Traditionally, the skin was believed to be devoid of B cells, and studies of the skin immune system have largely focused on other types of leukocytes. Exciting recent data show that B cells localize to the healthy skin of humans and other mammalian species with likely homeostatic functions in host defense, regulation of microbial communities, and wound healing. Distinct skin-associated B cell subsets drive or suppress cutaneous inflammatory responses with important clinical implications. Localized functions of skin-associated B cell subsets during inflammation comprise Ab production, interactions with skin T cells, tertiary lymphoid tissue formation, and production of proinflammatory cytokines but also include immunosuppression by providing IL-10. In this review, we delve into the intriguing new roles of skin-associated B cells in homeostasis and inflammation.

The skin is an essential barrier organ exposed to a variety of daily insults that range from simple physical injuries like cuts and UV damage to infections with sophisticated skin pathogens such as HSV or Staphylococcus aureus. In addition to these external threats, the skin is the frequent target of allergy, autoimmunity, and cancer. The skin is colonized with microbes and consists of protective layers that encompass physical and cellular components as well as antimicrobial proteins and microbial metabolites (reviewed in Ref. 1). The outermost layer of the skin is the keratinized epidermis, and the dermis and s.c. adipose tissue lay below it, and leukocytes populate all of these anatomical skin sites (reviewed in Ref. 2). To enter the skin from blood, leukocytes migrate through postcapillary venules in the dermis from where they can access microenvironments within the dermis or traverse further into the epidermis (reviewed in detail in Ref. 3). To egress from skin, leukocytes have to reach dermal afferent lymph vessels, which will transport them via larger collecting lymph vessels to the draining lymph node (reviewed in Ref. 4).

In the past decades, knowledge about the skin immune system has been rapidly expanding by defining the protective and pathological roles of different skin-associated leukocyte subsets (e.g., T cells) as well as other skin cells, such as keratinocytes (reviewed in Ref. 5). In contrast, the existence of skin-localized B cell functions remained largely unknown, despite a recognized systemic importance of B cells for skin immunity and inflammation. Specifically, B cells that differentiate into Ab-secreting cells give rise to systemic Ab titers that enforce defense against cutaneous infections but also support cutaneous allergies and autoimmune diseases. Today, there is growing evidence for a localized function of B cells within skin in driving disease through multiple mechanisms. Subsets of skin-associated B cells have also emerged as critical negative regulators of skin inflammation. In addition, there is evidence that skin-associated B cells are involved in skin homeostasis and repair by regulating wound healing and the cutaneous microbiome. In this article, we review skin-associated B cells as novel players in cutaneous homeostasis and inflammation as illustrated in Figs. 1 and 2, respectively. A role for B cells in skin cancers has been reviewed elsewhere (6).

FIGURE 1.

Skin-associated B cells maintain normal healthy skin. B-1 and B-2 lineage cells as well as Ab-secreting cells (ASC) localize to healthy skin. (AC) Secretory IgM and IgA are produced in healthy skin (A) and bind skin commensals (B) as well as invading microbes after barrier breach (C). (D) Secreted natural IgM binds to and enhances uptake of apoptotic cells (efferocytosis) by macrophages, a process that induces anti-inflammatory programming in macrophages. (E) Skin-associated B cells, primarily B-1–like B cells, produce IL-10 to limit inflammation. (F) Additional cytokines and growth factors are produced by B cells and support wound healing. Inset, Summary of mechanisms by which B cells influence skin homeostasis. bFGF, basic fibroblast growth factor; PDGF, platelet-derived growth factor.

FIGURE 1.

Skin-associated B cells maintain normal healthy skin. B-1 and B-2 lineage cells as well as Ab-secreting cells (ASC) localize to healthy skin. (AC) Secretory IgM and IgA are produced in healthy skin (A) and bind skin commensals (B) as well as invading microbes after barrier breach (C). (D) Secreted natural IgM binds to and enhances uptake of apoptotic cells (efferocytosis) by macrophages, a process that induces anti-inflammatory programming in macrophages. (E) Skin-associated B cells, primarily B-1–like B cells, produce IL-10 to limit inflammation. (F) Additional cytokines and growth factors are produced by B cells and support wound healing. Inset, Summary of mechanisms by which B cells influence skin homeostasis. bFGF, basic fibroblast growth factor; PDGF, platelet-derived growth factor.

Close modal
FIGURE 2.

Skin-associated B cells are critical to driving and suppressing inflammation. Skin-associated B cells play several important roles in skin inflammation. (A) B cell subsets with various functions accumulate in inflamed skin. (B) Ab-secreting cells (ASC) accumulate in inflamed skin and secrete Abs with reactivity to cutaneous Ags, including autoantigens and skin-associated allergens. (C) Aggregates of collaborating B and T cells form, and B cells initiate further development of TLOs that support activation and differentiation of pathogenic B and T cells. (D) High-affinity autoreactive plasma cells and IgGs emerge from GCs in TLOs, exacerbating inflammation. (E) B cells act as APCs, inducing T cell activation and proinflammatory cytokine production. (F) B cells produce proinflammatory cytokines, like IL-6, and potentially GM-CSF, IFN-γ, or IL-4, all of which promote local inflammation and, in case of IL-6, fibrosis via stimulation of fibroblasts. (G) B cells secrete IL-10 that suppresses activation of other leukocytes, including T cells and macrophages, thereby counteracting inflammation. (H) Pathogenic autoantibody deposition can destroy cell–cell junctions, creating acantholysis or gaps between cells as in pemphigus. (I) Autoantibodies can accumulate at the dermoepidermal junction, as in cutaneous lupus erythematosus, leading to complement activation and other inflammatory downstream effector functions. Inset, Summary of disease settings in which skin-associated B cells modulate skin inflammation.

FIGURE 2.

Skin-associated B cells are critical to driving and suppressing inflammation. Skin-associated B cells play several important roles in skin inflammation. (A) B cell subsets with various functions accumulate in inflamed skin. (B) Ab-secreting cells (ASC) accumulate in inflamed skin and secrete Abs with reactivity to cutaneous Ags, including autoantigens and skin-associated allergens. (C) Aggregates of collaborating B and T cells form, and B cells initiate further development of TLOs that support activation and differentiation of pathogenic B and T cells. (D) High-affinity autoreactive plasma cells and IgGs emerge from GCs in TLOs, exacerbating inflammation. (E) B cells act as APCs, inducing T cell activation and proinflammatory cytokine production. (F) B cells produce proinflammatory cytokines, like IL-6, and potentially GM-CSF, IFN-γ, or IL-4, all of which promote local inflammation and, in case of IL-6, fibrosis via stimulation of fibroblasts. (G) B cells secrete IL-10 that suppresses activation of other leukocytes, including T cells and macrophages, thereby counteracting inflammation. (H) Pathogenic autoantibody deposition can destroy cell–cell junctions, creating acantholysis or gaps between cells as in pemphigus. (I) Autoantibodies can accumulate at the dermoepidermal junction, as in cutaneous lupus erythematosus, leading to complement activation and other inflammatory downstream effector functions. Inset, Summary of disease settings in which skin-associated B cells modulate skin inflammation.

Close modal

Both recirculating (mobile) and resident T cell subsets exist in skin; T cells resident for prolonged periods of time are termed “tissue-resident memory T cells” and have distinct surface markers (i.e., CD103 and CD69 on CD8+ T cells) as well as differentiation programming (reviewed in Ref. 7). Although it is clear that subsets of B cells egress from skin by entering afferent lymph vessels after some time of residency (8), it is currently unknown how long B cells dwell in skin. In this review, we use the term “skin-resident” for B cells that have left the vascular space and reside in the skin and “skin-associated” for B cells that are either skin recirculating (present in skin-draining lymph) or skin resident.

Although initial histological studies were unable to detect B cells in healthy human skin (9, 10), cannulation of afferent lymph vessels draining healthy skin in sheep (8, 11) and humans (12, 13) demonstrated the existence of skin-recirculating B cells. More recent studies that employ flow cytometry identified and characterized B cells residing in healthy skin of mice and humans (14, 15). Skin-associated B cells and Ig-secreting cells are also conserved in the skin of lower vertebrates, such as teleost fish (16). In mammalian skin, B cells localize to the dermis where they are sparsely scattered as individual cells during homeostasis and often as cell clusters or more-organized lymphoid structures during inflammation (14, 1720)

Skin-associated B cells are a heterogenous population with both conventional (B-2) and innate-like (B-1–like) B cells (8, 14). Relative to their blood-borne counterparts, IgG+ skin-infiltrating human B-2 B cells have a lower representation of the IgG1 subclass and show a preferred usage of certain Vh genes, suggesting the existence of skin-specific B cell subsets (15). Whereas the majority of skin B cells resemble conventional B-2 B cells, innate-like (B-1–like) B cells are enriched in skin compared with blood in humans and mice (14). Also, a population of ovine skin-recirculating B cells in afferent lymph expresses high levels of IgM and myeloid markers such as CD11b, indicative of B-1–like B cells (8). Innate-like B cells often recognize conserved pathogen structures and produce natural Abs without previous antigenic exposure as well as mount rapid T cell–independent Ab responses early postinfection (reviewed in Refs. 21 and 22). Their often polyreactive Abs (mostly IgM and IgA) are, therefore, an effective first line of defense against infection (21, 22). In mice, B-1 B cells have a high propensity to migrate into skin (14) and other barrier sites such as the respiratory tract or the small intestinal mucosa, where they limit early infections by secreting IgA and IgM and innate-stimulatory cytokines such as GM-CSF (2325). Thus, innate-like B cell localization to mammalian barrier organs represents a form of barrier enforcement and first line of defense against invading pathogens following barrier breach.

Whereas most Abs act systemically by blood distribution throughout the body and diffusion into tissues, tissue titers can be enhanced by localized Ab production. For example, tissue-resident plasma cells secrete multimeric IgA and IgM, which are transcytosed through epithelial barriers from abluminal to luminal side via polymeric IgR (reviewed in Ref. 26). As a result, they reach glandular excretions and coat mucosal surfaces as secretory Igs (26). The localized role of IgA, and to some degree, that of IgM, is well appreciated as a first line of defense against toxins and pathogen invasion of mucosal barriers (2628). In contrast, studies analyzing Ab production in mammalian skin are limited. To this end, Ab secretion in healthy skin has been demonstrated for humans and sheep (8, 29). In human skin, IgA is secreted in eccrine sweat glands and appears in sweat and sebum, and Abs of IgG and IgM isotype reach the skin surface by undefined mechanisms (29, 30). Our own studies found that the isotype predominantly produced by skin-resident plasma cells of naive mice is IgM (G.F. Debes and S.E. McGettigan, submitted for publication), which may reflect the relative lack of eccrine sweat glands in mouse skin. More extensive studies are necessary to determine how each Ig isotype reaches distinct skin compartments in different mammalian species.

During inflammation, the presence of Ab-secreting plasma cells in lesional human skin has been well documented for decades. Moreover, plasma cells, identified histologically, facilitate dermatopathological diagnosis of several inflammatory entities (e.g., necrobiosis lipoidica and acne keloidalis nuchae) (31). The isotype (i.e., subclasses of IgG, IgA, IgM, and/or IgE) produced by plasma cells in lesional skin varies depending on the skin site and type of inflammation (for examples, see Refs. 3234). Ig-secreting cell accumulation in inflamed skin leads to higher Ig titers in lymph draining the skin site, reflecting an increase in tissue titers (8). Although localized Ab production is likely an effective, or sometimes desperate, attempt to control skin pathogens, it is destructive in the case of pathogenic autoantibody production to cutaneous autoantigens [e.g., in pemphigus (18); further discussed below].

Diverse microbes colonize throughout the skin, including epidermal surfaces and appendages such as hair follicles (35), and colonization reportedly reaches as deep as the dermis and dermal (s.c.) adipose tissue (36). The microbial communities of the skin are critically involved in orchestrating local cutaneous physiological and pathophysiologic processes, including wound healing, inflammation, immune responses to skin pathogens (3739), and even susceptibility to skin cancer (40). In contrast, skin colonization with pathogenic bacteria, such as methicillin-resistant S. aureus, can cause serious disease upon skin barrier breach (41). In the intestine, Abs (in particular, thymus-independent secretory IgM and IgA) bind to commensal bacteria and facilitate symbiotic host–microbe interactions (4244), and the question arises of whether there is a similar role for Abs in shaping skin microbiomes. A study in RAG-1–deficient mice, which lack all B and T cells as well as Abs, was unable to detect alterations in the ear skin surface microbiome using 16S rRNA sequencing (45). However, electron microscopy studies using immunogold labeling of human skin samples showed that skin microbes are coated with IgA, IgG, and IgM (46), supporting a role for cutaneous Abs in regulating microbial colonization of skin microenvironments. In addition, IgM in human sweat from healthy skin can bind S. aureus (46). Thus, it is possible that, akin to their role in the intestine, secretory Igs influence microbial communities in the skin. Future studies are warranted that test the role of Abs for their ability to modulate microbial colonization of distinct skin compartments, including that of deeper skin layers.

In a number of autoimmune diseases with cutaneous manifestations, the role of B cells and Abs has generally been considered systemic (i.e., pathogenic autoantibodies produced in lymphoid tissue enter blood and diffuse into skin where they cause disease). Recently, B cells are increasingly recognized as an important part of the infiltrating immune compartment in autoimmune and inflammatory skin diseases. B cell numbers are elevated in lesional skin relative to control skin specimens in a number of inflammatory diseases, including, but not limited to, psoriasis (47), pemphigus (18), lupus profundus (19, 20), systemic sclerosis (scleroderma) (48), discoid lupus erythematosus (49, 50), Sjögren’s syndrome (51), IgG4-related skin diseases (33, 34), atopic dermatitis (52), and allergic contact dermatitis (53). Although an increase in skin-infiltrating B cells already suggested a role for B cells in cutaneous inflammation, the therapeutic success of systemic B cell depletion with rituximab confirmed a pathogenic role for B cells in a number of these diseases (5456). In pemphigus, atopic dermatitis, and scleroderma, disease severity and progression are positively correlated with the number of skin-infiltrating B cells (18, 48, 52). Specifically, in scleroderma, a chronic connective tissue disorder that often progresses to extensive cutaneous and vascular fibrosis, only those patients with skin-infiltrating B cells advanced in disease severity over a period of 12 mo (48). Furthermore, a pathogenic role for B cells in the initiation phase of skin fibrosis was confirmed using B cell depletion therapy in scleroderma patients and mouse models of the disease (5759). Collectively, the recent reports of skin-infiltrating B lineage cells in skin pathologies raise the question of whether B cells directly contribute to cutaneous inflammation through local effector functions (as discussed in the sections below). Of note, skin-infiltrating B cells are not always pathogenic (e.g., psoriasis does not usually improve with B cell depletion) (60), and rituximab treatment can induce psoriasis in some individuals (61), suggesting a protective role of B cells (discussed below). Czarnowicki et al. (52) investigated B cell subsets in two skin diseases in which B cells appear to have opposing roles: atopic dermatitis and psoriasis, which are driven by Th2 and Th17/Th1 cell responses, respectively (62). The authors suggested that a higher B cell activation status reflected accumulative Ag exposure and ability to contribute to T cell activation and other pathogenic mechanisms, such as Ab production and IgE switching (52). Thus, it is likely that BCR specificity for skin-associated Ags (autoantigens, allergens, or microbiota) in combination with B cell effector functions (discussed below) dictates the distinct roles of skin-associated B cells in different types of cutaneous inflammation.

A prominent example in which skin-infiltrating B cells may drive disease is pemphigus vulgaris, a severe autoimmune blistering disease that affects mucosal membranes and skin. Pemphigus is characterized by circulating anti-desmoglein 1/3 (Dsg1/3) autoantibodies, which recognize desmosomal adhesion proteins on epidermal keratinocytes. In humans, placental transfer of the anti-Dsg autoantibody from diseased mothers induces pemphigus symptoms in their newborns (reviewed in Ref. 63). Likewise, passive transfer of the autoantibodies into newborn mice is sufficient to induce pemphigus-like disease, confirming that the pathogenic autoantibody is the main etiology of the disease (63). The vast majority of pemphigus patients accumulate anti-Dsg1/3 autoantibodies in skin; however, circulating Dsg1/3 autoantibodies can be found in patients without skin lesions and in some healthy controls, indicating that in addition to systemic autoantibodies, other factors determine disease (reviewed in Ref. 64). The pathogenic potential of anti-Dsg1/3 Abs after transfer into mice correlates with serum titers (65). An increase in cutaneous autoantibody titers could be due to enhanced localized production and/or diffusion from systemic Abs, ultimately boosting localized binding to cutaneous autoantigens. In support of this notion, Yuan et al. (18) recently showed that autoreactive Dsg1- and Dsg3-specific Ab-secreting cells and B cells localize to lesional skin relative to healthy skin, suggesting a pathogenic role in situ by these autoreactive cells. Akin to pemphigus, the majority of scleroderma patients present with both systemic and local autoantibodies as well as skin lesion–infiltrating B cells and plasma cells (48). Finally, IgG4-related skin diseases, which are fibroinflammatory disorders affecting multiple organs, are induced by accumulation of terminally differentiated IgG4+ plasma cells and Ab deposition in affected skin (33, 34). Taken together, localized Ab production is a feature of several skin diseases, including those in which Ab binding to cutaneous autoantigens is a known driver of disease.

Although in some instances, such as pemphigus vulgaris, dissociation of tissue due to autoantibody binding alone causes disease (63), many times, complement fixation and/or Fc-mediated Ab effector functions are necessary for tissue destruction. For example, in cutaneous manifestations of the autoimmune disease lupus erythematosus, the lupus band test on skin biopsies reveals the deposition of autoantibody–Ag complexes as well as complement proteins along the dermoepidermal junction (66). In cutaneous vasculitis, Igs and complement proteins are deposited around dermal vessel walls (67). Complement activation in the skin has the potential to exacerbate inflammatory damage by direct cell lysis and/or via recruitment and activation of innate immune cells, such as macrophages and neutrophils. Thus, autoantibody production in skin is a powerful means for localized destruction and inflammation through complement and/or other downstream mechanisms.

Formation of ectopic lymphoid tissue or tertiary lymphoid organs (TLOs) in the skin may provide a local microenvironment for skin-associated B cells to promote skin inflammation. TLOs are considered outposts of secondary lymphoid organs because they possess similar characteristics, such as organized B cell and T cell areas with Ag-presenting dendritic cells on a network of fibroblasts as well as high endothelial venules, lymph vessels, and germinal center (GC)-like structures (68). TLOs are found in or near sites of chronic inflammation (e.g., during autoimmunity, infection, or transplant rejection). In extracutaneous lesions of autoimmunity, such as in systemic lupus erythematosus (SLE) and rheumatoid arthritis, TLOs are considered pathogenic because they perpetuate local inflammation by enhancing autoantibody production and autoreactive T cell activation (reviewed in Refs. 69 and 70). Even though TLO-like structures that support interactions between skin B and T cells exist in certain inflammatory skin diseases, little is known about the role of TLOs in the pathogenesis of cutaneous pathologies. In lupus erythematosus panniculitis (lupus profundus; a form of cutaneous lupus that is typified by inflammation of the s.c. adipose tissue), aggregates of B cells and plasma cells or lymphoid follicles containing distinct GCs are found in the majority of cutaneous lesions (19, 20). Kogame et al. (20) showed that additional components of a TLO are present in these lupus profundus lesions, including CXCL13+ cells, peripheral node addressin+ (PNAd+) high endothelial venules, and podoplanin+ lymphatic vessels. These findings confirm that TLO structures develop in lupus profundus. In pemphigus skin lesions, B cells and IL-21+ CD4+ T cells interact closely in aggregate structures that resemble developing TLOs (18). Aggregates of B cells and T cells have also been noted in skin manifestations of Sjögren’s syndrome (51), an autoimmune disorder of exocrine glands. Skin-localized GC-like structures likely enhance pathogenic Ig affinity maturation and class switching to different IgG subclasses, as well as Ab production, thereby augmenting humoral responses to skin-associated Ags. Localized activation of effector T cells and Ab production in lymphoid aggregates and TLOs could help explain why in one individual with systemic autoantibodies that target skin, some skin areas are affected, whereas other areas are spared from disease symptoms. Notably, B cells are drivers of lymphoid neogenesis, including TLOs, by provision of lymphotoxin (70). Therefore, when skin-resident B cells initiate formation of a local TLO, this may be the tipping point that accelerates disease in many cases of chronic skin inflammation.

B cells are increasingly recognized as key effector cells in inflammation and infection by secretion of inflammatory cytokines such as IL-6, GM-CSF, IFN-γ, and IL-4 (71), and B cell cytokine secretion in skin is a new area of investigation. Recently, Matsushita et al. (59) showed that uninflamed skin of mice harbors IL-6+ B cells, which drastically increase in inflamed skin in the bleomycin-induced scleroderma model. Importantly, the authors found that skin pathology, including skin fibrosis and thickening, were significantly reduced in mice whose B cells lack the ability to express IL-6, demonstrating a pathogenic role for IL-6+ B cells in fibrosing skin inflammation (59). Furthermore, blockade of the cytokine BAFF [a B cell–targeting approach used to treat SLE (72)] inhibited expansion of IL-6+ but not that of anti-inflammatory IL-10+ B cells, leading to reduced pathological changes in bleomycin-induced skin fibrosis (59). These findings stress the importance to further define skin-associated B cells and their functions because this may reveal novel tools to selectively target distinct B cell subsets.

Ag presentation to effector T cells is another Ab-independent effector function of B cells that is increasingly recognized to drive inflammation. Specifically, effector T cells that recognize Ags on B cell MHC class II (MHCII) release potent inflammatory cytokines and subsequently expand. For example, mice with B cells that cannot present Ags to T cells are protected from neuroinflammation in a mouse model of multiple sclerosis because of reduced pathogenic T cell responses (73). Studies by our laboratory showed that skin-resident and skin-recirculating B cells express high levels of MHCII and costimulatory molecules (CD80/86 and CD1) and are therefore well equipped for T cell activation (8). Although studies of B cells as APCs in skin inflammation are limited, Young et al. (74) showed a pathogenic role for B cells in a mouse model of chronic graft-versus-host disease, a serious complication of allogeneic hematopoietic stem cell transplantation. Specifically, donor B cells upregulated MHCII and costimulatory molecules and augmented donor CD4+ T cell expansion, alloreactivity, and survival, which in turn promoted chronic graft-versus-host disease in skin and lung (74). Although the authors concluded that B cell–T cell cross-talk occurred in secondary lymphoid tissues (74), it is possible that such interactions were also present directly in skin and/or skin-associated TLOs. Effector T cell activation by skin-associated B cells and downstream inflammation caused by T cell cytokines, such as IFN-γ, IL-17, and IL-4, have the potential to exacerbate many inflammatory skin disorders. Therefore, additional studies into the APC function of skin-associated B cells are highly warranted.

B cells with the capacity to suppress immune responses, termed regulatory B cells (Bregs), have recently received much attention because they limit inflammatory and autoimmune processes in many organ systems and diseases (reviewed in Refs. 75 and 76). Notably, B cells with the ability to suppress skin inflammation were already described more than 40 y ago (77, 78). The suppressive mechanisms employed by Bregs are varied and include expansion and maintenance of regulatory T cells, adenosine generation, and secretion of the suppressive cytokines IL-35 and IL-10 (reviewed in Refs. 79 and 80). In skin, B cells that produce IL-10 are critical to limiting inflammation, and the significance of other Breg functions has not been explored to date.

In mouse models, IL-10+ Bregs (also known as B10 cells) limit inflammation in cutaneous hypersensitivity (81, 82), scleroderma (59), and psoriasis-like inflammation (83, 84). Earlier studies suggested that splenic Bregs suppress T cell priming and/or polarization in the initiation phase and that innate-like Bregs (e.g., peritoneal B-1 B cells) are important in the remission phase of inflammation, with all Bregs acting within lymphoid tissues (81, 82). Our laboratory recently showed that IL-10+ B cells reside in normal mouse and human skin, and analysis of IL-10 reporter mice revealed that B cells actively transcribe IL-10 in the skin even in the absence of inflammation (14). Moreover, IL-10+ B-1 B cells, which are potent suppressors of inflammation in both cutaneous and intestinal inflammation (82, 85), are recruited into inflamed skin (14). Matsushita et al. (59) also showed that IL-10+ Bregs reside in skin and are critical in limiting inflammation and fibrosis in a scleroderma model. Therefore, IL-10+ Bregs are well positioned to suppress inflammation in the skin itself in addition to acting in lymphoid tissue. Breg presence and active IL-10 transcription in uninflamed skin (14) suggest that skin Bregs fulfill a steady-state function by suppressing aberrant responses to physical insults and/or skin commensal microbiota.

A potential pathway to induce skin Bregs is through cutaneous UV exposure. Specifically, moderate UV light exposure of skin leads to enrichment of skin lymph node–resident B cells with the capacity to inhibit dendritic cell activation of T cells in cutaneous hypersensitivity models in an IL-10–dependent manner (86, 87) as well as IL-10–negative Bregs that suppress experimental autoimmune encephalitis (88). It will be interesting to further investigate this pathway as well as potential differences between individual mammalian species with inherent differences in sunlight (UV) exposure (e.g., caused by hair density and nocturnal versus diurnal chronotypes).

In humans, B cell depleting therapy (i.e., rituximab, anti-CD20) is able to induce or exacerbate the inflammatory skin disease psoriasis (reviewed in Ref. 61). This apparent paradoxical effect of an immunosuppressive therapy supports a protective role for B cells in this largely Th1 and Th17 cell–mediated disease. Interestingly, psoriasis is associated with low levels of dermal IL-10 and is clinically responsive to locally administered IL-10 (89, 90), possibly reflecting a niche for regulation by IL-10+ cell types such as IL-10+ Bregs. Extending the mouse studies that demonstrate a critical role for IL-10+ B cells in suppressing psoriasiform inflammation (83, 84), individuals with psoriasis have reduced numbers of IL-10+ B cells in their blood (91, 92). Other cutaneous inflammatory diseases, such as the skin-blistering disease pemphigus and scleroderma, can also be associated with reduced numbers of circulating IL-10+ Bregs during active disease (9396) and an increase in disease remission (97). Notably, IL-10+ B cells localize to the inflamed skin in SLE (98). It is therefore possible that skin-associated Bregs limit skin inflammation even in B cell– and autoantibody-mediated inflammation.

Collectively, there is accumulating evidence that IL-10+ B cells suppress skin inflammation not only in mouse models but also in humans. Additional studies are needed to dissect the Breg subsets at cutaneous and extracutaneous sites and IL-10–dependent and –independent mechanisms by which they suppress different types and phases (i.e., acute versus chronic; initiation versus remission) of skin inflammation. In particular, in B cell–mediated autoimmune diseases, it would be advantageous to be able to deplete disease-driving B cells selectively, while sparing Bregs. Conversely, tools to selectively target Bregs that impede effective cutaneous immune responses, based on differential surface marker expression, for example, could potentially be desirable when treating cutaneous tumors or chronic infections. To complicate matters, Bregs that make IL-10 do not appear to be a separate B cell lineage and are rather an activation or differentiation state of several traditional B cells subsets (99). Similarly, in T cells, IL-10 is produced not only by bona fide Foxp3+ regulatory T cells but also by effector T cells and represents an essential mechanism for limiting immunopathology at effector sites (100, 101). For example, during influenza virus infection, IFN-γ+ CD4 and CD8 effector T cells acquire the ability to secrete IL-10, which is critical for host survival because it limits tissue destruction and pulmonary inflammation (102). Therefore, further studies are needed to decode the signals that induce IL-10 or other anti-inflammatory properties in distinct skin-associated B cell subsets, allowing for selective therapeutic manipulation.

As a barrier organ with various physical insults that happen on a daily basis, there is a vast need for repair of small and larger skin wounds. B cells localize to cutaneous wounds (103, 104) and are potentially involved in wound healing (105). Specifically, mice that overexpress BCR coreceptor CD19 (CD19-transgenic mice) exhibit accelerated, whereas CD19-deficent mice have delayed, wound healing (105). The authors of the study suggested a TLR4-initiated increase in cytokines, such as IL-6, IL-10, TGF-β, platelet-derived growth factor, and basic fibroblast growth factor, as the mechanism of B cell–enhanced wound healing (105). However, CD19-deficient and -transgenic mice have greatly altered B cell subsets with a relative lack or increase of innate-like B cells (i.e., MZ and B-1 B cells), respectively (106108). As B-1–like B cells, which are CD19hi, localize to skin (14), it is tempting to speculate that these cells promote wound healing. In further support of a role of B cells in wound healing is an observation that exogenous application of splenic B cells to wounds accelerates the healing process in wild-type and diabetic mice (109). Future studies are needed to dissect the mechanism by which different subsets of skin-associated B cells may contribute to the wound healing process and how this may be targeted for therapeutic purposes.

With the relatively recent discovery that B cells are part of the skin immune system, we are just beginning to understand the capacity of skin-associated B cells in modulating cutaneous immune responses. The newly uncovered pathogenic and protective functions of B cells in skin and those of B cells at other extralymphoid sites (e.g., the joints and synovia, the CNS, and intestinal mucosa) offer a glimpse of the power of skin-associated B cells in disease settings. Thus, further illuminating the functions of skin B cell subsets provides the opportunity to develop novel approaches to modulate cutaneous immune responses in homeostasis, inflammation, infection, and cancer. We anticipate that exciting new developments in cutaneous B cell biology are on the horizon.

We thank Neda Nikbakht, Gregg Silverman, and members of the Debes Lab for critical comments on the manuscript and stimulating discussions. We also thank John Stanley for advice and discussion of pemphigus pathogenesis. The authors are indebted to Paul Schiffmacher and Tim Flanagan for figure illustrations.

This work was supported in part by National Institutes of Health Grants R01AR067751 and R01AI127389 (to G.F.D.) and a Percival E. and Ethel Brown Foerderer Foundation Fellowship from Thomas Jefferson University (to S.E.M).

Abbreviations used in this article:

Breg

regulatory B cell

Dsg1/3

desmoglein 1/3

GC

germinal center

MHCII

MHC class II

SLE

systemic lupus erythematosus

TLO

tertiary lymphoid organ.

1
Chen
,
Y. E.
,
M. A.
Fischbach
,
Y.
Belkaid
.
2018
.
Skin microbiota-host interactions.
Nature
553
:
427
436
.
2
Lian
,
C. G.
,
G. F.
Murphy
2015
.
Histology of the skin.
In
Lever’s Histopathology of the Skin
, 11th Ed.
D. E.
Elder
, ed.
Wolters Kluwer
,
Philadelphia
, p.
8
75
.
3
Schön
,
M. P.
,
T. M.
Zollner
,
W. H.
Boehncke
.
2003
.
The molecular basis of lymphocyte recruitment to the skin: clues for pathogenesis and selective therapies of inflammatory disorders.
J. Invest. Dermatol.
121
:
951
962
.
4
Lund
,
A. W.
,
T. R.
Medler
,
S. A.
Leachman
,
L. M.
Coussens
.
2016
.
Lymphatic vessels, inflammation, and immunity in skin cancer.
Cancer Discov.
6
:
22
35
.
5
Pasparakis
,
M.
,
I.
Haase
,
F. O.
Nestle
.
2014
.
Mechanisms regulating skin immunity and inflammation.
Nat. Rev. Immunol.
14
:
289
301
.
6
Egbuniwe
,
I. U.
,
S. N.
Karagiannis
,
F. O.
Nestle
,
K. E.
Lacy
.
2015
.
Revisiting the role of B cells in skin immune surveillance.
Trends Immunol.
36
:
102
111
.
7
Gebhardt
,
T.
,
U.
Palendira
,
D. C.
Tscharke
,
S.
Bedoui
.
2018
.
Tissue-resident memory T cells in tissue homeostasis, persistent infection, and cancer surveillance.
Immunol. Rev.
283
:
54
76
.
8
Geherin
,
S. A.
,
S. R.
Fintushel
,
M. H.
Lee
,
R. P.
Wilson
,
R. T.
Patel
,
C.
Alt
,
A. J.
Young
,
J. B.
Hay
,
G. F.
Debes
.
2012
.
The skin, a novel niche for recirculating B cells.
J. Immunol.
188
:
6027
6035
.
9
Bos
,
J. D.
,
M. B.
Teunissen
.
2008
.
Innate and adaptive immunity.
In
Clinical and Basic Immunodermatology.
A. A.
Gaspari
,
S. K.
Tyring
, eds.
Springer
,
London
, p.
17
30
.
10
Bos
,
J. D.
,
I.
Zonneveld
,
P. K.
Das
,
S. R.
Krieg
,
C. M.
van der Loos
,
M. L.
Kapsenberg
.
1987
.
The skin immune system (SIS): distribution and immunophenotype of lymphocyte subpopulations in normal human skin.
J. Invest. Dermatol.
88
:
569
573
.
11
Mackay
,
C. R.
,
W. G.
Kimpton
,
M. R.
Brandon
,
R. N.
Cahill
.
1988
.
Lymphocyte subsets show marked differences in their distribution between blood and the afferent and efferent lymph of peripheral lymph nodes.
J. Exp. Med.
167
:
1755
1765
.
12
Olszewski
,
W. L.
,
I.
Grzelak
,
A.
Ziolkowska
,
A.
Engeset
.
1995
.
Immune cell traffic from blood through the normal human skin to lymphatics.
Clin. Dermatol.
13
:
473
483
.
13
Yawalkar
,
N.
,
R. E.
Hunger
,
W. J.
Pichler
,
L. R.
Braathen
,
C. U.
Brand
.
2000
.
Human afferent lymph from normal skin contains an increased number of mainly memory / effector CD4(+) T cells expressing activation, adhesion and co-stimulatory molecules.
Eur. J. Immunol.
30
:
491
497
.
14
Geherin
,
S. A.
,
D.
Gómez
,
R. A.
Glabman
,
G.
Ruthel
,
A.
Hamann
,
G. F.
Debes
.
2016
.
IL-10+ innate-like B cells are part of the skin immune system and require α4β1 integrin to migrate between the peritoneum and inflamed skin.
J. Immunol.
196
:
2514
2525
.
15
Saul
,
L.
,
K. M.
Ilieva
,
H. J.
Bax
,
P.
Karagiannis
,
I.
Correa
,
I.
Rodriguez-Hernandez
,
D. H.
Josephs
,
I.
Tosi
,
I. U.
Egbuniwe
,
S.
Lombardi
, et al
.
2016
.
IgG subclass switching and clonal expansion in cutaneous melanoma and normal skin.
Sci. Rep.
6
:
29736
.
16
Salinas
,
I.
,
Y. A.
Zhang
,
J. O.
Sunyer
.
2011
.
Mucosal immunoglobulins and B cells of teleost fish.
Dev. Comp. Immunol.
35
:
1346
1365
.
17
Lafyatis
,
R.
,
E.
Kissin
,
M.
York
,
G.
Farina
,
K.
Viger
,
M. J.
Fritzler
,
P. A.
Merkel
,
R. W.
Simms
.
2009
.
B cell depletion with rituximab in patients with diffuse cutaneous systemic sclerosis.
Arthritis Rheum.
60
:
578
583
.
18
Yuan
,
H.
,
S.
Zhou
,
Z.
Liu
,
W.
Cong
,
X.
Fei
,
W.
Zeng
,
H.
Zhu
,
R.
Xu
,
Y.
Wang
,
J.
Zheng
,
M.
Pan
.
2017
.
Pivotal role of lesional and perilesional T/B lymphocytes in pemphigus pathogenesis.
J. Invest. Dermatol.
137
:
2362
2370
.
19
Massone
,
C.
,
K.
Kodama
,
W.
Salmhofer
,
R.
Abe
,
H.
Shimizu
,
A.
Parodi
,
H.
Kerl
,
L.
Cerroni
.
2005
.
Lupus erythematosus panniculitis (lupus profundus): clinical, histopathological, and molecular analysis of nine cases.
J. Cutan. Pathol.
32
:
396
404
.
20
Kogame
,
T.
,
R.
Yamashita
,
M.
Hirata
,
T. R.
Kataoka
,
H.
Kamido
,
C.
Ueshima
,
M.
Matsui
,
T.
Nomura
,
K.
Kabashima
.
2018
.
Analysis of possible structures of inducible skin-associated lymphoid tissue in lupus erythematosus profundus.
J. Dermatol.
45
:
1117
1121
.
21
Baumgarth
,
N.
2011
.
The double life of a B-1 cell: self-reactivity selects for protective effector functions.
Nat. Rev. Immunol.
11
:
34
46
.
22
Kearney
,
J. F.
2005
.
Innate-like B cells.
Springer Semin. Immunopathol.
26
:
377
383
.
23
Choi
,
Y. S.
,
N.
Baumgarth
.
2008
.
Dual role for B-1a cells in immunity to influenza virus infection.
J. Exp. Med.
205
:
3053
3064
.
24
Suzuki
,
K.
,
M.
Maruya
,
S.
Kawamoto
,
S.
Fagarasan
.
2010
.
Roles of B-1 and B-2 cells in innate and acquired IgA-mediated immunity.
Immunol. Rev.
237
:
180
190
.
25
Weber
,
G. F.
,
B. G.
Chousterman
,
I.
Hilgendorf
,
C. S.
Robbins
,
I.
Theurl
,
L. M.
Gerhardt
,
Y.
Iwamoto
,
T. D.
Quach
,
M.
Ali
,
J. W.
Chen
, et al
.
2014
.
Pleural innate response activator B cells protect against pneumonia via a GM-CSF-IgM axis.
J. Exp. Med.
211
:
1243
1256
.
26
Brandtzaeg
,
P.
2013
.
Secretory IgA: designed for anti-microbial defense.
Front. Immunol.
4
:
222
.
27
Mantis
,
N. J.
,
N.
Rol
,
B.
Corthésy
.
2011
.
Secretory IgA’s complex roles in immunity and mucosal homeostasis in the gut.
Mucosal Immunol.
4
:
603
611
.
28
Ehrenstein
,
M. R.
,
C. A.
Notley
.
2010
.
The importance of natural IgM: scavenger, protector and regulator.
Nat. Rev. Immunol.
10
:
778
786
.
29
Okada
,
T.
,
H.
Konishi
,
M.
Ito
,
H.
Nagura
,
J.
Asai
.
1988
.
Identification of secretory immunoglobulin A in human sweat and sweat glands.
J. Invest. Dermatol.
90
:
648
651
.
30
Metze
,
D.
,
W.
Jurecka
,
W.
Gebhart
,
J.
Schmidt
,
M.
Mainitz
,
G.
Niebauer
.
1989
.
Immunohistochemical demonstration of immunoglobulin A in human sebaceous and sweat glands.
J. Invest. Dermatol.
92
:
13
17
.
31
Elston
,
D. M.
,
T.
Ferringer
,
C. J.
Ko
,
S.
Peckham
,
W. A.
High
,
D. J.
DiCaudo
,
S.
Bhuta
.
2014
.
Dermatopathology.
Elsevier
,
Amsterdam, the Netherlands
.
32
Lai A Fat
,
R. F.
,
R. H.
Cormane
,
R.
van Furth
.
1974
.
An immunohistopathological study on the synthesis of immunoglobulins and complement in normal and pathological skin and the adjacent mucous membranes.
Br. J. Dermatol.
90
:
123
136
.
33
Tokura
,
Y.
,
H.
Yagi
,
H.
Yanaguchi
,
Y.
Majima
,
A.
Kasuya
,
T.
Ito
,
M.
Maekawa
,
H.
Hashizume
.
2014
.
IgG4-related skin disease.
Br. J. Dermatol.
171
:
959
967
.
34
Hsiao
,
P. F.
,
Y. H.
Wu
.
2016
.
Characterization of cutaneous plasmacytosis at different disease stages.
Dermatology (Basel)
232
:
738
747
.
35
SanMiguel
,
A.
,
E. A.
Grice
.
2015
.
Interactions between host factors and the skin microbiome.
Cell. Mol. Life Sci.
72
:
1499
1515
.
36
Nakatsuji
,
T.
,
H. I.
Chiang
,
S. B.
Jiang
,
H.
Nagarajan
,
K.
Zengler
,
R. L.
Gallo
.
2013
.
The microbiome extends to subepidermal compartments of normal skin.
Nat. Commun.
4
:
1431
.
37
Grice
,
E. A.
,
E. S.
Snitkin
,
L. J.
Yockey
,
D. M.
Bermudez
,
K. W.
Liechty
,
J. A.
Segre
;
NISC Comparative Sequencing Program
.
2010
.
Longitudinal shift in diabetic wound microbiota correlates with prolonged skin defense response. [Published erratum appears in 2010 Proc. Natl. Acad. Sci. USA 107: 17851.]
Proc. Natl. Acad. Sci. USA
107
:
14799
14804
.
38
Naik
,
S.
,
N.
Bouladoux
,
C.
Wilhelm
,
M. J.
Molloy
,
R.
Salcedo
,
W.
Kastenmuller
,
C.
Deming
,
M.
Quinones
,
L.
Koo
,
S.
Conlan
, et al
.
2012
.
Compartmentalized control of skin immunity by resident commensals.
Science
337
:
1115
1119
.
39
Weyrich
,
L. S.
,
S.
Dixit
,
A. G.
Farrer
,
A. J.
Cooper
,
A. J.
Cooper
.
2015
.
The skin microbiome: associations between altered microbial communities and disease.
Australas. J. Dermatol.
56
:
268
274
.
40
Nakatsuji
,
T.
,
T. H.
Chen
,
A. M.
Butcher
,
L. L.
Trzoss
,
S. J.
Nam
,
K. T.
Shirakawa
,
W.
Zhou
,
J.
Oh
,
M.
Otto
,
W.
Fenical
,
R. L.
Gallo
.
2018
.
A commensal strain of Staphylococcus epidermidis protects against skin neoplasia.
Sci. Adv.
4
:
eaao4502
.
41
Yang
,
E. S.
,
J.
Tan
,
S.
Eells
,
G.
Rieg
,
G.
Tagudar
,
L. G.
Miller
.
2010
.
Body site colonization in patients with community-associated methicillin-resistant Staphylococcus aureus and other types of S. aureus skin infections.
Clin. Microbiol. Infect.
16
:
425
431
.
42
Bunker
,
J. J.
,
T. M.
Flynn
,
J. C.
Koval
,
D. G.
Shaw
,
M.
Meisel
,
B. D.
McDonald
,
I. E.
Ishizuka
,
A. L.
Dent
,
P. C.
Wilson
,
B.
Jabri
, et al
.
2015
.
Innate and adaptive humoral responses coat distinct commensal bacteria with immunoglobulin A.
Immunity
43
:
541
553
.
43
Kato
,
L. M.
,
S.
Kawamoto
,
M.
Maruya
,
S.
Fagarasan
.
2014
.
The role of the adaptive immune system in regulation of gut microbiota.
Immunol. Rev.
260
:
67
75
.
44
Magri
,
G.
,
L.
Comerma
,
M.
Pybus
,
J.
Sintes
,
D.
Lligé
,
D.
Segura-Garzón
,
S.
Bascones
,
A.
Yeste
,
E. K.
Grasset
,
C.
Gutzeit
, et al.
2017
.
Human secretory IgM emerges from plasma cells clonally related to gut memory B cells and targets highly diverse commensals.
Immunity
47
:
118
134.e8
.
45
Scholz
,
F.
,
B. D.
Badgley
,
M. J.
Sadowsky
,
D. H.
Kaplan
.
2014
.
Immune mediated shaping of microflora community composition depends on barrier site.
PLoS One
9
:
e84019
.
46
Metze
,
D.
,
A.
Kersten
,
W.
Jurecka
,
W.
Gebhart
.
1991
.
Immunoglobulins coat microorganisms of skin surface: a comparative immunohistochemical and ultrastructural study of cutaneous and oral microbial symbionts.
J. Invest. Dermatol.
96
:
439
445
.
47
Mahmoud
,
F.
,
H.
Abul
,
Q.
al Saleh
,
H.
Hassab-el Naby
,
M.
Kajeji
,
D.
Haines
,
J.
Burleson
,
G.
Morgan
.
1999
.
Elevated B-lymphocyte levels in lesional tissue of non-arthritic psoriasis.
J. Dermatol.
26
:
428
433
.
48
Bosello
,
S.
,
C.
Angelucci
,
G.
Lama
,
S.
Alivernini
,
G.
Proietti
,
B.
Tolusso
,
G.
Sica
,
E.
Gremese
,
G.
Ferraccioli
.
2018
.
Characterization of inflammatory cell infiltrate of scleroderma skin: B cells and skin score progression.
Arthritis Res. Ther.
20
:
75
.
49
O’Brien
,
J. C.
,
G. A.
Hosler
,
B. F.
Chong
.
2017
.
Changes in T cell and B cell composition in discoid lupus erythematosus skin at different stages.
J. Dermatol. Sci.
85
:
247
249
.
50
Hussein
,
M. R.
,
N. M.
Aboulhagag
,
H. S.
Atta
,
S. M.
Atta
.
2008
.
Evaluation of the profile of the immune cell infiltrate in lichen planus, discoid lupus erythematosus, and chronic dermatitis.
Pathology
40
:
682
693
.
51
Roguedas
,
A. M.
,
J. O.
Pers
,
G.
Lemasson
,
V.
Devauchelle
,
G. J.
Tobón
,
A.
Saraux
,
L.
Misery
,
P.
Youinou
.
2010
.
Memory B-cell aggregates in skin biopsy are diagnostic for primary Sjögren’s syndrome.
J. Autoimmun.
35
:
241
247
.
52
Czarnowicki
,
T.
,
J.
Gonzalez
,
K. M.
Bonifacio
,
A.
Shemer
,
P.
Xiangyu
,
N.
Kunjravia
,
D.
Malajian
,
J.
Fuentes-Duculan
,
H.
Esaki
,
S.
Noda
, et al.
2016
.
Diverse activation and differentiation of multiple B-cell subsets in patients with atopic dermatitis but not in patients with psoriasis.
J. Allergy Clin. Immunol.
137
:
118
129.e5
.
53
van Beek
,
N.
,
F. S.
Schulze
,
D.
Zillikens
,
E.
Schmidt
.
2016
.
IgE-mediated mechanisms in bullous pemphigoid and other autoimmune bullous diseases.
Expert Rev. Clin. Immunol.
12
:
267
277
.
54
Nagel
,
A.
,
M.
Hertl
,
R.
Eming
.
2009
.
B-cell-directed therapy for inflammatory skin diseases.
J. Invest. Dermatol.
129
:
289
301
.
55
Thiebaut
,
M.
,
D.
Launay
,
S.
Rivière
,
T.
Mahévas
,
S.
Bellakhal
,
E.
Hachulla
,
O.
Fain
,
A.
Mekinian
.
2018
.
Efficacy and safety of rituximab in systemic sclerosis: French retrospective study and literature review.
Autoimmun. Rev.
17
:
582
587
.
56
Joly
,
P.
,
M.
Maho-Vaillant
,
C.
Prost-Squarcioni
,
V.
Hebert
,
E.
Houivet
,
S.
Calbo
,
F.
Caillot
,
M. L.
Golinski
,
B.
Labeille
,
C.
Picard-Dahan
, et al
French study group on autoimmune bullous skin diseases
.
2017
.
First-line rituximab combined with short-term prednisone versus prednisone alone for the treatment of pemphigus (Ritux 3): a prospective, multicentre, parallel-group, open-label randomised trial.
Lancet
389
:
2031
2040
.
57
Hasegawa
,
M.
,
Y.
Hamaguchi
,
K.
Yanaba
,
J. D.
Bouaziz
,
J.
Uchida
,
M.
Fujimoto
,
T.
Matsushita
,
Y.
Matsushita
,
M.
Horikawa
,
K.
Komura
, et al
.
2006
.
B-lymphocyte depletion reduces skin fibrosis and autoimmunity in the tight-skin mouse model for systemic sclerosis.
Am. J. Pathol.
169
:
954
966
.
58
Jordan
,
S.
,
J. H.
Distler
,
B.
Maurer
,
D.
Huscher
,
J. M.
van Laar
,
Y.
Allanore
,
O.
Distler
;
EUSTAR Rituximab study group
.
2015
.
Effects and safety of rituximab in systemic sclerosis: an analysis from the European Scleroderma Trial and Research (EUSTAR) group.
Ann. Rheum. Dis.
74
:
1188
1194
.
59
Matsushita
,
T.
,
T.
Kobayashi
,
K.
Mizumaki
,
M.
Kano
,
T.
Sawada
,
M.
Tennichi
,
A.
Okamura
,
Y.
Hamaguchi
,
Y.
Iwakura
,
M.
Hasegawa
, et al
.
2018
.
BAFF inhibition attenuates fibrosis in scleroderma by modulating the regulatory and effector B cell balance.
Sci. Adv.
4
:
eaas9944
.
60
Jimenez-Boj
,
E.
,
T. A.
Stamm
,
M.
Sadlonova
,
J.
Rovensky
,
H.
Raffayová
,
B.
Leeb
,
K. P.
Machold
,
W. B.
Graninger
,
J. S.
Smolen
.
2012
.
Rituximab in psoriatic arthritis: an exploratory evaluation.
Ann. Rheum. Dis.
71
:
1868
1871
.
61
Kersh
,
A. E.
,
R. J.
Feldman
.
2018
.
Autoimmune sequelae following rituximab therapy: a review of the literature and potential immunologic mechanisms.
J. Clin. Rheumatol.
24
:
427
435
.
62
Guttman-Yassky
,
E.
,
J. G.
Krueger
.
2017
.
Atopic dermatitis and psoriasis: two different immune diseases or one spectrum?
Curr. Opin. Immunol.
48
:
68
73
.
63
Hammers
,
C. M.
,
J. R.
Stanley
.
2016
.
Mechanisms of disease: pemphigus and bullous pemphigoid.
Annu. Rev. Pathol.
11
:
175
197
.
64
Ahmed
,
A. R.
,
M.
Carrozzo
,
F.
Caux
,
N.
Cirillo
,
M.
Dmochowski
,
A. E.
Alonso
,
R.
Gniadecki
,
M.
Hertl
,
M. J.
López-Zabalza
,
R.
Lotti
, et al
.
2016
.
Monopathogenic vs multipathogenic explanations of pemphigus pathophysiology.
Exp. Dermatol.
25
:
839
846
.
65
Anhalt
,
G. J.
,
R. S.
Labib
,
J. J.
Voorhees
,
T. F.
Beals
,
L. A.
Diaz
.
1982
.
Induction of pemphigus in neonatal mice by passive transfer of IgG from patients with the disease.
N. Engl. J. Med.
306
:
1189
1196
.
66
Wallim
,
L. R.
,
R.
Nisihara
,
T.
Skare
,
V.
Mocelin
,
I. J.
Messias-Reason
.
2014
.
Mannose binding lectin deposition in skin of lupus erythematosus patients: a case series.
Hum. Immunol.
75
:
629
632
.
67
Grunwald
,
M. H.
,
I.
Avinoach
,
B.
Amichai
,
S.
Halevy
.
1997
.
Leukocytoclastic vasculitis--correlation between different histologic stages and direct immunofluorescence results.
Int. J. Dermatol.
36
:
349
352
.
68
Ruddle
,
N. H.
2016
.
High endothelial venules and lymphatic vessels in tertiary lymphoid organs: characteristics, functions, and regulation.
Front. Immunol.
7
:
491
.
69
Jones
,
G. W.
,
D. G.
Hill
,
S. A.
Jones
.
2016
.
Understanding immune cells in tertiary lymphoid organ development: it is all starting to come together.
Front. Immunol.
7
:
401
.
70
Alsughayyir
,
J.
,
G. J.
Pettigrew
,
R.
Motallebzadeh
.
2017
.
Spoiling for a fight: B lymphocytes as initiator and effector populations within tertiary lymphoid organs in autoimmunity and transplantation.
Front. Immunol.
8
:
1639
.
71
Shen
,
P.
,
S.
Fillatreau
.
2015
.
Antibody-independent functions of B cells: a focus on cytokines.
Nat. Rev. Immunol.
15
:
441
451
.
72
Hahn
,
B. H.
2013
.
Belimumab for systemic lupus erythematosus.
N. Engl. J. Med.
368
:
1528
1535
.
73
Molnarfi
,
N.
,
U.
Schulze-Topphoff
,
M. S.
Weber
,
J. C.
Patarroyo
,
T.
Prod’homme
,
M.
Varrin-Doyer
,
A.
Shetty
,
C.
Linington
,
A. J.
Slavin
,
J.
Hidalgo
, et al
.
2013
.
MHC class II-dependent B cell APC function is required for induction of CNS autoimmunity independent of myelin-specific antibodies.
J. Exp. Med.
210
:
2921
2937
.
74
Young
,
J. S.
,
T.
Wu
,
Y.
Chen
,
D.
Zhao
,
H.
Liu
,
T.
Yi
,
H.
Johnston
,
J.
Racine
,
X.
Li
,
A.
Wang
, et al
.
2012
.
Donor B cells in transplants augment clonal expansion and survival of pathogenic CD4+ T cells that mediate autoimmune-like chronic graft-versus-host disease.
J. Immunol.
189
:
222
233
.
75
Candando
,
K. M.
,
J. M.
Lykken
,
T. F.
Tedder
.
2014
.
B10 cell regulation of health and disease.
Immunol. Rev.
259
:
259
272
.
76
Mauri
,
C.
,
M.
Menon
.
2017
.
Human regulatory B cells in health and disease: therapeutic potential.
J. Clin. Invest.
127
:
772
779
.
77
Katz
,
S. I.
,
D.
Parker
,
J. L.
Turk
.
1974
.
B-cell suppression of delayed hypersensitivity reactions.
Nature
251
:
550
551
.
78
Neta
,
R.
,
S. B.
Salvin
.
1974
.
Specific suppression of delayed hypersensitivity: the possible presence of a suppressor B cell in the regulation of delayed hypersensitivity.
J. Immunol.
113
:
1716
1725
.
79
Mauri
,
C.
,
M.
Menon
.
2015
.
The expanding family of regulatory B cells.
Int. Immunol.
27
:
479
486
.
80
Ray
,
A.
,
B. N.
Dittel
.
2017
.
Mechanisms of regulatory B cell function in autoimmune and inflammatory diseases beyond IL-10.
J. Clin. Med.
DOI: 10.3390/JCM6010012.
81
Yanaba
,
K.
,
J. D.
Bouaziz
,
K. M.
Haas
,
J. C.
Poe
,
M.
Fujimoto
,
T. F.
Tedder
.
2008
.
A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses.
Immunity
28
:
639
650
.
82
Nakashima
,
H.
,
Y.
Hamaguchi
,
R.
Watanabe
,
N.
Ishiura
,
Y.
Kuwano
,
H.
Okochi
,
Y.
Takahashi
,
K.
Tamaki
,
S.
Sato
,
T. F.
Tedder
,
M.
Fujimoto
.
2010
.
CD22 expression mediates the regulatory functions of peritoneal B-1a cells during the remission phase of contact hypersensitivity reactions.
J. Immunol.
184
:
4637
4645
.
83
Yanaba
,
K.
,
M.
Kamata
,
N.
Ishiura
,
S.
Shibata
,
Y.
Asano
,
Y.
Tada
,
M.
Sugaya
,
T.
Kadono
,
T. F.
Tedder
,
S.
Sato
.
2013
.
Regulatory B cells suppress imiquimod-induced, psoriasis-like skin inflammation.
J. Leukoc. Biol.
94
:
563
573
.
84
Alrefai
,
H.
,
K.
Muhammad
,
R.
Rudolf
,
D. A.
Pham
,
S.
Klein-Hessling
,
A. K.
Patra
,
A.
Avots
,
V.
Bukur
,
U.
Sahin
,
S.
Tenzer
, et al
.
2016
.
NFATc1 supports imiquimod-induced skin inflammation by suppressing IL-10 synthesis in B cells. [Published erratum appears in 2016 Nat. Commun. 7: 12421.]
Nat. Commun.
7
:
11724
.
85
Maseda
,
D.
,
K. M.
Candando
,
S. H.
Smith
,
I.
Kalampokis
,
C. T.
Weaver
,
S. E.
Plevy
,
J. C.
Poe
,
T. F.
Tedder
.
2013
.
Peritoneal cavity regulatory B cells (B10 cells) modulate IFN-γ+CD4+ T cell numbers during colitis development in mice.
J. Immunol.
191
:
2780
2795
.
86
Byrne
,
S. N.
,
G. M.
Halliday
.
2005
.
B cells activated in lymph nodes in response to ultraviolet irradiation or by interleukin-10 inhibit dendritic cell induction of immunity.
J. Invest. Dermatol.
124
:
570
578
.
87
Matsumura
,
Y.
,
S. N.
Byrne
,
D. X.
Nghiem
,
Y.
Miyahara
,
S. E.
Ullrich
.
2006
.
A role for inflammatory mediators in the induction of immunoregulatory B cells.
J. Immunol.
177
:
4810
4817
.
88
Kok
,
L. F.
,
F.
Marsh-Wakefield
,
J. E.
Marshall
,
C.
Gillis
,
G. M.
Halliday
,
S. N.
Byrne
.
2016
.
B cells are required for sunlight protection of mice from a CNS-targeted autoimmune attack.
J. Autoimmun.
73
:
10
23
.
89
Nickoloff
,
B. J.
,
D. P.
Fivenson
,
S. L.
Kunkel
,
R. M.
Strieter
,
L. A.
Turka
.
1994
.
Keratinocyte interleukin-10 expression is upregulated in tape-stripped skin, poison ivy dermatitis, and Sezary syndrome, but not in psoriatic plaques.
Clin. Immunol. Immunopathol.
73
:
63
68
.
90
Asadullah
,
K.
,
W.
Sterry
,
K.
Stephanek
,
D.
Jasulaitis
,
M.
Leupold
,
H.
Audring
,
H. D.
Volk
,
W. D.
Döcke
.
1998
.
IL-10 is a key cytokine in psoriasis. Proof of principle by IL-10 therapy: a new therapeutic approach.
J. Clin. Invest.
101
:
783
794
.
91
Hayashi
,
M.
,
K.
Yanaba
,
Y.
Umezawa
,
Y.
Yoshihara
,
S.
Kikuchi
,
Y.
Ishiuji
,
H.
Saeki
,
H.
Nakagawa
.
2016
.
IL-10-producing regulatory B cells are decreased in patients with psoriasis.
J. Dermatol. Sci.
81
:
93
100
.
92
Mavropoulos
,
A.
,
A.
Varna
,
E.
Zafiriou
,
C.
Liaskos
,
I.
Alexiou
,
A.
Roussaki-Schulze
,
M.
Vlychou
,
C.
Katsiari
,
D. P.
Bogdanos
,
L. I.
Sakkas
.
2017
.
IL-10 producing Bregs are impaired in psoriatic arthritis and psoriasis and inversely correlate with IL-17- and IFNγ-producing T cells.
Clin. Immunol.
184
:
33
41
.
93
Kabuto
,
M.
,
N.
Fujimoto
,
T.
Tanaka
.
2016
.
Increase of interleukin-10-producing B cells associated with long-term remission after i.v. immunoglobulin treatment for pemphigus.
J. Dermatol.
43
:
815
818
.
94
Kabuto
,
M.
,
N.
Fujimoto
,
T.
Takahashi
,
T.
Tanaka
.
2017
.
Decreased level of interleukin-10-producing B cells in patients with pemphigus but not in patients with pemphigoid.
Br. J. Dermatol.
176
:
1204
1212
.
95
Matsushita
,
T.
,
Y.
Hamaguchi
,
M.
Hasegawa
,
K.
Takehara
,
M.
Fujimoto
.
2016
.
Decreased levels of regulatory B cells in patients with systemic sclerosis: association with autoantibody production and disease activity.
Rheumatology (Oxford)
55
:
263
267
.
96
Mavropoulos
,
A.
,
T.
Simopoulou
,
A.
Varna
,
C.
Liaskos
,
C. G.
Katsiari
,
D. P.
Bogdanos
,
L. I.
Sakkas
.
2016
.
Breg cells are numerically decreased and functionally impaired in patients with systemic sclerosis.
Arthritis Rheumatol.
68
:
494
504
.
97
Colliou
,
N.
,
D.
Picard
,
F.
Caillot
,
S.
Calbo
,
S.
Le Corre
,
A.
Lim
,
B.
Lemercier
,
B.
Le Mauff
,
M.
Maho-Vaillant
,
S.
Jacquot
, et al
.
2013
.
Long-term remissions of severe pemphigus after rituximab therapy are associated with prolonged failure of desmoglein B cell response.
Sci. Transl. Med.
5
:
175ra30
.
98
Yang
,
X.
,
J.
Yang
,
Y.
Chu
,
Y.
Xue
,
D.
Xuan
,
S.
Zheng
,
H.
Zou
.
2014
.
T follicular helper cells and regulatory B cells dynamics in systemic lupus erythematosus.
PLoS One
9
:
e88441
.
99
Lykken
,
J. M.
,
K. M.
Candando
,
T. F.
Tedder
.
2015
.
Regulatory B10 cell development and function.
Int. Immunol.
27
:
471
477
.
100
Trinchieri
,
G.
2007
.
Interleukin-10 production by effector T cells: Th1 cells show self control.
J. Exp. Med.
204
:
239
243
.
101
Maynard
,
C. L.
,
C. T.
Weaver
.
2008
.
Diversity in the contribution of interleukin-10 to T-cell-mediated immune regulation.
Immunol. Rev.
226
:
219
233
.
102
Sun
,
J.
,
R.
Madan
,
C. L.
Karp
,
T. J.
Braciale
.
2009
.
Effector T cells control lung inflammation during acute influenza virus infection by producing IL-10.
Nat. Med.
15
:
277
284
.
103
Cowin
,
A. J.
,
M. P.
Brosnan
,
T. M.
Holmes
,
M. W.
Ferguson
.
1998
.
Endogenous inflammatory response to dermal wound healing in the fetal and adult mouse.
Dev. Dyn.
212
:
385
393
.
104
Richards
,
A. M.
,
D. C.
Floyd
,
G.
Terenghi
,
D. A.
McGrouther
.
1999
.
Cellular changes in denervated tissue during wound healing in a rat model.
Br. J. Dermatol.
140
:
1093
1099
.
105
Iwata
,
Y.
,
A.
Yoshizaki
,
K.
Komura
,
K.
Shimizu
,
F.
Ogawa
,
T.
Hara
,
E.
Muroi
,
S.
Bae
,
M.
Takenaka
,
T.
Yukami
, et al
.
2009
.
CD19, a response regulator of B lymphocytes, regulates wound healing through hyaluronan-induced TLR4 signaling.
Am. J. Pathol.
175
:
649
660
.
106
Rickert
,
R. C.
,
K.
Rajewsky
,
J.
Roes
.
1995
.
Impairment of T-cell-dependent B-cell responses and B-1 cell development in CD19-deficient mice.
Nature
376
:
352
355
.
107
Martin
,
F.
,
J. F.
Kearney
.
2000
.
Positive selection from newly formed to marginal zone B cells depends on the rate of clonal production, CD19, and btk.
Immunity
12
:
39
49
.
108
Sato
,
S.
,
N.
Ono
,
D. A.
Steeber
,
D. S.
Pisetsky
,
T. F.
Tedder
.
1996
.
CD19 regulates B lymphocyte signaling thresholds critical for the development of B-1 lineage cells and autoimmunity.
J. Immunol.
157
:
4371
4378
.
109
Sîrbulescu
,
R. F.
,
C. K.
Boehm
,
E.
Soon
,
M. Q.
Wilks
,
I.
Ilieş
,
H.
Yuan
,
B.
Maxner
,
N.
Chronos
,
C.
Kaittanis
,
M. D.
Normandin
, et al
.
2017
.
Mature B cells accelerate wound healing after acute and chronic diabetic skin lesions.
Wound Repair Regen.
25
:
774
791
.

The authors have no financial conflicts of interest.