Skip to main content

The role of regulatory B cells in immune regulation and childhood allergic asthma

Abstract

Background

As the most common chronic disease in childhood, asthma displays a major public health problem worldwide with the incidence of those affected rising. As there is currently no cure for allergic asthma, it is mandatory to get a better understanding of the underlying molecular mechanism.

Main body

By producing IgE antibodies upon allergen contact, B cells play a pivotal role in allergic asthma. Besides that, IL-10-secreting B cell subsets, namely regulatory B cells (Bregs), are reported in mice and humans to play a role in allergic asthma. In humans, several Breg subsets with distinct phenotypic and functional properties are identified among B cells at different maturational and differentiation stages that exert anti-inflammatory functions by expressing several suppressor molecules. Emerging research has focused on the role of Bregs in allergic asthma as well as their role for future diagnostic and preventive strategies.

Conclusion

Knowledge about the exact function of human Bregs in allergic asthma is still very limited. This review aims to summarize the current knowledge on Bregs. We discuss different human Breg subsets, several ways of Breg induction as well as the mechanisms through which they exert immunoregulatory functions, and their role in (childhood) allergic asthma.

Background

Asthma is an inflammatory airway disease, affecting both children and adults, being the most common chronic disease in childhood. It is characterized by airway hyperresponsiveness, acute and chronic bronchoconstriction, airway edema, and mucus plugging [1]. There are two main forms of asthma: allergic and non-allergic asthma [2]. The World Health Organization (WHO) estimated that 262 million people were affected by asthma in 2019 [3], and with current trends rising, it is expected to reach 400 million by 2025 [4]. To this end, understanding the underlying disease mechanisms for further treatment is of great importance. Currently, there is no cure for asthma and therapy, which is focused on inhaled corticosteroids and bronchodilators, suppresses symptoms rather than changing the natural history of the disease [5]. Additional asthma therapies, also approved for children, use biologicals such as Omalizumab or Dupilumab which target IgE or asthma-associated cytokines such as IL-4 and IL-13, respectively [6]. Nevertheless, depending on the severity, asthma results in a diminished quality of life and an important economic burden on public health care systems, thus representing a major public health problem worldwide with a high socio-economic impact [7].

The immune system has several mechanisms to defend itself against viral, bacterial, fungal, and protozoal infections. For this, the mammalian immune system consists of the innate and the adaptive immune system. The latter is highly specific and relies on diverse antigen-specific receptors expressed on the surface of T- and B-lymphocytes [8]. For healthy immune regulation and to avoid excessive responses, the immune system needs to be tolerant against self and external innocuous antigens, referred to as immune tolerance. In healthy individuals, there is a subtle balance between anti-inflammatory (prevention of chronic inflammation and tissue damage) and pro-inflammatory (counter infections) responses [9]. Dysregulation can result in asthma, allergy, and autoimmune diseases [10].

The peripheral B cell compartment comprises a heterogeneous population of cells at different maturational stages along the lineage, each with distinctive functional properties [11]. They produce antibodies, cytokines, and act as antigen-presenting cells (APCs). In addition, B cells have the capability to regulate immune responses through regulatory B cells (Bregs), which are B cells with anti-inflammatory functions [12]. They exhibit their anti-inflammatory role by producing mainly anti-inflammatory cytokines, e.g., IL-10, transforming growth factor (TGF)-β, and IL-35, as well as by induction of other regulatory cells [13, 14]. In humans, Bregs are found among B-lymphocytes at different stages of maturation and differentiation, ranging from early transitional B cells to highly differentiated plasmablasts [15, 16].

Knowledge about the impact of B cells and Bregs on asthma is still rather limited. IgE production upon allergen contact is the main described role of B cells in allergic asthma, which is mandatory for the initiation of the allergic cascade [17]. Habener et al. revealed that regulatory B cells control airway hyperresponsiveness and airway remodeling in a murine asthma model, suggesting a potential role for B cells in future diagnostic and preventive strategies in asthma [18]. They also showed differences in human B cell populations between asthma and controls as well as between mild-moderate and severe asthma [19]. Moreover, Breg numbers contribute to the regulation of the immune system, which was confirmed by multiple human studies showing lower frequencies of some Breg subsets in allergic asthmatics [20,21,22,23]. This review aims to summarize the current knowledge about Bregs and their role in childhood allergic asthma to get new mechanistic insights into childhood asthma development and therapeutic and preventive strategies.

Immune mechanisms underlying childhood allergic asthma

Allergic asthma is the predominant form of asthma in childhood, which has been characterized by sensitization to specific allergens, high IgE levels, eosinophilia, a type 2 shifted immune response, and decreased innate immunity gene expression [24]. Both the innate and the adaptive immune system play a role in type 2 immune response and involve several immune cells such as T-helper (Th) 2 cells, group 2 innate lymphoid cells (ILCs), B cells, natural killer (NK) cells, NK T cells, basophils, eosinophils, and mast cells, as well as their major cytokines [25].

Repetitive exposure of children to environmental allergens such as house dust mites (HDM) results in the development of allergen-specific memory T and B cell responses following expression of inflammatory mediators to recruit immune effector cells such as T and B cells [26, 27]. Upon allergen presentation by APCs to naïve CD4+ T cells, they get activated and release type 2 cytokines such as IL-3 and IL-5, which drive the proliferation and differentiation of basophils, eosinophils, and mast cells, which in turn contribute to asthmatic bronchial hyperresponsiveness. In addition, IL-4 and IL-13 are released, which play a key role in IgE production of B cells. IgE in turn binds to high-affinity IgE surface receptors (FcεRI) on immune cells such as dendritic cells (DCs), basophils, and mast cells [28, 29]. Upon cross-linking of the IgE-FcεRI complexes by the allergen, it activates the cells resulting in the release of inflammatory mediators like histamine, leukotrienes, and the type 2 cytokines IL-4, IL-5, and IL-13. Consequently, an influx of CD4+ T cells and eosinophils into the airways maintains type 2 inflammation. To this end, higher levels of lymphocytes, eosinophils, basophils, and mast cells are found in the airways of allergic asthmatics [28, 30]. Moreover, higher levels of airway epithelial desquamation, goblet cell hyperplasia, and thicker basement membranes are characteristics of asthmatics [29]. Since sensitization to allergens plays a key role in the development of allergic asthma [31], targeting of allergen sensitization seems to be promising in allergic asthma therapy [32].

B cells

As part of the adaptive immune system, B cells play a pivotal role in the protection against pathogens. Moreover, they play important roles in several diseases such as autoimmune disorders [33, 34], cancer [35], allergy [36], and asthma [37, 38].

The peripheral B cell compartment consists of several cells at different maturational stages with different functions [11]. In humans, B cells develop in the bone marrow from hematopoietic precursors derived from the fetal liver [39]. Early B cell development includes (early and late) pro-B, pre-B, and immature B cells in which specific surface markers are expressed and are characterized by rearrangements of the immunoglobulin (Ig) heavy and light gene segments to generate diverse antigen receptors [40]. Immature B cells then exist in the bone marrow for several days until they enter the circulation as transitional B cells [41]. Depending on their surface markers and function, transitional B cells can be subdivided into T1, T2, and T3 B cells [42]. Depending on signals received through the B cell receptor (BCR) and other receptors, transitional B cells further differentiate into either follicular (FO) or marginal zone (MZ) B cells, now considered mature or naïve B cells [39, 43]. After specific antigen-recognition via BCR, selected B cells become either antibody-producing plasmablasts or plasma cells, or memory B cells [39]. Memory B cells are B cells, which circulate through the body [44]. Upon binding of the specific antigen, that originally activated the parent B cell that led to the production of the memory B cell, to the BCR, a strong and more rapid antibody response is initiated by the memory B cell [45]. In addition to the generation of immunological memory through memory B cells [45], B cells also contribute to immune responses through cytokine production [46], and antigen presentation, as well as through antibody production, which is their main contribution to allergy [47]. These multifaceted roles of B cells were also shown in several studies using allergic mouse models of asthma [48,49,50].

Regulatory B cells (Bregs)

B cells can also regulate immune responses by other mechanism than antibody production, antigen presentation, and cytokine production. In the 1970s, allergen sensitization studies described an immunosuppressive potential of B cells for the first time [51, 52]. At that time, the precise mechanisms of suppression were not identified, and for more than 20 years, the study of immunosuppressive B cells did not receive significant attention [9, 53]. However, during the past two decades, the knowledge of regulatory B cell (Breg) phenotype and function increased [12]. In 2002, Mitzoguchi et al. described interleukin 10 (IL-10) producing B cells with regulatory functions that are characterized by CD1d upregulation in murine models of intestinal inflammation [54]. Other studies in murine models also showed an essential role of IL-10-producing B cells in controlling autoimmunity [55] and the prevention of arthritis [56]. Regulatory B cells (Bregs) were not only described in mice but they were also identified in humans: In 1998, Akdis et al. described IL-10-producing B cells in humans in the context of high-allergen exposure models and allergen immunotherapy (AIT). In AIT, high doses of the allergen are administered, which significantly changed IL-10 production after 7 days of bee venom (BV)-AIT in epitope-specific T cells, but not in B cells [57]. In 2007, Goetz et al. and Dass et al. demonstrated that therapy with Rituximab, an anti-CD20-antibody that depletes B cells, leads to severe exacerbation of ulcerative colitis and new onset of psoriasis in humans. Furthermore, they showed an association with the suppression of local IL-10 production suggesting an important anti-inflammatory role of IL-10-producing B cells in humans [58, 59].

Human Breg subsets

Several murine and human disease models were used to study the phenotype of Bregs as well as the underlying molecular mechanisms of Breg-mediated immune suppression. This resulted in the identification of different Breg subsets in both mice and humans with distinct phenotypic and functional properties [9]. Until now, there is no clear consensus on the classification and definition of Bregs. Here, we discuss the major Breg subsets that were identified in humans (Table 1).

Table 1 Different subsets and functions of human Bregs

Human Bregs are found among B-lymphocytes at different stages of maturation and differentiation: Human CD27+CD24hi B10 cells produce IL-10 and thereby regulate cytokine production by monocytes after in vitro stimulation with lipopolysaccharide (LPS) and CpG, indicating a functional link between Bregs and the innate immune system. The mean frequencies of these cells are significantly increased in patients with autoimmune diseases [60]. Human CD19+CD24hiCD38hi immature transitional B cells also have regulatory functions. After stimulation with CD40, these cells produce IL-10 thereby suppressing the differentiation of naïve T cells into TH1 and TH17 cells while inducing the conversion into Tregs [15, 61]. Moreover, this Breg subset was shown to regulate T cell immunity in chronic HBV (CHB) infection [62]. Human CD19+CD25+CD71+CD73− BR1 cells are characterized by high expression of IL-10 and suppression of antigen-specific CD4+ T cell proliferation. Also, anti-inflammatory IgG4 antibody production was shown by these BR1 cells [63]. Lindner et al. described IL-21-induced granzyme B-expressing B cells (GrB+ B cells) that are found in the microenvironment of solid tumors next to IL-21-providing T cells. They are characterized by a CD19+CD38+CD1d+IgM+CD147+ expression signature and express regulatory molecules such as GrB, IL-10, CD25, and indolamine-2,3-dioxygenase (IDO). It was shown that they suppress T cell proliferation through GrB-dependent T cell receptor (TCR) degradation [65]. Another human Breg subset was described in 2017 by Brosseau et al. showing that CD9+ B cells induce effector T cell apoptosis in both mice and humans by IL-10 secretion. Moreover, they showed that these CD9+ B cells with regulatory properties are reduced in patients with severe asthma [66]. Human CD19+CD27intCD38+ plasmablasts produce IL-10 and secrete IgM. Interestingly, Matsumoto et al. showed that human CD19+CD27intCD38+ plasmablasts derived from naïve and especially immature B cells, but not human CD19+CD27hiCD38+ plasmablasts derived from memory B cells, are the major IL-10-producing B cells [16]. For human IgA+PD-L1+IL-10+ plasma cells, suppression of anti-tumor immunity was described [64]. Patients with tuberculosis were shown to have an increased number of CD5+Cd1d+ B cells with a stronger suppressive activity by inhibiting Th17 cell activation and IL-22 production [67, 68].

Due to this heterogeneity of Breg subsets, it was not possible until now to identify a Breg-specific transcription factor [9]. Since this is an emerging field of research, many questions still need to be answered with regard to plasticity, ontogeny, and the Breg-mediated suppression mechanism.

Several ways of Breg induction

The heterogeneity of Breg subsets leads to the assumption that there are either various distinct Breg lineages or that IL-10 can be induced by external stimuli at different stages of B cell development [9]. The precise mechanisms and required signals for Breg differentiation still need to be elucidated. However, several murine and human studies revealed that B cells of different maturational and differentiation stages are able to differentiate into Bregs upon antigen recognition and/or several kinds of stimuli [69,70,71]. CpG stimulation results in the generation of human CD19+CD27intCD38+ plasmablasts. Additionally treating B cells with IL-2, IL-6, and especially with the type I interferon IFN-α results in the generation of IL-10 secreting human CD19+CD27intCD38+ plasmablasts [16]. IFN-α was described to induce CD38+ expression on human naïve B cells [72] and promote differentiation into plasma cells [73]. In this regard, it is likely that IFN receptor signaling is required for IL-10-producing human plasmablasts [16]. Indeed, treatment with IFN-β, which is another type I IFN, increases IL-10 expression of human B cells after BCR and CD40 ligation [74]. Another study in mice revealed that stimulation with IL-1β, IL-6, and anti-CD40 in combination promotes the differentiation into IL-10-producing Bregs. Interestingly, B cell stimulation with TNF-α or IL-17 with or without anti-CD40 has no effect on IL-10 production [70]. It was also demonstrated that the TLR-MyD88-STAT3 pathway not only leads to antibody production of human B cells, but also regulates IL-10 production of human B cells by TLR7/8, which is enhanced by IFN-α [75]. The BATF/IRF-4/IRF-8-axis was also shown to play a role in IL-10 and IL-35 expression of murine regulatory B cells [76]. Matsumoto et al. also showed that IRF4 is required for the differentiation into IL-10-expressing murine plasmablasts, both in vitro and in vivo [16]. Moreover, IL-21 was described to induce IL-10 expression of murine B10 cells [71] and human GrB+ Bregs [65]. Also, in vitro stimulation of murine B cells with other molecules such as LPS, phorbol myristate acetate (PMA), and ionomycin generates IL-10-expressing B cells [77,78,79]. Menon et al. revealed that also plasmacytoid dendritic cells (pDCs) can drive the differentiation of human CD19+CD24hiCD38hi Bregs and plasmablasts that express IL-10, IL-6, and TNF-α [69].

As another mechanism for Breg induction, overexpression of IL-10 in primary human B cells was shown to upregulate suppressor of cytokine signaling 3 (SOCS3), glycoprotein A repetitions predominant (GARP), IL-2 receptor α chain (CD25), and programmed cell death ligand 1 (PD-L1). Moreover, IL-10 overexpressing human B cells secrete less pro-inflammatory cytokines such as TNF-α or IL-8, whereas production of anti-inflammatory IL-1 receptor antagonists (IL-1RN) and vascular endothelial growth factor (VEGF) is increased [80]. In addition, the aryl hydrocarbon receptor (AhR) was shown to regulate the differentiation and function of IL-10-producing murine Bregs [81] and A proliferation-inducing ligand (APRIL) drives the differentiation of naïve human B cells to IL-10-producing Bregs [82]. Human type 3 innate lymphoid cells (ILC3s) were also shown to play a role in the induction of human IL-10-producing immature transitional Bregs [83], whereas IL-35 was described to induce IL-35-producing human Bregs (IL-35+ Bregs) through activation of STAT1/STAT3 [84].

Breg suppressor molecules

Regulatory B cells mainly act by secreting immune-modulatory cytokines. Although research mainly focused on the role of IL-10 as the hallmark cytokine of Bregs [54,55,56, 63, 85], three main cytokines were identified to be expressed by Bregs as suppressor molecules: IL-10, TGF-β [86], and IL-35 [87, 88] (Fig. 1). The anti-inflammatory cytokine IL-10 is involved in the maintenance of homeostasis by dampening inflammatory responses and can be produced by many cell types such as monocytes, T cells, DCs, NK cells, macrophages, mast cells, and B cells. It induces immune tolerance in patients with chronic inflammatory diseases such as allergy, autoimmunity, organ transplantation, and tumor tolerance and has direct and indirect suppressive effects on cytokine production and proliferation of effector T cells [89]. Moreover, IL-10 has an effect on B cells, including B cell survival, class-switch recombination, and plasma cell differentiation [25, 89]. IL-10 is also thought to have a positive effect on asthma pathophysiology by suppressing the IgE-mediated allergic cascade and decreasing airway inflammation [37]. Production of IL-10 was shown to be negatively regulated by B cell lymphoma-3 (Bcl-3) [90, 91], which is an atypical member of the inhibitor of NF-κB (IκB) protein family and regulates NF-κB-mediated gene expression, thereby regulating TLR signaling [92, 93]. TGF-β is an anti-inflammatory cytokine that has an important role in immune regulation, wound healing, and tissue remodeling and is involved in the conversion of naïve CD4+ T cells into functional regulatory T cells (Tregs) [25, 89]. IL-35 is primarily produced by Tregs and induces Treg proliferation while suppressing TH17 and TH1 responses [25, 84].

Fig. 1
figure 1

Breg suppressor molecules and their effect on other immune cells. Bregs mainly secrete the anti-inflammatory cytokines IL-10, IL-35, and TGF-β that in turn have a suppressive function on TH1 and TH17 cells. In addition, the conversion of CD4+ T cells into Tregs is induced. The transmembrane protein PD-L1 regulates cell expansion and differentiation of TFH cells. Also, other surface-bound molecules such as MHC-II, CD19, CD73, CD39, FasL, and TIM1 as well as AhR and GrB are described to play a role in Breg-mediated immunosuppressive effects. IL, interleukin; TGF-β, transforming growth factor β; TH, T helper cell; Treg, regulatory T cell; PD-L1, programmed cell death ligand 1; TFH, follicular helper T cells, MHC-II, major histocompatibility complex II; FasL, Fas ligand; TIM1, T cell immunoglobulin and mucin domain 1; AhR, aryl hydrocarbon receptor; GrB, granzyme B

In addition to the expression of these anti-inflammatory cytokines, other anti-inflammatory molecules such as PD-L1 are associated with suppressive effects of Bregs. PD-L1, also known as CD274, is a transmembrane protein that regulates induced Treg cell function, development, and maintenance [94]. Bregs expressing PD-L1, described as PD-L1hi B cells, regulate cell expansion and differentiation of follicular helper T cells (TFH), thereby suppressing autoimmune disease [95]. Also other surface-bound molecules like the Fas ligand (FasL) [96], CD39 [97], CD73 [97], CD19 [78], major histocompatibility complex II (MHC-II) [71], T cell immunoglobulin, and mucin domain 1 (TIM1) [98] as well as GrB [65], AhR [99], and intracellular signaling molecules such as STAT3 and MyD88 [100] are described to play a role in Breg-mediated immunosuppressive effects. This shows that there is a huge variety of suppressor molecules that are expressed by Bregs to fulfill their immunosuppressive function (Fig. 1).

Role of Bregs in (childhood) allergic asthma

Recent research has concentrated on the role of Bregs in allergic asthma [26]. As mentioned above, Bregs express the anti-inflammatory cytokine IL-10, which is thought to have a positive effect on asthma pathophysiology by suppressing the IgE-mediated allergic cascade and decreasing airway inflammation [37]. Parasite infection in allergic mice models for asthma reveals protection from lung inflammation and airway hyperresponsiveness through IL-10-producing Bregs [37, 85]. Moreover, in human studies a higher number of allergen-specific, IL-10-producing Bregs is found upon allergen immunotherapies for cow milk and bee venom, indicating that higher Breg numbers are characteristic for induced tolerance to allergens [63, 101]. Braza et al. showed that upon allergen exposure the number of IL-10-producing Bregs is decreased in the lungs of asthmatic mice, indicating that the homeostasis of Bregs is altered by asthma [102]. Recently, Qian et al. showed that asthmatic patients have higher levels of IL-10 but lower levels of Bcl-3, suggesting that they have an important role in asthma pathogenesis. They also revealed that mice lacking Bcl-3 have increased eosinophilic airway inflammation, augmented airway goblet cell hyperplasia, elevated airway hyperresponsiveness, and increased levels of epithelial chemokines in the lungs after stimulation with HDM compared to control mice. These data demonstrate that Bcl-3 limits the IL-10 expression during allergic sensitization, thereby preventing lung inflammation and asthma pathogenesis in HDM-induced mice [103]. Since it was already shown that Bcl-3 is a negative regulator for IL-10 [91], these results indicate that Bcl-3 is a critical inhibitor of IL-10 in allergic asthma and targeting the Bcl-3/IL-10 axis may be a promising approach for allergic asthma therapy [103].

In adults, patients with allergic asthma show a lower percentage and absolute number of CD19+CD24hiCD27+ circulating Bregs [20, 21]. Also, these Bregs express less IL-10 upon LPS [104], but not CpG [105] stimulation. Moreover, frequencies of CD5+ and Cd1d+CD5+ Bregs are decreased in adult allergic asthmatics [22]. Recently, Sheehan et al. revealed that pediatric patients with allergic asthma also have significantly lower levels of circulating IL-10 expressing CD24+CD38+ Bregs, compared to the healthy control group. This could indicate that the lower Breg levels are associated with suboptimal control of allergic inflammation leading to asthma development or excess morbidity from asthma. To this end, it seems that Bregs are important for both children and adults with allergic asthma and that the appropriate number and function of Bregs may be mandatory to control asthma by releasing suppressive signals to decrease TH2 inflammation [23].

Conclusion

During the past two decades, several Breg subsets were described in mice and humans, each with distinct phenotypical and functional characteristics. However, in contrast to regulatory T cells, which can be clearly defined by the expression of the transcription factor Foxp3, the identification of specific markers or transcription factors that clearly define Bregs is still missing. In addition, the precise mechanism and required signals for Breg differentiation are not completely understood. The way by which Bregs mainly exert their immunosuppressive function is through anti-inflammatory cytokines such as IL-10, TGF-β, and IL-35. Other anti-inflammatory molecules such as PD-L1, FasL, or intracellular signaling molecules like STAT3 and MyD88 are also associated with Breg function; however, this varies between different human Breg subsets. Several studies showed that IL-10 expression has a positive effect on asthma pathophysiology and that higher Breg numbers are characteristic of allergen tolerance. Moreover, it has been shown that asthma alters the homeostasis of Bregs: in adult as well as pediatric patients with allergic asthma, a lower percentage and absolute number of several IL-10 expressing Breg subsets are found. This suggests that lower Breg levels are associated with suboptimal control of allergic inflammation, which in turn may lead to the development of asthma. Since this is an emerging field of research, many questions still need to be answered with regard to plasticity, ontogeny, and Breg-mediated suppression mechanism as well as their exact role especially in childhood allergic asthma.

Availability of data and materials

Not applicable.

Abbreviations

AA:

Allergic asthma

AhR:

Aryl hydrocarbon receptor

AIT:

Allergen immunotherapy

APC:

Antigen-presenting cell

APRIL:

A proliferation-inducing ligand

BATF:

Basic leucine zipper transcription factor

Bcl-3:

B cell lymphoma 3

BCR:

B cell receptor

Breg:

Regulatory B cell

CD:

Cluster of differentiation

CHB:

Chronic HBV

DC:

Dendritic cell

FasL:

Fas ligand

FcεRI:

High-affinity IgE receptor

FO:

Follicular

GARP:

Glycoprotein A repetitions predominant

GrB:

Granzyme B

HDM:

House dust mite

IDO:

Indolamine-2,3-dioxygenase

IFN:

Interferon

Ig:

Immunoglobulin

IgE:

Immunoglobulin E

IκB:

Inhibitor of NF-κB

IL:

Interleukin

IL-1RN:

IL-1 receptor antagonist

ILC:

Innate lymphoid cell

ILC3:

Type 3 innate lymphoid cell

IRF:

Immunoregulatory factor

LPS:

Lipopolysaccharide

MHC-II:

Major histocompatibility complex II

MyD88:

Myeloid differentiation primary response 88

MZ:

Marginal zone

NF-κB:

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

NK:

Natural killer

PD-L1:

Programmed cell death ligand 1

pDCs:

Plasmacytoid dendritic cells

PMA:

Phorbol myristate acetate

SOCS3:

Suppressor of cytokine signaling 3

STAT:

Signal transducer and activator of transcription

TCR:

T cell receptor

TFH :

Follicular helper T cells

TGF-β:

Transforming growth factor β

TH :

T helper cell

TIM1:

T cell immunoglobulin and mucin domain 1

TLR:

Toll-like receptor

TNF:

Tumor necrosis factor

Treg:

Regulatory T cell

VEGF:

Vascular endothelial growth factor

WHO:

World Health Organization

References

  1. Castagnoli R, Brambilla I, Giovannini M, Marseglia GL, Licari A (2023) New approaches in childhood asthma treatment. Curr Opin Allergy Clin Immunol 23(4):319–326

    Article  PubMed  PubMed Central  Google Scholar 

  2. Fainardi V, Esposito S, Chetta A, Pisi G (2022) Asthma phenotypes and endotypes in childhood. Minerva Med 113(1):94–105

    Article  PubMed  Google Scholar 

  3. Vos T et al. (2020) Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 396(10258):1204–22

  4. Masoli M, Fabian D, Holt S, Beasley R (2004) The global burden of asthma: executive summary of the GINA Dissemination Committee report. Allergy 59(5):469–478

    Article  PubMed  Google Scholar 

  5. El-Husseini ZW, Gosens R, Dekker F, Koppelman GH (2020) The genetics of asthma and the promise of genomics-guided drug target discovery. Lancet Respir Med 8(10):1045–1056

    Article  PubMed  CAS  Google Scholar 

  6. Salvermoser M, Zeber K, Boeck A, Klucker E, Schaub B (2021) Childhood asthma: novel endotyping by cytokines, validated through sensitization profiles and clinical characteristics. Clin Exp Allergy 51(5):654–665

    Article  PubMed  CAS  Google Scholar 

  7. Bousquet J, Hellings PW, Agache I, Bedbrook A, Bachert C, Bergmann KC et al (2016) ARIA 2016: care pathways implementing emerging technologies for predictive medicine in rhinitis and asthma across the life cycle. Clin Transl Allergy 6:47

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Janeway C (2005) Immunobiology: the immune system in health and disease, 6th edn. Garland Science, New York

    Google Scholar 

  9. van de Veen W, Stanic B, Wirz OF, Jansen K, Globinska A, Akdis M (2016) Role of regulatory B cells in immune tolerance to allergens and beyond. J Allergy Clin Immunol 138(3):654–665

    Article  PubMed  Google Scholar 

  10. Akdis M, Akdis CA (2009) Therapeutic manipulation of immune tolerance in allergic disease. Nat Rev Drug Discov 8(8):645–660

    Article  PubMed  CAS  Google Scholar 

  11. Pieper K, Grimbacher B, Eibel H (2013) B-cell biology and development. J Allergy Clin Immunol 131(4):959–971

    Article  PubMed  CAS  Google Scholar 

  12. Jansen K, Cevhertas L, Ma S, Satitsuksanoa P, Akdis M, van de Veen W (2021) Regulatory B cells. A to Z Allergy 76(9):2699–2715

    Article  CAS  Google Scholar 

  13. Mauri C, Bosma A (2012) Immune regulatory function of B cells. Annu Rev Immunol 30:221–241

    Article  PubMed  CAS  Google Scholar 

  14. Mauri C, Nistala K (2014) Interleukin-35 takes the ‘B’ line. Nat Med 20(6):580–581

    Article  PubMed  CAS  Google Scholar 

  15. Blair PA, Noreña LY, Flores-Borja F, Rawlings DJ, Isenberg DA, Ehrenstein MR et al (2010) CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic lupus erythematosus patients. Immunity 32(1):129–140

    Article  PubMed  CAS  Google Scholar 

  16. Matsumoto M, Baba A, Yokota T, Nishikawa H, Ohkawa Y, Kayama H et al (2014) Interleukin-10-producing plasmablasts exert regulatory function in autoimmune inflammation. Immunity 41(6):1040–1051

    Article  PubMed  CAS  Google Scholar 

  17. Akdis CA, Arkwright PD, Brüggen M-C, Busse W, Gadina M, Guttman-Yassky E et al (2020) Type 2 immunity in the skin and lungs. Allergy 75(7):1582–1605

    Article  PubMed  CAS  Google Scholar 

  18. Habener A, Happle C, Grychtol R, Skuljec J, Busse M, Dalüge K et al (2021) Regulatory B cells control airway hyperreactivity and lung remodeling in a murine asthma model. J Allergy Clin Immunol 147(6):2281-2294.e7

    Article  PubMed  CAS  Google Scholar 

  19. Habener A, Grychtol R, Gaedcke S, DeLuca D, Dittrich AM, Happle C et al (2022) IgA+ memory B-cells are significantly increased in patients with asthma and small airway dysfunction. Eur Respir J 60(5):2102130

  20. Oliveria J-P, El-Gammal AI, Yee M, Obminski CD, Scime TX, Watson RM et al (2018) Changes in regulatory B-cell levels in bone marrow, blood, and sputum of patients with asthma following inhaled allergen challenge. J Allergy Clin Immunol 141(4):1495-1498.e9

    Article  PubMed  Google Scholar 

  21. Miyajima S, Shigehara K, Kamekura R, Takaki H, Yabe H, Ikegami I et al (2020) Activated circulating T follicular helper cells and skewing of T follicular helper 2 cells are down-regulated by treatment including an inhaled corticosteroid in patients with allergic asthma. Allergol Int 69(1):66–77

    Article  PubMed  CAS  Google Scholar 

  22. Wiest M, Upchurch K, Hasan MM, Cardenas J, Lanier B, Millard M et al (2019) Phenotypic and functional alterations of regulatory B cell subsets in adult allergic asthma patients. Clin Exp Allergy 49(9):1214–1224

    Article  PubMed  CAS  Google Scholar 

  23. Sheehan WJ, Maghzian N, Rastogi D, Bollard CM, Lin AA (2023) Decreased regulatory B cells in pediatric patients with asthma. Ann Allergy Asthma Immunol 131(1):120–121

    Article  PubMed  CAS  Google Scholar 

  24. Krusche J, Basse S, Schaub B (2020) Role of early life immune regulation in asthma development. Semin Immunopathol 42(1):29–42

    Article  PubMed  CAS  Google Scholar 

  25. Akdis M, Aab A, Altunbulakli C, Azkur K, Costa RA, Crameri R et al (2016) Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: receptors, functions, and roles in diseases. J Allergy Clin Immunol 138(4):984–1010

    Article  PubMed  CAS  Google Scholar 

  26. Oliveria JP, Agayby R, Gauvreau GM (2021) Regulatory and IgE+ B cells in allergic asthma. Methods Mol Biol 2270:375–418

    Article  PubMed  CAS  Google Scholar 

  27. Akdis CA, Akdis M (2015) Mechanisms of allergen-specific immunotherapy and immune tolerance to allergens. World Allergy Organ J 8(1):17

    Article  PubMed  PubMed Central  Google Scholar 

  28. Wills-Karp M (1999) Immunologic basis of antigen-induced airway hyperresponsiveness. Annu Rev Immunol 17:255–281

    Article  PubMed  CAS  Google Scholar 

  29. Wills-Karp M (2004) Interleukin-13 in asthma pathogenesis. Immunol Rev 202:175–190

    Article  PubMed  CAS  Google Scholar 

  30. Smith SG, Gauvreau GM (2007) IL-13 is a novel therapeutic target in allergic asthma. Expert Rev Clin Immunol 3(5):671–675

    Article  PubMed  CAS  Google Scholar 

  31. Stoltz DJ, Jackson DJ, Evans MD, Gangnon RE, Tisler CJ, Gern JE et al (2013) Specific patterns of allergic sensitization in early childhood and asthma & rhinitis risk. Clin Exp Allergy 43(2):233–241

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Moon H-G, Kim S-J, Lee MK, Kang H, Choi HS, Harijith A et al (2020) Colony-stimulating factor 1 and its receptor are new potential therapeutic targets for allergic asthma. Allergy 75(2):357–369

    Article  PubMed  CAS  Google Scholar 

  33. Matsushita T, Yanaba K, Bouaziz J-D, Fujimoto M, Tedder TF (2008) Regulatory B cells inhibit EAE initiation in mice while other B cells promote disease progression. J Clin Invest 118(10):3420–3430

    PubMed  PubMed Central  CAS  Google Scholar 

  34. Watanabe R, Ishiura N, Nakashima H, Kuwano Y, Okochi H, Tamaki K et al (2010) Regulatory B cells (B10 cells) have a suppressive role in murine lupus: CD19 and B10 cell deficiency exacerbates systemic autoimmunity. J Immunol 184(9):4801–4809

    Article  PubMed  CAS  Google Scholar 

  35. Jurlander J, Lai C-F, Tan J, Chou C-C, Geisler CH, Schriber J et al (1997) Characterization of interleukin-10 receptor expression on B-cell chronic lymphocytic leukemia cells. Blood 89(11):4146–4152

    Article  PubMed  CAS  Google Scholar 

  36. Hulse KE, Norton JE, Suh L, Zhong Q, Mahdavinia M, Simon P et al (2013) Chronic rhinosinusitis with nasal polyps is characterized by B-cell inflammation and EBV-induced protein 2 expression. J Allergy Clin Immunol 131(4):1075–83 (1083.e1-7)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Amu S, Saunders SP, Kronenberg M, Mangan NE, Atzberger A, Fallon PG (2010) Regulatory B cells prevent and reverse allergic airway inflammation via FoxP3-positive T regulatory cells in a murine model. J Allergy Clin Immunol 125(5):1114-1124.e8

    Article  PubMed  CAS  Google Scholar 

  38. Gould HJ, Sutton BJ (2008) IgE in allergy and asthma today. Nat Rev Immunol 8(3):205–217

    Article  PubMed  CAS  Google Scholar 

  39. Eibel H, Kraus H, Sic H, Kienzler A-K, Rizzi M (2014) B cell biology: an overview. Curr Allergy Asthma Rep 14(5):434

    Article  PubMed  Google Scholar 

  40. Ghia P, ten Boekel E, Rolink AG, Melchers F (1998) B-cell development: a comparison between mouse and man. Immunol Today 19(10):480–485

    Article  PubMed  CAS  Google Scholar 

  41. Chung JB, Silverman M, Monroe JG (2003) Transitional B cells: step by step towards immune competence. Trends Immunol 24(6):343–349

    Article  PubMed  CAS  Google Scholar 

  42. Palanichamy A, Barnard J, Zheng B, Owen T, Quach T, Wei C et al (2009) Novel human transitional B cell populations revealed by B cell depletion therapy. J Immunol 182(10):5982–5993

    Article  PubMed  CAS  Google Scholar 

  43. Batten M, Groom J, Cachero TG, Qian F, Schneider P, Tschopp J et al (2000) BAFF mediates survival of peripheral immature B lymphocytes. J Exp Med 192(10):1453–1466

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Seifert M, Küppers R (2016) Human memory B cells. Leukemia 30(12):2283–2292

    Article  PubMed  CAS  Google Scholar 

  45. Good KL, Avery DT, Tangye SG (2009) Resting human memory B cells are intrinsically programmed for enhanced survival and responsiveness to diverse stimuli compared to naive B cells. J Immunol 182(2):890–901

    Article  PubMed  CAS  Google Scholar 

  46. Harris DP, Haynes L, Sayles PC, Duso DK, Eaton SM, Lepak NM et al (2000) Reciprocal regulation of polarized cytokine production by effector B and T cells. Nat Immunol 1(6):475–482

    Article  PubMed  CAS  Google Scholar 

  47. Allen CDC (2022) Features of B cell responses relevant to allergic disease. J Immunol 208(2):257–266

    Article  PubMed  CAS  Google Scholar 

  48. Wypych TP, Marzi R, Wu GF, Lanzavecchia A, Sallusto F (2018) Role of B cells in TH cell responses in a mouse model of asthma. J Allergy Clin Immunol 141(4):1395–1410

    Article  PubMed  CAS  Google Scholar 

  49. Dullaers M, Schuijs MJ, Willart M, Fierens K, van Moorleghem J, Hammad H et al (2017) House dust mite-driven asthma and allergen-specific T cells depend on B cells when the amount of inhaled allergen is limiting. J Allergy Clin Immunol 140(1):76-88.e7

    Article  PubMed  CAS  Google Scholar 

  50. Korsgren M, Erjefält JS, Korsgren O, Sundler F, Persson CG (1997) Allergic eosinophil-rich inflammation develops in lungs and airways of B cell-deficient mice. J Exp Med 185(5):885–892

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Katz SI, Parker D, Turk JL (1974) B-cell suppression of delayed hypersensitivity reactions. Nature 251(5475):550–551

    Article  PubMed  CAS  Google Scholar 

  52. Neta R, Salvin SB (1974) Specific suppression of delayed hypersensitivity: the possible presence of a suppressor B cell in the regulation of delayed hypersensitivity. J Immunol 113(6):1716–1725

    Article  PubMed  CAS  Google Scholar 

  53. Palomares O, Akdis M, Martín-Fontecha M, Akdis CA (2017) Mechanisms of immune regulation in allergic diseases: the role of regulatory T and B cells. Immunol Rev 278(1):219–236

    Article  PubMed  CAS  Google Scholar 

  54. Mizoguchi A, Mizoguchi E, Takedatsu H, Blumberg RS, Bhan AK (2002) Chronic intestinal inflammatory condition generates IL-10-producing regulatory B cell subset characterized by CD1d upregulation. Immunity 16(2):219–230

    Article  PubMed  CAS  Google Scholar 

  55. Fillatreau S, Sweenie CH, McGeachy MJ, Gray D, Anderton SM (2002) B cells regulate autoimmunity by provision of IL-10. Nat Immunol 3(10):944–950

    Article  PubMed  CAS  Google Scholar 

  56. Mauri C, Gray D, Mushtaq N, Londei M (2003) Prevention of arthritis by interleukin 10-producing B cells. J Exp Med 197(4):489–501

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Akdis CA, Blesken T, Akdis M, Wüthrich B, Blaser K (1998) Role of interleukin 10 in specific immunotherapy. J Clin Invest 102(1):98–106

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Goetz M, Atreya R, Ghalibafian M, Galle PR, Neurath MF (2007) Exacerbation of ulcerative colitis after rituximab salvage therapy. Inflamm Bowel Dis 13(11):1365–1368

    Article  PubMed  Google Scholar 

  59. Dass S, Vital EM, Emery P (2007) Development of psoriasis after B cell depletion with rituximab. Arthritis Rheum 56(8):2715–2718

    Article  PubMed  CAS  Google Scholar 

  60. Iwata Y, Matsushita T, Horikawa M, Dilillo DJ, Yanaba K, Venturi GM et al (2011) Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood 117(2):530–541

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Flores-Borja F, Bosma A, Ng D, Reddy V, Ehrenstein MR, Isenberg DA et al (2013) CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation. Sci Transl Med 5(173):173ra23

    Article  PubMed  Google Scholar 

  62. Das A, Ellis G, Pallant C, Lopes AR, Khanna P, Peppa D et al (2012) IL-10-producing regulatory B cells in the pathogenesis of chronic hepatitis B virus infection. J Immunol 189(8):3925–3935

    Article  PubMed  CAS  Google Scholar 

  63. van de Veen W, Stanic B, Yaman G, Wawrzyniak M, Söllner S, Akdis DG et al (2013) IgG4 production is confined to human IL-10-producing regulatory B cells that suppress antigen-specific immune responses. J Allergy Clin Immunol 131(4):1204–1212

    Article  PubMed  Google Scholar 

  64. Shalapour S, Font-Burgada J, Di Caro G, Zhong Z, Sanchez-Lopez E, Dhar D et al (2015) Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy. Nature 521(7550):94–98

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Lindner S, Dahlke K, Sontheimer K, Hagn M, Kaltenmeier C, Barth TFE et al (2013) Interleukin 21-induced granzyme B-expressing B cells infiltrate tumors and regulate T cells. Cancer Res 73(8):2468–2479

    Article  PubMed  CAS  Google Scholar 

  66. Brosseau C, Durand M, Colas L, Durand E, Foureau A, Cheminant M-A et al (2018) CD9+ regulatory B cells induce T cell apoptosis via IL-10 and are reduced in severe asthmatic patients. Front Immunol 9:3034

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Zhang M, Zheng X, Zhang J, Zhu Y, Zhu X, Liu H et al (2012) CD19(+)CD1d(+)CD5(+) B cell frequencies are increased in patients with tuberculosis and suppress Th17 responses. Cell Immunol 274(1–2):89–97

    Article  PubMed  CAS  Google Scholar 

  68. Zhang M, Zeng G, Yang Q, Zhang J, Zhu X, Chen Q et al (2014) Anti-tuberculosis treatment enhances the production of IL-22 through reducing the frequencies of regulatory B cell. Tuberculosis (Edinb) 94(3):238–244

    Article  PubMed  CAS  Google Scholar 

  69. Menon M, Blair PA, Isenberg DA, Mauri C (2016) A regulatory feedback between plasmacytoid dendritic cells and regulatory B cells is aberrant in systemic lupus erythematosus. Immunity 44(3):683–697

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Rosser EC, Oleinika K, Tonon S, Doyle R, Bosma A, Carter NA et al (2014) Regulatory B cells are induced by gut microbiota-driven interleukin-1β and interleukin-6 production. Nat Med 20(11):1334–1339

    Article  PubMed  CAS  Google Scholar 

  71. Yoshizaki A, Miyagaki T, Dilillo DJ, Matsushita T, Horikawa M, Kountikov EI et al (2012) Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions. Nature 491(7423):264–268

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Giordani L, Sanchez M, Libri I, Quaranta MG, Mattioli B, Viora M (2009) IFN-alpha amplifies human naive B cell TLR-9-mediated activation and Ig production. J Leukoc Biol 86(2):261–271

    Article  PubMed  CAS  Google Scholar 

  73. Jego G, Palucka AK, Blanck J-P, Chalouni C, Pascual V, Banchereau J (2003) Plasmacytoid dendritic cells induce plasma cell differentiation through type I interferon and interleukin 6. Immunity 19(2):225–234

    Article  PubMed  CAS  Google Scholar 

  74. Ramgolam VS, Sha Y, Marcus KL, Choudhary N, Troiani L, Chopra M et al (2011) B cells as a therapeutic target for IFN-β in relapsing-remitting multiple sclerosis. J Immunol 186(7):4518–4526

    Article  PubMed  CAS  Google Scholar 

  75. Liu B-S, Cao Y, Huizinga TW, Hafler DA, Toes REM (2014) TLR-mediated STAT3 and ERK activation controls IL-10 secretion by human B cells. Eur J Immunol 44(7):2121–2129

    Article  PubMed  CAS  Google Scholar 

  76. Yu C-R, Choi JK, Uche AN, Egwuagu CE (2018) Production of IL-35 by Bregs is mediated through binding of BATF-IRF-4-IRF-8 complex to il12a and ebi3 promoter elements. J Leukoc Biol 104(6):1147–1157

    Article  PubMed  CAS  Google Scholar 

  77. Yanaba K, Bouaziz J-D, Haas KM, Poe JC, Fujimoto M, Tedder TF (2008) A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses. Immunity 28(5):639–650

    Article  PubMed  CAS  Google Scholar 

  78. Yanaba K, Bouaziz J-D, Matsushita T, Tsubata T, Tedder TF (2009) The development and function of regulatory B cells expressing IL-10 (B10 cells) requires antigen receptor diversity and TLR signals. J Immunol 182(12):7459–7472

    Article  PubMed  CAS  Google Scholar 

  79. Haas KM, Watanabe R, Matsushita T, Nakashima H, Ishiura N, Okochi H et al (2010) Protective and pathogenic roles for B cells during systemic autoimmunity in NZB/W F1 mice. J Immunol 184(9):4789–4800

    Article  PubMed  CAS  Google Scholar 

  80. Stanic B, van de Veen W, Wirz OF, Rückert B, Morita H, Söllner S et al (2015) IL-10-overexpressing B cells regulate innate and adaptive immune responses. J Allergy Clin Immunol 135(3):771–80.e8

    Article  PubMed  CAS  Google Scholar 

  81. Piper CJM, Rosser EC, Oleinika K, Nistala K, Krausgruber T, Rendeiro AF et al (2019) Aryl hydrocarbon receptor contributes to the transcriptional program of IL-10-producing regulatory B cells. Cell Rep 29(7):1878-1892.e7

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Fehres CM, van Uden NO, Yeremenko NG, Fernandez L, Franco Salinas G, van Duivenvoorde LM et al (2019) APRIL induces a novel subset of IgA+ regulatory B cells that suppress inflammation via expression of IL-10 and PD-L1. Front Immunol 10:1368

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Komlósi ZI, Kovács N, van de Veen W, Kirsch AI, Fahrner HB, Wawrzyniak M et al (2018) Human CD40 ligand-expressing type 3 innate lymphoid cells induce IL-10-producing immature transitional regulatory B cells. J Allergy Clin Immunol 142(1):178-194.e11

    Article  PubMed  Google Scholar 

  84. Wang R-X, Yu C-R, Dambuza IM, Mahdi RM, Dolinska MB, Sergeev YV et al (2014) Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat Med 20(6):633–641

    Article  PubMed  PubMed Central  Google Scholar 

  85. van der Vlugt LEPM, Labuda LA, Ozir-Fazalalikhan A, Lievers E, Gloudemans AK, Liu K-Y et al (2012) Schistosomes induce regulatory features in human and mouse CD1d(hi) B cells: inhibition of allergic inflammation by IL-10 and regulatory T cells. PLoS ONE 7(2):e30883

    Article  PubMed  PubMed Central  Google Scholar 

  86. Parekh VV, Prasad DVR, Banerjee PP, Joshi BN, Kumar A, Mishra GC (2003) B cells activated by lipopolysaccharide, but not by anti-Ig and anti-CD40 antibody, induce anergy in CD8+ T cells: role of TGF-beta 1. J Immunol 170(12):5897–5911

    Article  PubMed  CAS  Google Scholar 

  87. Rosser EC, Mauri C (2015) Regulatory B cells: origin, phenotype, and function. Immunity 42(4):607–612

    Article  PubMed  CAS  Google Scholar 

  88. Shen P, Roch T, Lampropoulou V, O’Connor RA, Stervbo U, Hilgenberg E et al (2014) IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature 507(7492):366–370

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Palomares O, Martín-Fontecha M, Lauener R, Traidl-Hoffmann C, Cavkaytar O, Akdis M et al (2014) Regulatory T cells and immune regulation of allergic diseases: roles of IL-10 and TGF-β. Genes Immun 15(8):511–520

    Article  PubMed  CAS  Google Scholar 

  90. Reißig S, Tang Y, Nikolaev A, Gerlach K, Wolf C, Davari K et al (2017) Elevated levels of Bcl-3 inhibits Treg development and function resulting in spontaneous colitis. Nat Commun 8:15069

    Article  PubMed  PubMed Central  Google Scholar 

  91. Riemann M, Endres R, Liptay S, Pfeffer K, Schmid RM (2005) The IkappaB protein Bcl-3 negatively regulates transcription of the IL-10 gene in macrophages. J Immunol 175(6):3560–3568

    Article  PubMed  CAS  Google Scholar 

  92. Carmody RJ, Ruan Q, Palmer S, Hilliard B, Chen YH (2007) Negative regulation of toll-like receptor signaling by NF-kappaB p50 ubiquitination blockade. Science 317(5838):675–678

    Article  PubMed  CAS  Google Scholar 

  93. Zhang X, Wang H, Claudio E, Brown K, Siebenlist U (2007) A role for the IκB family member Bcl-3 in the control of central immunologic tolerance. Immunity 27(3):438–452

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Kuchroo VK et al (2009) PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med 206(13):3015–3029

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Khan AR, Hams E, Floudas A, Sparwasser T, Weaver CT, Fallon PG (2015) PD-L1hi B cells are critical regulators of humoral immunity. Nat Commun 6:5997

    Article  PubMed  CAS  Google Scholar 

  96. Klinker MW, Reed TJ, Fox DA, Lundy SK (2013) Interleukin-5 supports the expansion of fas ligand-expressing killer B cells that induce antigen-specific apoptosis of CD4(+) T cells and secrete interleukin-10. PLoS ONE 8(8):e70131

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Figueiró F, Muller L, Funk S, Jackson EK, Battastini AMO, Whiteside TL (2016) Phenotypic and functional characteristics of CD39high human regulatory B cells (Breg). Oncoimmunology 5(2):e1082703

    Article  PubMed  PubMed Central  Google Scholar 

  98. Ding Q, Yeung M, Camirand G, Zeng Q, Akiba H, Yagita H et al (2011) Regulatory B cells are identified by expression of TIM-1 and can be induced through TIM-1 ligation to promote tolerance in mice. J Clin Invest 121(9):3645–3656

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Rosser EC, Piper CJM, Matei DE, Blair PA, Rendeiro AF, Orford M et al (2020) Microbiota-derived metabolites suppress arthritis by amplifying aryl-hydrocarbon receptor activation in regulatory B cells. Cell Metab 31(4):837-851.e10

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Neves P, Lampropoulou V, Calderon-Gomez E, Roch T, Stervbo U, Shen P et al (2010) Signaling via the MyD88 adaptor protein in B cells suppresses protective immunity during Salmonella typhimurium infection. Immunity 33(5):777–790

    Article  PubMed  CAS  Google Scholar 

  101. Noh J, Lee JH, Noh G, Bang SY, Kim HS, Choi WS et al (2010) Characterisation of allergen-specific responses of IL-10-producing regulatory B cells (Br 1) in Cow Milk Allergy. Cell Immunol 264(2):143–149

    Article  PubMed  CAS  Google Scholar 

  102. Braza F, Chesne J, Durand M, Dirou S, Brosseau C, Mahay G et al (2015) A regulatory CD9(+) B-cell subset inhibits HDM-induced allergic airway inflammation. Allergy 70(11):1421–1431

    Article  PubMed  CAS  Google Scholar 

  103. Qian G, Jiang W, Sun D, Sun Z, Chen A, Fang H et al (2023) B-cell-derived IL-10 promotes allergic sensitization in asthma regulated by Bcl-3. Cell Mol Immunol 20(11):1313–1327

  104. van der Vlugt LEPM, Mlejnek E, Ozir-Fazalalikhan A, Janssen Bonas M, Dijksman TR, Labuda LA et al (2014) CD24(hi)CD27(+) B cells from patients with allergic asthma have impaired regulatory activity in response to lipopolysaccharide. Clin Exp Allergy 44(4):517–528

    Article  PubMed  Google Scholar 

  105. Wirz OF, Głobińska A, Ochsner U, van de Veen W, Eller E, Christiansen ES et al (2019) Comparison of regulatory B cells in asthma and allergic rhinitis. Allergy 74(4):815–818

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Open access funding enabled and organized by Projekt DEAL. This work was supported by the DFG grants Sch 997/10–1 (CK) and DFG SCHA 997/11–1 (BS).

Author information

Authors and Affiliations

Authors

Contributions

C.K. and B.S. conceived the study. C.K. drafted the manuscript; C.K. and B.S. have revised and approved the final manuscript.

Corresponding author

Correspondence to Bianca Schaub.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kliem, C.V., Schaub, B. The role of regulatory B cells in immune regulation and childhood allergic asthma. Mol Cell Pediatr 11, 1 (2024). https://doi.org/10.1186/s40348-023-00174-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40348-023-00174-2

Keywords