Next Article in Journal
Understanding Amyotrophic Lateral Sclerosis: Pathophysiology, Diagnosis, and Therapeutic Advances
Previous Article in Journal
Mechanistic Insight into the Enantioselective Degradation of Esterase QeH to (R)/(S)–Quizalofop–Ethyl with Molecular Dynamics Simulation Using a Residue-Specific Force Field
Previous Article in Special Issue
Mitochondrial Dysfunction in Systemic Lupus Erythematosus with a Focus on Lupus Nephritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Immune Mechanisms and Biomarkers in Systemic Lupus Erythematosus

by
Ioannis Parodis
1,2,* and
Christopher Sjöwall
3
1
Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, SE-171 77 Stockholm, Sweden
2
Department of Rheumatology, Faculty of Medicine and Health, Örebro University, SE-701 82 Örebro, Sweden
3
Division of Inflammation and Infection/Rheumatology, Department of Biomedical and Clinical Sciences, Linköping University, SE-581 85 Linköping, Sweden
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(18), 9965; https://doi.org/10.3390/ijms25189965
Submission received: 25 August 2024 / Accepted: 13 September 2024 / Published: 15 September 2024
(This article belongs to the Special Issue Immune Mechanisms and Biomarkers in Systemic Lupus Erythematosus)
The immense heterogeneity of the chronic, inflammatory, autoimmune disease systemic lupus erythematosus (SLE), both with regard to immunological aberrancies and clinical manifestations, poses diagnostic difficulties and challenges in the management of patients [1,2,3]. This is underlined by the lack of generally accepted diagnostic criteria and the numerous clinical trial failures. However, the treatment landscape has witnessed substantial changes during the last decade, facilitated by advances in biotechnology and hence new knowledge on the pathophysiology of SLE.
SLE predominantly affects women, with the onset of the disease typically occurring during their reproductive years. Early diagnosis and treatment initiation are important for the prevention of organ damage accrual [4]. The chronic nature of the disease and its varying course necessitate regular monitoring. The treatment of SLE mainly consists of antimalarial agents, glucocorticoids, non-biological immunosuppressants, and, more recently, biological agents, including B-cell-targeting therapies and a monoclonal antibody against the type I interferon receptor [5]. The recent approvals of new targeted therapies for SLE and lupus nephritis (LN), one of the most severe clinical manifestations of SLE [6], and the increasing awareness of the long-term adverse effects of glucocorticoids changed the focus of research towards the optimisation of therapeutic decision making, surveillance, and treatment evaluations, and technological advances paved the way for a cellular and molecular characterisation of SLE to serve as a basis for disease management [7,8]. In this context, identifying reliable biomarkers is imperative [9], and significant progress has been made in this area over the past few decades [10,11,12].
Historically, biomarker studies in SLE have focused on serum biomarkers [9,10,13,14]. Nevertheless, urinary, cerebrospinal fluid, and tissue biomarkers for organ-specific monitoring and prognostication are gaining increasing interest [12,15]. In this Special Issue titled “Immune Mechanisms and Biomarkers in Systemic Lupus Erythematosus”, we welcomed original works and review articles focusing on the cellular and molecular mechanisms underlying SLE in its different phases, with the goal of contributing novel knowledge of the pathogenesis of SLE and improved diagnostics, surveillance, prevention of long-term damage, and overall patient management. We also welcomed original works or review articles that evaluated immune components that could serve as diagnostic biomarkers, biomarkers of disease activity, or biomarkers of long-term outcomes. We received several contributions, and we hereby summarise the final content of the Special Issue, which is also visually represented in Figure 1.
In a cross-sectional study by Irene Carrión-Barberà et al., levels of advanced glycation end-products (AGEs) were found to be significantly higher in SLE patients compared to healthy controls. AGEs showed positive associations with the degree of SLE activity using the SLE disease activity index (SLEDAI) and the degree of organ damage using the Systemic Lupus International Collaborating Clinics (SLICC)/American College of Rheumatology (ACR) damage index (SDI), as well as various clinical features, indicating some potential of AGEs as biomarkers of monitoring and prognosis in SLE [16]. In a study by Bethany Wolf et al., significant differences in urine glycosphingolipids and N-glycans were observed between LN patients and healthy controls, with men showing more pronounced differences [17].
In a cohort study by Agnieszka Winikajtis-Burzyńska et al., elevated serum levels of soluble transferrin receptor (sTfR) were linked to an increased risk of cardiovascular, pulmonary, and haematological manifestations of SLE, while elevated interleukin (IL)-4 levels were associated with a decreased risk of mucocutaneous manifestations, collectively suggesting that sTfR and IL-4 could be useful for differentiating the risk of affliction across organ systems in SLE [18]. In an observational study of 284 SLE patients, Julia Mercader-Salvans et al. found that circulating levels of IL-6 were associated with a higher cardiovascular risk and disruption of the complement system, but not with SLE disease activity or organ damage [19]. Contrary to some prior observations [20,21], a study by Lina Wirestam et al. found no strong association between anti-oxidised low-density lipoprotein (anti-oxLDL) antibodies and vascular affliction in SLE patients [22]. Although a significant correlation was observed with intima media thickness in the common femoral artery, the overall findings do not support anti-oxLDL antibodies as reliable biomarkers for vascular involvement in SLE.
Using data from multiple phase III clinical trials of belimumab, Ioannis Parodis et al. investigated the role of early alterations in circulating B cell and plasma cell subsets in relation to renal flares in SLE patients treated for active extra-renal disease with non-biological standard therapy plus belimumab or placebo. A rapid decrease in short-lived plasma cells or plasmablasts with a subsequent return was associated with renal flares. Rapid decreases in transitional B cells and long-lived plasma cells upon belimumab therapy indicated greater protection against renal flares, which was not the case for the placebo-treated group of patients [23]. Collectively, B cell and plasma cell kinetics during the early phases of treatment with belimumab for active SLE might be useful early indicators of the need for therapeutic adjustments [24].
In a review by Matthieu Halfon et al., mitochondrial dysfunction was detailed as a significant factor in SLE pathogenesis, particularly in LN. Altered mitochondrial homeostasis and defective mitophagy appears to contribute to immune dysregulation, suggesting mitochondria as potential biomarkers and therapeutic targets in SLE and LN [25]. Based on the current literature, Alessandra Maria Vitale et al. proposed that molecular mimicry between human heat shock proteins (HSPs) belonging to the chaperone system and proteins and metabolites of gut commensal bacteria could link gut microbiota dysbiosis with SLE pathogenesis. The production of autoantibodies against HSPs, which are known to associate with SLE onset and progression, due to shared epitopes between human HSPs and those of gut commensal bacteria, may contribute to disease pathogenesis, warranting a coordinated study of these factors in SLE [26].
In a comprehensive review by Susannah von Hofsten et al., the roles of Toll-like receptors (TLRs) in SLE pathogenesis were discussed, particularly TLR7, TLR8, and TLR9. In brief, the authors detailed the link between the overexpression of TLR7 and SLE severity and further stated that TLR8 and TLR9 may regulate TLR7 activity, offering insights into potential therapeutic strategies targeting these receptors [27]. Last but not least, Fatima K. Alduraibi and George C. Tsokos provided a critical evaluation of biomarkers for LN. Despite improvements in kidney and patient survival, complete clinical and histological remission rates remain limited in LN, highlighting the need for the timely detection of kidney affliction due to SLE and the prompt initiation of therapy, as well as understanding of the specific attributes of proposed LN biomarkers, to further improve patient outcomes and guide disease management [28]. Importantly, despite advancements in biomarker research, kidney biopsy remains the gold standard for the determination of LN activity and the identification of histological features that dictate the pharmacotherapeutic need and guide management.
In summary, our Special Issue provides valuable insights into immune mechanisms and biomarkers relevant to SLE and LN, highlighting recent advancements in understanding the pathogenesis and improving diagnostics and patient management. The collected original works and reviews offer a comprehensive view of the potential biomarkers and therapeutic targets, paving the way for enhanced surveillance, the prevention of long-term organ damage, and optimised therapeutic decision making in SLE.

Author Contributions

I.P. and C.S. have contributed equally to this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

I.P. has received grants from the Swedish Rheumatism Association (R-995882), King Gustaf V’s 80-year Anniversary Foundation (FAI-2023-1055), Swedish Society of Medicine (SLS-974449), Nyckelfonden (OLL-1000881), Professor Nanna Svartz Foundation (2021-00436), Ulla and Roland Gustafsson Foundation (2024-43), Region Stockholm (FoUI-1004114), and Karolinska Institutet. C.S. has received grants from the Swedish Rheumatism Association (R-993724), Region Östergötland (RÖ-960604), Njurfonden (F2023-0030), Ulla and Roland Gustafsson Foundation (2023-36), the King Gustaf V’s 80-year Anniversary Foundation (FAI-2020-0663), and the King Gustaf V and Queen Victoria’s Freemasons Foundation (2021).

Acknowledgments

The authors express gratitude to Lina Wirestam, Linköping University, for her help with the graphical illustration.

Conflicts of Interest

I.P. has received research funding and/or honoraria from Amgen, AstraZeneca, Aurinia, Bristol Myers Squibb, Eli Lilly, Gilead, GSK, Janssen, Novartis, Otsuka, and Roche. C.S. discloses speaker’s bureau from Bristol Myers Squibb, Fresenius Kabi, Novartis, and AstraZeneca. Since April 2024, C.S. has been an employee of Bristol Myers Squibb. The funders had no role in the writing of the manuscript or in the decision to publish.

References

  1. Kaul, A.; Gordon, C.; Crow, M.K.; Touma, Z.; Urowitz, M.B.; van Vollenhoven, R.; Ruiz-Irastorza, G.; Hughes, G. Systemic lupus erythematosus. Nat. Rev. Dis. Primers 2016, 2, 16039. [Google Scholar] [CrossRef] [PubMed]
  2. Hoi, A.; Igel, T.; Mok, C.C.; Arnaud, L. Systemic lupus erythematosus. Lancet 2024, 403, 2326–2338. [Google Scholar] [CrossRef] [PubMed]
  3. Siegel, C.H.; Sammaritano, L.R. Systemic Lupus Erythematosus: A Review. JAMA 2024, 331, 1480–1491. [Google Scholar] [CrossRef] [PubMed]
  4. Frodlund, M.; Reid, S.; Wetterö, J.; Dahlström, Ö.; Sjöwall, C.; Leonard, D. The majority of Swedish systemic lupus erythematosus patients are still affected by irreversible organ impairment: Factors related to damage accrual in two regional cohorts. Lupus 2019, 28, 1261–1272. [Google Scholar] [CrossRef] [PubMed]
  5. Fanouriakis, A.; Kostopoulou, M.; Andersen, J.; Aringer, M.; Arnaud, L.; Bae, S.C.; Boletis, J.; Bruce, I.N.; Cervera, R.; Doria, A.; et al. EULAR recommendations for the management of systemic lupus erythematosus: 2023 update. Ann. Rheum. Dis. 2024, 83, 15–29. [Google Scholar] [CrossRef]
  6. Anders, H.J.; Saxena, R.; Zhao, M.H.; Parodis, I.; Salmon, J.E.; Mohan, C. Lupus nephritis. Nat. Rev. Dis. Primers 2020, 6, 7. [Google Scholar] [CrossRef]
  7. Lindblom, J.; Toro-Dominguez, D.; Carnero-Montoro, E.; Beretta, L.; Borghi, M.O.; Castillo, J.; Enman, Y.; Consortium, P.C.; Mohan, C.; Alarcon-Riquelme, M.E.; et al. Distinct gene dysregulation patterns herald precision medicine potentiality in systemic lupus erythematosus. J. Autoimmun. 2023, 136, 103025. [Google Scholar] [CrossRef]
  8. Parodis, I.; Lindblom, J.; Toro-Domínguez, D.; Beretta, L.; Borghi, M.O.; Castillo, J.; Carnero-Montoro, E.; Enman, Y.; Mohan, C.; Alarcón-Riquelme, M.E.; et al. Interferon and B-cell Signatures Inform Precision Medicine in Lupus Nephritis. Kidney Int. Rep. 2024, 9, 1817–1835. [Google Scholar] [CrossRef]
  9. Lindblom, J.; Mohan, C.; Parodis, I. Diagnostic, predictive and prognostic biomarkers in systemic lupus erythematosus: Current insights. Curr. Opin. Rheumatol. 2022, 34, 139–149. [Google Scholar] [CrossRef]
  10. Enocsson, H.; Wirestam, L.; Dahle, C.; Padyukov, L.; Jönsen, A.; Urowitz, M.B.; Gladman, D.D.; Romero-Diaz, J.; Bae, S.C.; Fortin, P.R.; et al. Soluble urokinase plasminogen activator receptor (suPAR) levels predict damage accrual in patients with recent-onset systemic lupus erythematosus. J. Autoimmun. 2020, 106, 102340. [Google Scholar] [CrossRef]
  11. Chalmers, S.A.; Ayilam Ramachandran, R.; Garcia, S.J.; Der, E.; Herlitz, L.; Ampudia, J.; Chu, D.; Jordan, N.; Zhang, T.; Parodis, I.; et al. The CD6/ALCAM pathway promotes lupus nephritis via T cell-mediated responses. J. Clin. Investig. 2022, 132. [Google Scholar] [CrossRef] [PubMed]
  12. Fava, A.; Buyon, J.; Magder, L.; Hodgin, J.; Rosenberg, A.; Demeke, D.S.; Rao, D.A.; Arazi, A.; Celia, A.I.; Putterman, C.; et al. Urine proteomic signatures of histological class, activity, chronicity, and treatment response in lupus nephritis. JCI Insight 2024, 9. [Google Scholar] [CrossRef] [PubMed]
  13. Palazzo, L.; Lindblom, J.; Mohan, C.; Parodis, I. Current Insights on Biomarkers in Lupus Nephritis: A Systematic Review of the Literature. J. Clin. Med. 2022, 11, 5759. [Google Scholar] [CrossRef]
  14. Lindblom, J.; Mohan, C.; Parodis, I. Biomarkers in Neuropsychiatric Systemic Lupus Erythematosus: A Systematic Literature Review of the Last Decade. Brain Sci. 2022, 12, 192. [Google Scholar] [CrossRef]
  15. Parodis, I.; Tamirou, F.; Houssiau, F.A. Prediction of prognosis and renal outcome in lupus nephritis. Lupus Sci. Med. 2020, 7, e000389. [Google Scholar] [CrossRef]
  16. Carrión-Barberà, I.; Triginer, L.; Tío, L.; Pérez-García, C.; Ribes, A.; Abad, V.; Pros, A.; Bermúdez-López, M.; Castro-Boqué, E.; Lecube, A.; et al. Role of Advanced Glycation End Products as New Biomarkers in Systemic Lupus Erythematosus. Int. J. Mol. Sci. 2024, 25, 3022. [Google Scholar] [CrossRef]
  17. Wolf, B.; Blaschke, C.R.K.; Mungaray, S.; Weselman, B.T.; Stefanenko, M.; Fedoriuk, M.; Bai, H.; Rodgers, J.; Palygin, O.; Drake, R.R.; et al. Metabolic Markers and Association of Biological Sex in Lupus Nephritis. Int. J. Mol. Sci. 2023, 24, 16490. [Google Scholar] [CrossRef]
  18. Winikajtis-Burzyńska, A.; Brzosko, M.; Przepiera-Będzak, H. Elevated Serum Levels of Soluble Transferrin Receptor Are Associated with an Increased Risk of Cardiovascular, Pulmonary, and Hematological Manifestations and a Decreased Risk of Neuropsychiatric Manifestations in Systemic Lupus Erythematosus Patients. Int. J. Mol. Sci. 2023, 24, 17340. [Google Scholar] [CrossRef]
  19. Mercader-Salvans, J.; García-González, M.; Gómez-Bernal, F.; Quevedo-Abeledo, J.C.; de Vera-González, A.; González-Delgado, A.; López-Mejías, R.; Martín-González, C.; González-Gay, M.Á.; Ferraz-Amaro, I. Relationship between Disease Characteristics and Circulating Interleukin 6 in a Well-Characterized Cohort of Patients with Systemic Lupus Erythematosus. Int. J. Mol. Sci. 2023, 24, 14006. [Google Scholar] [CrossRef]
  20. Iseme, R.A.; McEvoy, M.; Kelly, B.; Agnew, L.; Walker, F.R.; Handley, T.; Oldmeadow, C.; Attia, J.; Boyle, M. A role for autoantibodies in atherogenesis. Cardiovasc. Res. 2017, 113, 1102–1112. [Google Scholar] [CrossRef]
  21. van den Berg, V.J.; Vroegindewey, M.M.; Kardys, I.; Boersma, E.; Haskard, D.; Hartley, A.; Khamis, R. Anti-Oxidized LDL Antibodies and Coronary Artery Disease: A Systematic Review. Antioxidants 2019, 8, 484. [Google Scholar] [CrossRef]
  22. Wirestam, L.; Jönsson, F.; Enocsson, H.; Svensson, C.; Weiner, M.; Wetterö, J.; Zachrisson, H.; Eriksson, P.; Sjöwall, C. Limited Association between Antibodies to Oxidized Low-Density Lipoprotein and Vascular Affection in Patients with Established Systemic Lupus Erythematosus. Int. J. Mol. Sci. 2023, 24, 8987. [Google Scholar] [CrossRef] [PubMed]
  23. Parodis, I.; Gomez, A.; Lindblom, J.; Chow, J.W.; Sjöwall, C.; Sciascia, S.; Gatto, M. B Cell Kinetics upon Therapy Commencement for Active Extrarenal Systemic Lupus Erythematosus in Relation to Development of Renal Flares: Results from Three Phase III Clinical Trials of Belimumab. Int. J. Mol. Sci. 2022, 23, 13941. [Google Scholar] [CrossRef] [PubMed]
  24. Parodis, I.; Gatto, M.; Sjöwall, C. B cells in systemic lupus erythematosus: Targets of new therapies and surveillance tools. Front. Med. 2022, 9, 952304. [Google Scholar] [CrossRef] [PubMed]
  25. Halfon, M.; Tankeu, A.T.; Ribi, C. Mitochondrial Dysfunction in Systemic Lupus Erythematosus with a Focus on Lupus Nephritis. Int. J. Mol. Sci. 2024, 25, 6162. [Google Scholar] [CrossRef] [PubMed]
  26. Vitale, A.M.; Paladino, L.; Caruso Bavisotto, C.; Barone, R.; Rappa, F.; Conway de Macario, E.; Cappello, F.; Macario, A.J.L.; Marino Gammazza, A. Interplay between the Chaperone System and Gut Microbiota Dysbiosis in Systemic Lupus Erythematosus Pathogenesis: Is Molecular Mimicry the Missing Link between Those Two Factors? Int. J. Mol. Sci. 2024, 25, 5608. [Google Scholar] [CrossRef]
  27. von Hofsten, S.; Fenton, K.A.; Pedersen, H.L. Human and Murine Toll-like Receptor-Driven Disease in Systemic Lupus Erythematosus. Int. J. Mol. Sci. 2024, 25, 5351. [Google Scholar] [CrossRef]
  28. Alduraibi, F.K.; Tsokos, G.C. Lupus Nephritis Biomarkers: A Critical Review. Int. J. Mol. Sci. 2024, 25, 805. [Google Scholar] [CrossRef]
Figure 1. Visual representation of the content of the Special Issue. AGEs: advanced glycation end-products; anti-oxLDL: anti-oxidised low-density lipoprotein; HSP: heat shock protein; IL: interleukin; sTfR: soluble transferrin.
Figure 1. Visual representation of the content of the Special Issue. AGEs: advanced glycation end-products; anti-oxLDL: anti-oxidised low-density lipoprotein; HSP: heat shock protein; IL: interleukin; sTfR: soluble transferrin.
Ijms 25 09965 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Parodis, I.; Sjöwall, C. Immune Mechanisms and Biomarkers in Systemic Lupus Erythematosus. Int. J. Mol. Sci. 2024, 25, 9965. https://doi.org/10.3390/ijms25189965

AMA Style

Parodis I, Sjöwall C. Immune Mechanisms and Biomarkers in Systemic Lupus Erythematosus. International Journal of Molecular Sciences. 2024; 25(18):9965. https://doi.org/10.3390/ijms25189965

Chicago/Turabian Style

Parodis, Ioannis, and Christopher Sjöwall. 2024. "Immune Mechanisms and Biomarkers in Systemic Lupus Erythematosus" International Journal of Molecular Sciences 25, no. 18: 9965. https://doi.org/10.3390/ijms25189965

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop