New Therapeutics Approaches in the Autoimmune Field
© 2025 Giada Rossi, Sharon Failla, Giorgia Isinelli, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Abstract
The immune system is critical in defending the body against pathogens and foreign substances through innate and adaptive responses. However, dysregulation of these immune reactions can result in autoimmune diseases, where the immune system erroneously targets self-tissues. Autoimmunity is influenced by multiple factors, with specific susceptibility genes and environmental triggers contributing to disease onset and progression. Traditional treatments for autoimmune diseases, such as immunosuppressants and biologic agents, aim to manage symptoms but often fail to address the root cause of immune tolerance breakdown, leading to limited efficacy and adverse effects. In recent years, nanoparticles have emerged as potential immunomodulatory agents, offering novel therapeutic strategies targeting key immune components. Cationic nanoparticles, particularly poly(amidoamine) (PAMAM) dendrimers, can interact with extracellular nucleic acids, modulate macrophage polarization, and induce immune tolerance, thereby reducing inflammation and autoimmune activity. These nanoparticles hold promise in diseases such as rheumatoid arthritis, systemic lupus erythematosus, and type 1 diabetes. Despite their potential, challenges remain in optimizing nanoparticle formulations, understanding their long-term effects, and ensuring clinical safety. Future research focusing on improving targeting efficiency, reducing toxicity, and validating clinical outcomes will translate these innovative therapies into effective treatments for autoimmune diseases.
Introduction
Understanding Immunity: The Body’s Defense System
The immune system comprises a series of cells and molecules that collectively contribute to protection against foreign elements by a coordinated reaction known as the immune response [1]. The primary physiological role of the immune system is to defend against infectious microorganisms. Nevertheless, the immune response can also be triggered by non-infectious foreign substances and byproducts of damaged cells [2]. Immune responses are finely modulated by a network of positive feedback loops that enhance the reaction and mechanisms of control that curtail inappropriate or pathological responses [3]. Indeed, failure in the regulation of immune response can lead to tissue damage and diseases, such as severe infections, tumors, allergies, and autoimmune diseases [4].
Innate vs. Adaptive Immunity: The Two Lines of Defense
The immune response consists of a series of sequential and coordinated reactions. Based on the response’s speed and specificity, it is categorized into innate (natural) and adaptive (acquired) immunity [5].
Innate immunity includes physical and anatomical barriers (e.g. skin, mucosa) as well as effector cells (e.g. monocytes/macrophages, neutrophils, eosinophils, basophils, mast cells, natural killer cells, dendritic cells), antimicrobial peptides, soluble mediators (e.g. cytokines, acute phase proteins, complement system), and cell receptors (e.g. Toll-Like Receptors - TLR) [5,6]. Natural immunity is a highly conserved nonspecific response that plays a critical role in safeguarding against microbes during the initial hours or days following infection, serving as a precursor to the development of adaptive immune responses [7]. Molecules produced during innate immune reactions serve as second signals for lymphocyte activation, proliferation, and differentiation. Moreover, some innate effector cells (e.g. monocytes/macrophages, dendritic cells) collaborate with antigen presentation to stimulate antigen-specific T and B lymphocyte responses [8]. This two-signal mechanism ensures a well-coordinated and selective immune response [9].
The adaptive immunity is mediated by cells called lymphocytes and their products. There are two major populations of lymphocytes, called B and T lymphocytes, which mediate different types of adaptive immune responses [10]. Lymphocytes express highly diverse membrane receptors that are capable of identifying and responding to a wide array of substances, both microbial and non-microbial, referred to as antigens. The specific regions of complex antigens recognized by lymphocytes are referred to as determinants or epitopes [11]. The adaptive immune response is highly specific and it usually takes several weeks or days to start, progressing through a series of stages [10]. The inception of adaptive immune responses hinges on the capture and presentation of antigens to specific naïve lymphocytes. Cells designated as antigen-presenting cells (APCs) fulfill this pivotal role [12].
Subsequently, a process called clonal expansion ensues. Within this process, numerous distinct clones of lymphocytes come into play, each equipped with a unique antigen receptor, resulting in singular antigen specificity for each clone. Following clonal expansion, activated lymphocytes undergo differentiation. These activated lymphocytes, known as effector cells, play a central role in executing the ultimate effect of the immune response [13,14]. Throughout the initiation and execution phases of both innate and adaptive immune responses, immune system cells engage in interactions with one another and with other cells within the host [15]. These interactions are often mediated by molecules known as cytokines. Virtually all immune system cells release certain cytokines and possess specific signaling receptors for several cytokines [16].
In addition to effector cells, memory cells are generated, persisting over extended periods and mounting more robust and swifter responses upon encountering the same antigen anew [17]. Once the adaptive immune response has effectively eradicated the infection, the stimuli driving lymphocyte activation wane. Consequently, a majority of effector cells die out, resulting in the decline of the immune response [18]. Memory cells remain, ready to respond vigorously if the same infection recurs in the so-called secondary immune response [19,20]. Adaptive immunity is further divided into two distinct categories: humoral immunity and cell-mediated immunity. These forms of immunity are initiated by distinct classes of lymphocytes and function to counteract various types of microbes and insults [21,22]. Humoral immunity hinges on molecules present in the blood and mucosal secretions, known as antibodies. These antibodies are generated by plasma cells originating from B lymphocytes. Their role involves recognizing microbial antigens, thereby neutralizing the infectious capabilities of the microbes. Additionally, antibodies facilitate the elimination of microbes by tagging them for destruction through phagocytosis by specialized cells and by triggering the activity of the complement system [23]. Cell-mediated immunity, also known as cellular immunity, is orchestrated by T lymphocytes (T-cells). T-cells identify and react to antigens bound to host proteins localized on the surfaces of various cells and known as major histocompatibility complex (MHC) molecules [24]. Within the realm of T lymphocytes, distinct functional subsets exist, with helper T-cells and cytotoxic T lymphocytes (CTLs) being the most well-defined. Helper T-cells primarily exert their functions through the secretion of cytokines, whereas CTLs produce molecules that facilitate the destruction of other cells. An additional subset, known as regulatory T-cells (Tregs), mainly functions to suppress immune responses [25]. Distinguishing between different types of lymphocytes is possible through the expression of unique cell surface proteins, often designated by specific Cluster of Differentiation (CD) numbers, such as CD4 or CD8. These markers serve as identifiers for the different classes of lymphocytes [26].
The primary subsets of effector T-cells include CD8+ cytotoxic T lymphocytes and CD4+ helper T-cells. Additionally, among helper T-cells, there are other different subsets of cells such as Th4, Th2, and Th47, each characterized by the expression of specific cytokines and offering protection against different types of microbes and tumors(27). Th4 and Th2 cells play a pivotal role in immunity [28]. For example, Th4 cells promote cellular immune responses, by participating in the inhibition of macrophage activation, and by stimulating B-cells to produce immunoglobulins (Ig) M and IgG1. On the other hand, Th2 cells stimulate humoral immune responses, facilitating B-cell proliferation, and inducing antibody production [29].
An essential balance exists in terms of this T-cell polarization. Research has indicated that directing the polarization towards Th4 is crucial for effectively combating cancerous cells, whereas a polarization towards Th2 may aid cancer in evading the immune system [30,31]. CD4+ helper T-cells also offer activating signals to B-cells in response to protein antigens. However, B-cells can react to numerous non-protein antigens independently of helper T-cells [32,33]. After activation, each plasma cell, a specialized type of B-cell, starts to release antibodies that possess identical antigen-binding sites as the antigen receptors initially engaged on the cell surface [34]. More in detail, polysaccharides and lipids predominantly trigger the secretion of antibodies belonging to the immunoglobulin class IgM, while protein antigens prompt the generation of antibodies from a single B-cell clone in different classes, such as IgG, IgA, and IgE to fulfill specific functions [e.g. immune function in mucous membranes (IgA), protection against parasites (IgE), etc [35]. Finally, helper T-cells also stimulate the production of antibodies with heightened affinity for the antigen, enhancing the quality of the humoral immune response, in the so-called affinity maturation process [36]. A clearer picture of the immune system is displayed in Figure 1.
Understanding and manipulating these complex mechanisms is of primary importance to shape the immune response and optimize therapeutic strategies in immune-related disorders.
Figure 1: Immune System Landscape
Autoimmunity: When the Immune System Turns Against Itself The process of recognition between self and non-self mediated by the immune system is not absolute. In certain circumstances, the immune system can mistakenly direct itself against itself, generating aberrant responses implicated in inflammatory disorders, collectively defined as autoimmune diseases [37]. Although the mechanisms underlying autoimmunity and autoimmune diseases are not yet fully understood, growing evidence demonstrates the influence of genetic and environmental interactions. It is currently believed that for the rise of an autoimmune disease, there is a period in which there might be an interaction between the genetic and environmental factors, followed by the autoimmune activation that leads to diagnostic clinical symptoms [38]. In normal physiological conditions, the immune system takes care of eliminating pathogens after their identification. However, there are two main cases in which the immune system functions defectively: through autoimmune diseases and in immunodeficiency disorders. The main cause of autoimmune diseases therefore lies in the failure of recognizing the self. This phenomenon is often described as a breakdown of immunological tolerance [39]. Immune tolerance is divided into central and peripheral, and a deficiency can cause autoimmunity. Central tolerance aims to eliminate reactive B or T cells, promoting self-tolerance. This process occurs mainly in the bone marrow for B-cells and in the thymus for T-cells. Cells that bind to autoantigens thus undergo apoptosis [40]. Peripheral tolerance, in contrast to central tolerance, aims to control the excessive reactivity of B and T cells [41].
In central tolerance, T-cells that are responsible for recognizing self-antigens with a very high affinity will be eliminated in the thymus through the process of negative selection, to prevent autoimmune processes. Tregs instead will aim to suppress the activity of self-reactive T-cells, thus preventing autoimmunity [42]. Despite the thymus-mediated control mechanism, numerous autoreactive T-cells may elude thymic selection, triggering the risk of an autoimmune response. A peripheral mechanism is therefore needed to maintain self-tolerance. Subsequently, the peripheral tolerance starts its action, mediated by multiple cell classes belonging to the innate and adaptive response [43,44].
Hyperactive innate immune cells are responsible for responding to microbial triggers or high-danger signals, generating and modifying autoantigens, neoantigens, and Damage Associated Molecular Patterns (DAMPs). This modification can, in turn, be triggered by different mechanisms such as increased oxidative stress, causing damage to nucleic acids, proteins, lipids, and carbohydrates. In predisposed subjects, neoantigens can generate a cascade of inflammatory mechanisms. When cellular debris and dead cells are not properly eliminated by innate immune cells, mitochondrial and nuclear DNA and RNA are exposed to possible modifications. This entire process has a proinflammatory effect, activating TLR [45,46].
Autoimmune diseases can target different organs varying their clinical manifestations. Some are limited to specific tissues, while others have systemic characteristics. Regardless of these variations, all autoimmune diseases are believed to develop through phases. These are represented by an initial phase in which a genetic predisposition is influenced by environmental triggers. This phase is usually characterized by subclinical symptoms, in which patients are generally unaware. This is followed by a propagation phase, characterized by autoinflammatory processes and tissue damage, related to the production of cytokines and disruption of the balance between effector and regulatory T-cells. This is followed by a final resolution phase, in which there is a partial and usually short-term ability, in which the body attempts to restore the balance between effector and regulatory T-cells. This last phase is usually characterized by the relapsing of the disease [47]. In most cases, the genetic component of autoimmunity derives from changes found in class II molecules of the major histocompatibility complex (MHC II), which are essentially responsible for modulating the efficiency of antigen presentation. The remaining part of the genetic component derives from additive effects in which the cooperation of multiple genetic loci is recorded [48].
Genetic studies have shown that the same genes can increase the risk of different autoimmune diseases. Many genes contribute to autoimmune predisposition. For diseases such as Systemic Lupus Erythematosus (SLE) and rheumatoid arthritis (RA), for example, more than 100 different loci have been identified, responsible for an increased risk. The number of genes identified also suggests that pathological susceptibility is multigenic. An example is the PTPN22 gene, whose contribution has been demonstrated in several diseases including RA, SLE, and autoimmune (Type 1) diabetes mellitus. It represents one of the main susceptibility genes in addition to those of the MHC region [49-51].
RA is an organ-specific autoimmune disease caused by the presence of autoantibodies such as Rheumatoid Factor (RF) and Anti-Citrullinated Protein Antibodies (ACPA). This disease has been identified as having a familial clustering with more than 100 loci. Several environmental factors contribute to the risk, including cigarette smoking, lack of physical exercise, stress, and diet [52].
SLE is a multiorgan systemic autoimmune disease related to a dysregulated tolerance in B cells and increased synthesis of autoantibodies. Several susceptibility loci have been highlighted, such as the Human Leukocyte Antigen (HLA) and non-HLA genes. Smoking is associated with increased production of anti-double- stranded DNA (anti-dsDNA) antibodies [52,53].
In Type 1 diabetes mellitus (T1D), autoimmunity affects pancreatic beta cells and the genetic component plays a significant role. It has been observed that familial risk is mainly related to HLA genes [54]. Low physical activity, stress, infections, diet, and psychological trauma are associated with a higher risk of incidence [55].
The standard treatment for autoimmune diseases has been based for a long time on immunosuppressants. Subsequently, there was the advent of biological immunomodulatory drugs, which targeted inflammatory mediators including cytokines. An example of this class is TNF-α inhibitors that inhibit the proinflammatory activity of this mediator, for RA. This therapy has thus represented the reference treatment for a long time, allowing disease control and reduced adverse effects compared to broad-spectrum immunosuppressants. However, anti-TNF-α therapies have also shown limitations, such as the loss of efficacy due to the triggering of resistance mechanisms as in multiple sclerosis [56].
Currently approved immunomodulatory therapies aim to treat symptoms, but they do not eradicate the underlying problem, namely the loss of immune tolerance. Refractory autoimmune diseases are not susceptible to these treatments, which, while aiming to reduce systemic inflammation, can subject the patient to opportunistic infections and harmful side effects [58-62].
B-cells and plasma cells contribute to the pathogenesis of autoimmune diseases by stimulating the production of autoantibodies. Therefore, therapeutic strategies have been designed to eliminate these cell classes. Monoclonal antibodies (mAb) interrupt B-cell and plasma cell-mediated signaling and trigger complement-dependent cytotoxicity and antibody- dependent cytotoxicity. Another strategy has been the creation of engineered T-cells via Chimeric Antigen Receptors (CAR-T therapy). Nevertheless, even these therapies have important limitations, such as the need for repeated administrations in the first case, and difficulties in the administration and the exact control of the dosage in the second case [57]. Therefore, although B-cell depletion strategies have been shown to be useful in the treatment of autoimmune diseases such as RA, SLE, multiple sclerosis, and many other immune-mediated inflammatory diseases, not all patients respond to these treatments or achieve drug-free remission [58].
Given the increasing number of autoimmune diseases in the last decades, there has been an urge to renovate therapies to reduce the side effects of classical treatments.
Nanomedicine in Action: Current Research on Nanoparticles for Autoimmune Diseases
In recent years, nanoparticles have emerged as powerful immunomodulators in treating autoimmune diseases due to their ability to interact with key immune components. Engineered nanomaterials can modulate immune recognition, either enhancing or suppressing immune responses and may alter how the immune system perceives and detects foreign substances [59]. These tiny particles, typically less than 100 nanometers in size, can be engineered to interact specifically with the components of the immune system, offering targeted therapeutic strategies, as displayed in Figure 2.
Figure 2: Schematic of Nanoparticles Employed in Autoimmune Diseases
Cationic nanoparticles can scavenge extracellular nucleic acids, such as cell-free DNA, thereby preventing Toll-like receptor activation and reducing inflammation in conditions like SLE and RA(60–67). Additionally, nanoparticle-based strategies can regulate macrophage polarization, shifting pro-inflammatory M1 macrophages to the anti-inflammatory M2 phenotype, while also depleting hyperactive macrophages involved in chronic inflammation [68-72]. Targeting lymphocytes, nanoparticles can induce immune tolerance by promoting regulatory T-cells, suppressing autoreactive T-cells(76), and inhibiting antibody- producing B-cells, showing potential in diseases such as multiple sclerosis and Type 1 diabetes(64,67,73–80). Furthermore, nanoparticles can neutralize inflammatory cytokines like TNF-α and IL-1β or scavenge reactive oxygen species, offering a comprehensive strategy for autoimmune therapy [63,81-88].
One of the most studied cationic nanoparticles is poly(amidoamine) (PAMAM) dendrimers, which are extensively studied for their ability to bind nucleic acids, making them valuable for gene delivery and as nucleic acid scavengers [89]. Their effectiveness is influenced by surface charge density, with higher-generation dendrimers (G3-G10) displaying enhanced nucleic acid binding and transfection efficiency [90,91]. Notably, PAMAM-G3 has been explored for its ability to mitigate inflammation and thrombosis by neutralizing nucleic acids that activate TLRs, a key mechanism in immune system dysregulation [92-94].
Circulating cell-free DNA (cfDNA), first identified by Mandel and Metais (1948), has been linked to autoimmune disorders, with increased levels observed in conditions such as SLE and RA [95-97]. cfDNA originates from various sources, including apoptotic cells and neutrophil extracellular traps (NETs), and can trigger inflammatory pathways through endosomal TLRs such as TLR7, TLR8, and TLR9 [98,99]. Given the role of nucleic acids in autoimmune activation, cationic nanoparticles have been explored as potential therapeutic agents to prevent excessive immune stimulation.
Cationic nanomaterials, widely used for non-viral nucleic acid delivery, have recently been investigated for their ability to suppress immune activation by scavenging extracellular nucleic acids [100]. Some of these materials, including PAMAM-G3, have been shown to reduce TLR activation by binding inflammatory nucleic acids and altering their intracellular trafficking [101]. Additionally, they can interfere with lupus-associated immune complexes by displacing autoantibodies from DNA, thereby mitigating disease progression without inducing general immunosuppression [102,103]. Beyond autoimmune diseases, cationic nanoparticles have also effectively reduced inflammatory responses in sepsis models [104,105].
Conclusions and Future Perspectives
Nanoparticles have demonstrated significant potential as immunomodulators in treating autoimmune diseases by targeting key immune components, scavenging inflammatory nucleic acids, and modulating immune cell activity. Advances in nanotechnology have enabled the design of nanoparticles with enhanced specificity, reduced toxicity, and improved therapeutic efficacy, offering promising alternatives to conventional immunosuppressive therapies. Despite these advances, challenges remain in understanding their long-term effects, immune interactions, and clinical safety.
Future research should focus on optimizing nanoparticle formulations to improve targeting efficiency, reduce off-target effects, and ensure biocompatibility. Large-scale clinical studies are needed to validate preclinical findings and assess the long-term safety of these innovative therapies. Furthermore, interdisciplinary collaborations between immunologists, materials scientists, and clinicians will be essential to drive the development of next-generation nanoparticle-based treatments. Expanding our understanding of nanoparticle-immune interactions could unlock new therapeutic possibilities and pave the way for more effective and personalized therapies for autoimmune diseases. Further research in this rapidly evolving field will be crucial to translating these promising nanotechnologies into clinical applications.
Acknowledgments
We thank BioRender(106) for the illustrations of our figures.
Fundings
No fundings were received.
Availability of data and materials
Not applicable.
Authors’ contributions
GR, SF, and GI conceptualized the review. GR contributed to the study by creating a figure, collecting data from the literature, and editing the text. SF contributed to the study by collecting data from the literature and structuring and editing the manuscript.
GI contributed to the study by creating a figure, collecting data from the literature, editing the text, and finalizing the manuscript.
Competing interests
The authors declare that they have no competing interests.
References
- Delves PJ, Roitt IM (2000) The immune First of two parts. N Engl J Med 343: 37-49.
- Sattler S (2017) The Role of the Immune System Beyond the Fight Against Adv Exp Med Bio l1003: 3-14.
- Scheiermann C, Kunisaki Y, Frenette PS (2013) Circadian control of the immune Nat Rev Immunol 13: 190-198.
- McEwen BS (1998) Stress, adaptation, and disease. Allostasis and allostatic load. Ann N Y Acad Sci. 840: 33-44.
- Artis D, Spits H (2015) The biology of innate lymphoid Nature 517: 293-301.
- Mantovani A, Garlanda C (2023) Humoral Innate Immunity and Acute-Phase Proteins. N Engl J Med 388: 439-452.
- Tomar N, De RK (2014) A brief outline of the immune Methods Mol Biol Clifton NJ 1184: 3-12.
- Iwasaki A, Medzhitov R (2010) Regulation of adaptive immunity by the innate immune system. Science 327: 291-
- Bretscher PA (1999) A two-step, two-signal model for the primary activation of precursor helper T cells. Proc Natl Acad Sci USA 96: 185-190.
- Bonilla FA, Oettgen HC (2010) Adaptive J Allergy Clin Immunol 125: S33-S40.
- Pishesha N, Harmand TJ, Ploegh HL (2022) A guide to antigen processing and Nat Rev Immunol 22: 751-764.
- Cresswell P, Roche PA (2017) Editorial Overview: Antigen Processing and Presentation; many fingers in many pies. Curr Opin Immunol 46: 5-7.
- Chapman NM, Boothby MR, Chi H (2020) Metabolic coordination of T cell quiescence and activation. Nat Rev Immunol 20: 55-70.
- Wik JA, Skålhegg BS (2022) T Cell Metabolism in Front Immunol 13: 840610.
- Sun L, Wang X, Saredy J, Yuan Z, Yang X, et al. (2020) Innate-adaptive immunity interplay and redox regulation in immune response. Redox Biol 37: 101759.
- Dong C (2021) Cytokine Regulation and Function in T Annu Rev Immunol 39: 51-76.
- Laidlaw BJ, Craft JE, Kaech SM (2016) The multifaceted role of CD4(+) T cells in CD8(+) T cell memory. Nat Rev Immunol 16: 102-111.
- Marrack P, Scott-Browne J, MacLeod MKL (2010) Terminating the immune Immunol Rev 236: 5-10.
- Netea MG, Schlitzer A, Placek K, Joosten LAB, Schultze JL (2019) Innate and Adaptive Immune Memory: an Evolutionary Continuum in the Host’s Response to Cell Host Microbe 25: 13-26.
- Cancro MP, Tomayko MM (2021) Memory B cells and plasma cells: The differentiative continuum of humoral immunity. Immunol Rev 303: 72-82.
- Sebina I, Pepper M (2018) Humoral immune responses to infection: common mechanisms and unique strategies to combat pathogen immune evasion Curr Opin Immunol 51: 46-54.
- Kaufmann SH (1999) Cell-mediated immunity: dealing a direct blow to pathogens. Curr Biol CB 9: R97-99.
- Daëron M (2024) The function of Immunol Rev 328: 113-125.
- Neefjes J, Jongsma MLM, Paul P, Bakke O (2011) Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat Rev Immunol 11: 823-836.
- Ashby KM, Hogquist KA (2024) A guide to thymic selection of T cells. Nat Rev Immunol 24: 103-117.
- Mousset CM, Hobo W, Woestenenk R, Preijers F, Dolstra H, et (2019) Comprehensive Phenotyping of T Cells Using Flow Cytometry. Cytom Part J Int Soc Anal Cytol 95: 647-654.
- Luckheeram RV, Zhou R, Verma AD, Xia B (2012) CD4+T cells: differentiation and Clin Dev Immunol 2012: 925135.
- Zhang Y, Zhang Y, Gu W, Sun B (2014) Th4/TH2 cell differentiation and molecular signals. Adv Exp Med Biol 841: 15-44.
- Infante-Duarte C, Kamradt T (1999) Th4/Th2 balance in infection. Springer Semin Immunopathol 21: 317-338.
- Shang Q, Yu X, Sun Q, Li H, Sun C, et al. (2024) Polysaccharides regulate Th4/Th2 balance: A new strategy for tumor Biomed Pharmacother Biomedecine Pharmacother 170: 115976.
- Basu A, Ramamoorthi G, Albert G, Gallen C, Beyer A, et al. (2021) Differentiation and Regulation of TH Cells: A Balancing Act for Cancer Immunotherapy. Front Immunol 12: 669474.
- Burstein HJ, Abbas AK (1991) T-cell-mediated activation of B cells. Curr Opin Immunol 3: 345-349.
- Pike BL, Alderson MR, Nossal GJ (1987) T-independent activation of single B cells: an orderly analysis of overlapping stages in the activation Immunol Rev 99: 119-152.
- Eibel H, Kraus H, Sic H, Kienzler AK, Rizzi M (2014) B cell biology: an Curr Allergy Asthma Rep 14: 434.
- Bashford-Rogers RJM, Bergamaschi L, McKinney EF, Pombal DC, Mescia F, et al. (2019) Analysis of the B cell receptor repertoire in six immune-mediated diseases. Nature 574: 122-126.
- Victora GD, Nussenzweig MC (2022) Germinal Centers. Annu Rev Immunol 40: 413-442.
- Theofilopoulos AN, Kono DH, Baccala R (2017) The multiple pathways to autoimmunity. Nat Immunol 18: 716-724.
- Miller FW (2023) The Increasing Prevalence of Autoimmunity and Autoimmune Diseases: An Urgent Call to Action for Improved Understanding, Diagnosis, Treatment and Prevention. Curr Opin Immunol 80: 102266.
- Yasmeen F, Pirzada RH, Ahmad B, Choi B, Choi S (2024) Understanding Autoimmunity: Mechanisms, Predisposing Factors, and Cytokine Therapies. Int J Mol Sci 25: 7666.
- Kannan AK, Su Z, Gauvin DM, Paulsboe SE, Duggan R, et al. (2019) IL-23 induces regulatory T cell plasticity with implications for inflammatory skin Sci Rep 9: 17675.
- Nd S, T de B, Kv W, Sa J, K van M, et (2008) Human anti- inflammatory macrophages induce Foxp3+ GITR+ CD25+ regulatory T cells, which suppress via membrane-bound TGFbeta-1. J Immunol Baltim Md 1950 181.
- Suvas S, Singh V, Sahdev S, Vohra H, Agrewala JN (2002) Distinct role of CD80 and CD86 in the regulation of the activation of B cell and B cell J Biol Chem 277: 7766-7775.
- Hui R, Garon EB, Goldman JW, Leighl NB, Hellmann MD, et (2017) Pembrolizumab as first-line therapy for patients with PD-L1-positive advanced non-small cell lung cancer: a phase 1 trial. Ann Oncol Off J Eur Soc Med Oncol 28: 874-881.
- Curran MA, Montalvo W, Yagita H, Allison JP (2010) PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A 107: 4275-4280.
- Blanco LP, Kaplan MJ (2023) Metabolic alterations of the immune system in the pathogenesis of autoimmune diseases PLoS Biol 21: e3002084.
- Leinardi R, Longo Sanchez-Calero C, Huaux F (2022) Think Beyond Particle Cytotoxicity: When Self-Cellular Components Released After Immunogenic Cell Death Explain Chronic Disease Development. Front Toxicol 4: 887228.
- Rosenblum MD, Remedios KA, Abbas AK (2015) Mechanisms of human autoimmunity. J Clin Invest 125: 2228-2233.
- Alriyami M, Polychronakos C (2021) Somatic Mutations and Cells 10: 2056.
- Pisetsky DS (2023) Pathogenesis of autoimmune disease. Nat Rev Nephrol 19: 509-524.
- Stanford SM, Bottini N (2014) PTPN22: the archetypal non- HLA autoimmunity Nat Rev Rheumatol 10: 602-611.
- Mustelin T, Bottini N, Stanford SM (2019) The Contribution of PTPN22 to Rheumatic Disease. Arthritis Rheumatol Hoboken NJ 71: 486-495.
- Nandakumar KS, Nündel K (2022) Editorial: Systemic lupus erythematosus - predisposition factors, pathogenesis, diagnosis, treatment and disease models. Front Immunol 13: 1118180.
- Choi MY, Hahn J, Malspeis S, Stevens EF, Karlson EW, et (2022) Association of a Combination of Healthy Lifestyle Behaviors With Reduced Risk of Incident Systemic Lupus Erythematosus. Arthritis Rheumatol Hoboken NJ 74: 274- 283.
- Handel AE, Handunnetthi L, Ebers GC, Ramagopalan SV (2009) Type 1 diabetes mellitus and multiple sclerosis: common etiological features. Nat Rev Endocrinol 5: 655-664.
- Rewers M, Ludvigsson J (2016) Environmental risk factors for type 1 diabetes. Lancet Lond Engl 387: 2340-2348.
- Moorman CD, Sohn SJ, Phee H (2021) Emerging Therapeutics for Immune Tolerance: Tolerogenic Vaccines, T cell Therapy, and IL-2 Therapy. Front Immunol 12: 657768.
- Zhang Z, Xu Q, Huang L (2023) B cell depletion therapies in autoimmune diseases: Monoclonal antibodies or chimeric antigen receptor-based therapy? Front Immunol 14:
- Ana Merino-Vico, Giulia Frazzei, Jan Piet van Hamburg, Sander W Tas (2023) Targeting B cells and plasma cells in autoimmune diseases: From established treatments to novel therapeutic approaches - PubMed 53: e2149675.
- Rezaei R, Safaei M, Mozaffari HR, Moradpoor H, Karami S, et (2019) The Role of Nanomaterials in the Treatment of Diseases and Their Effects on the Immune System. Open Access Maced J Med Sci 7: 1884-1890.
- Lee YY, Park HH, Park W, Kim H, Jang JG, et (2021) Long- acting nanoparticulate DNase-1 for effective suppression of SARS-CoV-2-mediated neutrophil activities and cytokine storm. Biomaterials 267: 120389.
- Mendelson A, Frenette PS (2014) Hematopoietic stem cell niche maintenance during homeostasis and Nat Med 20: 833-846.
- Heidt T, Sager HB, Courties G, Dutta P, Iwamoto Y, et al. (2014) Chronic variable stress activates hematopoietic stem cells. Nat Med 20: 754-758.
- Zhang Q, Dehaini D, Zhang Y, Zhou J, Chen X, et (2018) Neutrophil membrane-coated nanoparticles inhibit synovial inflammation and alleviate joint damage in inflammatory arthritis. Nat Nanotechnol 13: 1182-1190.
- Bednar KJ, Nycholat CM, Rao TS, Paulson JC, Fung-Leung WP, et al. (2019) Exploiting CD22 To Selectively Tolerize Autoantibody Producing B-Cells in Rheumatoid Arthritis. ACS Chem Biol 14: 644-654.
- Zhang Q, Yuan R, Li C, Wei W, Shen W, et al. (2019) Macrophage depletion with clodronate-containing liposomes affects the incidence and development of rheumatoid arthritis. Z Rheumatol 78: 996-1003.
- Thurlings RM, Wijbrandts CA, Bennink RJ, Dohmen SE, Voermans C, et al. (2009) Monocyte scintigraphy in rheumatoid arthritis: the dynamics of monocyte migration in immune-mediated inflammatory PloS One 4: e7865.
- Liu Q, Wang X, Liu X, Kumar S, Gochman G, et (2019) Use of Polymeric Nanoparticle Platform Targeting the Liver To Induce Treg-Mediated Antigen-Specific Immune Tolerance in a Pulmonary Allergen Sensitization Model. ACS Nano 13: 4778-4794.
- Kwon D, Cha BG, Cho Y, Min J, Park EB, et Extra-Large Pore Mesoporous Silica Nanoparticles for Directing in Vivo M2 Macrophage Polarization by Delivering IL-4. Nano Lett 17: 2747-2756.
- Ahamad N, Kar A, Mehta S, Dewani M, Ravichandran V, et al. (2021) Immunomodulatory nanosystems for treating inflammatory diseases. Biomaterials. 274: 120875.
- Brown BN, Ratner BD, Goodman SB, Amar S, Badylak SF (2012) Macrophage polarization: an opportunity for improved outcomes in biomaterials and regenerative medicine. Biomaterials 33: 3792-3802.
- McCloskey E, Paterson AH, Powles T, Kanis JA (2021) Clodronate. Bone 143: 115715.
- Bacci M, Capobianco A, Monno A, Cottone L, Di Puppo F, et al. (2009) Macrophages are alternatively activated in patients with endometriosis and required for growth and vascularization of lesions in a mouse model of Am J Pathol 175: 547-556.
- Ohno M, Nishida A, Sugitani Y, Nishino K, Inatomi O, et al. (2017) Nanoparticle curcumin ameliorates experimental colitis via modulation of gut microbiota and induction of regulatory T cells. PloS One 12: e0185999.
- Singha S, Shao K, Yang Y, Clemente-Casares X, Solé P, et (2017) Peptide-MHC-based nanomedicines for autoimmunity function as T-cell receptor microclustering devices. Nat Nanotechnol 12: 701-710.
- Yeste A, Takenaka MC, Mascanfroni ID, Nadeau M, Kenison JE, et al. (2016) Tolerogenic nanoparticles inhibit T cell- mediated autoimmunity through Sci Signal 9: ra61.
- Özcan A, Sahin D, Impellizzieri D, Nguyen TT, Hafner J, et al. (2020) Nanoparticle-Coupled Topical Methotrexate Can Normalize Immune Responses and Induce Tissue Remodeling in Psoriasis. J Invest Dermatol 140: 1003-1014.
- Carnasciali A, Amoriello R, Bonechi E, Mazzoni A, Ravagli C, et al. (2021) T Cell Delivery of Nanoparticles-Bound Anti- CD20 Monoclonal Antibody: Successful B Cell Depletion in the Spinal Cord during Experimental Autoimmune Encephalomyelitis. J Neuroimmune Pharmacol Off J Soc NeuroImmune Pharmacol 16: 376-389.
- Clemente-Casares X, Blanco J, Ambalavanan P, Yamanouchi J, Singha S, et al. (2016) Expanding antigen-specific regulatory networks to treat autoimmunity. Nature 530: 434-440.
- Leuschner F, Dutta P, Gorbatov R, Novobrantseva TI, Donahoe JS, et al. (2011) Therapeutic siRNA silencing in inflammatory monocytes in Nat Biotechnol 29: 1005- 1010.
- Huang H, Feng W, Chen Y, Shi J (2020) Inorganic nanoparticles in clinical trials and translations. Nano Today 35: 100972.
- Gao J, Wang S, Wang Z (2017) High yield, scalable and remotely drug-loaded neutrophil-derived extracellular vesicles (EVs) for anti-inflammation therapy. Biomaterials 135: 62-73.
- Pereira M, Francisco S, Varanda AS, Santos M, Santos MAS, et al. (2018) Impact of tRNA Modifications and tRNA- Modifying Enzymes on Proteostasis and Human Disease. Int J Mol Sci 19: 3738.
- Gao J, Chu D, Wang Z (2016) Cell membrane-formed nanovesicles for disease-targeted J Control Release Off J Control Release Soc 224: 208-216.
- Rodriguez-Nogales A, Lozano-Pérez AA, Aznar-Cervantes SD, Algieri F, Garrido-Mesa J, et al. (2016) Effect of aqueous and particulate silk fibroin in a rat model of experimental colitis. Int J Pharm 511: 1-9.
- Gou S, Huang Y, Sung J, Xiao B, Merlin D (2019) Silk fibroin-based nanotherapeutics: application in the treatment of colonic diseases. Nanomed 14: 2373-2378.
- Zhong G, Yang X, Jiang X, Kumar A, Long H, et (2019) Dopamine-melanin nanoparticles scavenge reactive oxygen and nitrogen species and activate autophagy for osteoarthritis therapy. Nanoscale 11: 11605-11616.
- Liao N, Su L, Zheng Y, Zhao B, Wu M, et (2021) In Vivo Tracking of Cell Viability for Adoptive Natural Killer Cell- Based Immunotherapy by Ratiometric NIR-II Fluorescence Imaging. Angew Chem Int Ed Engl 60: 20888-20896.
- Zhang C, Ling CL, Pang L, Wang Q, Liu JX, et
- (2017) Direct Macromolecular Drug Delivery to Cerebral Ischemia Area using Neutrophil-Mediated Nanoparticles. Theranostics 7: 3260-3275.
- Abedi-Gaballu F, Dehghan G, Ghaffari M, Yekta R, Abbaspour-Ravasjani S, et (2018) PAMAM dendrimers as efficient drug and gene delivery nanosystems for cancer therapy. Appl Mater Today 12: 177-190.
- Jensen LB, Pavan GM, Kasimova MR, Rutherford S, Danani A, et al. (2011) Elucidating the molecular mechanism of PAMAM-siRNA dendriplex self-assembly: effect of dendrimer charge density. Int J Pharm 416: 410-418.
- Palmerston Mendes L, Pan J, Torchilin VP (2017) Dendrimers as Nanocarriers for Nucleic Acid and Drug Delivery in Cancer Mol Basel Switz 22: 1401.
- Jain S, Pitoc GA, Holl EK, Zhang Y, Borst L, et al. (2012) Nucleic acid scavengers inhibit thrombosis without increasing Proc Natl Acad Sci U S A 109: 12938-12943.
- Naqvi I, Gunaratne R, McDade JE, Moreno A, Rempel RE, et al. (2018) Polymer-Mediated Inhibition of Pro-invasive Nucleic Acid DAMPs and Microvesicles Limits Pancreatic Cancer Mol Ther J Am Soc Gene Ther 26: 1020- 1031.
- Holl EK, Frazier V, Landa K, Boczkowski D, Sullenger B, et (2021) Controlling cancer-induced inflammation with a nucleic acid scavenger prevents lung metastasis in murine models of breast cancer. Mol Ther J Am Soc Gene Ther 29: 1772-1781.
- Tug S, Helmig S, Menke J, Zahn D, Kubiak T, et (2014) Correlation between cell free DNA levels and medical evaluation of disease progression in systemic lupus erythematosus patients. Cell Immunol 292: 32-39.
- Muñoz LE, Lauber K, Schiller M, Manfredi AA, Herrmann M (2010) The role of defective clearance of apoptotic cells in systemic Nat Rev Rheumatol 6: 280-289.
- Dong C, Liu Y, Sun C, Liang H, Dai L, et al. (2020) Identification of Specific Joint-Inflammatogenic Cell-Free DNA Molecules From Synovial Fluids of Patients With Rheumatoid Arthritis. Front Immunol 11: 662.
- Barrat FJ, Meeker T, Gregorio J, Chan JH, Uematsu S, et al. (2005) Nucleic acids of mammalian origin can act as endogenous ligands for Toll-like receptors and may promote systemic lupus J Exp Med 202: 1131-1139.
- Fillatreau S, Manfroi B, Dörner T (2021) Toll-like receptor signalling in B cells during systemic lupus erythematosus. Nat Rev Rheumatol 17: 98-108.
- Yonezawa S, Koide H, Asai T (2020) Recent advances in siRNA delivery mediated by lipid-based Adv Drug Deliv Rev 155: 64-78.
- Lee J, Sohn JW, Zhang Y, Leong KW, Pisetsky D, et (2011) Nucleic acid-binding polymers as anti-inflammatory agents. Proc Natl Acad Sci U S A 108: 14055-14060.
- Stearns NA, Lee J, Leong KW, Sullenger BA, Pisetsky DS (2012) The inhibition of anti-DNA binding to DNA by nucleic acid binding polymers. PloS One 7: e40862.
- Holl EK, Shumansky KL, Pitoc G, Ramsburg E, Sullenger BA (2013) Nucleic acid scavenging polymers inhibit extracellular DNA-mediated innate immune activation without inhibiting anti-viral responses. PloS One 8: e69413.
- Dawulieti J, Sun M, Zhao Y, Shao D, Yan H, et al. (2020) Treatment of severe sepsis with nanoparticulate cell-free DNA scavengers. Sci Adv 6: eaay7148.
- Liu F, Sheng S, Shao D, Xiao Y, Zhong Y, et al. (2021) A Cationic Metal-Organic Framework to Scavenge Cell-Free DNA for Severe Sepsis Management. Nano Lett 21: 2461-
- Scientific Image and Illustration Software | BioRender [Internet]. [cited 2025 Mar 3]. Available from: https://www. com/.