Skip to main content

The interconnected roles of TRIM21/Ro52 in systemic lupus erythematosus, primary Sjögren’s syndrome, cancers, and cancer metabolism

Abstract

Protein tripartite motif-containing 21 (TRIM21/Ro52), an E3 ubiquitin ligase, is an essential regulator of innate immunity, and its dysregulation is closely associated with the development of autoimmune diseases, predominantly systemic lupus erythematosus (SLE) and primary Sjögren’s syndrome (pSS). TRIM21 /Ro52 also features anti-cancer and carcinogenic functions according to different malignancies. The interconnected role of TRIM21/Ro52 in regulating autoimmunity and cell metabolism in autoimmune diseases and malignancies is implicated. In this review, we summarize current findings on how TRIM21/Ro52 affects inflammation and tumorigenesis, and investigate the relationship between TRIM21/Ro52 expression and the formation of lymphoma and breast cancer in SLE and pSS populations.

Introduction

TRIM21/Ro52, which is encoded by a gene on the short arm of chromosome 11, is a member of the TRIM protein family, the members of which are involved in multiple cellular functions, including apoptosis signaling, the regulation of innate immunity, and the suppression or activation of carcinogenesis [1, 2]. The structure of TRIM21/Ro52, a cytosolic Fc receptor, consists of an N-terminal RING domain with E3 ubiquitin ligase activity, a B-box domain, a central coiled-coil domain, and a PRY/SPRY domain at its C terminus [3, 4]. TRIM21/Ro52 is crucial in antigen presentation and the regulation of innate immunity in response to intracellular pathogens as a negative regulator of interferon production via the ubiquitination of interferon regulatory factor (IRF)3/5/7, which leads to subsequent proteasomal degradation and prevents further IFN transcription [5,6,7].

TRIM21/Ro52 and the antibodies that target it (anti-TRIM21/Ro52) are involved in many autoimmune diseases, especially rheumatic diseases, such as systemic lupus erythematosus (SLE) and primary Sjögren’s syndrome (pSS) [8]. The presence of anti-TRIM21/Ro52 is one of the key items to validate the diagnosis of pSS [9], and it can be detected in 42–50% of SLE patients [10]. In addition to the diagnostic role in SLE and pSS, TRIM21/Ro52 can have opposing effects in different cancers [11]. Higher expression of TRIM21/Ro52 is associated with better patient survival in some cancer types, such as diffuse large B-cell lymphoma (DLBCL), breast cancer, and renal cell carcinoma [12,13,14]. In contrast, TRIM21/Ro52 promotes cancer cell proliferation and migration in glioma and thyroid cancer, and it increases drug resistance in colorectal and pancreatic cancers [15,16,17].

Heterogeneity is a characteristic shared by SLE, pSS, and cancers at the phenotypic and genotypic levels. Meanwhile, growing evidence indicates that reprogramming cellular metabolism and immune dysfunction both contribute to autoimmune diseases and tumor development [18, 19]. Recent studies have suggested a higher risk of not only hematologic cancers but also solid tumors such as lung cancer and thyroid cancer in individuals with either SLE or pSS. On the other hand, the risk of certain types of malignancies, breast cancer, for example, is decreased compared with healthy controls in other data [20, 21].

The pathophysiologic mechanisms by which SLE and pSS patients have a greater risk of certain cancers have yet to be well understood, and the comprehension of the regulatory role of TRIM21/Ro52 in these diseases and malignancies remains partial. Herein, in this review, we summarize current evidence of TRIM21/Ro52 relevant connections to immune regulation and cellular metabolism in SLE, pSS, and cancers, then discuss how TRIM21/Ro52 may function in the tumorigenesis, especially lymphoma and breast cancer in SLE and pSS patients (Fig. 1).

Fig. 1
figure 1

Graphic summary of the interconnected roles of TRIM21/Ro52 in systemic lupus erythematosus, primary Sjögren’s syndrome, cancers, and metabolism

The role of TRIM21/Ro52 in SLE and pSS

TRIM21/Ro52 expression in individuals and animal models with SLE or pSS

SLE and pSS are both systemic rheumatic diseases characterized by abnormal B-cell activation and autoantibody production. The mRNA level and surface expression of TRIM21/Ro52 protein, which was initially noted in apoptotic cells [22], is higher in freshly isolated peripheral blood mononuclear cells of pSS patients as compared with those of healthy donors [23]. Transcription of TRIM21/Ro52 is upregulated by interferon regulatory factor (IRF)1 and 2 while suppressed by IRF4 and IRF8, which is consistent with the significantly higher level of IRF1 and IRF2 and overexpression of TRIM21/Ro52 seen in patients with Sjögren’s syndrome [24]. In addition, overexpression of TRIM21/Ro52 inhibits cell proliferation and enhances apoptosis in CD40-mediated cell death, which also results in a more significant amount of autoantigen that may trigger a greater autoimmune response [23].

However, deficiency in TRIM21/Ro52 expression is another SLE and pSS development mechanism. Ro52-null (Ro52–/–) mice, which are generated from one of the lupus mouse models and detected by green fluorescent protein(GFP) expression, develop progressive dermatitis from the site of their ear tag injury and later are positive for proteinuria detection and deposition of immune complexes as based on renal pathology [25]. The immune response of Ro52–/– mice to a contact-sensitizing agent is also significantly higher than that of Ro52+/+ mice, which includes a larger amount of T-cell activation and proinflammatory cytokine production. This TRIM21/Ro52 deficiency increases proinflammatory cytokine production, and the inflammatory infiltration depends on the IL-23–Th17 pathway [26].

The regulatory role of TRIM21/Ro52 in SLE and pSS

TRIM21/Ro52 plays a regulatory role in B-cell proliferation and differentiation. Ro52−/− mice show increased B-cell activation and markedly higher antibody production than wild-type [27]. Previous studies have reported that TRIM21/Ro52 directly regulates several IRFs that are involved in B-cell development, and the presence of TRIM21/Ro52 could suppress the canonical NF‐κB pathway via monoubiquitinating the phosphorylated IκB kinase subunit beta(IKKβ) and subsequent autophagy [28, 29]. Thus, the absence of TRIM21/Ro52 results in aberrant NF-κB activation, which is also explicitly required for the proliferation of activated B-cell DLBCL, the main subtype of DLBCL found in SLE patients [30,31,32]. In another study, researchers noted that Ro52–/– mice have enhanced resting B-cell differentiation and exhibit a lupus-like disease phenotype with increased urinary protein and serum double-stranded DNA antibody titers. In the same study, the authors demonstrated that SLE patients who are positive for anti-TRIM21/Ro52 have a significantly higher level of resting B-cell differentiation toward plasmablasts and increased antibody production in general as compared with seronegative SLE patients and healthy controls, indicating that anti-TRIM21/Ro52 impairs the TRIM21/Ro52 function of B-cell homeostasis [33].

Under normal physiologic circumstances, TRIM21/Ro52 negatively regulates type I IFN expression via degradation of IRF3/5; however, the IRF degradation process is downregulated in anti-TRIM21/Ro52 positive SLE patients with both increased TRIM21/Ro52 mRNA and IFN levels, whereas there is no correlation in the SLE population without these antibodies. This suggests that the autoantibody targeting TRIM21/Ro52 impairs TRIM21/Ro52-dependent IRF degradation, resulting in dysregulated IFN production and subsequent high TRIM21/Ro52 expression [34]. A study also showed that the presence and elevated serum level of anti-TRIM21/Ro52 in SLE and pSS patients are positively correlated with the abnormally increased titer of immunoregulatory cytokine, including IL-2, IL-4, IL-21, IL-22, and CXCL10 [35]. The incidence rate and the trigger associated with the production of nuclear-penetrating autoantibodies have not been sufficiently studied yet, whereas the mechanisms by which autoantibodies penetrate cells have been widely investigated and include both Fc receptor–dependent endocytosis and Fc receptor–independent pathways involving myosin and the nucleoside transporter, respectively [36].

Anti-TRIM21/Ro52 and the link to clinical presentations

Clinically, anti-TRIM21/Ro52 antibodies serve as a potential diagnostic and prognostic biomarker. In pSS patients, higher serum level of anti-TRIM21/Ro52 antibodies are associated with increased disease severity, including a greater rate of anemia and muscular involvement, despite no clear association between the presence of anti-TRIM21/Ro52 antibodies and xerostomia, one of the most notable clinical presentation found on pSS patients [37]. However, it is demonstrated in another study that higher anti-TRIM21/Ro52 antibody titers are actually correlated with severe involvement in parotid scintigraphy, parotid enlargement, and positive salivary gland biopsy, which are all important for pSS diagnostics, indicating a strong association of histopathological findings with anti-TRIM21/Ro52 antibodies [38]. Furthermore, in patients with suspected autoimmune diseases, higher isolated positive serum anti-TRIM21/Ro52 antibodies are associated with a higher diagnostic rate of immunologic disorders and malignancies as compared with the anti-Ro52 and anti-Ro60 double-positive population [39].

Association of TRIM21/Ro52 and cancers

TRIM21/Ro52 inhibits tumorigenesis in multiple cancers

TRIM21/Ro52 has a dual role in cancers, as it can promote or suppress tumor growth depending on cancer cell types. The downregulation of TRIM21/Ro52 is associated with a poor prognosis in patients with certain kinds of cancer, including DLBCL, breast cancer, gastric cancer, renal cell carcinoma, ovarian cancer, and colitis-associated cancer [12,13,14, 40,41,42,43] (Table 1).

Table 1 The role of TRIM21/Ro52 in different cancers and its effect on clinical outcomes

In breast cancer, overexpression of TRIM21/Ro52 promotes ubiquitination and degradation of Snail, resulting in the downregulation of E-cadherin transcription throughout the epithelial–mesenchymal transition process, thereby inhibiting migration and invasion capabilities in breast cancer cells [44]. TRIM21/Ro52 also negatively regulates the SET domain containing 7, histone lysine methyltransferase (SETD7, also known as SET7/9), which is involved in breast cancer cell proliferation, migration, and invasion, such that higher expression of TRIM21/Ro52 is associated with better outcomes in these patients [45]. In addition, degradation of spalt-like transcription factor 4 (SALL4), a transcription factor that promotes breast cancer cell proliferation and migration, is regulated by TRIM21/Ro52, and thus higher levels of TRIM21/Ro52 can reduce SALL4 levels [46]. The proteasomal degradation of transforming growth factor beta receptor 2 (TβRII) protein is also mediated by TRIM21/Ro52. By disrupting the TGF-β signaling pathway, TRIM21/Ro52 can suppress triple-negative breast cancer cell metastasis [47]. In addition, lower TRIM21/Ro52 expression in mutant p53 breast cancer patients is associated with a poorer clinical outcome as TRIM21/Ro52 deficiency leads to the accumulation of mutant p53 and the subsequent breast cancer progression [48].

In gastric cancer, higher TRIM21/Ro52 expression not only is correlated with a lower recurrence and a better 5-year survival rate but also enhances the chemosensitivity of the tumor cells to apatinib, an FDA-proved treatment for chemotherapy-refractory advanced gastric cancer [41, 49]. Similarly, colitis-associated and ovarian cancer studies indicate that TRIM21/Ro52 can suppress intestinal epithelial carcinogenesis and ovarian tumorigenesis, respectively [42, 43]. In addition, in renal cell carcinoma, overexpression of TRIM21/Ro52 destabilizes hypoxia-inducible factor 1 subunit alpha (HIF-1α), leading to the suppression of aerobic glycolysis and subsequent inhibition of both in vitro and in vivo tumor cell proliferation and migration [14]. A recent study, in contrast, found that TRIM21/Ro52 inhibits the expression of the lipogenic enzyme via the degradation of sterol regulatory element binding transcription factor 1 (SREBF1), attenuating lipogenesis and tumor growth in renal cell carcinoma [50]. These results also suggest the essential role of TRIM21/Ro52 in cancer metabolism, which will be further discussed in section "The role of TRIM21/Ro52 in cancer cell metabolism".

TRIM21/Ro52 acts as an oncogene in glioma, thyroid, and pancreatic cancer

In contrast to the findings described above, in the case of certain cancers—such as glioma, thyroid cancer, and pancreatic cancers—overexpression of TRIM21/Ro52 is associated with unfavorable clinical outcomes. In the context of glioma, TRIM21/Ro52 overexpression promotes cell cycling, proliferation, and migration of glioma cells by suppressing the p53–p21 pathway [15]. TRIM21/Ro52 plays a similar role in thyroid cancer, such that the expression of TRIM21/Ro52 is upregulated in thyroid cancer tissue, and higher TRIM21/Ro52 levels are associated with a higher risk of recurrence and lymph node metastasis, though the mechanism involved has not yet been investigated [16]. It was also observed in colorectal and pancreatic tumor cells that TRIM21/Ro52 overexpression in response to cisplatin, a widely used chemotherapeutic agent, would downregulate the level of pro-apoptotic WT1 regulator (PAWR), which is a tumor suppressor mediating apoptosis regulation in various cancer cells, and thus increases the resistance of colorectal and pancreatic tumor cells to cisplatin treatment. It is also demonstrated that high TRIM21/Ro52 expression in pancreatic tumor patients indicates worse survival outcome [17].

Cancer type with controversial TRIM21/Ro52 function

The function of TRIM21/Ro52 in colorectal cancer (CRC) and hepatocellular carcinoma (HCC) remains contradictory. TRIM21/Ro52 is found to interact with DLGAP1 antisense RNA 2 (DLGAP1-AS2), a long noncoding RNA, and promote the CRC cells' growth and metastasis [51]. In contrast, it is shown in CRC that TRIM21/Ro52 mediates the ubiquitination of MICAL-like 2 protein (MICALL2), which is proven to promote CRC cell proliferation and migration, and thereby the presence of TRIM21/Ro52 decrease the activity of MICALL2 in CRC tumorigenesis [52]. Another study also demonstrates that TRIM21/Ro52 ubiquitinates IGF2 mRNA binding protein 3 (IGF2BP3), which plays an essential role in CRC development, and that the use of the natural drug Berberine improves CRC by promoting the expression of TRIM21/Ro52 [53].

The study by Ding et al. showed that the downregulation of TRIM21/Ro52 contributes to hepatocarcinogenesis and is associated with a poor prognosis [40]. In contrast, Qi et al. found the opposite: the expression of TRIM21/Ro52 is higher in HCC tissues than in normal control tissues, and it is significantly correlated with tumor progression in HCC patients [54]. Furthermore, one target of the E3 ubiquitin ligase activity of TRIM21/Ro52 is p62, and the function of the downstream p62–Keap1–Nrf2 antioxidant pathway in HCC is also controversial; again, the expression of TRIM21/Ro52 has been shown to both involve in promoting and suppressing HCC progression [55, 56]. Further studies are needed to identify the functional role of TRIM21/Ro52 in HCC.

In addition, it is worth noting that TRIM21/Ro52 is an intracellular Fc receptor with extremely high affinity for IgG antibodies, which may lead to the co-precipitation of not just proteins directly interacting with TRIM21 but also the antibodies themselves, leading to a false positive result in co-immunoprecipitation (Co-IP) experiments for identifying potential interaction proteins of TRIM21 [57]. Future studies investigating on the proteins interplay involving TRIM21/Ro52 may require rigorous experimental design and validation steps using alternative methods such as bioluminescence resonance energy transfer (BRET) or a mutated PRY/SPRY domain of TRIM21/Ro52 with reduced affinity for IgG to rule out non-specific binding [58].

The role of anti-TRIM21/Ro52 antibodies in cancers

There have been conflicting results regarding the function of anti-TRIM21/Ro52 antibodies in different cancers [59, 60]. Anti-TRIM21/Ro52 positive is associated with poor survival in patients with esophageal squamous cell carcinoma [60]. In another study, although there was a significantly high prevalence of anti-TRIM21/Ro52 in ovarian cancer patients, the presence of this antibody was correlated with higher overall survival compared with the antibody-negative ovarian cancer population [61]. As increased TRIM21/Ro52 expression was associated with better outcomes in ovarian cancer in recent research, whether the antibody impairs TRIM21/Ro52 protein function or the presence of anti-TRIM21/Ro52 antibodies reflects the overexpression phenomenon of TRIM21/Ro52 in cancer requires further investigation [42].

Cancer profile and its pathophysiological mechanisms in SLE and pSS

Cancer epidemiology study and mechanisms of tumor development in SLE and pSS individuals

The overall cancer risk in SLE and pSS patients is slightly higher than that of their matched general population. Among all cancer types, hematologic cancers, especially non-Hodgkin lymphoma, are associated with the most significant increase in risk [62, 63]. SLE and pSS patients have a fourfold and an up to 44-fold higher risk of developing B-cell lymphoma, respectively [64, 65]. Concerning solid neoplasms, SLE patients have a higher incidence rate of lung cancer, cervical cancer, and cervical dysplasia. In contrast, the risk of mouth and throat cancer, thyroid cancer, and lung cancer is notably increased in patients with pSS [66, 67]. In contrast, the risk of hormonal-related cancers, which include breast, prostate, endometrial, and ovarian cancers, is lower in SLE and pSS patients than in their healthy counterparts [67, 68] (Table 2). This characteristic may be attributed to the shorter period of hormonal exposure in these patients due to premature ovarian insufficiency [69]; undiscovered genetic variants; and the use of nonsteroidal anti-inflammatory drugs (NSAIDs), which have a protective effect against breast cancer [70], in SLE and pSS patients [71]. However, there is still a lack of solid evidence, and the breast cancer risk in SLE and pSS patients shows striking geographical differences [72, 73].

Table 2 Cancer risk profiles in SLE and pSS patients

Pathophysiological mechanisms underlying the increased risk of non-hormonal-related malignancies, particularly hematologic cancers, in SLE or pSS patients may involve the use of certain immunosuppressive drugs, chronic inflammatory status, susceptibility of genes, cell-penetrating autoantibodies, and other conventional shared risk factors such as smoking and Epstein-Barr virus infection [74,75,76]. It has been confirmed that a dose-dependent use of cyclophosphamide increased the risk of hematologic malignancies in SLE patients. In contrast, hydroxychloroquine protects patients with SLE, and systemic glucocorticoid, cyclophosphamide, methotrexate, or azathioprine was not correlated with an increased cancer risk [77,78,79]. In pSS patients, there is limited study on the question of cancer risk and the use of immunosuppressive agents, but it has been reported that hydroxychloroquine presents a neutral effect on cancer development in the pSS population [80]. In addition, immunosuppressive treatments may indirectly promote oncogenic virus infections [81].

The pathogenesis of SLE involves the overexpression of a myriad of cytokines and the subsequent dysregulation of B-cell proliferation and differentiation, which overlaps with the formation of DLBCL, the most common type of lymphoma found in SLE patients [31, 82]. The production of free radicals due to neutrophil activation and the promotion of proinflammatory cytokines in SLE and pSS patients also lead to direct DNA damage, which may contribute to certain malignancies [83, 84]. On the other hand, it has been established that in pSS patients not only T and B lymphocytes, but the salivary epithelial cells aberrantly produce B cell-activating factor (BAFF) which leads to B cell hyperactivity and clonal expansion of B cells [85, 86]. Overactivation of B cells results in a wide spectrum of autoantibodies, such as rheumatoid factor, an antibody against the Fc portion of IgG, or autoantibodies against other autoantigens including TRIM21/Ro52 and SSA/Ro60, and the accumulation of these immune complexes may in return give rise to chronic antigenic stimulation and activation of NF‐κB pathway [87, 88]. As previously mentioned, TRIM21/Ro52, which is aberrantly expressed in SLE and pSS patients, regulates B cell homeostasis though NF‐κB pathway and thus drives the formation of lymphoma, especially mucosa-associated lymphoid tissue (MALT) lymphomas and DLBCL [89, 90].

The genomes of patients with SLE and pSS are more likely to have polymorphisms at specific loci that encode DNA repair factors and cytokine regulators, and these polymorphisms not only contribute to the development of autoimmune diseases but also may increase the risk of cancer [91, 92]. The gene that encodes A20, or TNF alpha-induced protein 3 (TNFAIP3), an immunoregulatory factor involved in the downregulation of the NF-κB pathway and the carcinogenesis of lymphoma and solid tumors, often includes a single-nucleotide polymorphism and one gene mutation in SLE patients that are absent in healthy individuals [93]. Furthermore, autoantibodies that penetrate cells in individuals with autoimmune diseases, may interfere with the tumorigenesis [94, 95]. It has been reported that 3E10, a cell-penetrating lupus anti-DNA antibody, is toxic to BRCA2-deficient cancer cells [94]. On the contrary, antiphospholipid antibodies, which bind to mitochondria through internalization and trigger cell death, are found in 30–40% of SLE patients and associated with an increased risk of hematologic cancers [95, 96].

In addition to the fact that SLE patients are associated with a greater risk of metabolic syndrome, which is a combination of well-acknowledged risk factors for cancer, including hypertension, diabetes, and obesity, changes in immunometabolism in these patients could act as another predisposing factor for developing cancer [97, 98]. Expression of glucose transporter 1 (GLUT1), a critical regulatory component in glucose metabolism, in immune cells is higher in SLE and pSS patients than in healthy controls [97, 99, 100]. The upregulation of GLUT1 is correlated with autoimmune disease severity, and various cancer studies have found it in common [101, 102]. Pyruvate kinase M2 (PKM2), an isoform of the pivotal regulatory enzyme, pyruvate kinase, in cell metabolism, is significantly elevated in the monocytes, dendritic cells, and B cells of SLE patients relative to that in the general population [103]. Activation of PKM2 is not only involved in toll-like receptors mediated inflammation and autoimmunity but also contributes to cancer formation [103, 104]. The growing amount of study regarding metabolic syndrome in individuals with SLE and pSS has shed light on the pathogenesis of the diseases themselves and provided new avenues for exploring the relationship between cancers and autoimmune diseases.

The expression of TRIM21/Ro52 and its association with lymphoma and breast cancer development in SLE and pSS patients

In SLE and pSS patients, the risk of developing lymphoma is significantly elevated. Ro52–/– mice, which serve as a model for SLE, have demonstrated both lupus-like symptoms and aberrant B-cell differentiation and proliferation [33]. TRIM21/Ro52 deficiency may be relevant to the increased risk of lymphoma because of NF‐κB pathway activation, and maintaining TRIM21 expression is also associated with a preferable clinical outcome in patients with lymphoma [12, 29]. It is, however, paradoxical that the expression of TRIM21 mRNA and protein in SLE and pSS populations is found to be higher than that in healthy controls, and this increased level of TRIM21 could result in increased cell death and enhanced autogenic antigen exposure with subsequent antigen stimulation of autoimmune B cells, thus serving as a connection with the development of lymphoma [105] (Fig. 2). These results suggest that lymphomagenesis in individuals with SLE and pSS is a multistep and multifactorial process and that maintaining the balanced expression of TRIM21 is essential for both the development of autoimmune diseases and the formation of lymphoma.

Fig. 2
figure 2

A summarized mechanism(s) that SLE or pSS may lead to an increased risk of lymphoma and a decreased risk of breast cancer. Down-regulation of TRIM21/Ro52 expression is associated with poor overall survival in both lymphoma and breast cancer. IKKβ: kappa‐B kinase subunit beta; SALL-4: spalt like transcription factor 4; SET7/9: SET domain containing 7, histone lysine methyltransferase; TβRII: growth factor beta receptor 2

The relationship between SLE and breast cancer represents an interesting contrast based on regional differences. Research from some Asian countries, such as Taiwan and Korea, demonstrates an increased risk of breast cancer in SLE patients [72, 106]. In contrast, other multi-center cohort studies, primarily conducted in the US and European countries, show decreased breast cancer risk among the SLE population [71]. Since it is demonstrated that there is higher TRIM21 expression among individuals with SLE or pSS and that several studies confirmed that overexpression of TRIM21 is associated with the inhibition of breast cancer development and a better prognosis for those individuals who develop breast cancer, it is worth further investigation of the TRIM21 expression level in SLE and pSS patients in these Asian countries.

The role of TRIM21/Ro52 in cancer cell metabolism

Cancer cells often exhibit a higher rate of glycolysis than normal cells to meet the energetic needs associated with rapid growth. This metabolic change is known as the Warburg effect [107]. In human glioblastoma, blocking of TRIM21/Ro52 activity due to protein kinase B (AKT) activation impairs the proteasomal degradation of phosphofructokinase-1 (PFK1), the rate-limiting glycolysis enzyme, resulting in the promotion of glycolysis and brain tumor proliferation [108]. Regardless of the changes in the cellular microenvironment, there is a tendency for TRIM21/Ro52 inactivation to result in higher PFK1 expression and higher glycolysis rates in human non-small-cell-lung-cancer cells [109].

TRIM21/Ro52 is not only essential for the regulation of glucose metabolism but also has a pivotal role in lipogenesis. Fatty acid synthase (FASN), a complex of critical enzymatic proteins in the fatty acid synthesis pathway, is commonly upregulated in tumor cells to support the need for lipids for active proliferation, and increased FASN expression is correlated with drug resistance, tumor metastasis, and reduced survival among individuals with cancer. TRIM21/Ro52 is responsible for the polyubiquitination and proteasome degradation of FASN [110]. A study by Gu et al., which examined the underlying mechanism of how FASN promotes lipogenesis and HCC progression, found that the acetylation of glyceronephosphate O-acyltransferase (GNPAT), a critical enzyme that regulates plasmalogens, stabilizes FASN via suppressing both TRIM21/Ro52-mediated GNPAT and FASN degradation [111].

The upregulation of the pentose phosphate pathway is another metabolic change that frequently occurs in cancer cells in response to the high level of reactive oxygen species [112]. TRIM21/Ro52 is responsible for the degradation of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme in the pentose phosphate pathway, and its feedback regulation on phosphatidylinositol 3-kinase (PI3K)/AKT pathway makes TRIM21/Ro52 a potential therapeutic target in PI3K/AKT activation cancer [113].

Conclusion

TRIM21/Ro52 is a commonly seen autoantigen in many systemic autoimmune diseases, especially SLE and pSS patients. Though the mechanisms of autoimmune disease development and cancer formation remain elusive, two significant characteristics shared by both diseases are chronic inflammatory status and metabolic dysfunction. An emerging role of TRIM21/Ro52 in the regulation of inflammation and the reprogramming of cellular metabolism has been indicated in previous studies.

The underlying mechanism of how TRIM21/Ro52 may lead to cancer development or provide a protective effect against cancer in SLE and pSS patients requires further studies. Identifying the interconnected role of TRIM21/Ro52 between SLE, pSS, and tumorigenesis would be beneficial, as this could help develop an appropriate cancer monitoring method for patients with autoimmune diseases and may shed light on the disease development of SLE and pSS themselves.

Data availability

Not applicable.

Abbreviations

BAFF:

B cell-activating factor

CRC:

Colorectal cancer

DLBCL:

Diffuse large B-cell lymphoma

DLGAP1-AS2:

DLGAP1 antisense RNA 2

FASN:

Fatty acid synthase

GFP:

Green fluorescent protein

GLUT1:

Glucose transporter 1

G6PD:

Glucose-6-phosphate dehydrogenase

GNPAT:

Glyceronephosphate O-acyltransferase

TβRII:

Growth factor beta receptor 2

HCC:

Hepatocellular carcinoma

HIF-1α:

Hypoxia-inducible factor 1 subunit alpha

IGF2BP3:

IGF2 mRNA binding protein 3

IRF:

Interferon regulatory factor

IKKβ:

Kappa‐B kinase subunit beta

MICALL2:

MICAL like two protein

MALT:

Mucosa-associated lymphoid tissue

NSAIDs:

Nonsteroidal anti-inflammatory drugs

NF-κB:

Nuclear factor kappa-light-chain-enhancer of activated B cells

PI3K:

Phosphatidylinositol 3-kinase

PFK1:

Phosphofructokinase-1

PSS:

Primary Sjögren’s syndrome

PAWR:

Pro-apoptotic WT1 regulator

AKT:

Protein kinase B

PKM2:

Pyruvate kinase M2

SET7/9:

SET domain containing 7, histone lysine methyltransferase

SALL-4:

Spalt-like transcription factor 4

SREBF1:

Sterol regulatory element binding transcription factor 1

SLE:

Systemic lupus erythematosus

TNFAIP3:

TNF alpha-induced protein 3

References

  1. Rajsbaum R, Stoye JP, O’Garra A. Type I interferon-dependent and -independent expression of tripartite motif proteins in immune cells. Eur J Immunol. 2008;38(3):619–30. https://doi.org/10.1002/eji.200737916.

    Article  CAS  PubMed  Google Scholar 

  2. Hatakeyama S. TRIM family proteins: roles in autophagy, immunity, and carcinogenesis. Trends Biochem Sci. 2017;42(4):297–311. https://doi.org/10.1016/j.tibs.2017.01.002.

    Article  CAS  PubMed  Google Scholar 

  3. James LC, Keeble AH, Khan Z, Rhodes DA, Trowsdale J. Structural basis for PRYSPRY-mediated tripartite motif (TRIM) protein function. Proc Natl Acad Sci U S A. 2007;104(15):6200–5. https://doi.org/10.1073/pnas.0609174104.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  4. Dickson C, Fletcher AJ, Vaysburd M, Yang JC, Mallery DL, Zeng J, et al. Intracellular antibody signalling is regulated by phosphorylation of the Fc receptor TRIM21. Elife. 2018. https://doi.org/10.7554/eLife.32660.

    Article  PubMed Central  PubMed  Google Scholar 

  5. Manocha GD, Mishra R, Sharma N, Kumawat KL, Basu A, Singh SK. Regulatory role of TRIM21 in the type-I interferon pathway in Japanese encephalitis virus-infected human microglial cells. J Neuroinflamm. 2014;11:24. https://doi.org/10.1186/1742-2094-11-24.

    Article  CAS  Google Scholar 

  6. Higgs R, Lazzari E, Wynne C, Ni Gabhann J, Espinosa A, Wahren-Herlenius M, et al. Self protection from anti-viral responses–Ro52 promotes degradation of the transcription factor IRF7 downstream of the viral Toll-Like receptors. PLoS ONE. 2010;5(7): e11776. https://doi.org/10.1371/journal.pone.0011776.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Lazzari E, Korczeniewska J, Ni Gabhann J, Smith S, Barnes BJ, Jefferies CA. TRIpartite motif 21 (TRIM21) differentially regulates the stability of interferon regulatory factor 5 (IRF5) isoforms. PLoS ONE. 2014;9(8): e103609. https://doi.org/10.1371/journal.pone.0103609.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Yoshimi R, Ishigatsubo Y, Ozato K. Autoantigen TRIM21/Ro52 as a possible target for treatment of systemic lupus erythematosus. Int J Rheumatol. 2012;2012: 718237. https://doi.org/10.1155/2012/718237.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, et al. 2016 American College of Rheumatology/European League Against Rheumatism Classification Criteria for Primary Sjogren’s Syndrome: a consensus and data-driven methodology involving three international patient cohorts. Arthritis Rheumatol. 2017;69(1):35–45. https://doi.org/10.1002/art.39859.

    Article  PubMed  Google Scholar 

  10. Decker P, Moulinet T, Pontille F, Cravat M, De Carvalho BM, Jaussaud R. An updated review of anti-Ro52 (TRIM21) antibodies impact in connective tissue diseases clinical management. Autoimmun Rev. 2022;21(3): 103013. https://doi.org/10.1016/j.autrev.2021.103013.

    Article  CAS  PubMed  Google Scholar 

  11. Alomari M. TRIM21 - a potential novel therapeutic target in cancer. Pharmacol Res. 2021;165: 105443. https://doi.org/10.1016/j.phrs.2021.105443.

    Article  CAS  PubMed  Google Scholar 

  12. Brauner S, Zhou W, Backlin C, Green TM, Folkersen L, Ivanchenko M, et al. Reduced expression of TRIM21/Ro52 predicts poor prognosis in diffuse large B-cell lymphoma patients with and without rheumatic disease. J Intern Med. 2015;278(3):323–32. https://doi.org/10.1111/joim.12375.

    Article  CAS  PubMed  Google Scholar 

  13. Zhou W, Zhang Y, Zhong C, Hu J, Hu H, Zhou D, et al. Decreased expression of TRIM21 indicates unfavorable outcome and promotes cell growth in breast cancer. Cancer Manag Res. 2018;10:3687–96. https://doi.org/10.2147/CMAR.S175470.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Chen X, Li Z, Yong H, Wang W, Wang D, Chu S, et al. Trim21-mediated HIF-1alpha degradation attenuates aerobic glycolysis to inhibit renal cancer tumorigenesis and metastasis. Cancer Lett. 2021;508:115–26. https://doi.org/10.1016/j.canlet.2021.03.023.

    Article  CAS  PubMed  Google Scholar 

  15. Zhao Z, Wang Y, Yun D, Huang Q, Meng D, Li Q, et al. TRIM21 overexpression promotes tumor progression by regulating cell proliferation, cell migration and cell senescence in human glioma. Am J Cancer Res. 2020;10(1):114–30.

    CAS  PubMed Central  PubMed  Google Scholar 

  16. Wu Z, Wang Y, Yu Z, Meng Z, Duan W, Zhang W, et al. TRIM21-a potential biomarker for the prognosis of thyroid cancer. Exp Ther Med. 2022;24(6):761. https://doi.org/10.3892/etm.2022.11697.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  17. Nguyen JQ, Irby RB. TRIM21 is a novel regulator of Par-4 in colon and pancreatic cancer cells. Cancer Biol Ther. 2017;18(1):16–25. https://doi.org/10.1080/15384047.2016.1252880.

    Article  CAS  PubMed  Google Scholar 

  18. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. https://doi.org/10.1016/j.cell.2011.02.013.

    Article  CAS  PubMed  Google Scholar 

  19. Medina G, Vera-Lastra O, Peralta-Amaro AL, Jimenez-Arellano MP, Saavedra MA, Cruz-Dominguez MP, et al. Metabolic syndrome, autoimmunity and rheumatic diseases. Pharmacol Res. 2018;133:277–88. https://doi.org/10.1016/j.phrs.2018.01.009.

    Article  CAS  PubMed  Google Scholar 

  20. Clarke AE, Pooley N, Marjenberg Z, Langham J, Nicholson L, Langham S, et al. Risk of malignancy in patients with systemic lupus erythematosus: systematic review and meta-analysis. Semin Arthritis Rheum. 2021;51(6):1230–41. https://doi.org/10.1016/j.semarthrit.2021.09.009.

    Article  PubMed  Google Scholar 

  21. Liang Y, Yang Z, Qin B, Zhong R. Primary Sjogren’s syndrome and malignancy risk: a systematic review and meta-analysis. Ann Rheum Dis. 2014;73(6):1151–6. https://doi.org/10.1136/annrheumdis-2013-203305.

    Article  PubMed  Google Scholar 

  22. Ohlsson M, Jonsson R, Brokstad KA. Subcellular redistribution and surface exposure of the Ro52, Ro60 and La48 autoantigens during apoptosis in human ductal epithelial cells: a possible mechanism in the pathogenesis of Sjogren’s syndrome. Scand J Immunol. 2002;56(5):456–69. https://doi.org/10.1046/j.1365-3083.2002.01072_79.x.

    Article  CAS  PubMed  Google Scholar 

  23. Espinosa A, Zhou W, Ek M, Hedlund M, Brauner S, Popovic K, et al. The Sjogren’s syndrome-associated autoantigen Ro52 is an E3 ligase that regulates proliferation and cell death. J Immunol. 2006;176(10):6277–85. https://doi.org/10.4049/jimmunol.176.10.6277.

    Article  CAS  PubMed  Google Scholar 

  24. Sjostrand M, Ambrosi A, Brauner S, Sullivan J, Malin S, Kuchroo VK, et al. Expression of the immune regulator tripartite-motif 21 is controlled by IFN regulatory factors. J Immunol. 2013;191(7):3753–63. https://doi.org/10.4049/jimmunol.1202341.

    Article  CAS  PubMed  Google Scholar 

  25. Espinosa A, Dardalhon V, Brauner S, Ambrosi A, Higgs R, Quintana FJ, et al. Loss of the lupus autoantigen Ro52/Trim21 induces tissue inflammation and systemic autoimmunity by disregulating the IL-23-Th17 pathway. J Exp Med. 2009;206(8):1661–71. https://doi.org/10.1084/jem.20090585.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Bolland S, Garcia-Sastre A. Vicious circle: systemic autoreactivity in Ro52/TRIM21-deficient mice. J Exp Med. 2009;206(8):1647–51. https://doi.org/10.1084/jem.20091507.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Brauner S, Ivanchenko M, Thorlacius GE, Ambrosi A, Wahren-Herlenius M. The Sjogren’s syndrome-associated autoantigen Ro52/TRIM21 modulates follicular B cell homeostasis and immunoglobulin production. Clin Exp Immunol. 2018;194(3):315–26. https://doi.org/10.1111/cei.13211.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Lu R. Interferon regulatory factor 4 and 8 in B-cell development. Trends Immunol. 2008;29(10):487–92. https://doi.org/10.1016/j.it.2008.07.006.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Wada K, Niida M, Tanaka M, Kamitani T. Ro52-mediated monoubiquitination of IKK{beta} down-regulates NF-{kappa}B signalling. J Biochem. 2009;146(6):821–32. https://doi.org/10.1093/jb/mvp127.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. Jost PJ, Ruland J. Aberrant NF-kappaB signaling in lymphoma: mechanisms, consequences, and therapeutic implications. Blood. 2007;109(7):2700–7. https://doi.org/10.1182/blood-2006-07-025809.

    Article  CAS  PubMed  Google Scholar 

  31. Tessier-Cloutier B, Twa DD, Baecklund E, Gascoyne R, Johnson NA, Backlin C, et al. Cell of origin in diffuse large B-cell lymphoma in systemic lupus erythematosus: molecular and clinical factors associated with survival. Lupus Sci Med. 2019;6(1): e000324. https://doi.org/10.1136/lupus-2019-000324.

    Article  PubMed Central  PubMed  Google Scholar 

  32. Hillen MR, Urso K, Koppe E, Lopes AP, Blokland SLM, Pandit A, et al. Autoantigen TRIM21/Ro52 is expressed on the surface of antigen-presenting cells and its enhanced expression in Sjogren’s syndrome is associated with B cell hyperactivity and type I interferon activity. RMD Open. 2020;6(2): e001184. https://doi.org/10.1136/rmdopen-2020-001184.

    Article  PubMed Central  PubMed  Google Scholar 

  33. Kunishita Y, Yoshimi R, Kamiyama R, Kishimoto D, Yoshida K, Hashimoto E, et al. TRIM21 dysfunction enhances Aberrant B-cell differentiation in autoimmune pathogenesis. Front Immunol. 2020;11:98. https://doi.org/10.3389/fimmu.2020.00098.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  34. Kamiyama R, Yoshimi R, Takeno M, Iribe Y, Tsukahara T, Kishimoto D, et al. Dysfunction of TRIM21 in interferon signature of systemic lupus erythematosus. Mod Rheumatol. 2018;28(6):993–1003. https://doi.org/10.1080/14397595.2018.1436028.

    Article  CAS  PubMed  Google Scholar 

  35. Amezcua-Guerra LM, Perez-Garcia LF, Jimenez-Rojas V, Marquez-Velasco R, Silveira LH. Anti-Ro52/TRIM21 antibodies are associated with aberrant inflammatory circuits in patients with systemic autoimmune rheumatic diseases. Gac Med Mex. 2023;159(1):55–64. https://doi.org/10.24875/GMM.M22000739.

    Article  PubMed  Google Scholar 

  36. Noble PW, Bernatsky S, Clarke AE, Isenberg DA, Ramsey-Goldman R, Hansen JE. DNA-damaging autoantibodies and cancer: the lupus butterfly theory. Nat Rev Rheumatol. 2016;12(7):429–34. https://doi.org/10.1038/nrrheum.2016.23.

    Article  CAS  PubMed  Google Scholar 

  37. Lee AYS. A review of the role and clinical utility of anti-Ro52/TRIM21 in systemic autoimmunity. Rheumatol Int. 2017;37(8):1323–33. https://doi.org/10.1007/s00296-017-3718-1.

    Article  CAS  PubMed  Google Scholar 

  38. Retamozo S, Akasbi M, Brito-Zeron P, Bosch X, Bove A, Perez-de-Lis M, et al. Anti-Ro52 antibody testing influences the classification and clinical characterisation of primary Sjogren’s syndrome. Clin Exp Rheumatol. 2012;30(5):686–92.

    PubMed  Google Scholar 

  39. Lee AYS, Lin MW, Reed JH. Anti-Ro52/TRIM21 serological subsets identify differential clinical and laboratory parameters. Clin Rheumatol. 2022. https://doi.org/10.1007/s10067-022-06299-5.

    Article  PubMed Central  PubMed  Google Scholar 

  40. Ding Q, He D, He K, Zhang Q, Tang M, Dai J, et al. Downregulation of TRIM21 contributes to hepatocellular carcinoma carcinogenesis and indicates poor prognosis of cancers. Tumour Biol. 2015;36(11):8761–72. https://doi.org/10.1007/s13277-015-3572-2.

    Article  CAS  PubMed  Google Scholar 

  41. Ping M, Wang S, Guo Y, Jia J. TRIM21 improves apatinib treatment in gastric cancer through suppressing EZH1 stability. Biochem Biophys Res Commun. 2022;586:177–84. https://doi.org/10.1016/j.bbrc.2021.07.040.

    Article  CAS  PubMed  Google Scholar 

  42. Sun J, Chen X, Ji X, Meng S, Wang W, Wang P, et al. TRIM21 deficiency promotes cell proliferation and tumorigenesis via regulating p21 expression in ovarian cancer. Bioengineered. 2022;13(3):6024–35. https://doi.org/10.1080/21655979.2022.2042134.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  43. Zhou G, Wu H, Lin J, Lin R, Feng B, Liu Z. TRIM21 Is decreased in colitis-associated cancer and negatively regulates epithelial carcinogenesis. Inflamm Bowel Dis. 2021;27(4):458–68. https://doi.org/10.1093/ibd/izaa229.

    Article  PubMed  Google Scholar 

  44. Jin Y, Zhang Y, Li B, Zhang J, Dong Z, Hu X, et al. TRIM21 mediates ubiquitination of Snail and modulates epithelial to mesenchymal transition in breast cancer cells. Int J Biol Macromol. 2019;124:846–53. https://doi.org/10.1016/j.ijbiomac.2018.11.269.

    Article  CAS  PubMed  Google Scholar 

  45. Si W, Zhou J, Zhao Y, Zheng J, Cui L. SET7/9 promotes multiple malignant processes in breast cancer development via RUNX2 activation and is negatively regulated by TRIM21. Cell Death Dis. 2020;11(2):151. https://doi.org/10.1038/s41419-020-2350-2.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Itou J, Li W, Ito S, Tanaka S, Matsumoto Y, Sato F, et al. Sal-like 4 protein levels in breast cancer cells are post-translationally down-regulated by tripartite motif-containing 21. J Biol Chem. 2018;293(17):6556–64. https://doi.org/10.1074/jbc.RA117.000245.

    Article  CAS  PubMed  Google Scholar 

  47. Liu Q, Sun H, Liu Y, Li X, Xu B, Li L, et al. HTR1A Inhibits the progression of triple-negative breast cancer via TGF-beta canonical and noncanonical pathways. Adv Sci (Weinh). 2022;9(12): e2105672. https://doi.org/10.1002/advs.202105672.

    Article  CAS  PubMed  Google Scholar 

  48. Liu J, Zhang C, Xu D, Zhang T, Chang CY, Wang J, et al. The ubiquitin ligase TRIM21 regulates mutant p53 accumulation and gain of function in cancer. J Clin Invest. 2023;133(6): e164354. https://doi.org/10.1172/JCI164354.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  49. Zhang Y, Han C, Li J, Zhang L, Wang L, Ye S, et al. Efficacy and safety for Apatinib treatment in advanced gastric cancer: a real world study. Sci Rep. 2017;7(1):13208. https://doi.org/10.1038/s41598-017-13192-8.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  50. Chen X, Yong H, Chen M, Deng C, Wang P, Chu S, et al. TRIM21 attenuates renal carcinoma lipogenesis and malignancy by regulating SREBF1 protein stability. J Exp Clin Cancer Res. 2023;42(1):34. https://doi.org/10.1186/s13046-022-02583-z.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  51. Wang X, Cheng H, Zhao J, Li J, Chen Y, Cui K, et al. Long noncoding RNA DLGAP1-AS2 promotes tumorigenesis and metastasis by regulating the Trim21/ELOA/LHPP axis in colorectal cancer. Mol Cancer. 2022;21(1):210. https://doi.org/10.1186/s12943-022-01675-w.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  52. Wen P, Wang H, Li Y, Sui X, Hou Z, Guo X, et al. MICALL2 as a substrate of ubiquitinase TRIM21 regulates tumorigenesis of colorectal cancer. Cell Commun Signal. 2022;20(1):170. https://doi.org/10.1186/s12964-022-00984-3.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  53. Gui Z, Li J, Li J, Li X, Chen L, Ma Z, et al. Berberine promotes IGF2BP3 ubiquitination by TRIM21 to induce G1/S phase arrest in colorectal cancer cells. Chem Biol Interact. 2023;374: 110408. https://doi.org/10.1016/j.cbi.2023.110408.

    Article  CAS  PubMed  Google Scholar 

  54. Qi Z, Yan F, Chen D, Xing W, Li Q, Zeng W, et al. Identification of prognostic biomarkers and correlations with immune infiltrates among cGAS-STING in hepatocellular carcinoma. 2020. Biosci Rep. https://doi.org/10.1042/BSR20202603.

  55. Feng X, Jiang T, Yang C, Pang S, Ding Z, Hu H, et al. RPRD1A stabilizes NRF2 and aggravates HCC progression through competing with p62 for TRIM21 binding. Cell Death Dis. 2021;13(1):6. https://doi.org/10.1038/s41419-021-04447-4.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  56. Wang F, Zhang Y, Shen J, Yang B, Dai W, Yan J, et al. The ubiquitin E3 ligase TRIM21 promotes hepatocarcinogenesis by suppressing the p62-Keap1-Nrf2 antioxidant pathway. Cell Mol Gastroenterol Hepatol. 2021;11(5):1369–85. https://doi.org/10.1016/j.jcmgh.2021.01.007.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  57. Foss S, Watkinson R, Sandlie I, James LC, Andersen JT. TRIM21: a cytosolic Fc receptor with broad antibody isotype specificity. Immunol Rev. 2015;268(1):328–39. https://doi.org/10.1111/imr.12363.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  58. James LC. Intracellular antibody immunity and the cytosolic Fc receptor TRIM21. Curr Top Microbiol Immunol. 2014;382:51–66. https://doi.org/10.1007/978-3-319-07911-0_3.

    Article  CAS  PubMed  Google Scholar 

  59. Hurley LC, Levin NK, Chatterjee M, Coles J, Muszkat S, Howarth Z, et al. Evaluation of paraneoplastic antigens reveals TRIM21 autoantibodies as biomarker for early detection of ovarian cancer in combination with autoantibodies to NY-ESO-1 and TP53. Cancer Biomark. 2020;27(3):407–21. https://doi.org/10.3233/CBM-190988.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  60. Kuboshima M, Shimada H, Liu TL, Nomura F, Takiguchi M, Hiwasa T, et al. Presence of serum tripartite motif-containing 21 antibodies in patients with esophageal squamous cell carcinoma. Cancer Sci. 2006;97(5):380–6. https://doi.org/10.1111/j.1349-7006.2006.00192.x.

    Article  CAS  PubMed  Google Scholar 

  61. Bogdanos DP, Gkoutzourelas A, Papadopoulos V, Liaskos C, Patrikiou E, Tsigalou C, et al. Anti-Ro52 antibody is highly prevalent and a marker of better prognosis in patients with ovarian cancer. Clin Chim Acta. 2021;521:199–205. https://doi.org/10.1016/j.cca.2021.07.006.

    Article  CAS  PubMed  Google Scholar 

  62. Zhong H, Liu S, Wang Y, Xu D, Li M, Zhao Y, et al. Primary Sjogren’s syndrome is associated with increased risk of malignancies besides lymphoma: a systematic review and meta-analysis. Autoimmun Rev. 2022;21(5): 103084. https://doi.org/10.1016/j.autrev.2022.103084.

    Article  PubMed  Google Scholar 

  63. Tessier-Cloutier B, Clarke AE, Ramsey-Goldman R, Gordon C, Hansen JE, Bernatsky S. Systemic lupus erythematosus and malignancies: a review article. Rheum Dis Clin North Am. 2014;40(3):497–506. https://doi.org/10.1016/j.rdc.2014.04.005.

    Article  PubMed  Google Scholar 

  64. Igoe A, Merjanah S, Scofield RH. Sjogren syndrome and cancer. Rheum Dis Clin North Am. 2020;46(3):513–32. https://doi.org/10.1016/j.rdc.2020.05.004.

    Article  PubMed  Google Scholar 

  65. Ladouceur A, Tessier-Cloutier B, Clarke AE, Ramsey-Goldman R, Gordon C, Hansen JE, et al. Cancer and systemic lupus erythematosus. Rheum Dis Clin North Am. 2020;46(3):533–50. https://doi.org/10.1016/j.rdc.2020.05.005.

    Article  PubMed  Google Scholar 

  66. Bernatsky S, Kale M, Ramsey-Goldman R, Gordon C, Clarke AE. Systemic lupus and malignancies. Curr Opin Rheumatol. 2012;24(2):177–81. https://doi.org/10.1097/BOR.0b013e32834ff258.

    Article  CAS  PubMed  Google Scholar 

  67. Goulabchand R, Malafaye N, Jacot W, WitkowskiDurandViel P, Morel J, Lukas C, et al. Cancer incidence in primary Sjogren’s syndrome: data from the French hospitalization database. Autoimmun Rev. 2021;20(12): 102987. https://doi.org/10.1016/j.autrev.2021.102987.

    Article  CAS  PubMed  Google Scholar 

  68. Cobo-Ibanez T, Urruticoechea-Arana A, Rua-Figueroa I, Martin-Martinez MA, Ovalles-Bonilla JG, Galindo M, et al. Hormonal dependence and cancer in systemic lupus erythematosus. Arthritis Care Res (Hoboken). 2020;72(2):216–24. https://doi.org/10.1002/acr.24068.

    Article  CAS  PubMed  Google Scholar 

  69. Gompel A. Systemic lupus erythematosus and menopause. Climacteric. 2020;23(2):109–15. https://doi.org/10.1080/13697137.2019.1679113.

    Article  CAS  PubMed  Google Scholar 

  70. Sherman ME, Vierkant RA, Kaggal S, Hoskin TL, Frost MH, Denison L, et al. Breast cancer risk and use of nonsteroidal anti-inflammatory agents after a benign breast biopsy. Cancer Prev Res (Phila). 2020;13(11):967–76. https://doi.org/10.1158/1940-6207.CAPR-20-0178.

    Article  CAS  PubMed  Google Scholar 

  71. Bernatsky S, Ramsey-Goldman R, Foulkes WD, Gordon C, Clarke AE. Breast, ovarian, and endometrial malignancies in systemic lupus erythematosus: a meta-analysis. Br J Cancer. 2011;104(9):1478–81. https://doi.org/10.1038/bjc.2011.115.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  72. Chen YJ, Chang YT, Wang CB, Wu CY. Malignancy in systemic lupus erythematosus: a nationwide cohort study in Taiwan. Am J Med. 2010;123(12):1150. https://doi.org/10.1016/j.amjmed.2010.08.006.

    Article  Google Scholar 

  73. Deng J, Liu M, Xiao R, Wang J, Liao X, Ye Z, et al. Risk, incidence, and mortality of breast cancer in primary sjögren’s syndrome: a systematic review and meta-analysis. Front Immunol. 2022. https://doi.org/10.3389/fimmu.2022.904682.

    Article  PubMed Central  PubMed  Google Scholar 

  74. Choi MY, Flood K, Bernatsky S, Ramsey-Goldman R, Clarke AE. A review on SLE and malignancy. Best Pract Res Clin Rheumatol. 2017;31(3):373–96. https://doi.org/10.1016/j.berh.2017.09.013.

    Article  PubMed Central  PubMed  Google Scholar 

  75. Quartuccio L, Baldini C, Bartoloni E, Priori R, Carubbi F, Corazza L, et al. Anti-SSA/SSB-negative Sjogren’s syndrome shows a lower prevalence of lymphoproliferative manifestations, and a lower risk of lymphoma evolution. Autoimmun Rev. 2015;14(11):1019–22. https://doi.org/10.1016/j.autrev.2015.07.002.

    Article  CAS  PubMed  Google Scholar 

  76. Hardenbergh D, Naik R, Manno R, Azar A, Monroy Trujillo JM, Adler B, et al. The cancer risk profile of systemic lupus erythematosus patients. J Clin Rheumatol. 2022;28(1):e257–62. https://doi.org/10.1097/RHU.0000000000001729.

    Article  PubMed  Google Scholar 

  77. Hsu CY, Lin MS, Su YJ, Cheng TT, Lin YS, Chen YC, et al. Cumulative immunosuppressant exposure is associated with diversified cancer risk among 14 832 patients with systemic lupus erythematosus: a nested case-control study. Rheumatology (Oxford). 2017;56(4):620–8. https://doi.org/10.1093/rheumatology/kew457.

    Article  PubMed  Google Scholar 

  78. Bernatsky S, Ramsey-Goldman R, Clarke AE. Malignancy in systemic lupus erythematosus: what have we learned? Best Pract Res Clin Rheumatol. 2009;23(4):539–47. https://doi.org/10.1016/j.berh.2008.12.007.

    Article  PubMed Central  PubMed  Google Scholar 

  79. Guo J, Ren Z, Li J, Li T, Liu S, Yu Z. The relationship between cancer and medication exposure in patients with systemic lupus erythematosus: a nested case-control study. Arthritis Res Ther. 2020;22(1):159. https://doi.org/10.1186/s13075-020-02228-6.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  80. Fang YF, Chen YF, Chung TT, See LC, Yu KH, Luo SF, et al. Hydroxychloroquine and risk of cancer in patients with primary Sjogren syndrome: propensity score matched landmark analysis. Oncotarget. 2017;8(46):80461–71. https://doi.org/10.18632/oncotarget.19057.

    Article  PubMed Central  PubMed  Google Scholar 

  81. Gonzalez LA, Alarcon GS. The evolving concept of SLE comorbidities. Expert Rev Clin Immunol. 2017;13(8):753–68. https://doi.org/10.1080/1744666X.2017.1327353.

    Article  CAS  PubMed  Google Scholar 

  82. Mockel T, Basta F, Weinmann-Menke J, Schwarting A. B cell activating factor (BAFF): Structure, functions, autoimmunity and clinical implications in Systemic Lupus Erythematosus (SLE). Autoimmun Rev. 2021;20(2): 102736. https://doi.org/10.1016/j.autrev.2020.102736.

    Article  CAS  PubMed  Google Scholar 

  83. Yilmaz CN, Gemcioglu E, Baser S, Erten S, Erel O. Thiol/disulfide homeostasis impaired in patients with primary Sjogren’s syndrome. J Med Biochem. 2021;40(3):270–6. https://doi.org/10.5937/jomb0-27281.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  84. Weckerle CE, Mangale D, Franek BS, Kelly JA, Kumabe M, James JA, et al. Large-scale analysis of tumor necrosis factor alpha levels in systemic lupus erythematosus. Arthritis Rheum. 2012;64(9):2947–52. https://doi.org/10.1002/art.34483.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  85. Daridon C, Devauchelle V, Hutin P, Le Berre R, Martins-Carvalho C, Bendaoud B, et al. Aberrant expression of BAFF by B lymphocytes infiltrating the salivary glands of patients with primary Sjogren’s syndrome. Arthritis Rheum. 2007;56(4):1134–44. https://doi.org/10.1002/art.22458.

    Article  CAS  PubMed  Google Scholar 

  86. Lavie F, Miceli-Richard C, Quillard J, Roux S, Leclerc P, Mariette X. Expression of BAFF (BLyS) in T cells infiltrating labial salivary glands from patients with Sjogren’s syndrome. J Pathol. 2004;202(4):496–502. https://doi.org/10.1002/path.1533.

    Article  CAS  PubMed  Google Scholar 

  87. Veenbergen S, Kozmar A, van Daele PLA, Schreurs MWJ. Autoantibodies in Sjogren’s syndrome and its classification criteria. J Transl Autoimmun. 2022;5: 100138. https://doi.org/10.1016/j.jtauto.2021.100138.

    Article  CAS  PubMed  Google Scholar 

  88. Bende RJ, Aarts WM, Riedl RG, de Jong D, Pals ST, van Noesel CJ. Among B cell non-Hodgkin’s lymphomas, MALT lymphomas express a unique antibody repertoire with frequent rheumatoid factor reactivity. J Exp Med. 2005;201(8):1229–41. https://doi.org/10.1084/jem.20050068.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  89. Solans-Laque R, Lopez-Hernandez A, Bosch-Gil JA, Palacios A, Campillo M, Vilardell-Tarres M. Risk, predictors, and clinical characteristics of lymphoma development in primary Sjogren’s syndrome. Semin Arthritis Rheum. 2011;41(3):415–23. https://doi.org/10.1016/j.semarthrit.2011.04.006.

    Article  PubMed  Google Scholar 

  90. Deutsch AJ, Aigelsreiter A, Staber PB, Beham A, Linkesch W, Guelly C, et al. MALT lymphoma and extranodal diffuse large B-cell lymphoma are targeted by aberrant somatic hypermutation. Blood. 2007;109(8):3500–4. https://doi.org/10.1182/blood-2006-06-030494.

    Article  CAS  PubMed  Google Scholar 

  91. Bernatsky S, Easton DF, Dunning A, Michailidou K, Ramsey-Goldman R, Gordon C, et al. Decreased breast cancer risk in systemic lupus erythematosus: the search for a genetic basis continues. Lupus. 2012;21(8):896–9. https://doi.org/10.1177/0961203312443992.

    Article  CAS  PubMed  Google Scholar 

  92. Bernatsky S, Velasquez Garcia HA, Spinelli JJ, Gaffney P, Smedby KE, Ramsey-Goldman R, et al. Lupus-related single nucleotide polymorphisms and risk of diffuse large B-cell lymphoma. Lupus Sci Med. 2017;4(1): e000187. https://doi.org/10.1136/lupus-2016-000187.

    Article  PubMed Central  PubMed  Google Scholar 

  93. Zhu L, Zhou L, Wang L, Li Z, Lu S, Yang L, et al. A20 SNP rs77191406 may be related to secondary cancer for rheumatoid arthritis and systemic lupus erythematosus patients. Asia Pac J Clin Oncol. 2016;12(4):409–14. https://doi.org/10.1111/ajco.12577.

    Article  PubMed  Google Scholar 

  94. Hansen JE, Chan G, Liu Y, Hegan DC, Dalal S, Dray E, et al. Targeting cancer with a lupus autoantibody. Sci Transl Med. 2012;4(157):157ra42. https://doi.org/10.1126/scitranslmed.3004385.

    Article  Google Scholar 

  95. Pantham P, Viall CA, Chen Q, Kleffmann T, Print CG, Chamley LW. Antiphospholipid antibodies bind syncytiotrophoblast mitochondria and alter the proteome of extruded syncytial nuclear aggregates. Placenta. 2015;36(12):1463–73. https://doi.org/10.1016/j.placenta.2015.10.006.

    Article  CAS  PubMed  Google Scholar 

  96. Dias C, Isenberg DA. Susceptibility of patients with rheumatic diseases to B-cell non-Hodgkin lymphoma. Nat Rev Rheumatol. 2011;7(6):360–8. https://doi.org/10.1038/nrrheum.2011.62.

    Article  CAS  PubMed  Google Scholar 

  97. Teng X, Brown J, Choi SC, Li W, Morel L. Metabolic determinants of lupus pathogenesis. Immunol Rev. 2020;295(1):167–86. https://doi.org/10.1111/imr.12847.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  98. Esposito K, Chiodini P, Colao A, Lenzi A, Giugliano D. Metabolic syndrome and risk of cancer: a systematic review and meta-analysis. Diabetes Care. 2012;35(11):2402–11. https://doi.org/10.2337/dc12-0336.

    Article  PubMed Central  PubMed  Google Scholar 

  99. Luo X, Wu X, Wang A, Chen Y, Peng Y, Deng C, et al. mTORC1-GLUT1-mediated glucose metabolism drives hyperactivation of B cells in primary Sjogren’s syndrome. Immunology. 2022. https://doi.org/10.1111/imm.13580.

    Article  PubMed Central  PubMed  Google Scholar 

  100. Tzeng HT, Chyuan IT. Immunometabolism in systemic lupus erythematosus: relevant pathogenetic mechanisms and potential clinical applications. J Formos Med Assoc. 2021;120(9):1667–75. https://doi.org/10.1016/j.jfma.2021.03.019.

    Article  CAS  PubMed  Google Scholar 

  101. Zhou D, Duan Z, Li Z, Ge F, Wei R, Kong L. The significance of glycolysis in tumor progression and its relationship with the tumor microenvironment. Front Pharmacol. 2022;13:1091779. https://doi.org/10.3389/fphar.2022.1091779.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  102. Sharabi A, Tsokos GC. T cell metabolism: new insights in systemic lupus erythematosus pathogenesis and therapy. Nat Rev Rheumatol. 2020;16(2):100–12. https://doi.org/10.1038/s41584-019-0356-x.

    Article  CAS  PubMed  Google Scholar 

  103. Zhang X, Yang Y, Jing L, Zhai W, Zhang H, Ma Q, et al. Pyruvate Kinase M2 contributes to TLR-mediated inflammation and autoimmunity by promoting Pyk2 activation. Front Immunol. 2021;12: 680068. https://doi.org/10.3389/fimmu.2021.680068.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  104. Zahra K, Dey T, Ashish, Mishra SP, Pandey U. Pyruvate kinase M2 and cancer: the role of PKM2 in promoting tumorigenesis. Front Oncol. 2020;10:159. https://doi.org/10.3389/fonc.2020.00159.

    Article  PubMed Central  PubMed  Google Scholar 

  105. Nocturne G, Pontarini E, Bombardieri M, Mariette X. Lymphomas complicating primary Sjogren’s syndrome: from autoimmunity to lymphoma. Rheumatology (Oxford). 2019. https://doi.org/10.1093/rheumatology/kez052.

    Article  Google Scholar 

  106. Han JY, Kim H, Jung SY, Jang EJ, Cho SK, Sung YK. Increased risk of malignancy in patients with systemic lupus erythematosus: population-based cohort study in Korea. Arthritis Res Ther. 2021;23(1):270. https://doi.org/10.1186/s13075-021-02648-y.

    Article  PubMed Central  PubMed  Google Scholar 

  107. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029–33. https://doi.org/10.1126/science.1160809.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  108. Lee JH, Liu R, Li J, Zhang C, Wang Y, Cai Q, et al. Stabilization of phosphofructokinase 1 platelet isoform by AKT promotes tumorigenesis. Nat Commun. 2017;8(1):949. https://doi.org/10.1038/s41467-017-00906-9.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  109. Park JS, Burckhardt CJ, Lazcano R, Solis LM, Isogai T, Li L, et al. Mechanical regulation of glycolysis via cytoskeleton architecture. Nature. 2020;578(7796):621–6. https://doi.org/10.1038/s41586-020-1998-1.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  110. Lin HP, Cheng ZL, He RY, Song L, Tian MX, Zhou LS, et al. Destabilization of fatty acid synthase by acetylation inhibits de novo lipogenesis and tumor cell growth. Cancer Res. 2016;76(23):6924–36. https://doi.org/10.1158/0008-5472.CAN-16-1597.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  111. Gu L, Zhu Y, Lin X, Tan X, Lu B, Li Y. Stabilization of FASN by ACAT1-mediated GNPAT acetylation promotes lipid metabolism and hepatocarcinogenesis. Oncogene. 2020;39(11):2437–49. https://doi.org/10.1038/s41388-020-1156-0.

    Article  CAS  PubMed  Google Scholar 

  112. Hayes JD, Dinkova-Kostova AT, Tew KD. Oxidative stress in cancer. Cancer Cell. 2020;38(2):167–97. https://doi.org/10.1016/j.ccell.2020.06.001.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  113. Cheng J, Huang Y, Zhang X, Yu Y, Wu S, Jiao J, et al. TRIM21 and PHLDA3 negatively regulate the crosstalk between the PI3K/AKT pathway and PPP metabolism. Nat Commun. 2020;11(1):1880. https://doi.org/10.1038/s41467-020-15819-3.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful for the language help provided by Biomeditor (http://biomeditor.com).

Funding

This research was funded by grants from the National Science and Technology Council, Taiwan (NSTC 110-2314-B-039-048-; NSTC 110-2314-B-039-034-MY3; NSTC 111-2314-B-039-070-; NSTC 112-2320-B-039-020-), the National Health Research Institute, Taiwan (NHRI-112A1-CACO-13222202), the China Medical University, Taiwan (CMU108-MF-01; CMU109-MF-03), the China Medical University Hospital, Taiwan (DMR-111-053; DMR-111-209; DMR-112-198; DMR-112-195).

Author information

Authors and Affiliations

Authors

Contributions

CHH and YLY conceived the manuscript. CHH wrote and contributed to the figures. YLY revised and edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Yung-Luen Yu.

Ethics declarations

Competing interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hsu, CH., Yu, YL. The interconnected roles of TRIM21/Ro52 in systemic lupus erythematosus, primary Sjögren’s syndrome, cancers, and cancer metabolism. Cancer Cell Int 23, 289 (2023). https://doi.org/10.1186/s12935-023-03143-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-023-03143-x

Keywords