Skip to content

This report is written by MaltSci based on the latest literature and research findings


How do regulatory T cells control immune tolerance?

Abstract

Regulatory T cells (Tregs) are specialized CD4+ T lymphocytes that play a crucial role in maintaining immune tolerance and preventing autoimmunity. Their primary function is to suppress the activation and proliferation of other immune cells, thereby ensuring that the immune system does not overreact to self-antigens or harmless foreign substances. Tregs achieve their regulatory effects through various mechanisms, including the production of inhibitory cytokines like IL-10 and TGF-β, cell-contact-dependent interactions, and metabolic modulation of effector T cells. Recent research has identified distinct Treg subsets, such as thymus-derived Tregs (tTregs) and peripherally induced Tregs (pTregs), which contribute to both central and peripheral tolerance. Tregs are essential in various physiological contexts, including pregnancy, where they help maintain tolerance to fetal antigens, and in pathological conditions like autoimmunity and cancer. Dysregulation of Tregs can lead to autoimmune diseases or facilitate tumor progression by suppressing anti-tumor immunity. Current therapeutic strategies targeting Tregs aim to enhance their function in autoimmune diseases or deplete them in cancer therapy. Future research directions include identifying novel biomarkers for Treg subsets and exploring gene therapy approaches to manipulate Treg functions. Understanding the complexities of Treg biology will be crucial for developing innovative therapeutic interventions to modulate immune responses in various diseases.

Outline

This report will discuss the following questions.

  • 1 Introduction
  • 2 The Role of Regulatory T Cells in Immune Tolerance
    • 2.1 Mechanisms of Treg Suppression
    • 2.2 Treg Subsets and Their Functional Diversity
  • 3 Factors Influencing Treg Development and Function
    • 3.1 Cytokine Environment
    • 3.2 Microbiota Influence
  • 4 Tregs in Health and Disease
    • 4.1 Tregs in Autoimmunity
    • 4.2 Tregs in Cancer Immunology
    • 4.3 Tregs in Transplantation
  • 5 Therapeutic Targeting of Tregs
    • 5.1 Enhancing Treg Function for Autoimmune Diseases
    • 5.2 Depleting Tregs in Cancer Therapy
  • 6 Future Directions in Treg Research
    • 6.1 Novel Biomarkers for Treg Identification
    • 6.2 Potential for Gene Therapy
  • 7 Summary

1 Introduction

Regulatory T cells (Tregs) are a specialized subset of T lymphocytes that play a crucial role in maintaining immune tolerance and preventing autoimmunity. Their primary function is to suppress the activation and proliferation of other immune cells, ensuring that the immune system does not overreact to self-antigens or harmless foreign substances. The importance of Tregs is underscored by their involvement in various physiological and pathological processes, including transplantation, cancer, and chronic infections. Recent advances in our understanding of Treg biology have highlighted their multifaceted roles beyond mere suppression, revealing their involvement in tissue repair and metabolic regulation [1]. This expanding knowledge base emphasizes the necessity of elucidating the mechanisms by which Tregs exert their regulatory functions, as such insights are essential for developing therapeutic strategies aimed at modulating immune responses.

The significance of Tregs in the context of immune tolerance cannot be overstated. Immune tolerance is a critical process that allows the immune system to distinguish between self and non-self, thereby preventing autoimmune diseases and maintaining homeostasis [2]. Tregs contribute to both central and peripheral tolerance mechanisms, with thymus-derived regulatory T cells (tTregs) mediating central tolerance and peripherally derived regulatory T cells (pTregs) functioning in peripheral tolerance [2]. Moreover, the discovery of various Treg subsets, including the recently characterized type 1 regulatory T cells (Tr1) and Th3 cells, has further elucidated the complexity of Treg functions and their roles in immune regulation [3].

Despite the established roles of Tregs in preventing autoimmunity and maintaining tolerance, their dysregulation has been implicated in a range of diseases. For instance, insufficient Treg activity is associated with autoimmune disorders, while an overabundance of Tregs can contribute to tumor progression by suppressing anti-tumor immunity [4]. Additionally, Tregs play a pivotal role in transplantation tolerance, where their presence can significantly improve graft survival [5]. These diverse roles underscore the dual nature of Tregs as both protectors of self-tolerance and potential facilitators of disease, illustrating the need for a nuanced understanding of their biology.

The organization of this review will provide a comprehensive overview of the current knowledge regarding Tregs and their role in controlling immune tolerance. The subsequent sections will delve into the mechanisms of Treg suppression and the functional diversity among Treg subsets, highlighting the intricacies of Treg biology. We will then explore the various factors influencing Treg development and function, including the cytokine environment and microbiota influences, which are critical for the proper functioning of these cells [6]. The discussion will also encompass the role of Tregs in health and disease, examining their involvement in autoimmunity, cancer immunology, and transplantation. Following this, we will assess therapeutic strategies targeting Tregs, considering approaches to enhance Treg function in autoimmune diseases and strategies for depleting Tregs in cancer therapy [4][5]. Finally, we will outline future directions in Treg research, including the identification of novel biomarkers and the potential for gene therapy to manipulate Treg functions [7].

In summary, the understanding of Tregs and their regulatory functions is essential for developing innovative therapeutic interventions aimed at modulating immune responses. As we explore the complexities of Treg biology, it becomes increasingly clear that harnessing their potential could pave the way for novel treatments in various immune-mediated diseases.

2 The Role of Regulatory T Cells in Immune Tolerance

2.1 Mechanisms of Treg Suppression

Regulatory T cells (Tregs) are pivotal in maintaining immune tolerance, ensuring that the immune system can distinguish between self and non-self antigens, thereby preventing autoimmunity while allowing effective responses to pathogens. The mechanisms by which Tregs exert their suppressive functions are multifaceted and involve several distinct pathways.

One of the primary mechanisms of Treg-mediated suppression is through the production of inhibitory cytokines. Tregs secrete cytokines such as interleukin-10 (IL-10) and transforming growth factor beta (TGF-β), which inhibit the activation and proliferation of effector T cells and other immune cells. IL-10, in particular, plays a crucial role in dampening inflammatory responses, while TGF-β is involved in promoting the development and maintenance of Tregs themselves[8].

In addition to cytokine production, Tregs employ cell-contact-dependent mechanisms to exert their effects. They express inhibitory receptors such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1), which interact with their ligands on antigen-presenting cells (APCs) and effector T cells. This interaction can lead to the downregulation of immune responses and promote an anergic state in effector T cells[8].

Tregs also influence metabolic pathways within the immune microenvironment. They can modulate the metabolism of effector T cells, thereby limiting their activation and proliferation. This metabolic reprogramming is crucial in environments where immune responses need to be tightly regulated, such as during chronic infections or in tumor microenvironments[8].

Moreover, Tregs play a significant role in peripheral tolerance through various mechanisms, including the induction of anergy in autoreactive T cells and the promotion of regulatory feedback loops within the immune system. The complexity of these regulatory networks reflects the necessity for precise control of immune responses, particularly in the context of infection and autoimmunity[9].

The development and function of Tregs are also influenced by their microenvironment. Factors such as the type of antigen, the presence of specific cytokines, and the nature of the immune challenge can dictate the activation and differentiation of Tregs. For instance, the induction of type 1 regulatory T cells (Tr1 cells), which produce high levels of IL-10, has been shown to be crucial for maintaining tolerance in various settings[2].

In summary, regulatory T cells orchestrate immune tolerance through a combination of inhibitory cytokine production, cell-contact-dependent mechanisms, metabolic modulation, and environmental influences. Their multifaceted roles underscore their importance in maintaining immune homeostasis and preventing pathological immune responses[1][10].

2.2 Treg Subsets and Their Functional Diversity

Regulatory T cells (Tregs) are a specialized subset of CD4+ T lymphocytes that play a crucial role in maintaining immune tolerance and preventing autoimmune diseases. They exert their immunosuppressive functions through various mechanisms, contributing significantly to the regulation of immune responses. Tregs are characterized by the expression of the transcription factor Foxp3, which is essential for their development and function.

One of the primary roles of Tregs is to establish and maintain self-tolerance, which is vital for preventing the immune system from attacking the body’s own tissues. They achieve this through multiple mechanisms, including the secretion of anti-inflammatory cytokines such as interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β), which inhibit the activation and proliferation of effector T cells. Additionally, Tregs can engage in direct cell-to-cell interactions with other immune cells, modulating their activity and promoting a state of immune quiescence [11].

Tregs are not a homogeneous population; they can be classified into different subsets based on their origin and functional characteristics. Naturally occurring Tregs (nTregs) arise in the thymus and are critical for maintaining peripheral tolerance. Induced Tregs (iTregs), on the other hand, are generated in the periphery from naive T cells in response to specific signals, such as those from antigen-presenting cells (APCs) in the presence of certain cytokines [12]. This functional diversity allows Tregs to adapt to various immune challenges and maintain homeostasis across different tissue environments.

The importance of Tregs in immune tolerance is further highlighted by their involvement in pregnancy, where they help establish maternal tolerance to fetal alloantigens. Elevated levels of pregnancy-related hormones promote the recruitment and expansion of Tregs, underscoring their pivotal role in this unique immunological context [13]. Conversely, defects in Treg function or number are associated with several reproductive diseases, including recurrent spontaneous abortion and pre-eclampsia, indicating that a balanced Treg response is crucial for successful pregnancy [13].

Moreover, Tregs are implicated in the pathogenesis of autoimmune diseases, where their dysfunction can lead to the breakdown of self-tolerance and the activation of autoreactive immune responses [14]. Research has shown that enhancing Treg function or increasing their numbers can restore immune tolerance and ameliorate autoimmune conditions. Strategies such as low-dose IL-2 therapy have been explored to promote Treg expansion and improve their regulatory capabilities [14].

In addition to their role in autoimmunity, Tregs also play a significant part in tissue repair and regeneration. They have been shown to contribute to wound healing and the maintenance of tissue integrity across various organs, further illustrating their multifaceted role in the immune system [15]. This dual function of Tregs—suppressing immune responses while facilitating tissue repair—highlights their importance in maintaining overall immune homeostasis.

In summary, regulatory T cells are essential for controlling immune tolerance through their diverse functional roles and subsets. Their ability to suppress effector T cell responses, modulate immune cell interactions, and promote tissue homeostasis underscores their critical position in both preventing autoimmunity and fostering recovery from tissue damage. Understanding the mechanisms by which Tregs operate provides valuable insights into potential therapeutic strategies for a range of immune-related disorders.

3 Factors Influencing Treg Development and Function

3.1 Cytokine Environment

Regulatory T cells (Tregs) are essential in maintaining immune tolerance and homeostasis, primarily through their interaction with the cytokine environment. The development, function, and stability of Tregs are significantly influenced by various cytokines, which can either promote or inhibit their activity.

One of the key mechanisms by which Tregs enforce immune tolerance is through the action of anti-inflammatory cytokines. These cytokines exert direct inhibitory effects on immune cells, thereby enhancing Treg-mediated suppression. For instance, interleukin-2 (IL-2) is recognized as the most potent cytokine for promoting the development and survival of Tregs. This cytokine not only aids in the differentiation of Tregs but also supports their functional capabilities in maintaining tolerance to self-antigens [16].

Additionally, Tregs can be induced through specific cytokines that promote their differentiation from naive T cells. Cytokines such as IL-35, which is primarily produced by Tregs themselves, also play a role in modulating Treg biology, although its precise biological functions remain to be fully elucidated [16]. Moreover, the presence of cytokines like transforming growth factor-beta (TGF-β) is crucial for Treg differentiation and function, enhancing their immunosuppressive capabilities [17].

Conversely, pro-inflammatory cytokines such as IL-6 and tumor necrosis factor-alpha (TNF-α) can undermine Treg suppressive functions, potentially leading to a breakdown in immune tolerance. These cytokines can drive Tregs towards a more effector-like phenotype, thus compromising their ability to maintain tolerance [17].

The environmental context, including the presence of microbial metabolites, also influences Treg function. For example, metabolites derived from the gut microbiota, such as short-chain fatty acids, have been shown to promote Treg differentiation and enhance their anti-inflammatory responses [6]. This interplay between Tregs and their surrounding cytokine milieu underscores the complexity of immune regulation, as Tregs must integrate signals from both innate and adaptive immune cells to effectively control immune responses.

In summary, the cytokine environment is a critical determinant of Treg development and function. Anti-inflammatory cytokines foster Treg differentiation and survival, while pro-inflammatory cytokines can compromise their suppressive capabilities. Understanding these dynamics is essential for elucidating the mechanisms underlying immune tolerance and for developing therapeutic strategies aimed at modulating Treg function in various diseases, including autoimmune disorders and cancer [6][16][17].

3.2 Microbiota Influence

Regulatory T cells (Tregs) play a critical role in maintaining immune tolerance and homeostasis by modulating immune responses. Their development and function are influenced by a variety of factors, including cytokines, metabolites, and notably, the gut microbiota. The interaction between gut microbiota and Tregs is particularly significant in understanding how immune tolerance is achieved.

The gut microbiota contributes to the enhancement of Treg immunosuppressive capacity through several mechanisms. Firstly, it has been observed that gut microbiota can promote the induction of Tregs, which are essential for maintaining immune tolerance in the gut environment. This is facilitated by the upregulation of specific transcription factors characteristic of Tregs, such as Foxp3, which is crucial for their development and function. Additionally, gut microbiota metabolites, including short-chain fatty acids (SCFAs), have been identified as key players in this process. These metabolites can influence Treg differentiation and function by enhancing their secretion of anti-inflammatory cytokines, thereby ameliorating inflammatory conditions associated with autoimmune diseases like rheumatoid arthritis (RA) [18].

Moreover, the relationship between Tregs and the intestinal epithelial cells is bidirectional, where Tregs not only help maintain the integrity of the intestinal barrier but also receive signals from the gut epithelium that influence their activity. This interaction is vital for immune tolerance, as it ensures that the immune system can differentiate between harmless antigens from the microbiota and potentially harmful pathogens [19].

The gut microbiota also plays a role in shaping the overall immunological landscape by affecting the environmental stimuli that Tregs encounter. These environmental factors are crucial for Treg stability and function, as Tregs must adapt to various conditions in the gut to effectively suppress unwanted immune responses [20]. Dysbiosis, or an imbalance in the gut microbiota, can lead to impaired Treg function, contributing to the pathogenesis of various inflammatory and autoimmune disorders [21].

In summary, the gut microbiota significantly influences Treg development and function through mechanisms that promote their differentiation, enhance their immunosuppressive capabilities, and maintain intestinal homeostasis. Understanding these interactions is essential for developing therapeutic strategies aimed at modulating Treg activity in autoimmune diseases and other conditions characterized by immune dysregulation.

4 Tregs in Health and Disease

4.1 Tregs in Autoimmunity

Regulatory T cells (Tregs) are a specialized subset of CD4+ T cells that play a crucial role in maintaining immune tolerance and homeostasis. They are characterized by the expression of forkhead box protein P3 (Foxp3), which is essential for their development and function. Tregs are pivotal in preventing excessive immune responses, thus protecting against autoimmunity and ensuring the body's ability to tolerate self-antigens.

Tregs exert their immunosuppressive effects through various mechanisms, including direct suppression of effector T cells (Teff) and modulation of antigen-presenting cells (APCs). By interacting with these immune cells, Tregs can inhibit the activation and proliferation of Teff, thereby reducing inflammatory responses and promoting tolerance to self-antigens. The dysfunction or deficiency of Tregs is implicated in the pathogenesis of several autoimmune diseases, where the failure to maintain tolerance leads to the immune system attacking the body's own tissues [22][23][24].

In the context of autoimmunity, Tregs can lose their suppressive capabilities, resulting in an imbalance between protective and pathogenic immune responses. For instance, autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) are associated with reduced Treg numbers and impaired function. This deficiency in Treg activity can lead to the activation of autoreactive T cells and the subsequent destruction of self-tissues [25][26].

Recent studies have highlighted the importance of Treg plasticity and heterogeneity in their function. Tregs can adapt their suppressive mechanisms based on the inflammatory environment, which is crucial for their role in different pathological contexts. For example, Tregs can be categorized into thymus-derived Tregs (tTregs) and peripheral Tregs (pTregs), each exhibiting distinct characteristics and functions. This diversity allows Tregs to respond effectively to various immune challenges while maintaining self-tolerance [27][28].

The therapeutic potential of Tregs in autoimmune diseases is being actively explored. Strategies aimed at enhancing Treg function or increasing their numbers have shown promise in preclinical and clinical settings. Approaches include the use of immunomodulatory drugs, antigen-specific Treg therapies, and cell-based therapies such as chimeric antigen receptor (CAR) Tregs. These therapies aim to restore immune homeostasis and promote tolerance, thereby alleviating the symptoms of autoimmune diseases [29][30].

In conclusion, Tregs are integral to the regulation of immune tolerance. Their ability to suppress effector immune responses and maintain homeostasis is vital for preventing autoimmunity. The ongoing research into Treg biology and therapeutic manipulation offers hope for new treatment strategies for autoimmune disorders, highlighting the critical balance they maintain in the immune system.

4.2 Tregs in Cancer Immunology

Regulatory T cells (Tregs) play a crucial role in maintaining immune tolerance, which is essential for preventing autoimmune diseases and ensuring proper immune responses to various physiological and pathological conditions, including cancer. Tregs are characterized by their ability to suppress immune responses, thus facilitating self-tolerance and modulating the immune system's activity.

In the context of health, Tregs are vital for preventing the immune system from attacking self-antigens. They achieve this by producing immunosuppressive cytokines, such as interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β), which help maintain immune homeostasis. The balance between effector T cells and Tregs is crucial for immune stability, as an overactive immune response can lead to tissue damage and autoimmune disorders, while insufficient Treg activity can result in unchecked immune responses against pathogens or tumors [4][31].

During pregnancy, for instance, Tregs are essential for fostering maternal-fetal tolerance. Low numbers of Tregs have been associated with complications such as pre-eclampsia and pregnancy failure, indicating their protective role in maintaining a tolerant immune environment [4]. This highlights the importance of Tregs in not only managing immune responses but also in supporting unique physiological states.

In cancer immunology, the role of Tregs becomes more complex. Tumors can exploit Tregs to create an immunosuppressive microenvironment that facilitates tumor growth and metastasis. Tregs can infiltrate tumor sites and suppress the anti-tumor immune response, thus allowing cancer cells to evade immune surveillance [32][33]. This immunosuppressive activity is often correlated with poor prognosis in cancer patients, as Treg density and location within tumors can serve as independent prognostic factors [34].

Recent research has shown that Tregs in the tumor microenvironment can inhibit effector T cell functions through various mechanisms, including the production of inhibitory cytokines and the expression of immune checkpoint molecules [33][35]. Consequently, targeting Tregs has emerged as a potential strategy in cancer immunotherapy, with the aim of enhancing anti-tumor immunity while managing the risks of autoimmunity [36].

Overall, Tregs are pivotal in regulating immune tolerance in both health and disease contexts. Their dual role as suppressors of autoimmunity and facilitators of tumor immune evasion underscores the complexity of immune regulation, highlighting the need for tailored therapeutic approaches that consider the context-dependent functions of Tregs [31][33].

4.3 Tregs in Transplantation

Regulatory T cells (Tregs), primarily characterized by the expression of the transcription factor forkhead box protein P3 (Foxp3), play a pivotal role in maintaining immune tolerance and homeostasis. These cells are essential for preventing excessive immune reactions that can lead to autoimmunity, as well as for controlling immune responses to non-self antigens, such as those encountered during transplantation.

Tregs function through various mechanisms, including cytokine secretion, metabolic regulation, and direct cell-to-cell interactions. They are instrumental in the induction of peripheral tolerance, which is crucial for preventing autoimmunity by protecting self-reactive lymphocytes from activation. This self-tolerance is particularly important in the context of transplantation, where the immune system must distinguish between self and non-self tissues to avoid graft rejection [22].

In transplantation, Tregs help establish and maintain tolerance to transplanted organs by modulating the immune response. They suppress the activation of effector T cells and other immune cells that could target the graft, thereby promoting graft acceptance. Experimental and clinical studies have shown that enhancing Treg function or increasing their numbers can lead to improved outcomes in transplant settings. For instance, strategies such as the use of histone deacetylase (HDAC) inhibitors to enhance Foxp3 expression in Tregs have been proposed to promote transplant tolerance [37].

The therapeutic potential of Tregs in transplantation is underscored by their ability to curb the immune response against the graft while preserving the overall immune function. This balance is critical; while excessive Treg activity can lead to insufficient immune responses against pathogens and tumors, a deficiency can result in graft rejection and autoimmunity [38]. Therefore, the manipulation of Treg populations is being explored as a strategy to achieve long-term tolerance in transplant recipients without the need for chronic immunosuppression, which is often associated with significant side effects [39].

Moreover, Tregs have been shown to be involved in the resolution of inflammation and tissue repair, further highlighting their importance in maintaining immune balance [40]. In the context of autoimmune diseases, the dysregulation of Treg function can lead to a breakdown of tolerance, contributing to disease pathology. Therefore, therapeutic strategies aimed at restoring Treg function or enhancing their numbers are being investigated as potential treatments for autoimmune conditions [24].

In summary, Tregs are central to the maintenance of immune tolerance through their immunosuppressive actions and regulatory mechanisms. Their role in transplantation is particularly crucial, as they help prevent graft rejection while maintaining a functional immune system. The ongoing research into Treg biology and their therapeutic applications holds promise for improving outcomes in both transplantation and autoimmune diseases [41].

5 Therapeutic Targeting of Tregs

5.1 Enhancing Treg Function for Autoimmune Diseases

Regulatory T cells (Tregs) are a specialized subset of CD4+ T cells characterized by their expression of the transcription factor forkhead box protein P3 (Foxp3). They play a crucial role in maintaining immune tolerance and homeostasis, thereby preventing autoimmune diseases and excessive immune responses. Tregs exert their immunosuppressive effects through various mechanisms, including cytokine secretion, metabolic control, and direct cell-to-cell contact inhibition. This multi-faceted approach enables Tregs to modulate both innate and adaptive immune responses, effectively regulating the activity of other immune cells such as effector T cells, B cells, and antigen-presenting cells[11][22][24].

The therapeutic targeting of Tregs has emerged as a promising strategy for treating autoimmune diseases. This involves enhancing Treg function or number to restore immune balance. Various approaches have been developed to promote Treg activity, including the use of low-dose interleukin-2 (IL-2) therapy, adoptive Treg cell transfer, and engineering Tregs with chimeric antigen receptors (CAR-Tregs) to enhance their specificity and function against autoantigens[29][42]. For instance, CAR-Tregs have shown potential in redirecting Tregs to specifically target and suppress pathogenic T cells in autoimmune conditions, thus providing a more precise therapeutic option[29].

Moreover, strategies to enhance Treg function include the administration of tolerogenic dendritic cells, which can promote the expansion of antigen-specific Tregs, and the use of antigen-specific therapies that directly induce Tregs to improve their functionality[24][28]. Such targeted approaches aim to overcome the challenges posed by Treg dysfunction, which is often characterized by a reduced frequency and impaired function of Tregs in autoimmune diseases[30].

Despite the promise of Treg-based therapies, several challenges remain. These include ensuring the stability and functional integrity of engineered Tregs, achieving tissue-specific targeting, and minimizing off-target effects[11]. Furthermore, the complexity of Treg biology, including the heterogeneity of Treg subpopulations and their variable immunosuppressive capacities, complicates the development of effective therapies[28].

In summary, Tregs play a vital role in controlling immune tolerance through diverse mechanisms. Therapeutic targeting of Tregs, particularly in the context of autoimmune diseases, holds significant potential. However, continued research is necessary to address the challenges associated with enhancing Treg function and to develop effective, personalized therapeutic strategies that can improve outcomes for patients with autoimmune conditions.

5.2 Depleting Tregs in Cancer Therapy

Regulatory T cells (Tregs) are a specialized subset of CD4+ T cells that play a pivotal role in maintaining immune tolerance and homeostasis within the immune system. Characterized by the expression of forkhead box protein P3 (Foxp3), Tregs are essential for preventing excessive immune responses, thereby avoiding autoimmunity and promoting self-tolerance. They achieve this through several mechanisms, including the secretion of immunosuppressive cytokines, metabolic regulation, and direct cell-cell interactions, which help to restrain the activation and proliferation of effector T cells (Teffs) and other immune cells.

In the context of cancer, Tregs can be particularly problematic. Tumor cells often exploit Tregs to evade immune surveillance, creating an immunosuppressive microenvironment that allows for tumor growth and metastasis. The abundance of Tregs within tumors is associated with poor prognosis and treatment resistance, as they inhibit the function of cytotoxic T lymphocytes (CTLs) and other effector immune cells that are critical for anti-tumor responses[11][43].

Given their dual role in maintaining tolerance and suppressing anti-tumor immunity, Tregs have become an attractive target for therapeutic interventions in cancer. Strategies aimed at depleting or inhibiting Tregs are being explored to enhance anti-tumor immune responses. For instance, the depletion of Tregs has been shown to improve the efficacy of immune checkpoint inhibitors (ICIs) by reducing the immunosuppressive environment within tumors[44][45].

However, targeting Tregs presents several challenges. While the depletion of Tregs can enhance anti-tumor immunity, it also carries the risk of inducing autoimmunity due to the loss of immune tolerance[46]. Therefore, precision targeting strategies that selectively inhibit tumor-infiltrating Tregs while sparing peripheral Tregs are crucial to mitigate the risk of adverse effects. Such strategies may include the use of monoclonal antibodies against specific Treg markers or the application of small molecules that can selectively modulate Treg function without completely depleting them[47][48].

Recent advances in understanding the molecular mechanisms governing Treg differentiation and function within the tumor microenvironment have opened new avenues for therapeutic targeting. For example, approaches that focus on reprogramming Tregs from an immunosuppressive to an immunostimulatory phenotype could potentially enhance the effectiveness of cancer immunotherapies[49][50]. By selectively altering the function of Tregs rather than completely depleting them, it may be possible to strike a balance that promotes anti-tumor immunity while preserving necessary immune tolerance[45].

In summary, Tregs play a critical role in controlling immune tolerance through various immunosuppressive mechanisms. In cancer therapy, depleting Tregs represents a promising strategy to enhance anti-tumor responses, but it must be approached with caution to avoid triggering autoimmune reactions. Future therapeutic strategies will likely focus on precision targeting and functional reprogramming of Tregs to optimize cancer treatment outcomes.

6 Future Directions in Treg Research

6.1 Novel Biomarkers for Treg Identification

Regulatory T cells (Tregs) play a pivotal role in maintaining immune tolerance, which is crucial for preventing autoimmune diseases and ensuring the body does not mount an inappropriate response against self-antigens. The mechanisms through which Tregs exert their regulatory functions are diverse and multifaceted, involving both direct and indirect pathways.

One of the primary mechanisms of Treg-mediated immune tolerance is the secretion of immunosuppressive cytokines such as IL-10, TGF-β, and IL-35. These cytokines help create an immunosuppressive environment that inhibits the activation and proliferation of effector T cells, thereby preventing autoimmunity and maintaining homeostasis [1].

Tregs can also induce tolerance through cell-contact-dependent mechanisms. For instance, the expression of the transcription factor Foxp3 is a hallmark of Tregs, and this factor is essential for their suppressive functions. Foxp3+ Tregs can suppress other immune cells by direct interaction, which may involve the modulation of dendritic cells and other antigen-presenting cells, thereby influencing the overall immune response [3].

In addition to their role in preventing autoimmunity, Tregs are crucial during pregnancy, where they help establish and maintain tolerance towards the fetus, which expresses paternal antigens. A deficiency in Tregs during pregnancy is associated with complications such as pre-eclampsia and pregnancy loss [4]. This highlights the importance of Tregs not only in peripheral tolerance but also in specific physiological contexts.

As for future directions in Treg research, there is a growing interest in identifying novel biomarkers that can help characterize and isolate Tregs more effectively. The identification of specific surface markers or gene expression profiles associated with Tregs could enhance our understanding of their functional status and aid in therapeutic applications [51]. Current research is exploring various potential biomarkers, including CD25 and CTLA-4, which are already known to be associated with Treg activity [52].

In summary, Tregs control immune tolerance through various mechanisms, including cytokine secretion and direct cell interactions, and their role is critical in diverse biological contexts such as autoimmunity and pregnancy. Future research focused on novel biomarkers for Treg identification will likely provide deeper insights into their biology and therapeutic potential in immune-mediated diseases.

6.2 Potential for Gene Therapy

Regulatory T cells (Tregs) are a specialized subset of CD4⁺ T cells that play a critical role in maintaining immune tolerance and homeostasis within the immune system. They achieve this through a variety of mechanisms, including cytokine secretion, metabolic regulation, and direct cell-to-cell interactions, which collectively suppress excessive immune responses and prevent autoimmunity [11]. Tregs are essential for the induction and maintenance of self-tolerance, which is vital for preventing the immune system from attacking the body's own tissues [53].

One of the key factors in Treg function is the transcription factor Forkhead box protein P3 (Foxp3), which is crucial for their immunosuppressive capabilities [11]. Tregs utilize multiple strategies to exert their suppressive effects, including the production of anti-inflammatory cytokines like IL-10 and TGF-β, which can inhibit the activation and proliferation of effector T cells [24]. Additionally, Tregs can modulate the activity of dendritic cells and other antigen-presenting cells, further enhancing their ability to maintain tolerance [54].

In terms of future directions in Treg research, there is a growing interest in harnessing the therapeutic potential of Tregs through gene modification. Recent advancements in gene therapy techniques have opened new avenues for enhancing Treg function and specificity. For instance, gene-modified Tregs could be engineered to express high-affinity T cell receptors (TCRs) or chimeric antigen receptors (CARs) that target specific antigens associated with autoimmune diseases or cancer [55]. This approach aims to increase the efficacy of Tregs in suppressing unwanted immune responses while minimizing off-target effects [41].

Moreover, the development of gene-modified Tregs presents opportunities to overcome challenges associated with their use in clinical settings. For example, issues such as Treg heterogeneity, poor persistence, and functional impairment in disease states can potentially be addressed through targeted genetic modifications [56]. By enhancing the antigen specificity and suppressive functions of Tregs, researchers hope to create more effective therapies for conditions like autoimmune diseases, transplant rejection, and cancer [11].

Overall, the future of Treg research is promising, particularly with the integration of gene therapy approaches that could significantly enhance the clinical applicability of Treg-based therapies. As researchers continue to unravel the complexities of Treg biology and develop innovative strategies for their manipulation, the potential for using Tregs as a therapeutic tool in various immune-mediated diseases becomes increasingly feasible.

7 Conclusion

This review highlights the essential role of regulatory T cells (Tregs) in maintaining immune tolerance and preventing autoimmunity. The multifaceted functions of Tregs, including their mechanisms of suppression through cytokine production, cell-contact interactions, and metabolic modulation, underscore their importance in immune regulation. The evaluation of current research indicates that Tregs are not only pivotal in autoimmunity but also play significant roles in cancer immunology and transplantation. As our understanding of Treg biology evolves, future research directions should focus on identifying novel biomarkers for Treg subsets, exploring gene therapy approaches to enhance Treg function, and developing precise therapeutic strategies that leverage Treg capabilities in various clinical contexts. By harnessing the potential of Tregs, we can pave the way for innovative treatments that improve outcomes in autoimmune diseases, cancer, and transplant medicine.

References

  • [1] Peter Georgiev;Mehdi Benamar;SeongJun Han;Marcia C Haigis;Arlene H Sharpe;Talal A Chatila. Regulatory T cells in dominant immunologic tolerance.. The Journal of allergy and clinical immunology(IF=11.2). 2024. PMID:37778472. DOI: 10.1016/j.jaci.2023.09.025.
  • [2] Hanyu Zeng;Rong Zhang;Boquan Jin;Lihua Chen. Type 1 regulatory T cells: a new mechanism of peripheral immune tolerance.. Cellular & molecular immunology(IF=19.8). 2015. PMID:26051475. DOI: 10.1038/cmi.2015.44.
  • [3] Shimon Sakaguchi;Kajsa Wing;Yasushi Onishi;Paz Prieto-Martin;Tomoyuki Yamaguchi. Regulatory T cells: how do they suppress immune responses?. International immunology(IF=3.2). 2009. PMID:19737784. DOI: 10.1093/intimm/dxp095.
  • [4] Nanna Jørgensen;Gry Persson;Thomas Vauvert F Hviid. The Tolerogenic Function of Regulatory T Cells in Pregnancy and Cancer.. Frontiers in immunology(IF=5.9). 2019. PMID:31134056. DOI: 10.3389/fimmu.2019.00911.
  • [5] Y Zhai;J W Kupiec-Weglinski. What is the role of regulatory T cells in transplantation tolerance?. Current opinion in immunology(IF=5.8). 1999. PMID:10508706. DOI: 10.1016/s0952-7915(99)00007-2.
  • [6] Madeline Edwards;Leonie Brockmann. Microbiota-dependent modulation of intestinal anti-inflammatory CD4+ T cell responses.. Seminars in immunopathology(IF=9.2). 2025. PMID:40167791. DOI: 10.1007/s00281-025-01049-6.
  • [7] G Rizzuto;J F Brooks;S T Tuomivaara;T I McIntyre;S Ma;D Rideaux;J Zikherman;S J Fisher;A Erlebacher. Establishment of fetomaternal tolerance through glycan-mediated B cell suppression.. Nature(IF=48.5). 2022. PMID:35236989. DOI: 10.1038/s41586-022-04471-0.
  • [8] Oliver Goldmann;Obiageli Vivian Nwofor;Qian Chen;Eva Medina. Mechanisms underlying immunosuppression by regulatory cells.. Frontiers in immunology(IF=5.9). 2024. PMID:38380317. DOI: 10.3389/fimmu.2024.1328193.
  • [9] Jane H Buckner;Steven F Ziegler. Regulating the immune system: the induction of regulatory T cells in the periphery.. Arthritis research & therapy(IF=4.6). 2004. PMID:15380036. DOI: 10.1186/ar1226.
  • [10] Hervé Groux. Type 1 T-regulatory cells: their role in the control of immune responses.. Transplantation(IF=5.0). 2003. PMID:12819483. DOI: 10.1097/01.TP.0000067944.90241.BD.
  • [11] Lingling Wang;Ying Liang;Chunxia Zhao;Peijun Ma;Shulin Zeng;Dongen Ju;Minggao Zhao;Min Yu;Yun Shi. Regulatory T cells in homeostasis and disease: molecular mechanisms and therapeutic potential.. Signal transduction and targeted therapy(IF=52.7). 2025. PMID:41087343. DOI: 10.1038/s41392-025-02326-4.
  • [12] Helmut Jonuleit;Andrea Tuettenberg;Kerstin Steinbrink; . Research in practice: Regulatory T cells - targets for therapeutic approaches?. Journal der Deutschen Dermatologischen Gesellschaft = Journal of the German Society of Dermatology : JDDG(IF=3.8). 2011. PMID:21166772. DOI: 10.1111/j.1610-0387.2010.07503.x.
  • [13] Ning Huang;Hongbin Chi;Jie Qiao. Role of Regulatory T Cells in Regulating Fetal-Maternal Immune Tolerance in Healthy Pregnancies and Reproductive Diseases.. Frontiers in immunology(IF=5.9). 2020. PMID:32676072. DOI: 10.3389/fimmu.2020.01023.
  • [14] Masayuki Mizui;George C Tsokos. Targeting Regulatory T Cells to Treat Patients With Systemic Lupus Erythematosus.. Frontiers in immunology(IF=5.9). 2018. PMID:29755456. DOI: 10.3389/fimmu.2018.00786.
  • [15] Abdul Raheem;Ilyas Khan;Iqbal Ahmad;Abdul Wajid;Mohammad Y Alshahrani;Fuad M Alzahrani;Khalid J Alzahrani;Abdul Qadeer;I-Chuang Liao;Chien-Chin Chen. The emerging role of tissue regulatory T cells in tissue repair and regeneration.. Frontiers in immunology(IF=5.9). 2025. PMID:40948794. DOI: 10.3389/fimmu.2025.1640113.
  • [16] Jie Wu;Aini Xie;Wenhao Chen. Cytokine regulation of immune tolerance.. Burns & trauma(IF=9.6). 2014. PMID:27574641. DOI: 10.4103/2321-3868.124771.
  • [17] Yuan Zong;Kaihang Deng;Wai Po Chong. Regulation of Treg cells by cytokine signaling and co-stimulatory molecules.. Frontiers in immunology(IF=5.9). 2024. PMID:38807592. DOI: 10.3389/fimmu.2024.1387975.
  • [18] Xuan Xu;Jingying Zhou;Haihua Xie;Ruhan Zhang;Bowen Gu;Li Liu;Mi Liu;Xiaorong Chang. Immunomodulatory mechanisms of the gut microbiota and metabolites on regulatory T cells in rheumatoid arthritis.. Frontiers in immunology(IF=5.9). 2025. PMID:40692793. DOI: 10.3389/fimmu.2025.1610254.
  • [19] Zhiqiang Jiang;Chuan Wu. Reciprocal Interactions Between Regulatory T Cells and Intestinal Epithelial Cells.. Frontiers in immunology(IF=5.9). 2022. PMID:35860233. DOI: 10.3389/fimmu.2022.951339.
  • [20] Romy E Hoeppli;Dan Wu;Laura Cook;Megan K Levings. The environment of regulatory T cell biology: cytokines, metabolites, and the microbiome.. Frontiers in immunology(IF=5.9). 2015. PMID:25741338. DOI: 10.3389/fimmu.2015.00061.
  • [21] Amit Sharma;Garima Sharma;Sin-Hyeog Im. Gut microbiota in regulatory T cell generation and function: mechanisms and health implications.. Gut microbes(IF=11.0). 2025. PMID:40517372. DOI: 10.1080/19490976.2025.2516702.
  • [22] Fatemeh Bayati;Mahsa Mohammadi;Maryam Valadi;Saeid Jamshidi;Arron Munggela Foma;Ehsan Sharif-Paghaleh. The Therapeutic Potential of Regulatory T Cells: Challenges and Opportunities.. Frontiers in immunology(IF=5.9). 2020. PMID:33519807. DOI: 10.3389/fimmu.2020.585819.
  • [23] Ximei Zhang;Nancy Olsen;Song Guo Zheng. The progress and prospect of regulatory T cells in autoimmune diseases.. Journal of autoimmunity(IF=7.0). 2020. PMID:32305296. DOI: 10.1016/j.jaut.2020.102461.
  • [24] Peter J Eggenhuizen;Boaz H Ng;Joshua D Ooi. Treg Enhancing Therapies to Treat Autoimmune Diseases.. International journal of molecular sciences(IF=4.9). 2020. PMID:32977677. DOI: 10.3390/ijms21197015.
  • [25] Sophia Giang;Antonio La Cava. Regulatory T Cells in SLE: Biology and Use in Treatment.. Current rheumatology reports(IF=3.9). 2016. PMID:27704250. DOI: 10.1007/s11926-016-0616-6.
  • [26] Konstantia-Maria Chavele;Michael R Ehrenstein. Regulatory T-cells in systemic lupus erythematosus and rheumatoid arthritis.. FEBS letters(IF=3.0). 2011. PMID:21827750. DOI: 10.1016/j.febslet.2011.07.043.
  • [27] Oscar Palomares;Dirk Elewaut;Peter M Irving;Xavier Jaumont;Paolo Tassinari. Regulatory T cells and immunoglobulin E: A new therapeutic link for autoimmunity?. Allergy(IF=12.0). 2022. PMID:35852798. DOI: 10.1111/all.15449.
  • [28] Dionne Y Honing;Rosalie M Luiten;Tiago R Matos. Regulatory T Cell Dysfunction in Autoimmune Diseases.. International journal of molecular sciences(IF=4.9). 2024. PMID:39000278. DOI: 10.3390/ijms25137171.
  • [29] Seyedeh Ameneh Beheshti;Karim Shamsasenjan;Majid Ahmadi;Batol Abbasi. CAR Treg: A new approach in the treatment of autoimmune diseases.. International immunopharmacology(IF=4.7). 2022. PMID:34863655. DOI: 10.1016/j.intimp.2021.108409.
  • [30] Janine Schlöder;Fatemeh Shahneh;Franz-Joseph Schneider;Björn Wieschendorf. Boosting regulatory T cell function for the treatment of autoimmune diseases - That's only half the battle!. Frontiers in immunology(IF=5.9). 2022. PMID:36032121. DOI: 10.3389/fimmu.2022.973813.
  • [31] Ziyu Liu;Jiajun Zhou;Shihui Wu;Zhihong Chen;Shuhong Wu;Ling Chen;Xiao Zhu;Zesong Li. Why Treg should be the focus of cancer immunotherapy: The latest thought.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2023. PMID:37806087. DOI: 10.1016/j.biopha.2023.115142.
  • [32] Rong-Fu Wang. Immune suppression by tumor-specific CD4+ regulatory T-cells in cancer.. Seminars in cancer biology(IF=15.7). 2006. PMID:16140545. DOI: 10.1016/j.semcancer.2005.07.009.
  • [33] María Iglesias-Escudero;Noelia Arias-González;Eva Martínez-Cáceres. Regulatory cells and the effect of cancer immunotherapy.. Molecular cancer(IF=33.9). 2023. PMID:36739406. DOI: 10.1186/s12943-023-01714-0.
  • [34] Cailin Moira Wilke;Ke Wu;Ende Zhao;Guobin Wang;Weiping Zou. Prognostic significance of regulatory T cells in tumor.. International journal of cancer(IF=4.7). 2010. PMID:20473951. DOI: 10.1002/ijc.25464.
  • [35] Weiping Zou. Regulatory T cells, tumour immunity and immunotherapy.. Nature reviews. Immunology(IF=60.9). 2006. PMID:16557261. DOI: 10.1038/nri1806.
  • [36] Yan Pan;Hanqiong Zhou;Zhenqiang Sun;Yichen Zhu;Zhe Zhang;Jing Han;Yang Liu;Qiming Wang. Regulatory T cells in solid tumor immunotherapy: effect, mechanism and clinical application.. Cell death & disease(IF=9.6). 2025. PMID:40216744. DOI: 10.1038/s41419-025-07544-w.
  • [37] Ran Tao;Wayne W Hancock. Regulating regulatory T cells to achieve transplant tolerance.. Hepatobiliary & pancreatic diseases international : HBPD INT(IF=4.4). 2007. PMID:17690028. DOI: .
  • [38] Mohammad Afzal Khan. T regulatory cell mediated immunotherapy for solid organ transplantation: A clinical perspective.. Molecular medicine (Cambridge, Mass.)(IF=6.4). 2017. PMID:27878210. DOI: 10.2119/molmed.2016.00050.
  • [39] S C Juvet;A G Whatcott;A R Bushell;K J Wood. Harnessing regulatory T cells for clinical use in transplantation: the end of the beginning.. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons(IF=8.2). 2014. PMID:24592900. DOI: 10.1111/ajt.12647.
  • [40] Amit Sharma;Dipayan Rudra. Emerging Functions of Regulatory T Cells in Tissue Homeostasis.. Frontiers in immunology(IF=5.9). 2018. PMID:29887862. DOI: 10.3389/fimmu.2018.00883.
  • [41] Larissa A Pikor;Sindhu Arivazhagan;Michael Mendicino;Sarmitha Sathiamoorthy. Navigating the manufacturing, testing and regulatory complexities of regulatory T cells for adoptive cell therapy.. Frontiers in immunology(IF=5.9). 2025. PMID:40740779. DOI: 10.3389/fimmu.2025.1626085.
  • [42] Marwa Hassan;Mohamed Elzallat;Dina Mostafa Mohammed;Mahmoud Balata;Walaa H El-Maadawy. Exploiting regulatory T cells (Tregs): Cutting-edge therapy for autoimmune diseases.. International immunopharmacology(IF=4.7). 2025. PMID:40215774. DOI: 10.1016/j.intimp.2025.114624.
  • [43] Julian Swatler;Marco De Luca;Ivano Rotella;Veronica Lise;Emilia Maria Cristina Mazza;Enrico Lugli. CD4+ Regulatory T Cells in Human Cancer: Subsets, Origin, and Molecular Regulation.. Cancer immunology research(IF=8.2). 2024. PMID:38562083. DOI: 10.1158/2326-6066.CIR-23-0517.
  • [44] Reem Saleh;Eyad Elkord. Treg-mediated acquired resistance to immune checkpoint inhibitors.. Cancer letters(IF=10.1). 2019. PMID:31078738. DOI: 10.1016/j.canlet.2019.05.003.
  • [45] Diyuan Qin;Yugu Zhang;Pei Shu;Yanna Lei;Xiaoyu Li;Yongsheng Wang. Targeting tumor-infiltrating tregs for improved antitumor responses.. Frontiers in immunology(IF=5.9). 2024. PMID:38500876. DOI: 10.3389/fimmu.2024.1325946.
  • [46] Joudi Feras Khudeir;Eyad Elkord. Unmasking cancer's silent saboteurs: T regulatory cells as therapeutic targets in immunotherapy.. Expert review of clinical immunology(IF=3.7). 2025. PMID:41199577. DOI: 10.1080/1744666X.2025.2585521.
  • [47] Mahmoud Singer;Ahmed M Elsayed;Mohamed I Husseiny. Regulatory T-cells: The Face-off of the Immune Balance.. Frontiers in bioscience (Landmark edition)(IF=3.1). 2024. PMID:39614434. DOI: 10.31083/j.fbl2911377.
  • [48] Feng Shan;Ashwin Somasundaram;Tullia C Bruno;Creg J Workman;Dario A A Vignali. Therapeutic targeting of regulatory T cells in cancer.. Trends in cancer(IF=17.5). 2022. PMID:35853825. DOI: 10.1016/j.trecan.2022.06.008.
  • [49] Keywan Mortezaee. Selective targeting or reprogramming of intra-tumoral Tregs.. Medical oncology (Northwood, London, England)(IF=3.5). 2024. PMID:38341821. DOI: 10.1007/s12032-024-02300-0.
  • [50] Masoud Najafi;Bagher Farhood;Keywan Mortezaee. Contribution of regulatory T cells to cancer: A review.. Journal of cellular physiology(IF=4.0). 2019. PMID:30317612. DOI: 10.1002/jcp.27553.
  • [51] Kankana Bardhan;Theodora Anagnostou;Vassiliki A Boussiotis. The PD1:PD-L1/2 Pathway from Discovery to Clinical Implementation.. Frontiers in immunology(IF=5.9). 2016. PMID:28018338. DOI: 10.3389/fimmu.2016.00550.
  • [52] Kingston H G Mills. Regulatory T cells: friend or foe in immunity to infection?. Nature reviews. Immunology(IF=60.9). 2004. PMID:15516964. DOI: 10.1038/nri1485.
  • [53] Jeffrey A Bluestone;Eleonora Trotta;Daqi Xu. The therapeutic potential of regulatory T cells for the treatment of autoimmune disease.. Expert opinion on therapeutic targets(IF=4.4). 2015. PMID:25881491. DOI: 10.1517/14728222.2015.1037282.
  • [54] Qizhi Tang;Jeffrey A Bluestone. Regulatory T-cell physiology and application to treat autoimmunity.. Immunological reviews(IF=8.3). 2006. PMID:16903917. DOI: 10.1111/j.0105-2896.2006.00421.x.
  • [55] Nicholas A J Dawson;Jens Vent-Schmidt;Megan K Levings. Engineered Tolerance: Tailoring Development, Function, and Antigen-Specificity of Regulatory T Cells.. Frontiers in immunology(IF=5.9). 2017. PMID:29163527. DOI: 10.3389/fimmu.2017.01460.
  • [56] Panayiota Christofi;Chrysoula Pantazi;Nikoleta Psatha;Ioanna Sakellari;Evangelia Yannaki;Anastasia Papadopoulou. Promises and Pitfalls of Next-Generation Treg Adoptive Immunotherapy.. Cancers(IF=4.4). 2023. PMID:38136421. DOI: 10.3390/cancers15245877.

MaltSci Intelligent Research Services

Search for more papers on MaltSci.com

Regulatory T cells · Immune tolerance · Autoimmunity · Cytokines · Tumor immunity


© 2025 MaltSci