Abstract
Our understanding of B cells as merely antibody producers is slowly changing. Alone or in concert with antibody, they control outcomes of seemingly different diseases such as cancer, rheumatoid arthritis, diabetes, and multiple sclerosis. While their role in activation of effector immune cells is beneficial in cancer but bad in autoimmune diseases, their immunosuppressive and regulatory subsets (Bregs) inhibit autoimmune and anticancer responses. These pathogenic and suppressive functions are not static and appear to be regulated by the nature and strength of inflammation. Although aging increases inflammation and changes the composition and function of B cells, surprisingly, little is known whether the change affects aging-associated neurodegenerative disease, such as Alzheimer’s disease (AD). Here, by analyzing B cells in cancer and autoimmune and neuroinflammatory diseases, we elucidate their potential importance in AD and other aging-associated neuroinflammatory diseases.
Keywords: B-cell, Bregs, Alzheimer’s disease, Neurodegeneration
Introduction
Aging often leads to decline in the brain functions and memory loss. It is a risk factor for the development of Alzheimer’s disease (AD), a progressive neurodegenerative disorder of the elderly. Per amyloid cascade theory, an aberrant cleavage of the amyloid precursor protein (APP) with the gamma (γ) secretase complex results in brain accumulation of amyloid β-peptides (Aβ), which in turn induces a chain of inflammatory events, such as deposition of neurofibrillary tangles (NFTs) of hyper phosphorylating tau protein and activation of microglia (MG) and astrocytes [1], leading to gradual synaptic dysfunction and neuronal degeneration in multiple interconnected brain regions [2, 3]. This is also an inflammatory disease that involves upregulated and sustained expression of IFN-I, IFNγ, IL-12, IL-23, TNFα, IL-6, and IL-1β [4, 5] produced locally by activated MG and astrocytes [6–9] and by peripheral innate immune cells, such as MO, neutrophils, NK cells, and perivascular macrophages [10–12]. They disrupt the integrity of blood-brain-barrier (BBB); impair MG functions, such as phagocytosis and removal of Aβ deposits; reduce adult hippocampal neurogenesis; and exacerbate cognitive decline [13–19]. As in cancer and autoimmune diseases, production and activity of proinflammatory cytokines is regulated by immunosuppressive cytokine IL-10 [20] and TGFβ1 [21, 22]. Healthy adult CNS also constitutively expresses TGFβ2 and 3 [23], while TGFβ1 is induced in AD brains [24] from Aβ-stimulated MG and astrocytes [25]. It protects neurons from Aβ toxicity [26] and its deficiency leads to neuronal death and microgliosis [21].
As a double-edged sword, inflammation is also needed for the activation of MG to clear Aβ [11, 27] and to promote adult hippocampal neurogenesis by monocytes [10]. T cells infiltrate the CNS [28, 29] as important regulators of the maintenance and adult neurogenesis [30]. However, upon their activation, for example, by myelin olygodendrocyte protein (MOG), they promote multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE, a murine model for MS). Similarly, infiltration of pathogenic IFNγ and TNFα-producing T cells (Th1) in the brain parenchyma [31] is thought to cause cerebral vasculature leakage [32] and meningoencephalitis in some patients with AD immunized with Aβ vaccine [33, 34]. The loss of IFNγ signaling in AD mice or T cells in RAG-deficient AD mice blocks MG activation and Aβ deposition [18]. Conversely, regulatory T cells (Tregs, which block Th1 responses) can reverse Aβ-induced brain inflammation and impaired cognition in AD mice [35], presumably explaining the inverse correlation between Treg presence and mild cognitive impairment in AD patients [36]. However, Tregs can instead promote AD in 5xfAD mice, as they block IFNγ-dependent beneficial gateway of the brain’s choroid plexus (CP) needed for the infiltration of inflammation-resolving immune cells in the CNS [37]. Tregs also crosstalk with B cells and their regulatory subsets (Bregs), an important group of adaptive immune cells that actively control outcomes of cancer and autoimmune diseases.
The role of B cells in AD remains poorly understood and even thought to be inessential [38] despite the fact that they are implicated in neuropsychiatric disorders [39] and depressive-like symptoms [40, 41] and that Aβ is immunogenic and induces antibody (Ab) response in AD patients and mice. Passive administration of Aβ-specific Ab [42, 43] or active immunization with Aβ can reduce Aβ deposits in AD patients [34, 44] and mice ameliorating severity of the disease [45–47]. Even non-amyloid IgG is recently shown to be beneficial, as it activates MG phagocytosis and thereby reduces Aβ deposits and, conversely, its loss in RAG-5xfAD (due to its inability to generate mature B cells besides T cells and NK cells) accelerates AD [48]. On the other hand, IgG and its immune complexes can induce harmful activation of MG and neuronal neurotoxicity, for example, upon inflammation in old age. Aging not only increases inflammation but also dysregulates the composition and function of immune cells. By reducing B-cell lymphopoiesis and naive B cells, it impairs Ab response to neoantigens and pathogens while enriching for B7-DC+ ABC B cells that induce polarization of Th17 and Th1 cells [49]. Recently, we found that old humans, macaques, and mice also accumulate a different type of potentially pathogenic B cells [50]. These cells (designated 4BL cells) express high levels of 4-1BBL and membrane (m)TNFα and belong to innate B1a cells, which upon aging lost their immunosuppressive function and instead acquired a superb ability to induce cytolytic CD45RA+ CD8+ T cells to self-antigens [51]. Contrary to young B1a cells that support tumor growth, 4BL cells efficiently retard tumors in old mice. Since young B1a cells and B1a-like B10 cells protect from experimental autoimmune encephalitis (EAE) and colitis [52, 53], our findings raise an interesting question whether aging B/4BL cells can contribute to aging-associated neurodegenerative diseases. To answer this question, here, we analyze the role of B cells in seemingly different diseases, such as cancer and autoimmune and neuroinflammatory diseases, as epidemiological studies positively and negatively correlate them with AD, respectively [54–56]. First, we discuss the role of peripheral inflammation. Then, we analyze the pathogenic and protective activities of humoral immunity in AD. Although almost no report exists on the immunoglobulin-independent function of B cells in AD, to extrapolate their potential importance, we also provide in-depth analysis of B cells and Bregs in clinically relevant diseases, such as cancer and autoimmune diseases, including multiple sclerosis (MS).
AD is an age-associated inflammatory disease
A growing number of studies link function of the CNS with peripheral inflammation and activation of innate immune cells. In healthy brain, monocyte (MO) infiltration in perivascular space, in choroid plexus (CP) and meninges is linked with protective functions [57]. They alone or upon differentiation into perivascular macrophages can reduce Aβ deposits [58] via Aβ efflux by BBB neonatal Fc receptor [59]. As important cells that surveil inflammation, CCR2+ MO activated by normal gut microbiome are thought to be necessary for adult hippocampal neurogenesis [10]. As such, adult hippocampal neurogenesis can be impaired upon loss of CCR2+ MO [10] and commensal microbiota in gem-free mice or after treatment with broad-spectrum antibiotics [60]. Similarly, the CCR2 deficiency reduces MO infiltration in the CNS causing brain microhemorrhages and mortality of transgenic AD mice [10]. However, peripheral inflammation can also disrupt adult hippocampal neurogenesis and induce aging-associated cognitive decline [13–15]. It is a risk factor for AD [61] and various neuropsychiatric disorders [39] and depressive-like symptoms [40, 41]. Peripheral blood (PB) and CNS fluids of AD patients is enriched in proinflammatory cytokines, such as IL-6, IFN, IL-1, and TNFα [4, 5], which activate reactive astrocytes and MG [6, 7]. Note that IFNγ exhibits both pathogenic and protective functions depending on where and what cells produce it. While it activates MG [17] impairing their phagocytosis of Aβ [62], it controls immune cell trafficking into the brain of mice by inhibiting the negative effects of IFN-I in CP [63]. CP [64, 65] and recently discovered brain lymphoid structures [66] are the main gateways of immune cells into the brain.
Neuroinflammation [67] and brain functions in rodents and humans [68, 69] are controlled by gut commensal microbiota, whose pathological role increases in aging. First, aging markedly affects microbial composition in the gut reducing their diversity and shifting the Firmicutes/Bacteroidetes ratio toward inducers of inflammation [70, 71]. A proinflammatory Escherichia/Shigella taxon is increased and an anti-inflammatory taxon, E. rectale is reduced in patients with cognitive impairment and brain Aβ amyloidosis [72]. The microbiome change disrupts intestinal barrier, which is also increased in elderly [73], causing leakage of endotoxin and other factors [74] that impair blood-brain-barrier (BBB), activate MG and astrocytes, and induce accumulation of Aβ deposits [1, 61]. Alone or with T cells in the brain parenchyma [31], peripheral inflammation also triggers production of inflammatory cytokines leading to long-lasting alterations of MG (upregulation of MHCI and II) [75] and cerebral vasculature [32]. As such, IFN-I [16], IFNγ [17, 18], and other proinflammatory cytokines (TNFα and IL-1) are linked with aging cognitive decline, at least in part, as inhibitors of adult hippocampal neurogenesis [13–15].
T cells also produce less IFNγ and do not proliferate in the brain of transgenic AD mice, suggesting that the loss of their immunosurveilling functions may lead to Aβ accumulation and progression of the disease [76]. Moreover, the normal function and maintenance of healthy brain need cytokines. For example, IL-4 from meningeal T cells supports memory maintenances [77] while neuronal and glia-derived TNFα regulates synaptic strength via exocytosis and endocytosis of AMPA and GABA receptors, respectively [78]. IFNγ regulates neuronal connectivity and social behavior via induction of GABA on inhibitory neurons [79]. This normal use of cytokines and their balance shifts from the maintenance to damage during neuroinflammation. While M2 macrophages and microglia produce anti-inflammatory cytokine IL-10 and mediate tissue remodeling, neurogeneration, and remyelination through stimulation of oligodendrocytes [80], peripheral inflammation induces MO-to-M1 macrophage differentiation that exacerbate brain inflammation via producing TNFα, IL-12, IL-23, CXCL9, CXCL10, reactive oxygen, and nitrogen species [81]. The M1/M2 balance and the nature of inflammation affect function of B cells. For example, proinflammatory cytokines IL-1β and IL-6 produced in response to gut microbiota can induce differentiation of IL-10+ Bregs in the spleen and in the mesenteric lymph nodes [82].
Protective and pathogenic roles of immunoglobulin
The B-cell compartment consists of multiple B-cell subsets classified in two main categories: conventional B cells (also known as B2 cells) and the innate-like B1 cells. Mature B2 cells consist of follicular B cells (FO B) and the marginal zone B cells (MZB). B1 cells are represented by B1a (mainly produce IgM) and B1b (produce IgA and IgM) cells that mostly located in the pleural and peritoneal cavities, although a minor population (<5% of B cells) can be found in the spleen. While antibody (Ab) production from B2 cell and some B1b cells requires help of T cells (TD), B1a cells produce Ab in T cell independent fashion. In fact, B1a and marginal zone B cells (MZB) are the main producers of so-called natural Ab that protect from blood-borne bacteria and maintain tissue homeostasis (i.e., removal of apoptotic bodies, mucosal commensal bacteria, intestinal microbiota [83–86]). The importance of B2 cells in production of Aβ neutralizing antibody (Ab) [87, 88] was first noted in HIV patients before the advent of antiretroviral therapies, where about 50% of patients suffered from dementia associated with Aβ accumulation in brain and cerebrospinal fluids [89]. Aging also impairs TD Ab production by reducing lymphopoiesis and subsequently, naïve T and B cells and increasing antigen-experienced memory and mature conventional B2 cells [90–93]. It also increases B1 cells and production of natural Ab [94, 95].
Ab can infiltrate into healthy brain (albeit inefficiently, only 0.1% of IgG in circulation) by passive diffusion through BBB [96, 97], which is enhanced upon BBB disruption in systemic inflammation, aging, and AD [98, 99]. The Ab influx in the brain is a double-edged sword. On one hand, as first proposed by Morris and Muller in the late 1960s [100], immunoglobulin (Ig) is a potent and beneficial “non-specific” immune suppressor in autoimmune diseases [101–103]. It can induce inhibitory signaling from FcγRIIB in target cells [104] or activate FcγR on DCs to produce immune-suppressive factors [105]. In AD mice, as shown in an elegant study by Marsh et al. [48], non-amyloid reactive IgG promotes Aβ clearance upon association and activation of MG phagocytosis. Thus, the ablation of B cells in RAG enzyme-deficient 5xfAD mice markedly increases β-amyloid pathology. This probably explains the benefit of intracranially injected non-specific IgG in Aβ clearance in AD transgenic mice [106] and patients with AD [107, 108]. Despite its failure to improve cognition in Phase II trial, infusion of IgG (the formulation designated Gammagard) significantly reduced Aβ load and cognitive decline in a subset of patients with ApoE4 risk allele [107, 108]. Similarly, a passive administration of Aβ-specific Ab [42, 43] or an active immunization with Aβ(1–42) vaccine can also be beneficial in AD patients [34, 44] and mice [45, 46]. For example, as we reported, Aβ vaccine alleviates AD pathology and thereby reverse the AD-associated mortality in 3xTg-AD mice that develop onset of the disease late in life [47]. On the other hand, it can also be expected that Ab is harmful in AD, as the disease as well as systemic inflammation and aging upregulate Fc receptors in perivascular macrophages and resident cells in the CNS, such as MG and neurons [97]. Their ligation with therapeutic and endogenous Ab can lead to neurodegeneration via inducing a harmful inflammation [109, 110]. Auto-Ab to glutamatergic and dopaminergic neurons can exacerbate AD and Parkinson disease (PD) [111], while Ab to the synaptic NMDA receptor is thought to contribute to dementia and psychosis in patients with progressive cognitive dysfunction [112, 113].
The pathogenic role of B cells
Independently of Ab, B cells exhibit effector functions depending on the nature and strength of a signal/inflammation and immune environment, such as differentially expressed Th1- and Th2-type cytokines [114]. By producing IFNγ and TNFα, B cells activate inflammatory macrophages (Mϕ) and inhibit regulatory immune cells [115, 116], while MZB and B1a cells use IL-10 in immunosuppression [117, 118]. As antigen-presenting cells [119], B cells present cognate antigens (Ag) in activation of CD4+ T cells and CD8+ T cells [120, 121] and lipid Ag on their CD1d in induction of NKT cells [122]. By producing cytokines, they facilitate differentiation of CD8+ T cells [123], Th1, Th2, Th17, Tfh, and Treg cells [124–128]. However, whether these functions are affected in aging remains poorly understood. Old mice are enriched in anergic and exhausted age-associated B cells (CD21−CD23−ABCs) [129] and B7-DC+ ABC B cells that induce Th17 and Th1 cells [49]. We recently discovered a different type of B cells that accumulate in elderly humans, macaques, and mice, the activated B1a cells expressing 4-1BBL and mTNFα (designated 4BL cells) [50, 51]. While 4-1BBL is only transiently expressed in normal B cells after a combined stimulation with BCR and CD40 [130], this TNF superfamily type 2 transmembrane protein is usually expressed by Mϕ and DC [131] to activate NK, CD8 T cells and splenic DCs upon engagement with its receptor 4-1BB/CD137 [132–135]. In synergy with B7 and ICAM, it can also induce proliferation of naïve CD4+ T cells when antigen is limiting [136]. Despite unaltered 4-1BBL+ antigen-presenting cells, such as DCs, surprisingly, 4BL cells function as the main inducers of antigen-specific perforin+ GrB+ CD8+ T cells in aging [50, 51], explaining the accumulation of highly differentiated GrB+ CD45RA+ CD8+ T cells often detected in elderly humans [90].
As in aging, PB of patients with moderate-severe AD is also enriched in memory B cells such as IgG+ CD27−IgD−B cells [137], although a clinical relevance of this increase remains unknown. B cells are pathogenic in MS/EAE, lupus erythematosus (SLE), rheumatoid arthritis (RA), type 1 (T1D) and type 2 (T2D) diabetes mellitus, and Graves disease (GD), as their depletion with anti-CD20 Ab (rituximab) ameliorates these diseases [138–140]. Similarly, the ablation of memory CD27+ B cells, CD19HighCXCR3+ B cells, or autoreactive memory IgM+ B cells [140–143] can also alleviate these diseases due to, at least in part, reduction of self-reactive and pathogenic Ab and T cells [144]. The CD27+ CD19+ CD138+ or CD138− memory B cells are present in CSF of MS patients, which can be found on both sides of BBB [145]. Ex vivo stimulation with myelin olygodendrocyte protein (MOG) induces higher levels of TNFα, IL-12, and IFNγ in B cells from primary progressive MS as compared to relapsing-remitting MS (RRMS) or benign MS patients [146]. By presenting soluble or attached to Mϕ and DC antigens [147, 148], B cell prime naïve CD4+ T cells [149] to low antigen concentrations [150] and induce Th1 and Th17 cells implicated in the CNS autoimmunity [151, 152]. The MHC class II+ B cell-induced activation of myelin-specific T cells [153] and subsequent demyelination and axonal loss in MS/EAE [154] are reversed upon B-cell depletion or if mice are B-cell deficient [119, 153]. B cells also promote stroke-associated dementia in mice [155] and humans even the decade following the stroke [156, 157].
The protective role of B cells
As originally reported by Shimamura and colleagues about 30 years ago in mice immunized with a high dose of sheep erythrocytes [158], B cells can also elicit immunosuppression and thereby protect from autoimmunity. Thus, loss of B cells [159] or their essential components (CD19) [160] can instead promote autoimmune diseases and EAE. This protective function is, at least in part, mediated by Bregs (the definition first used by Mizoguchi to describe protective B cells in mice with colitis [161]), a diverse group of B cells that regulate effector immune responses by promoting tolerance of CD4+ and CD8+ T cells [162, 163] and inducing Tregs and other immune suppressive cells [164–166] and Tr1 cells [167]. To date, they are represented by murine B10 regulatory cells (CD1dHighCD5+ B cells) [160], T2-MZP Bregs (CD43−CD21HighCD23LowCD2High IgD+ IgM+ CD93IntCD1dHighCD19+ transitional-2 marginal zone B-cell precursor) [168], CD1dHighB1b cells (CD5−B220LowCD11b+ CD23−CD24+ CD62L+ IgM+ CD1dHigh) [118, 161], Tim-1+ CD1dHighCD5+ Bregs [169], IL-33+ Bregs (CD19+ CD25+ CD1dHighIgMHigh CD5−CD23−Tim1−) [170], and IL-35+ Bregs (CD1dHighCD5+ B220+ B cells) [171]. The loss of Bregs or suppressive B cells in humans also lead to autoimmune diseases, such as CD19+ CD24High CD38High B cells [172], IL-10 producing memory CD24HighCD27+ B-cells (a human counterpart of B10 cells) [173], IL-10 and TGFβ-producing CD25HighCD27HighCD86HighCD1dHigh B cells [174], and GrB+ CD25+ B cells [175]. The cerebrospinal fluid of MS patients is enriched in perforin-expressing FoxP3+ CD25+ CD19+ B cells (Bregs) [176].
Cancer also uses Bregs, although not every subset reported in autoimmune protection. T2-MZP Bregs are increased and negatively associate with cytolytic CD8+ T cells in mice with squamous carcinoma induced by 7, 12-dimethylbenz[α] anthracene/terephtalic acid (MBA/TPA) [177] and human colorectal cancer [178]. It also generates unique cancer-associated Bregs, such as GrB+ Bregs in humans with breast, ovarian, colorectal, and prostate carcinomas [179]; IL-10 and TGFβ-expressing FoxP3+ CD5+ CD19+ B cells that accumulate in patients with esophageal cancer [180]; and CD25+ CD20Low tBregs we originally found in BALB/c mice with 4T1 breast cancer [128] and C57BL/6 mice with spontaneous ovarian cancer [181] and in humans with B-CLL [182]. tBregs differ from other Bregs and LPS- or BCR-activated suppressive B cells [183, 184], as they are poorly proliferative CD25HighB7-H1HighCD81HighCD86HighCD20LowCD62LLowIgMInt/Low B2 cells that express constitutively active Stat3 without detectable CD5. Their regulatory activity does not require B7-H1/PD1, Fas/FasL, IL-10, IL-27, and IL-35. While suppressive activity of most Bregs is enhanced after stimulation of Toll-like receptors (TLRs), the TLR-9 stimulation instead converts tBregs into inducers of cytolytic CD8+ T cells [128].
Most suppressive B cells/Bregs use IL-10 to directly or indirectly (via inducing other regulatory immune cells) regulate immune responses. Thus, IL-10 deficiency alone can lead to spontaneous autoimmunity due to hyper activation of T cells [185], as it is an important immunoregulatory cytokine that promotes Th2-type cytokine responses and inhibits Th1, Th17, CD8+ T, and NK cells [52, 160, 168, 172, 173, 186–188]. It is also a prosurvival factor for murine and human B1 cells [189–191]. B1a cells, B10 cells, and MZB require stimulation with TLR ligands, IL-21, or IL-35 to upregulate IL-10 and thereby efficiently protect mice from EAE, experimental autoimmune uveitis (EAU), lupus, and collagen-induced arthritis [117, 171, 192–195]. Bregs also inhibit autoimmune and antitumor CD8+ T cells utilizing IL-10 [188, 196]. B cells/Bregs use IL-10 in regulation of MO and Mϕ [197]. B10 cells downregulate surface FcγR on Mϕ using IL-10 and retard therapeutic efficacy of B-cell lymphoma-depleting antibody [198]. By utilizing IL-10 and natural Ab, cancer-associated B cells and B1 cells induce M2 polarization of Mϕ and TAMs [199, 200]. We recently reported that myeloid-derived suppressive cells (MDSCs), the key inhibitors of antitumor effector cells [201], also require cancer B cells/tBregs [181]. tBregs evoke regulatory and thereby prometastatic function of MDSCs via triggering the TgfβR1/TgfβR2 signaling axis [181]. Thus, loss of tBregs or their downstream Tregs is sufficient to abrogate lung metastasis despite unaltered expansion of MDSCs in response to cancer [128, 182, 202]. Bregs also mediate IL-10-independednt regulation [203, 204]. Human GrB+ Bregs disable antitumor T cells by degrading their TCR-ζ with granzyme B [179]. Activated B cells express TGFβ [186, 205], which alone or as a complex with Ig is immunosuppressive [206, 207]. They also produce galectin-1 and other potentially cytotoxic factors that induce apoptosis and anergy Th1 cells and CD8+ T cells [205, 208–210]. Most human CD25HighCD27HighCD86HighCD1dHighCD19+ B cells [174] and murine B cells and Bregs utilize TGFβ and IL-10 to induce FoxP3+ CD25+ CD4+ Tregs and iTregs, respectively [128, 166, 174, 205, 211, 212]. However, TGFβ is not needed in some CD1d+ Breg-mediated induction of FoxP3+ Tregs [164, 213]. They mostly used IL-10 [164, 213] possibly to maintain FoxP3 expression [214]. IL-10 also induces Tr1 cells (FoxP3−IL-10High Tregs [167] and effector-memory Tregs (TREM) [215, 216], although it remains unknown whether Bregs can induce them. Bregs also expand Tregs via IL-35 and suppress EAU by inhibiting pathogenic Th-17 and Th1 cells [171].
Despite importance of Bregs in autoimmune diseases and cancer, little is known whether Bregs participate neurodegenerative diseases and, particularly, in AD. The protective role of B cells in EAE, which is originally reported by Wolf and colleagues in 1996 [159], is now expanded to various suppressive B cells and Bregs, such as B10 cells, plasmablasts [52, 187, 217] and IgM+ CD138HighTACI+ CXCR4+ CD1dInt Tim1Int plasma cells [218]. Bregs inhibit activated Mϕ and MOG-specific Th17 and Th1 cells via IL-10 and IL-35 [52, 218], presumably explaining reduction of IL-10+ Bregs and memory CD27+ IgD−CD19+ B cells in PB of patients with RRMS [219]. Conversely, CD25+ CD19+, FoxP3+ CD25+ CD19+, FoxP3+ CD19+, and perforin+ CD25+ CD19+ Bregs increased in PB of non-active RRMS patients as compared to MS patients with active disease [176]. Interestingly, these Bregs appear to function as our tBregs CD25HighCD19+ that promote metastasis [128, 182], as both inhibit proliferation of TCR-activated CD4+ T cells [176] - the feature rarely linked to Bregs. Recently, a new type of BTLA+ Bregs is found in MS patient [146]. They increase upon disease remission and become reduced in active MS. Their function appears to control cytokine production from TCR-induced T cells via BTLA/HVEM axis [146]. Similarly, B10 cells (CD24HighCD38HighCD19+ Bregs) are increased upon rituximab therapy-induced amelioration of myasthenia gravi [220].
Concluding remarks
Peripheral inflammation plays an important role in the pathology of age-associated neurodegenerative diseases and AD, which is mostly thought to involve activated innate immune cells and Th1 cells. However, the role of B cells, which also actively sense and respond to the environmental cues, remains poorly understood and is even considered to be non-essential. The question is why to date almost nothing is known about B cells in AD when they participate in numerous clinically relevant diseases, such as cancer, autoimmune diseases, and MS/EAE and modulate neuroinflammation and CNS injury?
Can it be due to the dysregulation of B cells in aging humans and mice, which accumulate new types of potentially pathogenic B cells not seen in young? If so, the modeling studies in young mice cannot fully capture this change in the context of aging-associated diseases. Thus, unless tested in old mice, the role of 4BL cells (memory/innate B cells), which retard tumor growth by inducing cytolytic GrB+ CD8+ T cells via 4-1BBL/4-1BB axis [50] and B7-DC+ ABC B cells that induce Th17 and Th1 cells [49] in AD will be hidden. One may argue that B cells are not known to infiltrate the brains of mice and humans with AD. Bregs can be missed within the overwhelming numbers of other cells in the CNS due to their low numbers, as for example, they account for less than 10% of B cells in tumor-bearing mice. On the other hand, B cells can remotely affect AD, as they express a full array of cytokines and other factors implicated in this disease. They can induce or impair MG phagocytosis and clearance of Aβ and promote adult hippocampal neurogenesis or neurodegeneration. Thus, the inflammation-activated CNS is expected to differ in responses to humoral factors or/and innate and adaptive immune cells. Harmful Th1 cells can be reactivated by loss of CD19+ CD24High CD38High B cells (Bregs?) in humans with SLE [172] or after depletion of B cells/Bregs with rituximab, causing anecdotal exacerbation of ulcerative colitis and induction of psoriasis in patients with psoriatic arthropathy and colitis, GD, and non-Hodgkin lymphoma [221–224]. In summary, we propose that, as important activators and regulators of almost every cell type involved in AD, including Tregs (whose numbers increase in aging), cytotoxic CD8+ T cells, NK cells, and M1/M2 macrophages [128, 140, 199, 200, 225], B cells can affect the pathogenesis of AD and other age-associated neurodegenerative diseases.
Acknowledgments
This work was funded by Intramural Program of the National Institute on Aging, National Institutes of Health, USA, and Russian Science Foundation grant # 14-15-00077, Center for Brain Neurobiology and Neurogenetics, Russia.
Footnotes
Conflict of interest The authors do not have any conflict of interest.
Reference
- 1.Xie J, Brayne C, Matthews FE (2008) Survival times in people with dementia: analysis from population based cohort study with 14 year follow-up. BMJ 336:258–262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mattson MP (2004) Pathways towards and away from Alzheimer’s disease. Nature 430:631–639 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Golde TE, Dickson D, Hutton M (2006) Filling the gaps in the abeta cascade hypothesis of Alzheimer’s disease. Curr Alzheimer Res 3:421–430 [DOI] [PubMed] [Google Scholar]
- 4.Doecke JD, Laws SM, Faux NG et al. (2012) Blood-based protein biomarkers for diagnosis of Alzheimer disease. Arch Neurol 69: 1318–1325 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ray S, Britschgi M, Herbert C et al. (2007) Classification and prediction of clinical Alzheimer’s diagnosis based on plasma signaling proteins. Nat Med 13:1359–1362 [DOI] [PubMed] [Google Scholar]
- 6.Clark IA, Vissel B (2015) Amyloid beta: one of three danger-associated molecules that are secondary inducers of the proinflammatory cytokines that mediate Alzheimer’s disease. Br J Pharmacol 172:3714–3727 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Heneka MT, Golenbock DT, Latz E (2015) Innate immunity in Alzheimer’s disease. Nat Immunol 16:229–236 [DOI] [PubMed] [Google Scholar]
- 8.Mildner A, Schlevogt B, Kierdorf K et al. (2011) Distinct and non-redundant roles of microglia and myeloid subsets in mouse models of Alzheimer’s disease. J Neurosci 31:11159–11171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Aloisi F, Penna G, Cerase J, Menendez Iglesias B, Adorini L (1997) IL-12 production by central nervous system microglia is inhibited by astrocytes. J Immunol 159:1604–1612 [PubMed] [Google Scholar]
- 10.El Khoury J, Toft M, Hickman SE et al. (2007) Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nat Med 13:432–438 [DOI] [PubMed] [Google Scholar]
- 11.Hawkes CA, McLaurin J (2009) Selective targeting of perivascular macrophages for clearance of beta-amyloid in cerebral amyloid angiopathy. Proc Natl Acad Sci U S A 106:1261–1266 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Solerte SB, Cravello L, Ferrari E, Fioravanti M (2000) Overproduction of IFN-gamma and TNF-alpha from natural killer (NK) cells is associated with abnormal NK reactivity and cognitive derangement in Alzheimer’s disease. Ann N Y Acad Sci 917:331–340 [DOI] [PubMed] [Google Scholar]
- 13.Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O (2003) Inflammation is detrimental for neurogenesis in adult brain. Proc Natl Acad Sci U S A 100:13632–13637 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Monje ML, Toda H, Palmer TD (2003) Inflammatory blockade restores adult hippocampal neurogenesis. Science 302:1760–1765 [DOI] [PubMed] [Google Scholar]
- 15.Liu Q, Xin W, He P et al. (2014) Interleukin-17 inhibits adult hippocampal neurogenesis. Sci Rep. 4:7554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Taylor JM, Minter MR, Newman AG, Zhang M, Adlard PA, Crack PJ (2014) Type-1 interferon signaling mediates neuro-inflammatory events in models of Alzheimer’s disease. Neurobiol Aging 35:1012–1023 [DOI] [PubMed] [Google Scholar]
- 17.Meda L, Cassatella MA, Szendrei GI et al. (1995) Activation of microglial cells by beta-amyloid protein and interferon-gamma. Nature 374:647–650 [DOI] [PubMed] [Google Scholar]
- 18.Browne TC, McQuillan K, McManus RM, O’Reilly JA, Mills KH, Lynch MA (2013) IFN-gamma production by amyloid beta-specific Th1 cells promotes microglial activation and increases plaque burden in a mouse model of Alzheimer’s disease. J Immunol 190:2241–2251 [DOI] [PubMed] [Google Scholar]
- 19.Vom Berg J, Prokop S, Miller KR et al. (2012) Inhibition of IL-12/IL-23 signaling reduces Alzheimer’s disease-like pathology and cognitive decline. Nat Med 18:1812–1819 [DOI] [PubMed] [Google Scholar]
- 20.Bettelli E, Nicholson LB, Kuchroo VK (2003) IL-10, a key effector regulatory cytokine in experimental autoimmune encephalomyelitis. J Autoimmun 20:265–267 [DOI] [PubMed] [Google Scholar]
- 21.Brionne TC, Tesseur I, Masliah E, Wyss-Coray T (2003) Loss of TGF-beta 1 leads to increased neuronal cell death and microgliosis in mouse brain. Neuron 40:1133–1145 [DOI] [PubMed] [Google Scholar]
- 22.Wyss-Coray T, Lin C, Yan F et al. (2001) TGF-beta1 promotes microglial amyloid-beta clearance and reduces plaque burden in transgenic mice. Nat Med 7:612–618 [DOI] [PubMed] [Google Scholar]
- 23.Flanders KC, Ludecke G, Engels S et al. (1991) Localization and actions of transforming growth factor-beta s in the embryonic nervous system. Development 113:183–191 [DOI] [PubMed] [Google Scholar]
- 24.Flanders KC, Lippa CF, Smith TW, Pollen DA, Sporn MB (1995) Altered expression of transforming growth factor-beta in Alzheimer’s disease. Neurology 45:1561–1569 [DOI] [PubMed] [Google Scholar]
- 25.Tichauer JE, von Bernhardi R (2012) Transforming growth factor-beta stimulates beta amyloid uptake by microglia through Smad3-dependent mechanisms. J Neurosci Res 90:1970–1980 [DOI] [PubMed] [Google Scholar]
- 26.Ma D, Doi Y, Jin S et al. (2012) TGF-beta induced by interleukin-34-stimulated microglia regulates microglial proliferation and attenuates oligomeric amyloid beta neurotoxicity. Neurosci Lett 529:86–91 [DOI] [PubMed] [Google Scholar]
- 27.Colonna M, Wang Y (2016) TREM2 variants: new keys to decipher Alzheimer disease pathogenesis. Nat Rev Neurosci. 17:201–207 [DOI] [PubMed] [Google Scholar]
- 28.Hickey WF, Hsu BL, Kimura H (1991) T-lymphocyte entry into the central nervous system. J Neurosci Res 28:254–260 [DOI] [PubMed] [Google Scholar]
- 29.Kunis G, Baruch K, Rosenzweig N et al. (2013) IFN-gamma-dependent activation of the brain’s choroid plexus for CNS immune surveillance and repair. Brain 136:3427–3440 [DOI] [PubMed] [Google Scholar]
- 30.Radjavi A, Smirnov I, Kipnis J (2014) Brain antigen-reactive CD4+ T cells are sufficient to support learning behavior in mice with limited T cell repertoire. Brain Behav Immun 35:58–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Galea I, Bernardes-Silva M, Forse PA, van Rooijen N, Liblau RS, Perry VH (2007) An antigen-specific pathway for CD8 T cells across the blood-brain barrier. J Exp Med 204:2023–2030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Puntener U, Booth SG, Perry VH, Teeling JL (2012) Long-term impact of systemic bacterial infection on the cerebral vasculature and microglia. J Neuroinflammation 9:146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Boche D, Zotova E, Weller RO et al. (2008) Consequence of abeta immunization on the vasculature of human Alzheimer’s disease brain. Brain : a journal of neurology 131:3299–3310 [DOI] [PubMed] [Google Scholar]
- 34.Gilman S, Koller M, Black RS et al. (2005) Clinical effects of abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology 64:1553–1562 [DOI] [PubMed] [Google Scholar]
- 35.Yang H, Yang H, Xie Z, Wei L, Bi J (2013) Systemic transplantation of human umbilical cord derived mesenchymal stem cells-educated T regulatory cells improved the impaired cognition in AbetaPPswe/PS1dE9 transgenic mice. PLoS One 8:e69129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Saresella M, Calabrese E, Marventano I et al. (2010) PD1 negative and PD1 positive CD4+ T regulatory cells in mild cognitive impairment and Alzheimer’s disease. J Alzheimers Dis 21:927–938 [DOI] [PubMed] [Google Scholar]
- 37.Baruch K, Rosenzweig N, Kertser A, et al. (2015) Breaking immune tolerance by targeting Foxp3(+) regulatory T cells mitigates Alzheimer’s disease pathology. Nature Communications 6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Heppner FL, Ransohoff RM, Becher B (2015) Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci. 16:358–372 [DOI] [PubMed] [Google Scholar]
- 39.Potvin S, Stip E, Sepehry AA, Gendron A, Bah R, Kouassi E (2008) Inflammatory cytokine alterations in schizophrenia: a systematic quantitative review. Biol Psychiatry 63:801–808 [DOI] [PubMed] [Google Scholar]
- 40.Snyder JS, Cameron HA (2012) Could adult hippocampal neurogenesis be relevant for human behavior? Behav Brain Res 227:384–390 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Snyder JS, Soumier A, Brewer M, Pickel J, Cameron HA (2011) Adult hippocampal neurogenesis buffers stress responses and depressive behaviour. Nature 476:458–461 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Holmes C, Boche D, Wilkinson D et al. (2008) Long-term effects of Abeta42 immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet 372:216–223 [DOI] [PubMed] [Google Scholar]
- 43.Ryan DA, Mastrangelo MA, Narrow WC, Sullivan MA, Federoff HJ, Bowers WJ (2010) Abeta-directed single-chain antibody delivery via a serotype-1 AAV vector improves learning behavior and pathology in Alzheimer’s disease mice. Molecular therapy: the journal of the American Society of Gene Therapy 18:1471–1481 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bombois S, Maurage CA, Gompel M et al. (2007) Absence of beta-amyloid deposits after immunization in Alzheimer disease with Lewy body dementia. Arch Neurol 64:583–587 [DOI] [PubMed] [Google Scholar]
- 45.Morgan D, Diamond DM, Gottschall PE et al. (2000) A beta peptide vaccination prevents memory loss in an animal model of Alzheimer’s disease. Nature 408:982–985 [DOI] [PubMed] [Google Scholar]
- 46.Movsesyan N, Ghochikyan A, Mkrtichyan M et al. (2008) Reducing AD-like pathology in 3xTg-AD mouse model by DNA epitope vaccine—a novel immunotherapeutic strategy. PLoSONE 3:e2124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Olkhanud PB, Mughal M, Ayukawa K et al. (2012) DNA immunization with HBsAg-based particles expressing a B cell epitope of amyloid beta-peptide attenuates disease progression and prolongs survival in a mouse model of Alzheimer’s disease. Vaccine 30:1650–1658 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Marsh SE, Abud EM, Lakatos A et al. (2016) The adaptive immune system restrains Alzheimer’s disease pathogenesis by modulating microglial function. Proc Natl Acad Sci U S A 113:E1316–E1325 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Tomihara K, Shin T, Hurez VJ et al. (2012) Aging-associated B7-DC+ B cells enhance anti-tumor immunity via Th1 and Th17 induction. Aging Cell 11:128–138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lee-Chang C, Bodogai M, Moritoh K et al. (2014) Accumulation of 4-1BBL+ B cells in the elderly induces the generation of granzyme-B+ CD8+ T cells with potential antitumor activity. Blood 191:4141–4151 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lee-Chang C, Bodogai M, Moritoh K et al. (2016) Aging converts innate B1a cells into potent CD8+ T cell inducers. J Immunol 196: 3385–3397 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Matsushita T, Yanaba K, Bouaziz JD, Fujimoto M, Tedder TF (2008) Regulatory B cells inhibit EAE initiation in mice while other B cells promote disease progression. JClinInvest 118: 3420–3430 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Shimomura Y, Mizoguchi E, Sugimoto K et al. (2008) Regulatory role of B-1 B cells in chronic colitis. IntImmunol 20:729–737 [DOI] [PubMed] [Google Scholar]
- 54.Gisondi P, Sala F, Alessandrini F et al. (2014) Mild cognitive impairment in patients with moderate to severe chronic plaque psoriasis. Dermatology 228:78–85 [DOI] [PubMed] [Google Scholar]
- 55.Driver JA, Beiser A, Au R et al. (2012) Inverse association between cancer and Alzheimer’s disease: results from the Framingham heart study. BMJ 344:e1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Musicco M, Adorni F, Di Santo S et al. (2013) Inverse occurrence of cancer and Alzheimer disease: a population-based incidence study. Neurology 81:322–328 [DOI] [PubMed] [Google Scholar]
- 57.Kim WK, Alvarez X, Fisher J et al. (2006) CD163 identifies perivascular macrophages in normal and viral encephalitic brains and potential precursors to perivascular macrophages in blood. Am J Pathol 168:822–834 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Michaud JP, Bellavance MA, Prefontaine P, Rivest S (2013) Real-time in vivo imaging reveals the ability of monocytes to clear vascular amyloid beta. Cell Rep 5:646–653 [DOI] [PubMed] [Google Scholar]
- 59.Deane R, Sagare A, Hamm K et al. (2005) IgG-assisted age-dependent clearance of Alzheimer’s amyloid beta peptide by the blood-brain barrier neonatal fc receptor. J Neurosci 25:11495–11503 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Mohle L, Mattei D, Heimesaat MM et al. (2016) Ly6C(hi) monocytes provide a link between antibiotic-induced changes in gut microbiota and adult hippocampal neurogenesis. Cell Rep 15:1945–1956 [DOI] [PubMed] [Google Scholar]
- 61.Perry VH, Cunningham C, Holmes C (2007) Systemic infections and inflammation affect chronic neurodegeneration. Nat Rev Immunol. 7:161–167 [DOI] [PubMed] [Google Scholar]
- 62.Yamamoto M, Kiyota T, Horiba M et al. (2007) Interferon-gamma and tumor necrosis factor-alpha regulate amyloid-beta plaque deposition and beta-secretase expression in Swedish mutant APP transgenic mice. Am J Pathol 170:680–692 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Baruch K, Deczkowska A, David E et al. (2014) Aging. Aging-induced type I interferon response at the choroid plexus negatively affects brain function. Science 346:89–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ransohoff RM, Engelhardt B (2012) The anatomical and cellular basis of immune surveillance in the central nervous system. Nat Rev Immunol 12:623–635 [DOI] [PubMed] [Google Scholar]
- 65.Schwartz M, Baruch K (2014) The resolution of neuroinflammation in neurodegeneration: leukocyte recruitment via the choroid plexus. EMBO J 33:7–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Louveau A, Smirnov I, Keyes TJ et al. (2015) Structural and functional features of central nervous system lymphatic vessels. Nature 523:337–341 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Petra AI, Panagiotidou S, Hatziagelaki E, Stewart JM, Conti P, Theoharides TC (2015) Gut-microbiota-brain Axis and its effect on neuropsychiatric disorders with suspected immune dysregulation. Clin Ther 37:984–995 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bercik P, Denou E, Collins J et al. (2011) The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology 141:599–609 609 e591–593 [DOI] [PubMed] [Google Scholar]
- 69.Diaz Heijtz R, Wang S, Anuar F et al. (2011) Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A 108:3047–3052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Mariat D, Firmesse O, Levenez F et al. (2009) The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol 9:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zwielehner J, Liszt K, Handschur M, Lassl C, Lapin A, Haslberger AG (2009) Combined PCR-DGGE fingerprinting and quantitative-PCR indicates shifts in fecal population sizes and diversity of Bacteroides, Bifidobacteria and Clostridium cluster IV in institutionalized elderly. Exp Gerontol 44:440–446 [DOI] [PubMed] [Google Scholar]
- 72.Cattaneo A, Cattane N, Galluzzi S et al. (2016) Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging 49:60–68 [DOI] [PubMed] [Google Scholar]
- 73.Man AL, Bertelli E, Rentini S et al. (2015) Age-associated modifications of intestinal permeability and innate immunity in human small intestine. Clin Sci (Lond) 129:515–527 [DOI] [PubMed] [Google Scholar]
- 74.Kim KA, Jeong JJ, Yoo SY, Kim DH (2016) Gut microbiota lipopolysaccharide accelerates inflamm-aging in mice. BMC Microbiol 16:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Lucin KM, Wyss-Coray T (2009) Immune activation in brain aging and neurodegeneration: too much or too little? Neuron 64: 110–122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ferretti MT, Merlini M, Spani C et al. (2016) T-cell brain infiltration and immature antigen-presenting cells in transgenic models of Alzheimer’s disease-like cerebral amyloidosis. Brain Behav Immun 54:211–225 [DOI] [PubMed] [Google Scholar]
- 77.Derecki NC, Cardani AN, Yang CH et al. (2010) Regulation of learning and memory by meningeal immunity: a key role for IL-4. J Exp Med 207:1067–1080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Stellwagen D, Beattie EC, Seo JY, Malenka RC (2005) Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-alpha. J Neurosci 25:3219–3228 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Filiano AJ, Xu Y, Tustison NJ et al. (2016) Unexpected role of interferon-gamma in regulating neuronal connectivity and social behaviour. Nature 535:425–429 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Prinz M, Priller J (2014) Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease. Nat Rev Neurosci 15:300–312 [DOI] [PubMed] [Google Scholar]
- 81.Jiang Z, Jiang JX, Zhang GX (2014) Macrophages: a double-edged sword in experimental autoimmune encephalomyelitis. Immunol Lett 160:17–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Rosser EC, Oleinika K, Tonon S et al. (2014) Regulatory B cells are induced by gut microbiota-driven interleukin-1beta and interleukin-6 production. Nat Med 20:1334–1339 [DOI] [PubMed] [Google Scholar]
- 83.Chen Y, Khanna S, Goodyear CS et al. (2009) Regulation of dendritic cells and macrophages by an anti-apoptotic cell natural antibody that suppresses TLR responses and inhibits inflammatory arthritis. J Immunol 183:1346–1359 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Baumgarth N (2011) The double life of a B-1 cell: self-reactivity selects for protective effector functions. Nat Rev Immunol. 11:34–46 [DOI] [PubMed] [Google Scholar]
- 85.Magri G, Miyajima M, Bascones S et al. (2014) Innate lymphoid cells integrate stromal and immunological signals to enhance antibody production by splenic marginal zone B cells. Nat Immunol 15:354–364 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Puga I, Cols M, Barra CM et al. (2012) B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen. Nat Immunol 13:170–180 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Mawuenyega KG, Sigurdson W, Ovod V et al. (2010) Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science 330:1774–1774 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Tarasoff-Conway JM, Carare RO, Osorio RS et al. (2015) Clearance systems in the brain-implications for Alzheimer disease. Nat Rev Neurol 11:457–470 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Brew BJ, Pemberton L, Blennow K, Wallin A, Hagberg L (2005) CSF amyloid beta 42 and tau levels correlate with AIDS dementia complex. Neurology 65:1490–1492 [DOI] [PubMed] [Google Scholar]
- 90.Koch S, Larbi A, Derhovanessian E, Ozcelik D, Naumova E, Pawelec G (2008) Multiparameter flow cytometric analysis of CD4 and CD8 T cell subsets in young and old people. Immunity & ageing: I & A 5:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lerner A, Yamada T, Miller RA (1989) Pgp-1hi T lymphocytes accumulate with age in mice and respond poorly to concanavalin A. Eur J Immunol 19:977–982 [DOI] [PubMed] [Google Scholar]
- 92.Guerrettaz LM, Johnson SA, Cambier JC (2008) Acquired hematopoietic stem cell defects determine B-cell repertoire changes associated with aging. Proc Natl Acad Sci U S A 105:11898–11902 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Cancro MP, Allman DM (2005) Connecting the dots: revealing the interactions of lymphocyte development and homeostasis in the immunobiology of aging. Semin Immunol 17:319–320 [DOI] [PubMed] [Google Scholar]
- 94.Linehan E, Dombrowski Y, Snoddy R, Fallon PG, Kissenpfennig A, Fitzgerald DC (2014) Aging impairs peritoneal but not bone marrow-derived macrophage phagocytosis. Aging Cell 13:699–708 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Stall AM, Farinas MC, Tarlinton DM et al. (1988) Ly-1 B-cell clones similar to human chronic lymphocytic leukemias routinely develop in older normal mice and young autoimmune (New Zealand black-related) animals. Proc Natl Acad Sci U S A 85:7312–7316 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Poduslo JF, Curran GL, Berg CT (1994) Macromolecular permeability across the blood-nerve and blood-brain barriers. Proc Natl Acad Sci U S A 91:5705–5709 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Fuller JP, Stavenhagen JB, Teeling JL. (2014) New roles for Fc receptors in neurodegeneration—the impact on immunotherapy for Alzheimer’s disease. Frontiers in Neuroscience. 8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bouras C, Riederer BM, Kovari E, Hof PR, Giannakopoulos P (2005) Humoral immunity in brain aging and Alzheimer’s disease. Brain Res Brain Res Rev 48:477–487 [DOI] [PubMed] [Google Scholar]
- 99.Deane R, Sagare A, Hamm K et al. (2008) apoE isoform-specific disruption of amyloid beta peptide clearance from mouse brain. J Clin Investig 118:4002–4013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Morris A, Moller G (1968) Regulation of cellular antibody synthesis effect of adoptively transferred antibody-producing spleen cells on cellular antibody synthesis. J Immunol 101:439–445 [PubMed] [Google Scholar]
- 101.Mimouni D, Gdalevich M, Mimouni FB, Grotto I, Eldad A, Shpilberg O (2000) Does immune serum globulin confer protection against skin diseases? Int J Dermatol 39:628–631 [DOI] [PubMed] [Google Scholar]
- 102.Siragam V, Brinc D, Crow AR, Song S, Freedman J, Lazarus AH (2005) Can antibodies with specificity for soluble antigens mimic the therapeutic effects of intravenous IgG in the treatment of autoimmune disease? J Clin Invest 115:155–160 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Bruhns P, Samuelsson A, Pollard JW, Ravetch JV (2003) Colony-stimulating factor-1-dependent macrophages are responsible for IVIG protection in antibody-induced autoimmune disease. Immunity 18:573–581 [DOI] [PubMed] [Google Scholar]
- 104.Ravetch JV, Bolland S (2001) IgG Fc receptors. Annu Rev Immunol 19:275–290 [DOI] [PubMed] [Google Scholar]
- 105.Siragam V, Crow AR, Brinc D, Song S, Freedman J, Lazarus AH (2006) Intravenous immunoglobulin ameliorates ITP via activating fc gamma receptors on dendritic cells. Nat Med 12:688–692 [DOI] [PubMed] [Google Scholar]
- 106.Sudduth TL, Greenstein A, Wilcock DM (2013) Intracranial injection of gammagard, a human IVIg, modulates the inflammatory response of the brain and lowers a beta in APP/PS1 mice along a different time course than anti-a beta antibodies. J Neurosci 33: 9684–9692 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Knight EM, Gandy S (2014) Immunomodulation and AD—down but not out. J Clin Immunol 34:S70–S73 [DOI] [PubMed] [Google Scholar]
- 108.Relkin N (2014) Clinical trials of intravenous immunoglobulin for Alzheimer’s disease. J Clin Immunol 34:S74–S79 [DOI] [PubMed] [Google Scholar]
- 109.Peress NS, Fleit HB, Perillo E, Kuljis R, Pezzullo C (1993) Identification of fc gamma RI, II and III on normal human brain ramified microglia and on microglia in senile plaques in Alzheimer’s disease. J Neuroimmunol 48:71–79 [DOI] [PubMed] [Google Scholar]
- 110.Cribbs DH, Berchtold NC, Perreau V et al. (2012) Extensive innate immune gene activation accompanies brain aging, increasing vulnerability to cognitive decline and neurodegeneration: a microarray study. J Neuroinflammation 9:179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Orr CF, Rowe DB, Mizuno Y, Mori H, Halliday GM (2005) A possible role for humoral immunity in the pathogenesis of Parkinson’s disease. Brain 128:2665–2674 [DOI] [PubMed] [Google Scholar]
- 112.Pruss H, Holtje M, Maier N et al. (2012) IgA NMDA receptor antibodies are markers of synaptic immunity in slow cognitive impairment. Neurology 78:1743–1753 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Dalmau J, Gleichman AJ, Hughes EG et al. (2008) Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 7:1091–1098 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Harris DP, Haynes L, Sayles PC et al. (2000) Reciprocal regulation of polarized cytokine production by effector B and T cells. Nat Immunol 1:475–482 [DOI] [PubMed] [Google Scholar]
- 115.Martin F, Chan AC (2006) B cell immunobiology in disease: evolving concepts from the clinic. Annu Rev Immunol 24:467–496 [DOI] [PubMed] [Google Scholar]
- 116.Duddy ME, Alter A, Bar-Or A (2004) Distinct profiles of human B cell effector cytokines: a role in immune regulation? J Immunol 172:3422–3427 [DOI] [PubMed] [Google Scholar]
- 117.Lenert P, Brummel R, Field EH, Ashman RF (2005) TLR-9 activation of marginal zone B cells in lupus mice regulates immunity through increased IL-10 production. J Clin Immunol 25:29–40 [DOI] [PubMed] [Google Scholar]
- 118.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 16:219–230 [DOI] [PubMed] [Google Scholar]
- 119.Weber MS, Prod’homme T, Patarroyo JC et al. (2010) B-cell activation influences T-cell polarization and outcome of anti-CD20 B-cell depletion in central nervous system autoimmunity. Ann Neurol 68:369–383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Lanzavecchia A (1990) Receptor-mediated antigen uptake and its effect on antigen presentation to class II-restricted T lymphocytes. Annu Rev Immunol 8:773–793 [DOI] [PubMed] [Google Scholar]
- 121.Avalos AM, Ploegh HL (2014) Early BCR events and antigen capture, processing, and loading on MHC class II on B cells. Front Immunol 5:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Sonoda K-H, Stein-Streilein J (2002) CD1d on antigen-transporting APC and splenic marginal zone B cells promotes NKT cell-dependent tolerance. Eur J Immunol 32:848–857 [DOI] [PubMed] [Google Scholar]
- 123.Schneider R, Mohebiany AN, Ifergan I et al. (2011) B cell-derived IL-15 enhances CD8 T cell cytotoxicity and is increased in multiple sclerosis patients. J Immunol 187:4119–4128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kurt-Jones EA, Liano D, HayGlass KA, Benacerraf B, Sy MS, Abbas AK (1988) The role of antigen-presenting B cells in T cell priming in vivo. Studies of B cell-deficient mice. J Immunol 140: 3773–3778 [PubMed] [Google Scholar]
- 125.O’Neill SK, Cao Y, Hamel KM, Doodes PD, Hutas G, Finnegan A (2007) Expression of CD80/86 on B cells is essential for autoreactive T cell activation and the development of arthritis. J Immunol 179:5109–5116 [DOI] [PubMed] [Google Scholar]
- 126.van Essen D, Kikutani H, Gray D (1995) CD40 ligand-transduced co-stimulation of T cells in the development of helper function. Nature 378:620–623 [DOI] [PubMed] [Google Scholar]
- 127.Blair PJ, Riley JL, Harlan DM et al. (2000) CD40 ligand (CD154) triggers a short-term CD4(+) T cell activation response that results in secretion of immunomodulatory cytokines and apoptosis. J Exp Med 191:651–660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Olkhanud PB, Damdinsuren B, Bodogai M et al. (2011) Tumor-evoked regulatory B cells promote breast cancer metastasis by converting resting CD4+ T cells to T-regulatory cells. Cancer Res 71:3505–3515 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Hao Y, O’Neill P, Naradikian MS, Scholz JL, Cancro MP (2011) A B-cell subset uniquely responsive to innate stimuli accumulates in aged mice. Blood 118:1294–1304 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Futagawa T, Akiba H, Kodama T et al. (2002) Expression and function of 4-1BB and 4-1BB ligand on murine dendritic cells. Int Immunol 14:275–286 [DOI] [PubMed] [Google Scholar]
- 131.Wang C, Lin GH, McPherson AJ, Watts TH (2009) Immune regulation by 4-1BB and 4-1BBL: complexities and challenges. Immunol Rev 229:192–215 [DOI] [PubMed] [Google Scholar]
- 132.Cooper D, Bansal-Pakala P, Croft M (2002) 4-1BB (CD137) controls the clonal expansion and survival of CD8 T cells in vivo but does not contribute to the development of cytotoxicity. Eur J Immunol 32:521–529 [DOI] [PubMed] [Google Scholar]
- 133.Watts TH, DeBenedette MA (1999) T cell co-stimulatory molecules other than CD28. Curr Opin Immunol 11:286–293 [DOI] [PubMed] [Google Scholar]
- 134.Shuford WW, Klussman K, Tritchler DD et al. (1997) 4-1BB costimulatory signals preferentially induce CD8+ T cell proliferation and lead to the amplification in vivo of cytotoxic T cell responses. J Exp Med 186:47–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kim HJ, Lee JS, Kim JD et al. (2012) Reverse signaling through the costimulatory ligand CD137L in epithelial cells is essential for natural killer cell-mediated acute tissue inflammation. Proc Natl Acad Sci U S A 109:E13–E22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Gramaglia I, Cooper D, Miner KT, Kwon BS, Croft M (2000) Co-stimulation of antigen-specific CD4 Tcells by 4-1BB ligand. Eur J Immunol 30:392–402 [DOI] [PubMed] [Google Scholar]
- 137.Bulati M, Buffa S, Martorana A et al. (2015) Double negative (IgG + IgD−CD27−) B cells are increased in a cohort of moderate-severe Alzheimer’s disease patients and show a pro-inflammatory trafficking receptor phenotype. J Alzheimers Dis 44:1241–1251 [DOI] [PubMed] [Google Scholar]
- 138.Ansel KM, Ngo VN, Hyman PL et al. (2000) A chemokine-driven positive feedback loop organizes lymphoid follicles. Nature 406:309–314 [DOI] [PubMed] [Google Scholar]
- 139.Corcione A, Casazza S, Ferretti E et al. (2004) Recapitulation of B cell differentiation in the central nervous system of patients with multiple sclerosis. Proc Natl Acad Sci U S A 101:11064–11069 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Townsend MJ, Monroe JG, Chan AC (2010) B-cell targeted therapies in human autoimmune diseases: an updated perspective. Immunol Rev 237:264–283 [DOI] [PubMed] [Google Scholar]
- 141.Nicholas MW, Dooley MA, Hogan SL et al. (2008) A novel subset of memory B cells is enriched in autoreactivity and correlates with adverse outcomes in SLE. Clin Immunol 126:189–201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Sellam J, Rouanet S, Hendel-Chavez H et al. (2011) Blood memory B cells are disturbed and predict the response to rituximab in patients with rheumatoid arthritis. Arthritis Rheum 63:3692–3701 [DOI] [PubMed] [Google Scholar]
- 143.Maurer MA, Rakocevic G, Leung CS et al. (2012) Rituximab induces sustained reduction of pathogenic B cells in patients with peripheral nervous system autoimmunity. J Clin Invest 122:1393–1402 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Stroopinsky D, Katz T, Rowe JM, Melamed D, Avivi I (2012) Rituximab-induced direct inhibition of T-cell activation. Cancer immunology, immunotherapy : CII. 61:1233–1241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Cepok S, Rosche B, Grummel V et al. (2005) Short-lived plasma blasts are the main B cell effector subset during the course of multiple sclerosis. Brain 128:1667–1676 [DOI] [PubMed] [Google Scholar]
- 146.Piancone F, Saresella M, Marventano I et al. (2016) B lymphocytes in multiple sclerosis: Bregs and BTLA/CD272 expressing-CD19+ lymphocytes modulate disease severity. Sci Rep 6:29699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Carrasco YR, Batista FD (2007) B cells acquire particulate antigen in a macrophage-rich area at the boundary between the follicle and the subcapsular sinus of the lymph node. Immunity 27:160–171 [DOI] [PubMed] [Google Scholar]
- 148.Qi H, Egen JG, Huang AY, Germain RN (2006) Extrafollicular activation of lymph node B cells by antigen-bearing dendritic cells. Science 312:1672–1676 [DOI] [PubMed] [Google Scholar]
- 149.Constant S, Schweitzer N, West J, Ranney P, Bottomly K (1995) B lymphocytes can be competent antigen-presenting cells for priming CD4+ T cells to protein antigens in vivo. J Immunol 155: 3734–3741 [PubMed] [Google Scholar]
- 150.Rivera A, Chen CC, Ron N, Dougherty JP, Ron Y (2001) Role of B cells as antigen-presenting cells in vivo revisited: antigen-specific B cells are essential for T cell expansion in lymph nodes and for systemic T cell responses to low antigen concentrations. Int Immunol 13:1583–1593 [DOI] [PubMed] [Google Scholar]
- 151.Molnarfi N, Schulze-Topphoff U, Weber MS 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Bao Y, Cao X. (2014) The immune potential and immunopathology of cytokine-producing B cell subsets: a comprehensive review. Journal of autoimmunity [DOI] [PubMed] [Google Scholar]
- 153.Pierson ER, Stromnes IM, Goverman JM (2014) B cells promote induction of experimental autoimmune encephalomyelitis by facilitating reactivation of T cells in the central nervous system. J Immunol 192:929–939 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Goverman J (2009) Autoimmune T cell responses in the central nervous system. Nat Rev Immunol 9:393–407 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Doyle KP, Quach LN, Sole M et al. (2015) B-lymphocyte-mediated delayed cognitive impairment following stroke. J Neurosci 35:2133–2145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Desmond DW, Moroney JT, Sano M, Stern Y (2002) Incidence of dementia after ischemic stroke: results of a longitudinal study. Stroke 33:2254–2260 [DOI] [PubMed] [Google Scholar]
- 157.Mena H, Cadavid D, Rushing EJ (2004) Human cerebral infarct: a proposed histopathologic classification based on 137 cases. Acta Neuropathol 108:524–530 [DOI] [PubMed] [Google Scholar]
- 158.Shimamura T, Hashimoto K, Sasaki S (1982) Feedback suppression of the immune response in vivo. I. Immune B cells induce antigen-specific suppressor T cells. Cell Immunol 68:104–113 [DOI] [PubMed] [Google Scholar]
- 159.Wolf SD, Dittel BN, Hardardottir F, Janeway CA Jr (1996) Experimental autoimmune encephalomyelitis induction in genetically B cell-deficient mice. J Exp Med 184:2271–2278 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Yanaba K, Bouaziz JD, 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 28:639–650 [DOI] [PubMed] [Google Scholar]
- 161.Mizoguchi A, Mizoguchi E, Smith RN, Preffer FI, Bhan AK (1997) Suppressive role of B cells in chronic colitis of T cell receptor alpha mutant mice. J Exp Med 186:1749–1756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Eynon EE, Parker DC (1992) Small B cells as antigen-presenting cells in the induction of tolerance to soluble protein antigens. J Exp Med 175:131–138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Bennett SR, Carbone FR, Toy T, Miller JF, Heath WR (1998) B cells directly tolerize CD8(+) T cells. J Exp Med 188:1977–1983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.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. The Journal of allergy and clinical immunology 125:1114–1124.e1118 [DOI] [PubMed] [Google Scholar]
- 165.Sun JB, Czerkinsky C, Holmgren J (2012) B lymphocytes treated in vitro with antigen coupled to cholera toxin B subunit induce antigen-specific Foxp3(+) regulatory T cells and protect against experimental autoimmune encephalomyelitis. J Immunol 188: 1686–1697 [DOI] [PubMed] [Google Scholar]
- 166.Reichardt P, Dornbach B, Rong S et al. (2007) Naive B cells generate regulatory T cells in the presence of a mature immunologic synapse. Blood 110:1519–1529 [DOI] [PubMed] [Google Scholar]
- 167.Sayi A, Kohler E, Toller IM et al. (2011) TLR-2-activated B cells suppress helicobacter-induced preneoplastic gastric immunopathology by inducing T regulatory-1 cells. J Immunol 186:878–890 [DOI] [PubMed] [Google Scholar]
- 168.Evans JG, Chavez-Rueda KA, Eddaoudi A et al. (2007) Novel suppressive function of transitional 2 B cells in experimental arthritis. J Immunol 178:7868–7878 [DOI] [PubMed] [Google Scholar]
- 169.Ding Q, Yeung M, Camirand G 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: 3645–3656 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Sattler S, Ling GS, Xu D et al. (2014) IL-10-producing regulatory B cells induced by IL-33 (Breg(IL-33)) effectively attenuate mucosal inflammatory responses in the gut. J Autoimmun 50:107–122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Wang RX, CR Y, Dambuza IM et al. (2014) Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat Med 20: 633–641 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Blair PA, Norena LY, Flores-Borja F 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 32:129–140 [DOI] [PubMed] [Google Scholar]
- 173.Iwata Y, Matsushita T, Horikawa M et al. (2011) Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood 117:530–541 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Kessel A, Haj T, Peri R et al. (2012) Human CD19(+) CD25(high) B regulatory cells suppress proliferation of CD4(+) T cells and enhance Foxp3 and CTLA-4 expression in T-regulatory cells. Autoimmun Rev 11:670–677 [DOI] [PubMed] [Google Scholar]
- 175.Brisslert M, Bokarewa M, Larsson P, Wing K, Collins LV, Tarkowski A (2006) Phenotypic and functional characterization of human CD25+ B cells. Immunology 117:548–557 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.de Andrés C, Tejera-Alhambra M, Alonso B et al. (2014) New regulatory CD19(+) CD25(+) B-cell subset in clinically isolated syndrome and multiple sclerosis relapse. Changes after glucocorticoids. J Neuroimmunol 270:37–44 [DOI] [PubMed] [Google Scholar]
- 177.Schioppa T, Moore R, Thompson RG et al. (2011) B regulatory cells and the tumor-promoting actions of TNF-alpha during squamous carcinogenesis. Proc Natl Acad Sci U S A 108:10662–10667 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Shimabukuro-Vornhagen A, Schlosser HA, Gryschok L et al. (2014) Characterization of tumor-associated B-cell subsets in patients with colorectal cancer. Oncotarget 5:4651–4664 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Lindner S, Dahlke K, Sontheimer K et al. (2013) Interleukin 21-induced granzyme B-expressing B cells infiltrate tumors and regulate T cells. Cancer Res 73:2468–2479 [DOI] [PubMed] [Google Scholar]
- 180.Shi J, Li S, Zhou Y et al. (2014) Perioperative changes in peripheral regulatory B cells of patients with esophageal cancer. Mol Med Rep 10:1525–1530 [DOI] [PubMed] [Google Scholar]
- 181.Bodogai M, Moritoh K, Lee-Chang C et al. (2015) Immunosuppressive and prometastatic functions of myeloid-derived suppressive cells rely upon education from tumor-associated B cells. Cancer Res 75:3456–3465 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Bodogai M, Lee Chang C, Wejksza K et al. (2013) Anti-CD20 antibody promotes cancer escape via enrichment of tumor-evoked regulatory B cells expressing low levels of CD20 and CD137L. Cancer Res 73:2127–2138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Fuchs EJ, Matzinger PB (1992) Cells turn off virgin but not memory T cells. Science 258:1156–1159 [DOI] [PubMed] [Google Scholar]
- 184.Hussain S, Delovitch TL (2007) Intravenous transfusion of BCR-activated B cells protects NOD mice from type 1 diabetes in an IL-10-dependent manner. JImmunol 179:7225–7232 [DOI] [PubMed] [Google Scholar]
- 185.Xiao S, Brooks CR, Zhu C et al. (2012) Defect in regulatory B-cell function and development of systemic autoimmunity in T-cell Ig mucin 1 (Tim-1) mucin domain-mutant mice. Proc Natl Acad Sci U S A 109:12105–12110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Lampropoulou V, Hoehlig K, Roch T et al. (2008) TLR-activated B cells suppress T cell-mediated autoimmunity. J Immunol 180: 4763–4773 [DOI] [PubMed] [Google Scholar]
- 187.Fillatreau S, Sweenie CH, McGeachy MJ, Gray D, Anderton SM (2002) B cells regulate autoimmunity by provision of IL-10. Nat Immunol 3:944–950 [DOI] [PubMed] [Google Scholar]
- 188.Byrne SN, Halliday GM (2005) B cells activated in lymph nodes in response to ultraviolet irradiation or by interleukin-10 inhibit dendritic cell induction of immunity. JInvest Dermatol 124:570–578 [DOI] [PubMed] [Google Scholar]
- 189.Sindhava V, Woodman ME, Stevenson B, Bondada S (2010) Interleukin-10 mediated autoregulation of murine B-1 B-cells and its role in Borrelia hermsii infection. PLoS One 5:e11445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Balabanian K, Foussat A, Bouchet-Delbos L et al. (2002) Interleukin-10 modulates the sensitivity of peritoneal B lymphocytes to chemokines with opposite effects on stromal cell-derived factor-1 and B-lymphocyte chemoattractant. Blood 99:427–436 [DOI] [PubMed] [Google Scholar]
- 191.Gary-Gouy H, Harriague J, Bismuth G, Platzer C, Schmitt C, Dalloul AH (2002) Human CD5 promotes B-cell survival through stimulation of autocrine IL-10 production. Blood 100:4537–4543 [DOI] [PubMed] [Google Scholar]
- 192.Yoshizaki A, Miyagaki T, DiLillo DJ et al. (2012) Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions. Nature 491:264–268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Iwata Y, Yoshizaki A, Komura K et al. (2009) CD19, a response regulator of B lymphocytes, regulates wound healing through hyaluronan-induced TLR4 signaling. Am J Pathol 175:649–660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.O’Garra A, Howard M (1992) Cytokines and Ly-1 (B1) B cells. Int Rev Immunol 8:219–234 [DOI] [PubMed] [Google Scholar]
- 195.Brummel R, Lenert P (2005) Activation of marginal zone B cells from lupus mice with type a(D) CpG-oligodeoxynucleotides. J Immunol 174:2429–2434 [DOI] [PubMed] [Google Scholar]
- 196.Watt V, Ronchese F, Ritchie D (2007) Resting B cells suppress tumor immunity via an MHC class-II dependent mechanism. J Immunother 30:323–332 [DOI] [PubMed] [Google Scholar]
- 197.Moulin V, Andris F, Thielemans K, Maliszewski C, Urbain J, Moser M (2000) B lymphocytes regulate dendritic cell (DC) function in vivo: increased interleukin 12 production by DCs from B cell-deficient mice results in T helper cell type 1 deviation. J Exp Med 192:475–482 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Horikawa M, Minard-Colin V, Matsushita T, Tedder TF (2011) Regulatory B cell production of IL-10 inhibits lymphoma depletion during CD20 immunotherapy in mice. J Clin Invest 121: 4268–4280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Wong SC, Puaux AL, Chittezhath M et al. (2010) Macrophage polarization to a unique phenotype driven by B cells. Eur J Immunol 40:2296–2307 [DOI] [PubMed] [Google Scholar]
- 200.Affara NI, Ruffell B, Medler TR et al. (2014) B cells regulate macrophage phenotype and response to chemotherapy in squamous carcinomas. Cancer Cell 25:809–821 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Gabitass RF, Annels NE, Stocken DD, Pandha HA, Middleton GW (2011) Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer immunology, immunotherapy : CII 60:1419–1430 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Olkhanud PB, Baatar D, Bodogai M et al. (2009) Breast cancer lung metastasis requires expression of chemokine receptor CCR4 and regulatory T cells. Cancer Res 69:5996–6004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Teichmann LL, Kashgarian M, Weaver CT, Roers A, Müller W, Shlomchik MJ (2012) B cell-derived IL-10 does not regulate spontaneous systemic autoimmunity in MRL.Fas(lpr) mice. J Immunol 188:678–685 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Lee-Chang C, Bodogai M, Martin-Montalvo A et al. (2013) Inhibition of breast cancer metastasis by resveratrol-mediated inactivation of tumor-evoked regulatory B cells. J Immunol 191: 4141–4151 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Parekh VV, Prasad DV, 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 170:5897–5911 [DOI] [PubMed] [Google Scholar]
- 206.Stach RM, Rowley DA (1993) A first or dominant immunization. II. Induced immunoglobulin carries transforming growth factor beta and suppresses cytolytic T cell responses to unrelated alloantigens. J Exp Med 178:841–852 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Rowley DA, Stach RM (1998) B lymphocytes secreting IgG linked to latent transforming growth factor-beta prevent primary cytolytic T lymphocyte responses. Int Immunol 10:355–363 [DOI] [PubMed] [Google Scholar]
- 208.Tretter T, Venigalla RK, Eckstein V et al. (2008) Induction of CD4+ T-cell anergy and apoptosis by activated human B cells. Blood 112:4555–4564 [DOI] [PubMed] [Google Scholar]
- 209.Frommer F, Heinen TJ, Wunderlich FT et al. (2008) Tolerance without clonal expansion: self-antigen-expressing B cells program self-reactive Tcells for future deletion. J Immunol 181:5748–5759 [DOI] [PubMed] [Google Scholar]
- 210.Zuniga E, Rabinovich GA, Iglesias MM, Gruppi A (2001) Regulated expression of galectin-1 during B-cell activation and implications for T-cell apoptosis. J Leukoc Biol 70:73–79 [PubMed] [Google Scholar]
- 211.Sun JB, Flach CF, Czerkinsky C, Holmgren J (2008) B lymphocytes promote expansion of regulatory T cells in oral tolerance: powerful induction by antigen coupled to cholera toxin B subunit. J Immunol 181:8278–8287 [DOI] [PubMed] [Google Scholar]
- 212.Zheng SG, Wang J, Wang P, Gray JD, Horwitz DA (2007) IL-2 is essential for TGF-beta to convert naive CD4 + CD25- cells to CD25 + Foxp3+ regulatory T cells and for expansion of these cells. J Immunol 178:2018–2027 [DOI] [PubMed] [Google Scholar]
- 213.Scapini P, Lamagna C, Hu Y et al. (2011) B cell-derived IL-10 suppresses inflammatory disease in Lyn-deficient mice. Proc Natl Acad Sci U S A 108:E823–E832 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Murai M, Turovskaya O, Kim G et al. (2009) Interleukin 10 acts on regulatory Tcells to maintain expression of the transcription factor Foxp3 and suppressive function in mice with colitis. Nat Immunol 10:1178–1184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Levings MK, Roncarolo MG (2000) T-regulatory 1 cells: a novel subset of CD4 T cells with immunoregulatory properties. JAllergy ClinImmunol 106:S109–S112 [DOI] [PubMed] [Google Scholar]
- 216.Kleinewietfeld M, Puentes F, Borsellino G, Battistini L, Rotzschke O, Falk K (2005) CCR6 expression defines regulatory effector/memory-like cells within the CD25(+) CD4+ T-cell subset. Blood 105:2877–2886 [DOI] [PubMed] [Google Scholar]
- 217.Matsumoto M, Baba A, Yokota T et al. (2014) Interleukin-10-producing plasmablasts exert regulatory function in autoimmune inflammation. Immunity 41:1040–1051 [DOI] [PubMed] [Google Scholar]
- 218.Shen P, Roch T, Lampropoulou V et al. (2014) IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature 507:366–370 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Knippenberg S, Peelen E, Smolders J et al. (2011) Reduction in IL-10 producing B cells (Breg) in multiple sclerosis is accompanied by a reduced naive/memory Breg ratio during a relapse but not in remission. J Neuroimmunol 239:80–86 [DOI] [PubMed] [Google Scholar]
- 220.Sun F, Ladha SS, Yang L et al. (2014) Interleukin-10 producing-B cells and their association with responsiveness to rituximab in myasthenia gravis. Muscle Nerve 49:487–494 [DOI] [PubMed] [Google Scholar]
- 221.Mielke F, Schneider-Obermeyer J, Dorner T (2008) Onset of psoriasis with psoriatic arthropathy during rituximab treatment of non-Hodgkin lymphoma. Ann Rheum Dis 67:1056–1057 [DOI] [PubMed] [Google Scholar]
- 222.Dass S, Vital EM, Emery P (2007) Development of psoriasis after B cell depletion with rituximab. Arthritis Rheum 56:2715–2718 [DOI] [PubMed] [Google Scholar]
- 223.Goetz M, Atreya R, Ghalibafian M, Galle PR, Neurath MF (2007) Exacerbation of ulcerative colitis after rituximab salvage therapy. Inflamm Bowel Dis 13:1365–1368 [DOI] [PubMed] [Google Scholar]
- 224.Benedetti L, Franciotta D, Vigo T et al. (2007) Relapses after treatment with rituximab in a patient with multiple sclerosis and anti myelin-associated glycoprotein polyneuropathy. Arch Neurol 64: 1531–1533 [DOI] [PubMed] [Google Scholar]
- 225.Inoue S, Leitner WW, Golding B, Scott D (2006) Inhibitory effects of B cells on antitumor immunity. Cancer Res 66:7741–7747 [DOI] [PubMed] [Google Scholar]