofluorescence [2]. Renal involvement, or lupus nephritis (LN), can be silent or manifest with clinical patterns known as nephritic or nephrotic syndrome, high blood pressure, and progressive deterioration of kidney function, eventually leading to end-stage renal failure [3]. Approximately 75% of patients with SLE develop renal involvement at some point in the course of the disease, of which 25 to 50% develop nephropathy in the early stages of the condition [4]. The percentages for different types of LN are 25% for focal (class III according to the World Health Organization), 37% for diffuse (class IV), and 13% for membranous (class V) [5].

LN is a frequent and severe manifestation of SLE. The prevalence of LN varies depending on age, gender, ethnicity, and other factors, occurring in approximately 50–60% of SLE patients, of which up to 10% will develop end-stage renal disease [6, 7]. The incidence of SLE varies by region, with higher rates in the United States compared to Europe [8, 9], and by ethnicity, with higher rates in Black individuals (34–51%), followed by Hispanics (31–43%) and Asians (33–55%). There is also a relationship between ethnicity and the severity of symptoms, with Black individuals exhibiting more severe symptoms [10, 11].

The pathogenesis may involve a variety of mechanisms, including gene expression leading to neutrophil activation, increased production of interferon and other proinflammatory myeloid mediators, release of neutrophil extracellular traps (NETs), and complement activation. The glomerular lesion pattern observed in SLE is generally related to the binding of antibodies to multiple intrarenal autoantigens or the formation of circulating antigens, forming immune complexes containing autoantibodies deposited in various parts of the glomerulus, mainly due to antibodies against double-stranded DNA (anti-dsDNA or anti-DNA) [10, 12, 13]. Molecular phenotyping of the kidney or urine can identify the current immunological and/or injury process affecting each individual, leaving a metabolic footprint. This allows for the evaluation of diagnostic and/or monitoring biomarkers for LN and expands knowledge of pathogenesis [14, 15, 16].

Renal biopsy remains the gold standard for confirming the diagnosis of LN and determining the histological type and extent of damage, using the International Society of Nephrology/Renal Pathology Society (ISN/RPS) classification [17]. Fluid biomarkers, which are validated indicators of physiological or pathological processes or treatment response, are currently considered less invasive alternatives or complementary means of evaluating SLE-related kidney disease [18]. Traditional laboratory biomarkers include immunoserological tests such as anti-double-stranded DNA (dsDNA) levels, complement levels, and parameters associated with renal disease, such as 24-hour urinary protein or urinary protein/creatinine ratio, urine sediment, and glomerular filtration rate (GFR). They are well-established tools for clinical LN evaluation. However, they do not detect LN at early stages of the disease, presenting limited sensitivity and specificity for distinguishing between active disease and chronic lesions [19].

Recent technological advancements, including deep proteomic and metabolic analysis tests, have contributed to the evidence of new biomarkers, some of which have shown performance equivalent to traditional markers [20, 21].

The concept of metabolomics includes the systematic detection and quantification of low-molecular-weight molecules produced by cells (endogenous metabolites) that leave unique chemical traces [15, 22]. Metabonomics, on the other hand, is defined as the quantitative measurement of changes in these metabolic profiles [23]. The theoretical basis lies in the idea that disturbances in a biological system, caused by a disease, for example, will be detectable as changes in the concentrations of certain metabolites. This also provides the possibility of revealing associated underlying biochemical phenomena, thus offering insights that help in developing a better understanding of disease pathogenesis [15].

In some cases, it may be possible to identify a single robust diagnostic metabolite, as in congenital metabolic diseases. Still, in many cases, the disturbances are more subtle, involving the activation of multiple enzymatic (biochemical) pathways, making it unlikely that a single biomarker will be specific enough for diagnostic purposes [15]. However, by using multivariate statistics, it may be possible to describe changes in biomarker patterns rather than individual biomarkers [15]. Nuclear magnetic resonance spectroscopy (NMR) and liquid and gas chromatography coupled with tandem mass spectrometry (LC-MS/MS and GC-MS/MS) are the most widely used analytical techniques for metabolomic studies due to their great power to identify new metabolites in different biofluids in an unbiased manner. In this book chapter, we will review the literature related to the application of metabolomic techniques for the study of biomarkers in individuals with SLE and LN and the impact these technologies can have on personalized medicine.

Advertisement

2. Role of metabolism disturbances in the context of SLE

2.1 TCA cycle

Within the many possible explanations for the pathogenesis of SLE, it has been established that abnormal T cell activation and cell apoptosis, which are highly energy-dependent processes, play a major role in the progression of the disease. SLE patients exhibit mitochondrial abnormalities that affect energy production and cell survival [24, 25]. A study conducted by Yan et al. that applied gas chromatography–mass spectrometry to analyze serum samples found that SLE patients had lower levels of fumarate and citrate (Table 1) [26], which are intermediates of the energy-producing tricarboxylic acid (TCA) cycle. This suggests that SLE disrupts cellular energy metabolism, and the extent of this dysfunction may be related to disease activity. This same study also reported that patients with SLE had lower levels of glucose in the sera compared to healthy controls, attributed to an upregulation of the pentose phosphate pathway in SLE, which requires a higher rate of glucose consumption and a subsequent disturbance of glucose utilization [26, 34].

MetaboliteType of sampleDiseaseMethod appliedReferences
Fumarate, citrate, glucoseSerumSLEGC-MS[26]
GlutathioneSerumSLEHPLC[24]
Aminomalonate, threonate, alpha-tocopherolUrineSLEGC-MS[14]
L-pyroglutamic acid, adenosineSerumSLELC-MS-MS[27]
AdenosineFecesSLEUHPLC-MS-MS[28]
ArginineSerumSLELC-MS and GC-MS[29]
2-Hydroxyisovalerate, 2-keto-3-methylvalerate, and 2-ketoisocaproateSerumSLEGC-MS[26]
GlutamineBrain ImagingSLEH single-voxel spectroscopy[30]
Tryptophan, kynurenineSerumSLEUHPLC[31]
Kynurenine and Kynurenic acidPeripheral Blood LymphocytesSLELC-MS-MS[32]
2-hydroxyisobutyrate, glycerol, oleic acid, arachidonic acid, 1-monopalmitin, and linoleic acidSerumSLEGC-MS[26]
Deoxycholic acid, glycolic acid, ursodeoxycholic acid, and arachidonic acidFeces and serumSLELC-MS[33]

Table 1.

Metabolites in SLE.

2.2 Oxidative stress

SLE is characterized not only by abnormal T cell activation but also by an increase in oxidative stress [35, 36], which has been associated to alterations in the immune system and autoantibody production, as well as to the cardiovascular complications of the disease [26, 37]. Glutathione is an important antioxidant that has been found to have significant implications in nutrient metabolism and regulation of cellular events such as gene expression, synthesis of proteins and DNA, cellular proliferation, apoptosis, signal transduction, and protein glutathionylation [38, 39]. By the subsequent actions of glutamate cysteine ligase (GCL) and GSH synthetase, this antioxidant is created from constituent amino acids including cysteine, glutamate, and pyroglutamarate [26, 38]. Glutathione is responsible for suppressing the development of Th17 cells, a subset of T cells involved in autoimmune responses, and for reducing intracellular levels of reactive oxygen species (ROS), which are known to contribute to tissue damage [24, 40]. Additionally, glutathione regulates the rise of the mitochondrial transmembrane potential, thereby influencing the activation of the mammalian target of rapamycin (mTOR) pathway in T cells of SLE patients [41, 42]. The dysregulation of the mTOR pathway is connected to the development of SLE and will be further explored later in this chapter.

A study using high-performance liquid chromatography performed by Gergely et al. [24] reported decreased levels of glutathione in the peripheral blood of SLE patients, indicating a deficiency in this important antioxidant (Table 1). In an effort to adjust glutathione levels and modulate disease severity, a couple of studies administered N-acetylcysteine (NAC), a cell-permeable precursor of cysteine and a rate-limiting component of de novo reduced glutathione synthesis, to both human patients and mouse models of SLE [42, 43]. The administration of NAC resulted in the inhibition of the mTOR pathway in T cells, resulting in a reduction in disease severity and a decrease in organ damage [38, 39, 44]. In general, SLE patients exhibit insufficient levels of both glutathione and GCL activity. In nonpathogenic states, glutathione and GCL play vital roles in protecting against oxidative stress, detoxifying xenobiotics, and maintaining cellular homeostasis. However, in SLE, these crucial components of the antioxidant defense system are compromised [45].

In addition, a study using gas chromatography/mass spectrometry on urine samples revealed that individuals with SLE exhibited reduced levels of aminomalonate, threonate, and the antioxidant alpha-tocopherol, which are crucial in regulating oxidative stress [14, 46]. Furthermore, metabolic profiling based on liquid chromatography-tandem mass spectrometry detected elevated levels of L-pyroglutamic acid in serum samples from SLE patients, indicating its potential as a disease biomarker [27]. Also known as 5-oxoproline, L-pyroglutamic acid is a derivative of glutamate via the γ-glutamyl cycle, a process in which glutathione decomposes into a γ-glutamyl amino acid and is subsequently converted to pyroglutamic acid through γ-glutamyl cyclotransferase [47]. Given the confirmed decrease in serum glutathione in SLE patients [24], it is suggested that the elevated levels of L-pyroglutamic acid in their serum result from changes in glutathione metabolism. Zhang et al. found a positive correlation between the concentrations of L-pyroglutamic acid and the erythrocyte sedimentation rate (ESR) and anti-Sm antibody in SLE patients, indicating its potential as a marker for disease progression [27]. These findings collectively underscore the imbalanced oxidant/antioxidant system in SLE patients compared to healthy controls, as evidenced by multiple studies demonstrating alterations in glutathione metabolism.

2.3 Purine metabolism

Alterations in the metabolism of purines have also been correlated to the pathogenesis of SLE. A study performed by Zhang et al. that utilized liquid chromatography-tandem mass spectrometry revealed that serum adenosine levels were lower in SLE patients compared to healthy individuals [27]. This outcome aligns with a metabolomic investigation involving feces samples from SLE patients [28]. Adenosine, an endogenous purine nucleoside, holds great significance as Treg cells produce it as a potent immunosuppressive agent, thereby reducing immune responses to oneself, regulating tolerance to tissue grafts, and providing protection against autoimmune conditions [48, 49]. Gao et al.’s study [50] also revealed a notable rise in the blood adenosine deaminase activity, an enzyme involved in adenosine hydrolysis, among individuals with SLE, which may account for the reduced blood adenosine levels observed in these patients. The pathophysiology of SLE is frequently characterized by an imbalance between Th17 and Treg cells. This imbalance involves a reduction in Treg cells and an elevation in Th17 cells, which commonly exhibit functional abnormalities [51]. Consequently, the reduction in adenosine could in addition be linked to the imbalance of Treg cells and could be used to predict the population of Treg cells in individuals with SLE [27].

Finally, in relation to purine metabolism, a study investigating the intestinal dysbiosis in individuals with SLE [52] identified a clear association between purine metabolism and the prevalence of the genus Streptococcus, which was significantly higher in SLE patients. The known role of adenosine in facilitating intestinal epithelial repair and anti-inflammation has been established [53, 54], suggesting that fecal purine metabolism serves as an informative indicator of the gut microbiome in SLE patients. These results imply that adenosine or purine metabolism, which affects gut flora, inflammation, and systemic immunity, has a role in the pathogenesis of SLE [27, 28].

2.4 Amino acid metabolism

In the context of SLE, amino acids are key nutrients for proliferating T cells as they serve both as a source of energy and as precursors for protein and nucleic acid biosynthesis. Glutaminolysis is essential for the production of Th1 and Th17, pro-inflammatory effectors T cells. The enzymes involved in this pathway are of great interest and thus have been examined in the context of pathogenic states [38]. Several investigations have also suggested that amino acid transporters are essential components for T cell clonal proliferation and differentiation in response to antigen presentation. Particularly, it has been demonstrated that the loss of the Large Neutral Amino Acid Transporter 1 (LAT-1), also known as the L-system Slc7a5 transporter, which is a transporter devoted to the transport of essential amino acids, inhibits CD4+ and CD8+ T cell differentiation and proliferation but does not impair CD4+ T cell differentiation into regulatory T cells. T cells without Slc7a5 trancarrier do not increase glutamine and glucose uptake and do not change their metabolism to aerobic glycolysis after stimulation of the T cell receptor. This lack of availability of amino acids leads to inadequate mTORC1 activation, which is required for CD4+ cell differentiation into the T-helper (Th1) and Th17 subsets [55, 56].

Branched-chain amino acids (BCAAs) include valine, leucine, and isoleucine. More than half of the body’s overall metabolism is accounted for by the metabolism of amino acids in muscle tissue, with skeletal muscle being the primary site of BCAA catabolism [26, 57]. Branched amino acids are known to function as signaling molecules that control the synthesis of proteins, lipids, and glucose [38]. Through unique signaling networks, particularly the phosphoinositide 3-kinase signaling – protein kinase B – mTOR pathway (PI3K/AKT/mTOR), they can also contribute to gut health and immunity [58].

The activity of the target of rapamycin (mTOR) is regulated by several elements such as the availability of amino acids, the energy levels, and growth factors. In mammalian cells, mTOR forms two distinct complexes: the mTORC 1 complex (mTORC1) and the mTORC 2 complex (mTORC2). The mTORC1 complex detects several signs of stress, including the accumulation of amino acids such as leucine, isoleucine, and glutamine. According to studies, Th17 cells and T cells that produce IL-4 have increased mTORC activity, which contributes to the proinflammatory profile seen in SLE patients. Additionally, mTOR is necessary for Th 17 cell development because it promotes the synthesis of hypoxia inducible factor 1α (HIF1α), which enhances inflammatory cells’ glycolysis in the pseudo-hypoxia state that is characteristic of subjects with SLE [59].

A study performed on murine models showed that the leucine antagonist, N-acetyl-leucine amide (NALA), can inhibit mTORC1 activity and T cell function by compromising the production of IL-2 and IFγ in Th1 cells [60]. It has been reported that leucine is essential for Treg cell function since it promotes mTORC1 activity in Treg cells through the small G RagA/B and Rheb1/2 proteins, inducing its suppressive activity by boosting the expression of the inducible T cell co-stimulator (ICOS) and CTLA4. According to the study, mice with Rheb1-Rheb2- or RagA-RagB-deficient Treg cells experienced a diminished effector activity of Treg cells and developed an autoimmune illness resembling scurvy [58].

The first steps in the breakdown of BCAAs involve decarboxylation by the branched-chain alpha-keto acid dehydrogenase (BCKDH) complex or transamination by aminotransferases (BCATs). Following these processes, metabolites of BCAAs are changed into succinyl- and acetyl-CoA, which are then involved in the tricarboxylic acid cycle (TCA). BCAT has a negative regulatory effect on glycolysis and mTOR in CD4+ T cells. Activated T cells from mice with cytosolic branched chain aminotransferase (BCATc) deficiency showed an increase in mTORC1 activation compared to control healthy mouse T cells [61]. Furthermore, a different study found that oral delivery of the leucine analog ERG240 specifically suppressed BCAT1 activity, lessening the severity of proliferative glomerulonephritis and collagen-induced arthritis in mice [62].

A GC-MS metabolomics study additionally identified a decrease in the concentrations of BBCAs and their metabolites such as 2-hydroxyisovalerate, 2-keto-3-methylvalerate, and 2-ketoisocaproate, indicating SLE may be involved in increased energy production, gluconeogenesis, and protein synthesis or decreased protein degradation or both [26].

During amino acid catabolism, the carbon skeleton and amino groups are processed in separate but interconnected pathways: the TCA and the urea cycles, respectively. Amino acids entering the TCA cycle contribute to energy generation, but in humans, the oxidative energy derived from amino acid catabolism comprises only a small fraction. Thus far, peripheral blood samples from SLE patients have shown a reduction in most of the amino acids examined, including gluconeogenic and ketogenic amino acids. The amino group’s catabolic result is ammonia, which is then transformed into urea via the urea cycle and eliminated in the urine. A metabolomic study carried out the measurement of the metabolites related to the urea cycle, finding that arginine, the immediate precursor of urea, and urea were increased in subjects with SLE, suggesting increased urea cycle activity in SLE, compared to healthy individuals [29, 63]. On top of this, a GC-MS metabolomics study identified a decrease in the concentrations of BBCAs and their metabolites such as 2-hydroxyisovalerate, 2-keto-3-methylvalerate, and 2-ketoisocaproate. This suggests that SLE may be associated with increased energy production, gluconeogenesis, protein synthesis, decreased protein degradation, or both [26].

Glutamate appears to be a particularly critical amino acid for T cell activation because, upon their activation, T cells consume glutamine at rates that are comparable to or higher than those of glucose [64]. Through the amino acid transporter 2 (ASCT2), CD4+ T cells are able to bind glutamine, which in turn affects the in vivo and in vitro production of proinflammatory Th1 and Th17 cells. It has been noted, meanwhile, that neither Th2 nor regulatory T cell immunological responses are impacted by the genetic ablation of ASCT2. ASCT2−/− T cells exhibit a reduction in oxygen consumption, lactate generation, and glucose uptake, indicating that glutamine is crucial for the way T cells respond to abrupt changes in their metabolic needs [37, 65].

Glutamine is also a fundamental component of protein synthesis and has a role in the production of fatty acids, nucleotides, and redox regulation, among other critical activities that promote T cell proliferation. Citrate, which is produced from glycolytic pyruvate, is exported from the mitochondria and employed in the synthesis of lipids in activated lymphocytes. Through the cycle of tricarboxylic acids, α-ketoglutarate, which is produced from glutamine, contributes to the formation of citrate. After that, citrate is used to produce acetyl groups for fatty acid synthesis. By means of this process, α-ketoglutarate supplies the building blocks needed to synthesize polyamines, which are essential for the synthesis of nucleotides. Ultimately, glutamate—the initial byproduct of glutamine oxidation—acts as a metabolic bridge for the production of glutathione and has a significant impact on lymphocytes’ state of oxidative stress [37, 56].

Glutaminase, the enzyme that is responsible for converting glutamine into glutamate, promotes the proliferation and activation of Th17 cells through various mechanisms. The cAMP responsive element modulator, which is overexpressed in T cells of SLE patients and MRL/lpr mice prone to SLE, regulates the production of glutaminase [66]. Multiple investigations have been carried out to elucidate the impact of this enzyme on the pathophysiology of SLE. Bis-2-(5-phenylacetamide-1,3,4-thiadiazole-2-yl sulfide) ethyl (BPTES), a glutaminase 1 hybridizer, has been shown to reduce Th1 cell development and disease activity in mice with experimental autoimmune encephalomyelitis [67]. This inhibitor, BPTES, also improves disease activity SLE in MRL/lpr mice [55]. Lastly, another enzyme, glutamate oxaloacetate transaminase 1 (GOT1), which converts glutamate to α-ketoglutarate, helps to potentiate Th17 differentiation through epigenetic processes. Selective inhibition of GOT1 by small hairpin (shRNA) RNA silencing or aminooxyacetic acid (AOA) therapy decreased notably the Th17 differentiation of murine T cells [55].

Furthermore, a study based on H single-voxel spectroscopy specifically identified glutamine as a potential biomarker for cerebral disease activity in SLE patients since the levels of glutamine were found to be altered in the brain of SLE patients even before neurologic and imaging manifestations emerged [26, 30].

Another metabolite that has been associated with SLE pathogenesis is tryptophan. This is an essential amino acid whose bioavailability is strictly based on dietary supplement and the subsequent degradation by the intestinal microbiota [43]. Tryptophan is the precursor of several metabolites produced by microbial and endogenous enzymes. The tryptophanase enzymes (Tnase) in the gut microbiota convert dietary tryptophan into bioactive catabolites, including indole and other indole derivatives via several pathways. Moreover, tryptamine, which is produced by some bacteria via the enzyme tryptophan decarboxylase, controls intestinal transit by binding to serotonin receptors [68].

Multiple mechanisms have been proposed to explain how tryptophan contributes to the development and progression of lupus. Low tryptophan concentrations but high kynurenine levels in the serum of lupus-prone mice were indicative of a skewed tryptophan metabolism through the kynurenine pathway, as reported by Brown et al. [31]. In addition, Pearl et al. [32] showed, thanks to a quantitative metabolome analysis based on peripheral blood lymphocytes, that N-acetylcysteine (NAC) therapy may be able to reverse the accumulation of kynurenine and kynurenic acid in patients with SLE. Exogenous kynurenine has been shown in some studies to enhance Th1 polarization of CD4+ T cells and decrease Treg cell polarization of cytotoxic T cells, indicating that kynurenine supports pro-inflammatory T cell phenotypes. According to the same study [32], kynurenine causes the activation of the target of rapamycin (mTOR) in human T cells, which contributes to the high level of mTOR activation that characterizes CD4+ T cells in patients with SLE. Proinflammatory cytokines, such as interferon gamma (IFNγ) and interleukin 17 (IL-17), which are essential to the pathophysiology of SLE, have also been observed to be over-expressed and over-activated in CD4+ T cells with active hypomethylation of genes in the mTOR pathway [69].

Lastly, many tryptophan-derived metabolites, including indole-3-aldehyde, indole-3-acetic acid, 3-methylindole, and tryptamine, are aryl hydrocarbon receptor (AhR) ligands [31, 70]. Through the regulation of P53, FasR, Bcl-2, and cell cycle kinases, AhR activation controls numerous essential biological functions, including cell cycle progression, apoptosis, and cell proliferation. AhR stimulation increases the regulation of genes encoding cytokines such as IL-10, which regulate immune tolerance in SLE [31].

2.5 Gut-microbiome-derived metabolism

Lupus patients often experience gastrointestinal symptoms that are atypical and nonspecific. Until 2014, rheumatologists and researchers dismissed the idea that gut dysbiosis could be associated with SLE due to the lack of clear evidence. However, a breakthrough came in 2014 when Hevia et al. [71] demonstrated, for the first time in a group of 20 SLE patients, a distinct dysbiosis characterized by a higher abundance of Bacteroidetes and a significant decrease in the Firmicutes/Bacteroidetes ratio compared to healthy individuals. Since then, subsequent studies have revealed similarities in SLE-related gut microbiota across different regions, such as Spain and China. In these populations, SLE patients exhibited an enrichment of Prevotellaceae compared to their healthy counterparts [70, 72]. Moreover, He et al. identified an increased prevalence of specific genera, including Rhodococcus, Eggerthella, Klebsiella, Prevotella, Eubacterium, Flavonifractor, and Incertae sedis, along with a decrease in Dialister and Pseudobutyrivibrio [72]. A study by Azzouz et al. [73] found that SLE patients had reduced species richness diversity, particularly pronounced in those with a high SLE Disease Activity Index (SLEDAI). This study highlighted a greater representation of Ruminococcus gnavus (RG) of the Lachnospiraceae family in SLE patients, which is known to contribute to gut barrier impairment. The combination of increased intestinal barrier permeability and gut dysbiosis in SLE patients has been linked to the translocation of pathogenic bacteria, bacterial endotoxins, toxic metabolites, and an elevation in circulating microbial components. Although these changes may not manifest clinically as infections, they contribute to pathological alterations and immune system dysregulation [74].

The gut microbiota constitutes an intricate assembly of bacteria crucial for digestion, playing a key role in facilitating the breakdown of nutrients through enzymes absent in the human genome [75]. Consequently, bacterial metabolites significantly influence the metabolic processes related to nutrient absorption [70]. Numerous studies have substantiated this concept by delineating the abnormal metabolism of amino acids, lipids, and carbohydrates observed in individuals with SLE [76, 77, 78].

A metabolomic investigation using GC-MS revealed a notable elevation of 2-hydroxyisobutyrate in SLE patients experiencing active disease compared to both healthy individuals and SLE patients with inactive disease. This finding suggests a potential link between altered gut microbial metabolism and SLE morbidity, particularly in cases of active SLE, as 2-hydroxyisobutyrate is primarily derived from the degradation of valine by gut microbes [26]. In an ongoing study by our group, we also found a group of bacterial metabolites associated to SLE by urinary metabolomic profiling using LC/GC-MS-MS (unpublished data).

2.6 Lipid metabolism

Examinations of lipid metabolism, conducted in both the serum and feces of SLE patients, indicate a heightened susceptibility to developing lipid profile disorders compared to individuals without the disease [33, 79]. The proposition that dyslipidemia plays a role in the pathogenesis of SLE is substantiated by the elevated occurrence of ischemic heart disease among SLE patients, ranging from 3.8–16%, a figure nearly 10 times greater than that observed in the general population and 50 times higher than the prevalence seen in young women of reproductive age. Furthermore, it has been established that dyslipidemia is prevalent in 68–100% of adult individuals with SLE [80]. Studies exploring lipid metabolism and its association with SLE revealed that more than 60% of altered metabolites in the serum of SLE patients are lipids [33].

Metabolomic and lipidomic investigations have revealed elevated levels of medium-chain fatty acids (MCFA) and free fatty acids (FFA), along with a reduction in long-chain fatty acids (LCFA) in the profiles of individuals with SLE. Short-chain fatty acids (SCFA) can efficiently enter the mitochondrial matrix for beta oxidation directly through the inner mitochondrial membrane, whereas LCFA require assistance from carnitine transport, a process found to be diminished in SLE patients compared to healthy controls [63, 80, 81]. Regarding lipid membranes, a decrease in most phospholipids was observed in individuals with SLE, possibly indicating an increase in cellular turnover. Lipid metabolism in SLE has also been linked to heightened oxidative stress, evidenced by elevated levels of lipid peroxidation products such as malonaldehyde acid (MDA), 9-hydroxyctadecadienoic acid (9-HODE), and 13-HODE, coupled with a decrease in antioxidants such as α-tocopherol, glutathione, and their precursors. Lipid peroxidation, associated with increased free radical activity and damage to cellular membranes, organelles, and/or DNA, has been implicated in cardiovascular and renal complications in individuals with SLE [82, 83, 84].

In an investigation conducted by Yan et al. it was observed that individuals with SLE had notably lower levels of glycerol, oleic acid, and arachidonic acid, while 1-monopalmitin levels were significantly higher compared to control subjects. Glycerol is an essential constituent of triglycerides and phospholipids, whereas 1-monopalmitin falls under the category of monoacylglycerols. In addition, this study revealed that patients with active SLE exhibited lower levels of linoleic acid in comparison to the control group [26].

The impact of lipid metabolism on the functions of immune cells and the pro-inflammatory processes in SLE has been extensively documented. Cells of the innate immune system, like macrophages, engage with oxidized Low-Density Lipoprotein (LDL) particles through scavenger receptors in atherosclerotic plaques. This interaction results in lipid saturation, the production of pro-inflammatory cytokines, and the recruitment of immune cells. In SLE, this process is exacerbated due to elevated circulating levels of LDL and alterations in macrophage function. This dysfunction is induced by the direct activation of hepatic X receptors (LXRs) by lipids, which regulate cellular cholesterol levels and immune functions, influencing factors such as the production of IL-23 and IL-17 and phagocytic pathways [85, 86].

The hyperlipidic environment in individuals with SLE has also been reported to impact T cells. Elevated levels of oxidized LDL have been shown to indirectly enhance T cell activation through monocyte uptake [85, 87]. Furthermore, SLE patients exhibit increased cholesterol and glycosphingolipids in the membranes of T cells, leading to alterations in the composition of signaling platforms known as lipid rafts. These rafts are crucial for T cell receptors to provide stimulatory signals that regulate cell function and inflammation. This alteration is partly attributed to the expression of genes responsible for lipid metabolism in individuals with SLE, but it could also result from changes in the cellular uptake of LDL/VLDL cholesterol and modifications in cholesterol efflux to High-Density Lipoprotein (HDL) [85, 88].

A combined study utilizing 16S sequencing and LC-MS metabolomics characterized alterations in the gut microbiome, as well as fecal and serum metabolomes in SLE patients. The findings indicated a significant correlation between specific bile acids, including deoxycholic acid, glycolic acid, ursodeoxycholic acid, and arachidonic acid, and the SLE Disease Activity Index (SLEDAI) score in patients. Moreover, these bile acids exhibited a strong predictive ability for disease activity [33]. Besides their well-known role in lipid metabolism, bile acids are also signaling molecules that exercise their functions through the activation of the bile acid receptors. A study reported a decrease in FXR receptors in subjects with SLE and in MRL/lpr models of lupus with liver dysfunction [89]. Notably, the administration of chenodeoxycholic acid, an FXR receptor agonist, in mice demonstrated suppression in the expression of inflammatory cytokines such as TNF-α, IFN-γ, and IL-6.

In addition to their impact on the liver, bile acids were identified to modulate intestinal immunity in a separate study. This research revealed that certain metabolites derived from lithocholic acid (LCA), such as 3-oxoLCA and isoalloLCA, could inhibit the differentiation of Th17 cells by directly binding to a crucial transcription factor, retinoid-related orphan receptor-γt (RORγt). These metabolites were found to enhance FOXP3 gene expression by generating mitochondrial reactive oxygen species (mitoROS), leading to the expansion of regulatory T cells [70, 90].

Advertisement

3. Metabolomics in urine and its alteration in the context of LN

One of the studies in LN metabolomics was conducted by Kalantari et al., who used 1H nuclear magnetic resonance (NMR-1H) to compare the urinary metabolic profile of LN patients, patients with SLE without renal damage, and healthy controls (HC). In the aforementioned study, biomarkers were identified to assess their diagnostic effectiveness in search of operative characteristics of the receptor (ROC). Three metabolites were used including 3,4-dihydroxyphenylacetdehyde, beta-alanine, and 2,2-dimethylsuccinic acid of which the combination of 2,2-dimethylsuccinic and DOPAL was associated with greater diagnostic sensitivity when determining acute renal injury, suggested as a diagnostic panel for LN with an AUC of 0.89, sensitivity of 81%, and specificity of 100% [91].

Among the pathways impaired in LN pathogenesis, twenty-four pathways were significant. Nicotinate and nicotinamide metabolism, AXIN degradation, and DNA nucleotide excision repair pathways were the top three impaired pathways in LN patients [91]. Souliotis and colleagues also reported DNA repair issues in active LN patients, resulting in higher levels of DNA damage than those with inactive disease [92].

In another study based on gas chromatography/mass spectrometry in patients with SLE and LN, 81 metabolites were detected, of which mainly highlights amino acids, nucleotides, organic acids, amines, fatty acids, glyceric acid, creatinine, p-cresol, myoinositol, treonate, hypurate, and urate [22]. However, this study did not differentiate between patients with SLE and LN.

Β-alanine is one of the two amino acids (along with histidine) that produce carnosine. The synthesis and metabolism of carnosine develops mainly at the renal level. Its metabolism favors eliminating reactive oxygen species. A significant decrease in urinary excretion of beta-alanine has been observed in LN patients, which could lead to a decrease in the carnosine content of the kidneys in LN patients, making their kidneys susceptible to oxidative stress insults [91].

2,2-Dimethylsuccinate (2,2-DMS) performs a plasma and urinary function by selectively inhibiting sodium-dependent dicarboxylate transporters (NaDC-3 and NaDC-1), which are located at the level of the proximal renal tubule in the luminal and basolateral plasma membranes. Transporters import intermediates from the Krebs cycle for the cells of the proximal tubule to use as energy substrates, among these compounds is succinate. The import of 2,2-DMS is done with higher speed compared to the suction given at greater affinity with the conveyor. Therefore, a decrease in 2,2-DMS levels in the urine of patients with LN may be secondary to increased activity of these transporters in patients with LN [91].

Class IV of the LN favors the proliferation of endocapillary and extracapillary cells, which would explain the energy requirement. There is also evidence that increased levels of nicotinate and nicotinamide (NMN), which participate in the metabolism of vitamin B3, is directly related to energy metabolism. When increasing dicarboxylate transporters, there is a decrease in levels of 2,2-DMS [91].

3.1 Lipids

Within the sphingolipid family are glycosphingolipids that favor cell proliferation and inflammation. At the renal level, they are related to a variety of diseases including glomerulonephritis, renal neoplasia, and renal polycystosis [93]. Tamara and colleagues aimed to identify the role of glycosphingolipids in LN. They utilized the LMR/MpJ-FasLpr/J (LMR/lpr) mouse model of LN, as well as human renal biopsies and urine samples [93]. Elevations of renal and urinary glycosphingolipids occurred in mice prone to lupus; a relationship was determined with the positive transcriptional regulation of neuraminidase that initiates its function in early stages of the disease and progressively increases its activity favoring the elevation of glycosphingolipids. Urinary levels of neuraminidase and lactosilceramide are higher in patients with lupus and LN. Complex glycosphingolipids are formed with the participation of lactosilceramide, example of which is the formation of gangliosides with the addition of sialic acid residues in conjunction with lactosilceramide. Glycosphingolipids are transported within the cell and are mainly located in the plasma membrane. Neuraminidases are enzymes that eliminate sialic acids and proteins and are located in various cell structures at the level of the lysosome, cytosol, plasma membrane, and mitochondria (NEU 1, 2, 3, 4, respectively), of which NEU1 has the ability to secrete itself extracellularly to act on the plasma membrane. Given the above, there is a regeneration of lactosilceramide through NEU enzymes. These data strongly suggest that glycosphingolipids may serve as early markers of LN [93].

With the evolution of the disease, the histological characteristics of the LN are changing; in the follow-up, good or poor response to treatment is evidenced. However, at the time, it is not possible to determine changes in the LN dynamically and in real time, and it may affect the start of therapy and follow-up and thus the prognosis [23].

In the evaluation of the patient, the differentiation of proliferative LN (Class III/IV) and pure membranous LN (Class V) is complex, since they present signs and symptoms in common; both are associated with proteinuria, alterations in blood pressure and kidney function. Pronounced proteinuria is the hallmark of focal segmental glomerulosclerosis (FSGS). One histological characteristic of FSGS is podocyte injury, leading to varying degrees of proteinuria and potentially hypoalbuminemia. These clinical and histological characteristics can also occur with active LN [23].

Romick et al. conducted a study with the aim of identifying urinary metabolites that could discriminate between proliferative LN (Class III/IV), pure membranous LN (Class V), and primary FSGS, using metabolomics based on NMR spectroscopy [23]. It was determined that the urinary metabolites citrate and taurine allow differentiating between proliferative LN and pure membranous LN (Table 2). In Class V, citrate levels are up to eight times lower and taurine levels normal, compared to Class III/IV where citrate levels may be normal and taurine up to 10 times lower. Additionally, the levels of urinary hypurate were evaluated, which allowed us to distinguish with precision between Class V that presents normal levels of taurine, compared with FSGS, with the absence of hypurate [23]. In a study conducted by Ganguly et al., it was demonstrated that LN patients exhibit significantly low levels of urinary creatinine/citrate compared to clinically healthy individuals, levels that increased significantly after 6 months of cyclophosphamide treatment. In the same study, it was shown that LN patients had significantly high levels of the acetate/creatinine ratio in urine, levels that did not change after immunosuppressive therapy [95].

MetaboliteType of sampleDiseaseMethod appliedReferences
3,4-Dihydroxyphenylacetdehyde, beta-alanine
2,2-dimethylsuccinic acid
UrineLNNMR-1H[91]
Neuraminidase, glucosylceramide (GlcCer) and lactosylceramide (LacCer), Neu1Urine human renal biopsies
LMR/MpJ-FasLpr/J (LMR/lpr) mouse model of LN
LNMatrix-assisted laser desorption/ionization imaging MS (MALDI-IMS)[93]
Citrate and taurine hypurateUrineLNNMR spectroscopy[23]
LDL/VLDL lipoproteins, sorbitol, glycolic acid acetate, cortisol, creatinine, L-aspartyl-L-phenylalanineSerumLNUPLC-HRMS[94]

Table 2.

Metabolites in SLE and LN.

The relationship of the metabolites citrate and taurine with kidney function has been determined. Normally, the kidney filters and reabsorbs metabolites, which allows a metabolic balance, so in case of functional deterioration, an alteration in the metabolic profile will be observed [96].

3.2 Serum metabolomics

In LN, a wide range of alterations in metabolic pathways are observed, including glycolysis, amino acid metabolism, and lipid metabolism, resulting in a consequential elevation or reduction that generates a metabolic signature allowing the distinction of patients with SLE. However, variations are observed depending on the region, population, and disease state. In general terms, patients with LN exhibit elevated serum levels of lipid metabolites (including LDL/VLDL lipoproteins), sorbitol, and glycolic acid metabolites and reduced levels of acetate, cortisol, creatinine, and L-aspartyl-L-phenylalanine [94]. Compared to SLE, metabolic variations are observed, such as an increase in LDL/VLDL lipoproteins (triglycerides and fatty acids) and reduced serum levels of acetate [97].

In lupus nephritis, reduced levels of several glucogenic amino acids (such as glycine, alanine, valine, glutamate, proline, and histidine) and ketogenic amino acids (such as leucine) have been determined. This could lead to abnormal catabolism of amino acids and biosynthesis of important proteins in various biological processes, such as cell cycle advancement, genetic transcription, inflammatory responses, and autoimmunity. It has been described that lower serum histidine levels could be closely related to protein waste, energy, inflammation, and oxidative stress. In general, elevated serum glucose levels and reduced levels of most amino acids guide changes in energy production, such as reduced aerobic glycolysis and the use of metabolites as amino acids and ketone bodies as a source of energy [98, 99].

Li J, Xie X et al. conducted a metabolomic study on human serum that revealed a wide range of differential metabolic signatures in patients with LN and idiopathic nephrotic syndrome (INS). Significant metabolic alterations were found in five metabolites, including cortisol, creatinine, sorbitol, L-aspartyl-L-phenylalanine, and glycolic acid [94]. Furthermore, combined forms of biomarkers proved to be more effective in the diagnosis of LN than a single one.

The AUC (area under the curve – ROC) was 0.85, the highest obtained, when combining theophylline, oxidized glutathione, and capric acid, which indicates a very good diagnostic accuracy, with a sensitivity and specificity of 87.50 and 67.86%, respectively; according to the study data in general, the positive predictive value (PPV) and the predictive value negative (NPV) were 75.68 and 82.61%, respectively [94].

3.3 Cortisol

Low serum levels of cortisol play a proinflammatory role, being negatively correlated with the degree of systemic inflammation. Zietz et al. reported that serum cortisol is negatively correlated with the degree of systemic inflammation in SLE. The study by Li J, Xie X et al. indicated that a significant decrease in the amount of cortisol is associated with a high risk of LN and perhaps an impact on the patient’s prognosis [94].

Straub et al., in their study, related that a low level of serum cortisol may be due to a reduction in adrenal steroidogenesis secondary to the inflammatory process and adrenal insufficiency [97].

On the other hand, in the work carried out by Judd et al., it was concluded that proinflammatory cytokines (such as tumor necrosis factor (TNF)) repress relevant enzymatic steps in steroidogenesis that occurs in adrenalocortical cells. The decrease in cortisol levels in patients with LN may be due to inflammation and adrenal insufficiency in patients with LN [94, 97].

3.4 Glycolic acid

Glycolic acid, or glycocholic acid, is a crystalline bile acid that plays a role in the emulsification of fats. It is an acyl glycine produced during the enzymatic metabolism of bile acids in the colonic environment. This compound acts as a fat solubilizer, facilitating its absorption. Additionally, it contributes to the homeostasis of bile acids, which act as signaling molecules with endocrine functions, thereby promoting the homeostasis of triglycerides, cholesterol, and glucose. In a study, the level of glycolic acid was reduced in patients with LN. The decrease in the level of glycolic acid in plasma could be due to the reduced concentration of glycolic acid in the plasma of LN patients [94].

Advertisement

4. Conclusion

The pathogenesis of SLE and LN is of complex etiology. There are no sufficiently specific biomarkers that allow for the noninvasive or minimally invasive prediction of renal involvement in the context of SLE. With the advent of “omics” approaches, a series of metabolites, amino acids, lipids, and proteins, among others have been discovered that could potentially serve as biomarkers for SLE and LN in the future. Metabolomics has unique characteristics that make it a methodology with enormous potential to discover metabolites associated with imbalances in crucial metabolic pathways for the organism. Large-scale validation of these metabolites discovered by these non-targeted approaches in patient cohorts from different regions will be necessary to uncover their role and impact in personalized medicine.

Advertisement

Acknowledgments

We are truly grateful to the rheumatology department and the renal unit of the Clinica de la Costa in Barranquilla City (Colombia) for the recruitment and sample collection of the patients enrolled in the study that was support for this chapter book.

Advertisement

Conflict of interests

The authors declare no conflict of interests.

Advertisement

Funding

This chapter book was supported by the Ministerio de Ciencia, Tecnología e Innovación (MINCIENCIAS) (SIGP code: 125384467468, Contract: RC No. 852 of 2019).

Advertisement

Abbreviations

2,2-DMS

2,2-dimethylsuccinate

9-HODE

9-hydroxyoctadecadienoic acid

13-HODE

13-hydroxyoctadecadienoic acid

AhR

aryl hydrocarbon receptor

Anti-DNA

antibodies against DNA

Anti-dsDNA

antibodies against double-stranded DNA

Anti-Sm Antibody

anti-smith antibody

AOA

aminooxyacetic

ASCT2

alanine serine cysteine transporter 2

AUC

area under the curve - ROC

BCAAs

branched-chain amino acids

BCAT1

branched-chain amino acid transaminase 1

BCATs

branched-chain amino acid aminotransferases

BCKDH

branched-chain alpha-keto acid dehydrogenase

Bcl-2

B-cell lymphoma 2

BPTES

bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide

cAMP

cyclic adenosine monophosphate

CTLA4

cytotoxic T-lymphocyte-associated antigen 4

ERG240

leucine analogue

ESR

erythrocyte sedimentation rate

FasR

Fas receptor

FFA

free fatty acids

FOXP3

transcription factor forkhead box protein 3

FSGS

focal segmental glomerulosclerosis

FXR receptors

farsenoid X receptors

GC-MS-MS

gas chromatography with tandem mass spectrometry

GC-MS

gas chromatography - mass spectrometry

GCL

glutamate cysteine ligase

GFR

glomerular filtration rate

GOT1

glutamate oxaloacetate transaminase 1

GSH

glutathione

HC

healthy controls

HDL

high-density lipoprotein

HIF1α

hypoxia inducible factor 1 subunit alpha

HPLC

high-performance liquid chromatography

ICOS

inducible t cell co-stimulator

IFγ

interferon gamma

IL-2

interleukin 2

IL-4

interleukin 4

IL-6

interleukin 6

IL-10

interleukin 10

IL-17

interleukin 17

IL-23

interleukin 23

INS

idiopathic nephrotic syndrome

ISN/RPS

International Society of Nephrology/Renal Pathology Society

LacCer

lactosilceramide

LAT-1

large neutral amino acid transporter 1

LC-MS-MS

liquid chromatography with tandem mass spectrometry

LCA

lithocholic acid

LCFA

long-chain fatty acids

LDL

low-density lipoprotein

LMR/lpr

LMR/MpJ-FasLpr/J

LN

lupus nephritis

LXRs

liver X receptors

MCFA

medium-chain fatty acids

MDA

malonaldehyde acid

mitoROS

mitochondrial reactive oxygen species

MRL/lpr mice

mice are homozygous for the lymphoproliferation spontaneous mutation

mTOR

mammalian target of rapamycin

mTORC 1

mammalian target of rapamycin complex 1

mTORC 2

mammalian target of rapamycin complex 2

NAC

N-acetylcysteine

NaDC

sodium-dependent dicarboxylate transporters

NALA

N-acetyl-leucine Amide

NETs

neutrophil extracellular traps

NEU1

neuraminidase 1 lysosome

NEU2

neuraminidase 2 cytosol

NEU3

neuraminidase 3 plasma membrane

NEU4

neuraminidase 4 mitochondria

NMN

nicotinamide

NMR-1H

1H nuclear magnetic resonance

NMR

nuclear magnetic resonance spectroscopy

NPV

negative predictive value

p53

tumor protein p53

PI3K/AKT/mTOR pathway

phosphoinositide 3-kinase signaling - protein kinase B - mTOR pathway

PPV

positive predictive value

Rag A/B

Ras-related GTP-binding protein A/B

RG

ruminococcus gnavus

Rheb1/2

Ras homolog enriched in brain protein 1/2

ROC

operative characteristics of the receptor

RORγt

retineic-acid-receptor-related orphan nuclear receptor gamma

ROS

reactive oxygen species

SCFA

short-chain fatty acids

shRNA

short hairpin RNA

Slc7a5 transporter

large neutral amino acid transporter 1 (LAT-1)

SLE

systemic lupus erythematous

SLEDAI

systemic lupus erythematous disease activity index

TCA

tricarboxylic acid

Th1 cells

type 1 T helper cells

Th2 cells

type 2 T helper cells

Th17 cells

type 17 T helper cells

Tnase

tryptophanase enzymes

TNF-α

tumor necrosis factor alpha

TNF

tumor necrosis factor

Treg cells

regulatory T cells

UHPLC-MS/MS

ultra-high performance liquid chromatography tandem mass spectrometry

VLDL

very low-density lipoprotein

References

  1. 1. Ruiz-Irastorza G, Khamashta MA, Castellino G, Hughes GR. Systemic lupus erythematosus. The Lancet Lond Engl. 2001;357(9261):1027-1032. DOI: 10.1016/S0140-6736(00)04239-2
  2. 2. Austin HA. Clinical evaluation and monitoring of lupus kidney disease. Lupus. 1998;7(9):618-621. DOI: 10.1191/096120398678920749
  3. 3. Cameron JS. Lupus nephritis. Journal of the American Society of Nephrology JASN. 1999;10(2):413-424. DOI: 10.1681/ASN.V102413
  4. 4. Hricik DE, Chung-Park M, Sedor JR. Glomerulonephritis. The New England Journal of Medicine. 1998;339(13):888-899. DOI: 10.1056/NEJM199809243391306
  5. 5. Golbus J, McCune WJ. Lupus nephritis. Classification, prognosis, immunopathogenesis, and treatment. Rheumatic Diseases Clinics of North America. 1994;20(1):213-242
  6. 6. Anders HJ, Saxena R, Hui ZM, Parodis I, Salmon JE, Mohan C. Lupus nephritis. Nature Reviews Disease Primers. 2020;6(1):7. DOI: 10.1038/s41572-019-0141-9
  7. 7. Almaani S, Meara A, Rovin BH. Update on lupus nephritis. Clinical Journal of the American Society of Nephrology. 2017;12(5):825-835. DOI: 10.2215/CJN.05780616
  8. 8. Cervera R, Khamashta MA, Font J, Sebastiani GD, Gil A, Lavilla P, et al. Morbidity and mortality in systemic lupus erythematosus during a 10-year period: A comparison of early and late manifestations in a cohort of 1,000 patients. Medicine (Baltimore). 2003;82(5):299-308. DOI: 10.1097/01.md.0000091181.93122.55
  9. 9. Cervera R, Khamashta MA, Font J, Sebastiani GD, Gil A, Lavilla P, et al. Systemic lupus erythematosus: Clinical and immunologic patterns of disease expression in a cohort of 1,000 patients. The European working party on systemic lupus erythematosus. Medicine (Baltimore). 1993;72(2):113-124
  10. 10. Parikh SV, Almaani S, Brodsky S, Rovin BH. Update on lupus nephritis: Core curriculum 2020. American Journal of Kidney Diseases. 2020;76(2):265-281. DOI: 10.1053/j.ajkd.2019.10.017
  11. 11. Lim SS, Bayakly AR, Helmick CG, Gordon C, Easley KA, Drenkard C. The incidence and prevalence of systemic lupus erythematosus, 2002-2004: The Georgia lupus registry: The Georgia lupus registry. Arthritis & Rhematology. 2014;66(2):357-368. DOI: 10.1002/art.38239
  12. 12. Banchereau R, Hong S, Cantarel B, Baldwin N, Baisch J, Edens M, et al. Personalized immunomonitoring uncovers molecular networks that stratify lupus patients. Cell. 2016;165(3):551-565. DOI: 10.1016/j.cell.2016.03.008
  13. 13. Lech M, Anders HJ. The pathogenesis of lupus nephritis. Journal of the American Society of Nephrology. 2013;24(9):1357-1366. DOI: 10.1681/ASN.2013010026
  14. 14. Yan B, Huang J, Dong F, Yang L, Huang C, Gao M, et al. Urinary metabolomic study of systemic lupus erythematosus based on gas chromatography/mass spectrometry. Biomed Chromatogr BMC. 2016;30(11):1877-1881. DOI: 10.1002/bmc.3734
  15. 15. Bengtsson AA, Trygg J, Wuttge DM, Sturfelt G, Theander E, Donten M, et al. Metabolic profiling of systemic lupus erythematosus and comparison with primary Sjögren’s syndrome and systemic sclerosis. PLoS One. 2016;11(7):e0159384. DOI: 10.1371/journal.pone.0159384
  16. 16. Davidson A. What is damaging the kidney in lupus nephritis? Nature Reviews Rheumatology. 2016;12(3):143-153. DOI: 10.1038/nrrheum.2015.159
  17. 17. Fanouriakis A, Kostopoulou M, Cheema K, Anders HJ, Aringer M, Bajema I, et al. 2019 Update of the joint European league against rheumatism and European renal association–European dialysis and transplant association (EULAR/ERA–EDTA) recommendations for the management of lupus nephritis. Annals of the Rheumatic Diseases 2020;79(6):713-723. DOI: 10.1136/annrheumdis-2020-216924
  18. 18. Califf RM. Biomarker definitions and their applications. Experimental Biology and Medicine. 2018;243(3):213-221. DOI: 10.1177/1535370217750088
  19. 19. Soliman S, Mohan C. Lupus nephritis biomarkers. Clinical Immunology. 2017;185:10-20. DOI: 10.1016/j.clim.2016.08.001
  20. 20. Stanley S, Vanarsa K, Soliman S, Habazi D, Pedroza C, Gidley G, et al. Comprehensive aptamer-based screening identifies a spectrum of urinary biomarkers of lupus nephritis across ethnicities. Nature Communications. 2020;11(1):2197. DOI: 10.1038/s41467-020-15986-3
  21. 21. Palazzo L, Lindblom J, Mohan C, Parodis I. Current insights on biomarkers in lupus nephritis: A systematic review of the literature. Journal of Clinical Medicine. 2022;11(19):5759. DOI: 10.3390/jcm11195759
  22. 22. For the IMID Consortium, Alonso A, Julià A, Vinaixa M, Domènech E, Fernández-Nebro A, et al. Urine metabolome profiling of immune-mediated inflammatory diseases. BMC Medicine. 2016;14(1):133. DOI: 10.1186/s12916-016-0681-8
  23. 23. Romick-Rosendale LE, Brunner HI, Bennett MR, Mina R, Nelson S, Petri M, et al. Identification of urinary metabolites that distinguish membranous lupus nephritis from proliferative lupus nephritis and focal segmental glomerulosclerosis. Arthritis Research & Therapy. 2011;13(6):R199. DOI: 10.1186/ar3530
  24. 24. Gergely P Jr, Grossman C, Niland B, Puskas F, Neupane H, Allam F, et al. Mitochondrial hyperpolarization and ATP depletion in patients with systemic lupus erythematosus. Arthritis and Rheumatism. 2002;46(1):175-190. DOI: 10.1002/1529-0131(200201)46:1<175::AID-ART10015>3.0.CO;2-H
  25. 25. Perl A, Hanczko R, Doherty E. Assessment of mitochondrial dysfunction in lymphocytes of patients with systemic lupus erythematosus. Methods in Molecular Biology Clifton NJ. 2012;900:61-89. DOI: 10.1007/978-1-60761-720-4_4
  26. 26. Yan B, Huang J, Zhang C, Hu X, Gao M, Shi A, et al. Serum metabolomic profiling in patients with systemic lupus erythematosus by GC/MS. Modern Rheumatology. 2016;26:1-25. DOI: 10.3109/14397595.2016.1158895
  27. 27. Zhang Q , Li X, Yin X, Wang H, Fu C, Wang H, et al. Metabolomic profiling reveals serum L-pyroglutamic acid as a potential diagnostic biomarker for systemic lupus erythematosus. Rheumatology Oxford England. 2021;60(2):598-606. DOI: 10.1093/rheumatology/keaa126
  28. 28. Zhang Q , Yin X, Wang H, Wu X, Li X, Li Y, et al. Fecal metabolomics and potential biomarkers for systemic lupus erythematosus. Frontiers in Immunology. 2019;3(10):976. DOI: 10.3389/fimmu.2019.00976. DOI: 10.3389/fimmu.2019.00976
  29. 29. Wu T, Xie C, Han J, Ye Y, Weiel J, Li Q , et al. Metabolic disturbances associated with systemic lupus erythematosus. PLoS One. 2012;7(6):e37210. DOI: 10.1371/journal.pone.0037210
  30. 30. Cagnoli P, Harris RE, Frechtling D, Berkis G, Gracley RH, Graft CC, et al. Reduced insular glutamine and N-acetylaspartate in systemic lupus erythematosus: A single-voxel (1)H-MR spectroscopy study. Academic Radiology. 2013;20(10):1286-1296. DOI: 10.1016/j.acra.2013.07.011
  31. 31. Brown J, Abboud G, Ma L, Choi SC, Kanda N, Zeumer-Spataro L, et al. Microbiota-mediated skewing of tryptophan catabolism modulates CD4+ T cells in lupus-prone mice. iScience. 2022;25(5):104241. DOI: 10.1016/j.isci.2022.104241
  32. 32. Perl A, Hanczko R, Lai ZW, Oaks Z, Kelly R, Borsuk R, et al. Comprehensive metabolome analyses reveal N-acetylcysteine-responsive accumulation of kynurenine in systemic lupus erythematosus: Implications for activation of the mechanistic target of rapamycin. Metabolomics. 2015;11(5):1157-1174. DOI: 10.1007/s11306-015-0772-0
  33. 33. He J, Chan T, Hong X, Zheng F, Zhu C, Yin L, et al. Microbiome and metabolome analyses reveal the disruption of lipid metabolism in systemic lupus erythematosus. Frontiers in Immunology. 2020;31(11):1703. DOI: 10.3389/fimmu.2020.01703
  34. 34. Fernandez D, Bonilla E, Phillips P, Perl A. Signaling abnormalities in systemic lupus erythematosus as potential drug targets. Endocrine, Metabolic & Immune Disorders Drug Targets. 2006;6(4):305-311. DOI: 10.2174/187153006779025748
  35. 35. Perl A. Systems biology of lupus: Mapping the impact of genomic and environmental factors on gene expression signatures, cellular signaling, metabolic pathways, hormonal and cytokine imbalance, and selecting targets for treatment. Autoimmunity. 2010;43(1):32-47. DOI: 10.3109/08916930903374774
  36. 36. Shah D, Sah S, Nath SK. Interaction between glutathione and apoptosis in systemic lupus erythematosus. Autoimmunity Reviews. 2013;12(7):741-751. DOI: 10.1016/j.autrev.2012.12.007
  37. 37. Yang Z, Matteson EL, Goronzy JJ, Weyand CM. T-cell metabolism in autoimmune disease. Arthritis Research & Therapy. 2015;17(1):29. DOI: 10.1186/s13075-015-0542-4
  38. 38. Kono M, Yoshida N, Tsokos GC. Amino acid metabolism in lupus. Frontiers in Immunology. 2021;12:623844. DOI: 10.3389/fimmu.2021.623844
  39. 39. Wu G, Fang YZ, Yang S, Lupton JR, Turner ND. Glutathione metabolism and its implications for health. The Journal of Nutrition. 2004;134(3):489-492. DOI: 10.1093/jn/134.3.489
  40. 40. Johnson MO, Wolf MM, Madden MZ, Andrejeva G, Sugiura A, Contreras DC, et al. Distinct regulation of Th17 and Th1 cell differentiation by glutaminase-dependent metabolism. Cell. 2018;175(7):1780-1795.e19. DOI: 10.1016/j.cell.2018.10.001
  41. 41. Fernandez DR, Telarico T, Bonilla E, Li Q , Banerjee S, Middleton FA, et al. Activation of mammalian target of rapamycin controls the loss of TCRzeta in lupus T cells through HRES-1/Rab4-regulated lysosomal degradation. Journal of Immunology. 2009;182(4):2063-2073. DOI: 10.4049/jimmunol.0803600
  42. 42. Lai ZW, Hanczko R, Bonilla E, Caza TN, Clair B, Bartos A, et al. N-acetylcysteine reduces disease activity by blocking mammalian target of rapamycin in T cells from systemic lupus erythematosus patients: A randomized, double-blind, placebo-controlled trial. Arthritis and Rheumatism. 2012;64(9):2937-2946. DOI: 10.1002/art.34502
  43. 43. Piranavan P, Bhamra M, Perl A. Metabolic targets for treatment of autoimmune diseases. Immunometabolism. 2020;2(2):e200012. DOI: 10.20900/immunometab20200012
  44. 44. Perl A. Review: Metabolic control of immune system activation in rheumatic diseases. Arthritis & Rheumatology (Hoboken NJ). Dec 2017;69(12):2259-2270. DOI: 10.1002/art.40223
  45. 45. Song W, Yuan J, Zhang Z, Li L, Hu L. Altered glutamate cysteine ligase activity in peripheral blood mononuclear cells from patients with systemic lupus erythematosus. Experimental and Therapeutic Medicine. 2014;8(1):195-200. DOI: 10.3892/etm.2014.1689
  46. 46. Duarte-Delgado NP, Cala MP, Barreto A, Rodríguez CLS. Metabolites and metabolic pathways associated with rheumatoid arthritis and systemic lupus erythematosus. Journal of Translational Autoimmunity. 2022;5:100150. DOI: 10.1016/j.jtauto.2022.100150
  47. 47. Van der Werf P, Orlowski M, Meister A. Enzymatic conversion of 5-oxo-L-proline (L-pyrrolidone carboxylate) to L-glutamate coupled with cleavage of adenosine triphosphate to adenosine diphosphate, a reaction in the -glutamyl cycle. Proceedings of the National Academy of Sciences of the United States of America. 1971;68(12):2982-2985. DOI: 10.1073/pnas.68.12.2982
  48. 48. Sitkovsky M, Lukashev D, Deaglio S, Dwyer K, Robson SC, Ohta A. Adenosine A2A receptor antagonists: Blockade of adenosinergic effects and T regulatory cells. British Journal of Pharmacology. 2008;153(Suppl 1):S457-S464. DOI: 10.1038/bjp.2008.23
  49. 49. Mandapathil M, Hilldorfer B, Szczepanski MJ, Czystowska M, Szajnik M, Ren J, et al. Generation and accumulation of immunosuppressive adenosine by human CD4+CD25highFOXP3+ regulatory T cells. The Journal of Biological Chemistry. 2010;285(10):7176-7186. DOI: 10.1074/jbc.M109.047423
  50. 50. Gao ZW, Zhao GH, Zhang Z, Huang J, Li ZY, Zhang HZ, et al. Serum adenosine deaminase activity is increased in systemic lupus erythematosus patients and correlated with disease activity. Immunologic Research. 2018;66(2):299-304. DOI: 10.1007/s12026-018-8984-9
  51. 51. Alunno A, Bartoloni E, Bistoni O, Nocentini G, Ronchetti S, Caterbi S, et al. Balance between regulatory T and Th17 cells in systemic lupus erythematosus: The old and the new. Clinical & Developmental Immunology. 2012;2012:823085. DOI: 10.1155/2012/823085
  52. 52. Li Y, Wang HF, Li X, Li HX, Zhang Q , Zhou HW, et al. Disordered intestinal microbes are associated with the activity of systemic lupus erythematosus. Clinical Science London England 1979. 2019;133(7):821-838. DOI: 10.1042/CS20180841
  53. 53. Colgan SP, Fennimore B, Ehrentraut SF. Adenosine and gastrointestinal inflammation. Journal of Molecular Medicine Berl Ger. 2013;91(2):157-164. DOI: 10.1007/s00109-012-0990-0
  54. 54. Haskó G, Cronstein B. Regulation of inflammation by adenosine. Frontiers in Immunology. 2013;8(4):85. DOI: 10.3389/fimmu.2013.00085
  55. 55. Kono M, Yoshida N, Maeda K, Suárez-Fueyo A, Kyttaris VC, Tsokos GC. Glutaminase 1 inhibition reduces glycolysis and ameliorates lupus-like disease in MRL/lpr mice and experimental autoimmune encephalomyelitis. Arthritis & Rheumatology Hoboken NJ. 2019;71(11):1869-1878. DOI: 10.1002/art.41019
  56. 56. DeBerardinis RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, Wehrli S, et al. Beyond aerobic glycolysis: Transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(49):19345-19350. DOI: 10.1073/pnas.0709747104)
  57. 57. Wang XY, Li N, Gu J, Li WQ , Li JS. The effects of the formula of amino acids enriched BCAA on nutritional support in traumatic patients. World Journal of Gastroenterology. 2003;9(3):599-602. DOI: 10.3748/wjg.v9.i3.599
  58. 58. Shi H, Chapman NM, Wen J, Guy C, Long L, Dhungana Y, et al. Amino acids license kinase mTORC1 activity and Treg cell function via small G proteins rag and Rheb. Immunity. 2019;51(6):1012-1027.e7. DOI: 10.1016/j.immuni.2019.10.001
  59. 59. Kato H, Perl A. Mechanistic target of rapamycin complex 1 expands Th17 and IL-4+ CD4-CD8- double-negative T cells and contracts regulatory T cells in systemic lupus erythematosus. Journal of Immunology Baltimore Md. 2014;192(9):4134-4144. DOI: 10.4049/jimmunol.1301859
  60. 60. Zheng Y, Delgoffe GM, Meyer CF, Chan W, Powell JD. Anergic T cells are metabolically anergic. Journal of Immunology Baltimore Md. 2009;183(10):6095-6101. DOI: 10.4049/jimmunol.0803510
  61. 61. Ananieva EA, Patel CH, Drake CH, Powell JD, Hutson SM. Cytosolic branched chain aminotransferase (BCATc) regulates mTORC1 signaling and glycolytic metabolism in CD4+ T cells. The Journal of Biological Chemistry. 2014;289(27):18793-18804. DOI: 10.1074/jbc.M114.554113
  62. 62. Papathanassiu AE, Ko JH, Imprialou M, Bagnati M, Srivastava PK, Vu HA, et al. BCAT1 controls metabolic reprogramming in activated human macrophages and is associated with inflammatory diseases. Nature Communications. 2017;12(8):16040. DOI: 10.1038/ncomms16040
  63. 63. Zhang T, Mohan C. Caution in studying and interpreting the lupus metabolome. Arthritis Research & Therapy. 2020;22:172. DOI: 10.1186/s13075-020-02264-2
  64. 64. Ardawi MS. Glutamine and glucose metabolism in human peripheral lymphocytes. Metabolism. 1988;37(1):99-103. DOI: 10.1016/0026-0495(88)90036-4
  65. 65. Nakaya M, Xiao Y, Zhou X, Chang JH, Chang M, Cheng X, et al. Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation. Immunity. 2014;40(5):692-705. DOI: 10.1016/j.immuni.2014.04.007
  66. 66. Yoshida N, Comte D, Mizui M, Otomo K, Rosetti F, Mayadas TN, et al. ICER is requisite for Th17 differentiation. Nature Communications. 2016;29(7):12993. DOI: 10.1038/ncomms12993
  67. 67. Kono M, Yoshida N, Maeda K, Tsokos GC. Transcriptional factor ICER promotes glutaminolysis and the generation of Th17 cells. Proceedings of the National Academy of Sciences of the United States of America. 2018;115(10):2478-2483. DOI: 10.1073/pnas.1714717115
  68. 68. Leung LH. Systemic lupus erythematosus: A combined deficiency disease. Medical Hypotheses. 2004;62(6):922-924. DOI: 10.1016/j.mehy.2004.01.017
  69. 69. Oaks Z, Winans T, Huang N, Banki K, Perl A. Activation of the mechanistic target of rapamycin in SLE: Explosion of evidence in the last five years. Current Rheumatology Reports. 2016;18(12):73. DOI: 10.1007/s11926-016-0622-8
  70. 70. Zhang L, Qing P, Yang H, Wu Y, Liu Y, Luo Y. Gut microbiome and metabolites in systemic lupus erythematosus: Link, mechanisms and intervention. Frontiers in Immunology. 2021;15(12):686501. DOI: 10.3389/fimmu.2021.686501), 10.3389/fimmu.2021.686501)
  71. 71. Hevia A, Milani C, López P, Cuervo A, Arboleya S, Duranti S, et al. Intestinal dysbiosis associated with systemic lupus erythematosus. MBio. 2014;5(5):e01548-e01514. DOI: 10.1128/mBio.01548-14
  72. 72. He Z, Shao T, Li H, Xie Z, Wen C. Alterations of the gut microbiome in Chinese patients with systemic lupus erythematosus. Gut Pathogens. 2016;8:64. DOI: 10.1186/s13099-016-0146-9
  73. 73. Azzouz D, Omarbekova A, Heguy A, Schwudke D, Gisch N, Rovin BH, et al. Lupus nephritis is linked to disease-activity associated expansions and immunity to a gut commensal. Annals of the Rheumatic Diseases. 2019;78(7):947-956. DOI: 10.1136/annrheumdis-2018-214856
  74. 74. Chi M, Ma K, Wang J, Ding Z, Li Y, Zhu S, et al. The immunomodulatory effect of the gut microbiota in kidney disease. Journal of Immunology Research. 2021;15(2021):5516035. DOI: 10.1155/2021/5516035
  75. 75. Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I, et al. Gut microbiota functions: Metabolism of nutrients and other food components. European Journal of Nutrition. 2018;57(1):1-24. DOI: 10.1007/s00394-017-1445-8
  76. 76. Mok CC. Metabolic syndrome and systemic lupus erythematosus: The connection. Expert Review of Clinical Immunology. 2019;15(7):765-775. DOI: 10.1080/1744666X.2019.1620601
  77. 77. Parker B, Urowitz MB, Gladman DD, Lunt M, Bae SC, Sanchez-Guerrero J, et al. Clinical associations of the metabolic syndrome in systemic lupus erythematosus: Data from an international inception cohort. Annals of the Rheumatic Diseases. 2013;72(8):1308-1314. DOI: 10.1136/annrheumdis-2012-202106
  78. 78. Demir S, Artim-Esen B, Şahinkaya Y, Pehlivan Ö, Alpay-Kanıtez N, Omma A, et al. Metabolic syndrome is not only a risk factor for cardiovascular diseases in systemic lupus erythematosus but is also associated with cumulative organ damage: A cross-sectional analysis of 311 patients. Lupus. 2016;25(2):177-184. DOI: 10.1177/0961203315603140
  79. 79. Sarkissian T, Beyene J, Feldman B, McCrindle B, Silverman ED. Longitudinal examination of lipid profiles in pediatric systemic lupus erythematosus. Arthritis and Rheumatism. 2007;56(2):631-638. DOI: 10.1002/art.22332
  80. 80. Sun W, Li P, Cai J, Ma J, Zhang X, Song Y, et al. Lipid metabolism: Immune regulation and therapeutic prospectives in systemic lupus erythematosus. Frontiers in Immunology. 2022;18(13):860586. DOI: 10.3389/fimmu.2022.860586)
  81. 81. Li Y, Liang L, Deng X, Zhong L. Lipidomic and metabolomic profiling reveals novel candidate biomarkers in active systemic lupus erythematosus. International Journal of Clinical and Experimental Pathology. 2019;12(3):857-866
  82. 82. Frostegård J, Svenungsson E, Wu R, Gunnarsson I, Lundberg IE, Klareskog L, et al. Lipid peroxidation is enhanced in patients with systemic lupus erythematosus and is associated with arterial and renal disease manifestations. Arthritis and Rheumatism. 2005;52(1):192-200. DOI: 10.1002/art.20780
  83. 83. Hu C, Du Y, Xu X, Li H, Duan Q , Xie Z, et al. Lipidomics revealed aberrant metabolism of lipids including FAHFAs in renal tissue in the progression of lupus nephritis in a murine model. Metabolites. 2021;11(3):142. DOI: 10.3390/metabo11030142
  84. 84. Frostegård J. SLE, atherosclerosis and cardiovascular disease. Journal of Internal Medicine. 2005;257(6):485-495. DOI: 10.1111/j.1365-2796.2005.01502.x
  85. 85. Robinson GA, MGL W, Wincup C. The role of immunometabolism in the pathogenesis of systemic lupus erythematosus. Frontiers in Immunology. 2022;26(12):806560. DOI: 10.3389/fimmu.2021.806560
  86. 86. Kiss M, Czimmerer Z, Nagy L. The role of lipid-activated nuclear receptors in shaping macrophage and dendritic cell function: From physiology to pathology. The Journal of Allergy and Clinical Immunology. 2013;132(2):264-286. DOI: 10.1016/j.jaci.2013.05.044
  87. 87. Ryu H, Lim H, Choi G, Park YJ, Cho M, Na H, et al. Atherogenic dyslipidemia promotes autoimmune follicular helper T cell responses via IL-27. Nature Immunology. 2018;19(6):583-593. DOI: 10.1038/s41590-018-0102-6
  88. 88. Waddington KE, Papadaki A, Coelewij L, Adriani M, Nytrova P, Kubala Havrdova E, et al. Using serum metabolomics to predict development of anti-drug antibodies in multiple sclerosis patients treated with IFNβ. Frontiers in Immunology. 2020;17(11):1527. DOI: 10.3389/fimmu.2020.01527
  89. 89. Lian F, Wang Y, Chen J, Xu H, Yang X, Liang L, et al. Activation of farnesoid X receptor attenuates liver injury in systemic lupus erythematosus. Rheumatology International. 2012;32(6):1705-1710. DOI: 10.1007/s00296-011-1874-2
  90. 90. Hang S, Paik D, Yao L, Kim E, Trinath J, Lu J, et al. Bile acid metabolites control TH17 and Treg cell differentiation. Nature. 2019;576(7785):143-148. DOI: 10.1038/s41586-019-1785-z
  91. 91. Kalantari S, Chashmniam S, Nafar M, Zakeri Z, Parvin M. Metabolomics approach reveals urine biomarkers and pathways associated with the pathogenesis of lupus nephritis. Iranian Journal of Basic Medical Sciences. 2019;22(11):1288-1295. DOI: 10.22038/ijbms.2019.38713.9178)
  92. 92. Souliotis VL, Vougas K, Gorgoulis VG, Sfikakis PP. Defective DNA repair and chromatin organization in patients with quiescent systemic lupus erythematosus. Arthritis Research & Therapy. 2016;18(1):182. DOI: 10.1186/s13075-016-1081-3
  93. 93. Nowling TK, Mather AR, Thiyagarajan T, Hernández-Corbacho MJ, Powers TW, Jones EE, et al. Renal glycosphingolipid metabolism is dysfunctional in lupus nephritis. Journal of the American Society of Nephrology. 2015;26(6):1402-1413. DOI: 10.1681/ASN.2014050508
  94. 94. Li J, Xie XW, Zhou H, Wang B, Zhang MJ, Tang FY. Metabolic profiling reveals new serum biomarkers of lupus nephritis. Lupus. 2017;26(11):1166-1173. DOI: 10.1177/0961203317694256
  95. 95. Ganguly S, Kumar U, Gupta N, Guleria A, Majumdar S, Phatak S, et al. Nuclear magnetic resonance–based targeted profiling of urinary acetate and citrate following cyclophosphamide therapy in patients with lupus nephritis. Lupus. 2020;29(7):782-786. DOI: 10.1177/0961203320918011
  96. 96. Sui W, Hou X, Che W, Yang M, Dai Y. The applied basic research of systemic lupus erythematosus based on the biological omics. Genes and Immunity. 2013;14(3):133-146. DOI: 10.1038/gene.2013.3
  97. 97. Guleria A, Pratap A, Dubey D, Rawat A, Chaurasia S, Sukesh E, et al. NMR based serum metabolomics reveals a distinctive signature in patients with lupus nephritis. Scientific Reports. 2016;6(1):35309. DOI: 10.1038/srep35309
  98. 98. Ouyang X, Dai Y, Wen J, Wang L. 1H NMR-based metabolomic study of metabolic profiling for systemic lupus erythematosus. Lupus. 2011;20(13):1411-1420. DOI: 10.1177/0961203311418707
  99. 99. Watanabe M, Suliman ME, Qureshi AR, Garcia-Lopez E, Bárány P, Heimbürger O, et al. Consequences of low plasma histidine in chronic kidney disease patients: Associations with inflammation, oxidative stress, and mortality. The American Journal of Clinical Nutrition. 2008;87(6):1860-1866. DOI: 10.1093/ajcn/87.6.1860

Written By

Alejandra Rojo-Sánchez, Agustín Abuchaibe, Ada Carmona, Valentina Arrieta-Bravo, David Chica-Valle, Yirys Díaz-Olmos and Lisandro Pacheco-Lugo

Submitted: 27 November 2023 Reviewed: 27 January 2024 Published: 24 June 2024