- Review
- Open access
- Published:
The role of autoantibodies in bridging obesity, aging, and immunosenescence
Immunity & Ageing volume 21, Article number: 85 (2024)
Abstract
Antibodies are essential to immune homeostasis due to their roles in neutralizing pathogenic agents. However, failures in central and peripheral checkpoints that eliminate autoreactive B cells can undermine self-tolerance and generate autoantibodies that mistakenly target self-antigens, leading to inflammation and autoimmune diseases. While autoantibodies are well-studied in autoimmune and in some communicable diseases, their roles in chronic conditions, such as obesity and aging, are less understood. Obesity and aging share similar aspects of immune dysfunction, such as diminished humoral responses and heightened chronic inflammation, which can disrupt immune tolerance and foster autoantigen production, thus giving rise to autoreactive B cells and autoantibodies. In return, these events may also contribute to the pathophysiology of obesity and aging, to the associated autoimmune disorders linked to these conditions, and to the development of immunosenescence, an age-related decline in immune function that heightens vulnerability to infections, chronic diseases, and loss of self-tolerance. Furthermore, the cumulative exposure to antigens and cellular debris during obesity and aging perpetuates pro-inflammatory pathways, linking immunosenescence with other aging hallmarks, such as proteostasis loss and mitochondrial dysfunction. This review examines the mechanisms driving autoantibody generation during obesity and aging and discusses key putative antigenic targets across these conditions. We also explore the therapeutic potential of emerging approaches, such as CAR-T/CAAR-T therapies, vaccines, and BiTEs, to tackle autoimmune-related conditions in aging and obesity.
Introduction
Antibodies are essential components of the humoral immune response that neutralize pathogenic agents. However, approximately 5–8% of people develop autoimmune diseases in which autoantibodies (autoAbs) [1] bind to self-antigens, aggravating inflammation and disease progression [2]. While autoAbs have traditionally been associated with autoimmune diseases, recent findings are identifying increased levels of autoAbs across many different conditions, such as obesity and aging, not traditionally characterized as autoimmune conditions [3, 4]. Specifically, obesity and aging are commonly associated with systemic chronic inflammation and functional decline of the adaptive immune system [5, 6] which is reflected as attenuated humoral responses and amplified autoimmunity [7]. In addition, age-related declines in humoral immunity appear to be exacerbated by obesity [8], and obesity has been shown to accelerate an aging immune phenotype [9,10,11]. Thus, obesity and aging are risk factors for the development of chronic diseases [12], in which disease-associated autoAbs may play a role.
In fact, each individual may harbor a set of specific autoAbs, known as the autoAb reactome, capable of contributing to a wide variety of phenotypic traits [13]. The autoAb reactome has been shown to play important roles in the host response to viral pathogens [14], obesity [15], aging [16], cancer [17], and neurological disease [18]. In non-communicable diseases, such as obesity, increased levels of autoAbs in the serum are seen across a diverse array of antigens, ranging from lipids like malondialdehyde (MDA) to proteins like glial fibrillary acid protein (GFAP), to nucleic acids like double-stranded (ds)DNA [3, 19]. While autoAbs are generally associated with adverse outcomes in most conditions, autoAbs have been observed to be protective in cancer [20] and may also reduce infectious and non-infectious diseases and inflammation as cytokine-neutralizing antibodies [21, 22]. Aging is also associated with increased levels of certain autoAbs, such as antinuclear antibodies (ANA), which may predispose older individuals to higher risks of type 2 diabetes (T2D) and other age-related diseases (ARDs) [23]. Finally, healthy individuals share a common set of autoAbs that may play protective roles, such as helping B cell development and self-antigen recognition [24]. Thus, autoAb responses are highly variable among individuals or disease etiology and they can have both pathological and protective roles.
In this review, we summarize current knowledge regarding mechanisms underlying autoAb development, how autoAb production is exacerbated by aging and obesity, what key autoantigens are targeted in various tissues, and how these processes could represent future therapies in complications of obesity and aging.
Development of autoAbs
During B cell development, B cells undergo systematic rounds of expansion, selection, and differentiation to generate antibody-secreting cells (ASCs) that ultimately defend the host against pathogenic agents. However, despite existing checkpoints that prevent autoimmunity, autoreactive B cells that produce autoAbs can emerge because of altered selection processes [25]. Most of our understanding of autoAb development comes from studies in autoimmune diseases. However, the role of aging and obesity in autoAb production has been less explored. Here, we describe the mechanisms involved in the formation of autoAbs, with a focus on how the chronic inflammation commonly associated with aging and obesity facilitates the breaching of central and peripheral immune tolerance.
The first site where self-tolerance is established is the bone marrow (BM) and is referred to as central tolerance. B cell development begins prior to foreign antigen exposure, whereby immature B cells generate B cell receptors (BCR) through V(D)J recombination, a process involving programmed DNA damage and error-prone repair to increase antibody diversity [26]. Due to the randomness of this process, an estimated 55–75% of early immature B cells can display self-reactive behavior [27]. However, aging-linked genomic instability and chronic inflammation can disrupt the DNA damage response (DDR) and reduce the efficiency of V(D)J recombination [28, 29], leading to higher rates of autoreactive BCRs, though various inhibitory checkpoints act to reduce their frequency [25] (Fig. 1A). A key mechanism of self-tolerance in the central tolerance checkpoint involves negative selection, which relies on the strength of BCR signaling: Pre-B cells with moderate-affinity BCRs are allowed to proceed into immature B cells, whereas Pre-B cells with high-affinity BCRs for self-antigens are directed to undergo receptor editing and clonal deletion, though weak BCR signaling can also allow some Pre-B cells to escape tolerance [30] (Fig. 1A). While central tolerance checkpoints can limit the abundance of autoreactive immature B cells, roughly 40% of newly emigrated B cells into the periphery are self-reactive [27], a rate that can further increase during aging. For example, upon self-antigen binding, murine and human immature autoreactive B cells express 1.5-fold higher C-X-C chemokine receptor type 4 (CXCR4), which is conceived to aid in their retention in the BM for receptor editing [31]. However, elevated plasma C-X-C Motif Chemokine Ligand 12 (CXCL12) during aging [32] can disrupt this retention, potentially allowing autoreactive B cells to enter the periphery.
Aging- and obesity-related risk factors contributing to the generation of autoreactive B cells and autoantibodies. A) Autoreactive B cells can be generated throughout the B cell development, from hematopoietic stem cells in the BM to various mature, differentiated B cells. In the BM, central tolerance mechanisms, including receptor editing, clonal deletion, and anergy, reduce the autoreactivity of immature B cells, which resulted mostly from the random V(D)J recombination. In the spleen (shown here) and lymph nodes, clonal deletion or anergy remove transitional B cells (spleen) or naïve B cells that strongly bind self-antigens and B cells lacking the costimulatory signals from CD4 + TH cells or CD4 + TFH cells. However, obesity- and aging-related risk factors, including several hallmarks of aging, promote the release of autoantigens and compromise the self-tolerance, facilitating the generation of autoreactive B cells and augmenting the function of existing autoreactive B cells. B) In various tissues and organs, including the gastrointestinal tract and kidneys, ectopic lymphoid structures, such as TLS, can potentially provide a niche for autoreactive B cells, facilitating their ability to promote local and systemic inflammation. The resulting pro-inflammatory factors and autoantibodies drive tissue damage in multiple organs, further fueling the ongoing inflammation and systemic diseases. ANCA = anti-neutrophil cytoplasm antibody; Abeta = amyloid beta; ASCA = anti-Saccharomyces cerevisiae antibodies; BCR = B cell receptor; BTK = Bruton tyrosine kinase; COL1A1 = collagen 1 A; CSR = class switch recombination; DMD = Dystrophin; FN1 = fibronectin; FYN = tyrosine-protein kinase Fyn; GC = germinal center; GRP78 = Glucose regulated protein 78; GP2 = Glycoprotein 2; HSP60 = Heat shock protein 60; MyHC = Myosin Heavy Chain; OmpC = outer membrane porin C; PLIN1 = Perilipin 1; SASP = senescence-associated secretory phenotype; SC5D = Sterol-C5-desaturase; SHM = somatic hypermutation; TLS = tertiary lymphoid structures; USP4 = Ubiquitin specific protease 4
Transitional B cells exiting the BM migrate to secondary lymphoid organs (SLOs), such as the spleen and the gut-associated lymphoid tissue (GALT), where they undergo positive selection to produce antigen-specific BCRs, while maintaining self-tolerance [33, 34]. Prior to entering germinal centers (GCs), B cells undergo activation at the B-T cell border. This initiates the GC reaction resulting in affinity maturation with help from CD4 + follicular helper T cell (Tfh), follicular antigen-presenting cells, such as follicular dendritic cells (FDCs), and chemokines [35]. As a critical component of peripheral tolerance, self-tolerance in the GCs is achieved through mechanisms, such as clonal deletion, anergy induction, antigen exclusion [36], co-engagement with ITAM expressing Siglecs, activation of ITIM, and other inhibitory receptors and interaction with Tfh cells (Fig. 1A) [33, 37, 38]. Particularly, B cells with higher-affinity BCRs are more effective at presenting antigens to Tfh cells, gaining crucial co-stimulatory cues like CD40L-CD40 and ICOS-ICOSL singular signaling for proliferation [39, 40]. Meanwhile, Tfh-derived IL-21 promotes CSR and SHM via activation-induced cytidine deaminase (AID) and B lymphocyte-induced maturation protein-1 (BLIMP-1) [41, 42], enhancing BCR- and CD40-driven phosphorylation of AKT and S6, key components of the mammalian target of rapamycin complex 1 (mTORC1) pathway [43]. These series of selection, activation, and metabolic reprogramming act as self-tolerance checkpoints, supporting the rapid clonal expansion of GC B cells with proper BCR affinity, while reducing autoreactive B cells to a proportion below 20% [27, 44].
Notably, a pathophysiological form of the GC reaction can also occur in disorganized tertiary lymphoid structures (TLSs) in cases of cancer and autoimmune disease, where they drive opposing outcomes [45] (Fig. 1A). In locations such as the intestinal mucosal surface, TLSs initiate antigen-driven immune responses much like GCs. In the aging bladder and liver, some groups have reported the increased formation of TLSs [46, 47]. Specifically, in the context of kidney injury, aged-associated B cells (ABCs) and a subtype of senescent T cells interact to drive the expansion of TLSs [48]. In autoimmunity, malfunctioning TLS and GC reactions drive the production of pathogenic autoAbs [49], implicating their potential role in driving autoAb production that may exacerbate chronic inflammation during aging and obesity, as discussed below.
Overview of AutoAb formation during aging and obesity
While the breakdown of autoimmunity checkpoints can lead to autoAb development, additional pathways can also contribute to the loss of self-tolerance. Due to the importance of Tfh cell function on B cell maturation, the loss of, or perturbations in Tfh cells has been implicated in autoAb development [50]. Furthermore, the breakdown of self-tolerance can also arise from T-cell tolerance-related mechanisms, which ultimately affect B cells [51, 52]. During aging, shifts in pro- and anti-inflammatory signals can result in an overall immunosuppressive phenotype from some cells. Aged Tfh cells produce higher levels of IL-10, which is associated with reduced B cell responses to immunization [53]. In addition, dysregulation of CD40 and ICOS signaling in B and T cells in patients with SLE [54] suggests that autoimmunity may be contingent in part on costimulatory signaling. Notably, age-related perturbations in immunometabolism also contribute to an altered T-B crosstalk. For example, splenic B cells from aged mice polarized T cells towards an inflammatory phenotype, and co-culturing young T cells with aged B cells shifted T cell metabolism towards glycolysis, mediated by rises in lactate [55].
Meanwhile, dysfunctional immunometabolism can also directly impact the peripheral tolerance and autoAb development during aging and obesity, where altered insulin receptor expression and function, mTOR signaling, and other nutrient-sensing pathways may influence B cell autoAb production [5, 56]. For instance, Tfh cells induce mTORC1 signaling in GC B cells, leading to positive selection, activation, and migration from the light zone to dark zone. Although transient increases in mTORC1 signaling generated a proliferative boost in B cell proliferation needed for sustained proliferation in the dark zone, constitutive activation of mTORC1 impaired affinity maturation [57]. Hyperactivation of mTORC1 has been observed in aging and obesity [58], giving rise to a potential mechanism whereby constitutive activation of GC B cells by mTORC1 blunts affinity maturation, leading to polyreactive autoAbs. It should be noted that B cell lineage, as discussed above, occurs through many steps throughout multiple tissues. This could imply that altered nutrient signaling induced by aging or obesity in any of these compartments could modulate B cell autoreactivity.
It is becoming clear that autoAb production during aging and obesity is associated with inflammation, which impacts central and peripheral tolerance. For example, aging promotes an increased tumor necrosis factor (TNF-α) production by circulatory B cells in mice and humans [59, 60]. The increased TNF-α levels, primarily found in IgM memory B cells, were negatively correlated with AID mRNA expression, suggesting that increase in some age-related pro-inflammatory cytokines can hinder CSR and SHM [60]. In addition, aberrant macrophage function is also thought to contribute to autoAb development and has been implicated in a variety of autoimmune diseases [61]. The inability to clear damaged cells by macrophages can increase apoptotic antigens that become autoAb targets [62] and impaired macrophage phagocytic or efferocytosis capacity is seen with age [63, 64]. Additionally, macrophages isolated from synovial fluid of patients with rheumatoid arthritis (RA) or from aged individuals express higher levels of pro-inflammatory markers [65], suggesting a correlation between pro-inflammatory states and autoAb production. Thus, dysregulation in the crosstalk between B cells via other immune cells, cytokines, chemokines, and metabolism appear to be potential routes that increase autoAb development. Although more direct evidence is needed to establish causative mechanisms between inflammation and autoAb, modulating inflammation does appear to be involved in the development of autoAbs. Below we discuss in detail the potential mechanisms underlying autoAb production and self-targeting first in obesity and then in aging.
Mechanisms of AutoAb development in obesity
As a metabolic disease characterized by chronic inflammation, obesity is associated with comorbidities, such as insulin resistance and T2D, and a wide range of immune-mediated diseases, such as multiple sclerosis (MS) and RA [66]. Obesity is also a risk factor in defective immune responses to viruses, such as SARS-CoV-2 [67, 68]. As both the instigator and the product of chronic inflammation, autoAbs have long been known to play a role in mediating the pathogenesis of obesity. Multiple antibodies (e.g. anti-citrullinated protein antibodies (ACPA) and rheumatoid factors (RF) are found positively correlated with obesity [69, 70]. Research has indicated that obesity increased secretion of autoreactive IgG that can target specific self-antigens, which is associated with increased inflammation and dysfunctional glucose metabolism [3, 71]. Additionally, transferring purified serum IgG from high-fat diet (HFD)-fed mice was enough to induce altered glucose metabolism in mice deficient in T-bet + B cells [72]. How obesity can promote the breaching of immune tolerance, the dysfunction of immune checkpoints, and the generation autoAb-secreting B cells has been widely explored, and various mechanisms are discussed below.
B cell TLRs - drivers of autoimmunity during obesity
Harboring an array of innate and adaptive immune cells, the visceral adipose tissue (VAT) exhibits a shift towards a pro-inflammatory phenotype during obesity [6]. In addition to the pro-inflammatory effect of multiple adipokines, including leptin, resistin, and visfatin [73], adipocyte hypertrophy, hypoxia, and cell death result in the release danger-associated molecular patterns (DAMPs), including “self” protein antigens and cell-free DNA or RNA, which are potent activators of NOD-like receptors (NLRs) and TLRs in macrophages and B cells [66, 74]. For example, obesity induces an increase in VAT T-bet + B cells with marked capacity for IgG2C antibody and the pro-inflammatory chemokine, CXCL10, secretion following glycolipid or TLR7 activation [72]. Meanwhile, HFD-fed mice showed an increased level of autoAb against conserved nuclear antigens, including histone and dsDNA, and obese patients show increased ANA levels, indicating a pathogenic role of TLRs in mediating autoimmune conditions during obesity [74].
The combined TLR and BCR activation by common antigens has been proposed to be a major driving force for the development of autoimmune responses in systemic autoimmune diseases [75]. Numerous common antigens are shared by B cell TLRs and BCRs, including nuclear proteins, citrullinated fibrinogen, cardiolipin, and oxidated phospholipids [75]. In addition, upon BCR binding, self-antigens, such as nucleic acids, can be internalized and translocated to endosomes, where TLR7 and 9 can recognize the same antigens and induce a robust activation of B cells, resulting in a pathogenic GC response, production of autoAbs, and breaching of the peripheral tolerance [76] (Fig. 1A). Indeed, TLR8 KO mice (which can develop TLR7-mediated lupus [77]) fed with a HFD exhibited significantly increased GC B cells, anti-DNA and anti-RNA autoAbs [78]. Consistently, HFD-fed mice lacking TLR7 or TLR9 showed improved glucose tolerance, and TLR7 activation by an agonist can worsen glycemic control [74, 78, 79]. Future work is needed to better define VAT-sourced antigens that can be recognized by TLRs and BCRs.
Altered nutrient metabolism in B cell dysfunction and AutoAb production in obesity
Obesity is associated with dysregulated lipid metabolism, deregulated nutrient-sensing pathways, and altered mitochondrial energetics, which may contribute to the loss of self-tolerance [80,81,82]. Through excessive nutrient intake and subsequent meta-inflammation, metabolic reprogramming shifts B cell function towards a pathogenic, autoAb-secreting phenotype [83]. Indeed, B cells from VAT of obese individuals exhibit a hypermetabolic profile and autoAb repertoire, driving the senescence-associated secretory phenotype (SASP). Pathogenic B cells exhibit increased expression of enzymes involved in glycolysis and oxidative phosphorylation (OXPHOS) [84]. In line with this result, splenic B cells from obese mice have increased mitochondrial membrane potential, likely as a response to increased bioenergetic demands [82]. Notably, unlike pro-inflammatory T helper cells, whose bioenergy demand is met by glycolysis, B cell activation and differentiation rely on both glycolysis and OXPHOS [85], the dysregulation of which can lead to autoimmune responses [86]. Furthermore, obesity promotes dyslipidemia and deregulated fatty acid metabolism to facilitate metabolic reprogramming of other immune cells [87]. CD19 + IgD- CD27- B cells (DN) from obese individuals exhibited significantly greater anti-self autoAbs, including adipocyte-specific (anti-SAT) IgG, anti-dsDNA IgG, and anti-MDA IgG, compared to lean controls [16]. Further analysis revealed that the DN B cells had increased expression for enzymes involved in glycolysis, fatty acid oxidation and oxidative phosphorylation, along with higher levels of reactive oxygen species (ROS) [16]. This suggests that during obesity DN B cells have increased metabolic demand, possibly to sustain autoAb production, while supporting their functional requirements. In addition, during obesity B cell activating factor (BAFF) levels have been found elevated in both mice and humans [88,89,90], where it not only promotes breaking of the peripheral tolerance by suppressing Tregs, but also facilitates the metabolic reprogramming of B cells through increasing both glycolysis and OXPHOS, enhancing antibody production upon LPS stimulation [91], while allowing autoreactive B cells to evade deletion at post-GC checkpoints [92].
Structural and functional changes in AutoAbs in obesity
Obesity can also cause structural changes to antibodies, predisposing them to become self-reactive. One such modification of antibody function is Fc domain glycosylation, which influences antibody stability, conformation, and effector functions by modulating their affinity for FcγR receptors [93]. For instance, sialic acid and galactose residues are linked to anti-inflammatory activity, while their absence and presence of bisecting N-acetylglucosamine (GlcNAc) residues are associated with pro-inflammatory activity, which is also linked to higher BMI and central adiposity [94, 95]. Structural modifications can also occur to ligands during HFD-induced obesity. Tanigaki and co-workers found hyposialylated IgG2C in mice fed a HFD in addition to IgG in patients with T2D [96]. The hyposialylated IgG2C contributed to inflammation and insulin resistance in part through activation of endothelial FcyRllB [96]. Remarkably, a low-calorie diet significantly reduced the levels of IgG N-glycans with bisecting GlcNAc, which are typically elevated with aging and inflammatory conditions [97]. In addition, Arai and co-workers found that HFD increased the binding of IgM to the apoptosis inhibitor of macrophages (AIM), enabling it to evade Fcα/µR internalization by splenic dendritic follicular cells (FDCs). The IgM-AIM complex results in high levels of IgG in the plasma in obese mice [98]. Additionally, these authors noted higher AIM levels in people with BMIs > 25 with autoimmune disease, suggesting a direct link between obesity and autoimmune diseases [98]. Additional functional changes in antibodies are found in the IgA from HFD-fed mice, which exhibit lower SHM [99], implying an increased polyreactive and autoreactive potential. Therefore, dietary intake may enhance the metabolic adducts and post-translational modifications (PTM) of specific regions on immune cells, leading to structural and functional changes that increase the propensity for autoAb production. These changes may occur through metabolic reprogramming of cellular pathways or modification of the gut microbiota. Future research should focus on identifying the specific PTMs that facilitate the loss of self-tolerance and the mechanisms by which these PTMs arise.
Mechanisms of AutoAb development in aging
Aging is associated with a decline in immune function that increases the risk for infections, chronic disease, and loss of self-tolerance [100, 101]. The age-related decline in immune cell dysfunction, pro-inflammatory output and decreased antigen specificity is generally referred to as immunosenescence and is associated with endogenous and exogenous stresses that accumulate throughout the lifespan [102]. Key features of immunosenescence include basal chronic inflammation, thymus involution, naive and memory T/B cell imbalances, altered cellular metabolism, and increased senescence associated secretory phenotype SASP [102]. Immunosenescence is a risk factor for ARDs such as Alzheimer’s disease (AD), cardiovascular disease, T2D, and autoimmune diseases [102], and here we propose that it also contributes to dysregulated autoAb production. Aging is characterized by persistent low-grade inflammation, known as inflammaging [103]. Compared with young controls, healthy elderly subjects have higher levels of pro-inflammatory B cells and markers of inflammation [104]. The cumulative exposure to antigens, damage associated molecular patterns, and cellular debris sustains pro-inflammatory pathways [102], which in turn can compromise proteostasis (e.g. by overloading lysosomal pathways, leading to aggregates) and dysregulated mitochondria [105, 106] (Fig. 1A). Aging is also characterized by the dysregulation of nutrient-sensing pathways such as the insulin and insulin-like growth factor (IGF-1) pathways [5, 56]. Phosphoinositide 3-kinase-gamma (PI3Kγ), downstream of insulin and IGF-1, drives ASCs differentiation and potentially T cell-dependent autoAb generation [107]. As a result, insulin receptor signaling may promote autoreactivity, which in turn induces T cell dysfunction [108]. Aside from metabolic dysregulation, IgG antibodies from older humans (> 60 years) have a high propensity to bind to peptides containing two consecutive serine residues at the N-terminus [109], suggesting that autoAbs target selective amino acids. Therefore, the generation of autoAbs with age may be a result of comprehensive stressors placed on the immune system over a lifetime, leading to a set of age-related autoantigens. Driven by the natural progression of aging, immunosenescence is influenced by environmental and lifestyle factors [110]. Studies from mice and humans have shown that aging leads to higher secretion of IgG antibodies with autoreactive behavior [111]. Age-associated intra-tissue accumulation of IgG recently has been found to contribute to senescence development in multiple organs, including spleen, lymph node, hippocampus, lung, and heart [112]. These findings indicate that accretion of cellular defects leading to an immunosenescent phenotype in B cells may ultimately lead to the production of deleterious autoAbs, which may play an underlying role in driving the pathology of ARDs.
Aging disrupts B cell populations
Aged patients experience impaired B cell proliferation and activation, likely due to defects in their activation threshold. This can lead to a decrease in B cell heterogeneity driven by increased clonal expansion to only select antigens [113]. Moreover, aging induces a shift in the B cell repertoire, towards a B-1 cell phenotype [114]. The increase or expansion in the antigen experienced B-1 phenotype cells may represent a more autoreactive B cell population capable of producing autoAbs. B-1 cells taken from older patients had less spontaneous IgM secretion and no change in IgG secretion, potentially caused by reduced expression of immunoglobulin secreting transcription factors [115]. At the cellular level, aging is associated with decreased expression of the transcription factor, E47, necessary for B cell differentiation, and AID, which is required for SHM and CSR [116, 117]. The decrease in E47 and AID was associated with remarkably lower levels of memory B cells, and higher percentage of naive B cells [117]. As such, aging populations have blunted neutralizing responses to vaccination [118] and present disease modifying autoAbs, which exacerbate pathogen-induced inflammation [119]. Additionally, aged B cells have a reduced ability to differentiate self from non-self-antigens because of the oligoclonal expansion of B lymphocyte subpopulations rich in antigen-experienced cells. These subpopulations of B cells in mice have age related B cell features (CD21low/-, CD23low/-), and others express CD5, enabling the production of low-affinity antibodies [120]. CD5 + B cells are associated with various autoimmune diseases and can produce natural antibodies that typically target infectious agents. With age these natural antibodies may display a repertoire that is indicative of autoreactivity [121]. This characteristic is significant in the context of autoAb generation, as aging induced modifications to B cell populations can initiate an autoimmune response [122].
Related to these findings, the loss of immunological tolerance to auto-antigens in aging is also associated with an increased frequency of aged associated B cells (ABCs in mice) and double negative (DN2 in humans) populations [123, 124]. ABCs are considered antigen-experienced B cells, identified as CD21- and CD23- B cells, which also contain fractions of T-bet+, CD11b+, and CD11c + cells [108, 125, 126]. Heterogeneity in surface marker expression has been observed in a mouse model of SLE, suggesting ABC-like subsets can arise in autoimmune diseases [127]. ABCs are enriched in patients with autoimmune disease and appear in in-vivo models of autoimmunity, where they have been observed to drive inflammation, producing autoAb [126]. ABCs are thought to arise from the exhaustive expansion of the follicular B cell pool that occurs naturally through aging [125], through signaling pathways that induce T-bet expression [128] and are responsive to TLR7/9 stimulation [125]. The transcription factor ZEB2 was recently found to be required for ABC formation and production of IgG2c, an inflammatory isotype of ABCs [129]. This unique population of B cells is characterized as being pro-inflammatory and autoreactive [130] while producing a unique secretome and transcriptome in patients with RA [131]. In humans with SLE, the abundance of ABCs correlates with disease severity [132], suggesting a potential pathogenic function of this B cell subset. Notably, ABCs from old mice secrete higher levels of IgG antibodies against MDA and adipose tissue-derived antigens [111], suggesting that during aging, ABCs contribute to chronic inflammation and autoAb production. ABCs have been shown to accumulate in the spleen, liver, and salivary glands in old mice, where they induce inflammation and suppress B cell lymphpoiesis [133, 134]. These results suggest that ABCs can exert detrimental functions throughout a variety of tissues, while impairing B cell lineage progression. Future research will need to determine the full spectrum of ABCs induced pathology in immune and non-immune tissue.
Temporal dynamics of AutoAb during the lifespan
AutoAb development observed during aging appear to follow temporal patterns becoming enriched during ARDs. For example, one study found that the abundance of autoAbs was significantly greater in people aged 65 years and older compared to those younger than 45 [135]. AutoAb abundance was also greater in those diagnosed with AD, MS, and Parkinson’s disease (PD) [135]. Yin and co-workers found 3 waves of autoimmune signatures corresponding to ages 30, 50, and 62. 162 autoAbs were shared between the 30- and 62-year-old age groups, while each age group had 546 and 118 unique autoAbs, respectively [136]. Interestingly, those in the 50-year-old group had 12 unique autoAbs, suggesting that throughout aging, autoAb development occurs in waves, rather than linearly. Longitudinal sampling from 35 patients within the Swiss HIV Cohort Study revealed the presence of autoAbs for INFɑ, β, and ⍵ during the 6th decade of life, on average [137]. Interestingly, autoAb to IFN-I may have occurred in subjects with reduced IFN-stimulated gene (ISG) levels, indicating innate immune dysfunction, or people with pre-existing autoimmunity who reacted against IFN-I injection therapy; these data suggest that aging, along with other factors, including pre-existing or concomitant loss of self-tolerance plus past exogenous IFN-I therapy (i.e., past exposure to antigen) may contribute to IFN-I autoAb development [137]. A recent multi-omics analysis of 108 individuals found two crests of dysregulated molecules that occurred at approximately the age of 40 and 60 [138]. The biological function modules associated with crest 1 (aged 44) included skin/muscle, cardiovascular disease, alcohol, lipid and caffeine metabolism, while those associated with crest 2 (age 60) were skin/muscle, cardiovascular disease, kidney function, immunity, carbohydrate, and caffeine metabolism [138]. Notably, functional analysis indicated that pathways related to antioxidant activity, oxidative stress, and oxygen carrier activity showed marked nonlinear changes in the age of 60s, while pathways related to mRNA destabilization, mRNA processing, and macroautophagy showed marked nonlinear changes in the age of 70s [138]. These changes may provide insights into the formation of age-related pro-inflammatory environments that trigger generation of autoantigens and potentially autoimmune responses in specific decades of life. A similar study investigating the proteome also found crests of aging proteins at 34, 60 and 78 years. These crests were largely defined by unique protein signatures relating to cardiovascular disease, Alzheimer’s disease, and Down Syndrome enriched in middle age (60) and old (78) age [139]. Additionally, two-thirds of the proteins that changed with age also changed with sex [139]. While molecular events triggering these waves of aging and the associated autoAb development have not been determined in a temporal order, positive correlations between sex hormones, renal, and immune aging were found in a cohort of individuals aged 20–45 [140]. Other patterns of nonlinear aging have been observed in DNA methylation dynamics of mice colons [141]. Therefore, the nonlinear waves of aging may be a result of multiple contributing factors, including hormones, shifts in metabolism, and epigenetics, the alteration of which can eventually contribute to a niche of chronic inflammation, favoring the pathological actions of autoAbs. The autoAb signatures corresponding to specific decades of life could hence be used as predictive markers for aging to determine the onset of ARDs. These data also suggest specific decades of life when interventions may be more crucial compared to others. Future research needs to determine at what point throughout the lifespan interventions provide the most robust protection.
Effector actions of autoantibodies in obesity and aging
Once generated, autoAbs exert their pathological functions through a variety of mechanisms that lead to prolonged immune activation, inflammation, and tissue damage. Some of these mechanisms include binding to Fc receptors (FcR), activating complement, neutralization of antigens, opsonization, and cell-specific depletion and clearance, which all modulate inflammation [13, 142, 143]. Features of premature immunosenescence, such as accelerated telomeric attrition and excessive production of pro-inflammatory cytokines, have been observed in some autoimmune diseases, such as RA [144]. Notably, increased titers of autoAbs are detected before the onset of the disease [145], indicating a potential early role of autoAbs in promoting immunosenescence and chronic inflammation. For example, the immune complexes ACPA-IgG activate macrophages via TLR4 and FcR, while complement activation by autoAbs can boost inflammation in the synovium [146]. These events create an inflammatory niche through the production of age-related cytokines, such as TNF and IL-6 [147, 148]. Similarly, IgG accumulated during aging can prime macrophages to a senescent state through IgG FcR, as indicated by increases in SA-β-Gal activity, p21 and inducible nitric oxide synthase (iNOS) expression and the production of nitric oxide, all of which are considered hallmarks of inflammatory macrophages [112]. In another example, autoAbs increase in patients with long COVID [149], along with prolonged complement activation leading to persistent immune activation and T-cell exhaustion, risk factors of immunosenescence [150,151,152]. We next discuss how such effector mechanisms of autoAb function are potentially influenced with a focus on obesity and/or aging.
Effector actions through dysregulations in the complement system
As one important axis of the antibody effector function, the regulation of antibody-mediated complement activation becomes impaired during inflammaging, potentially due to alterations in complement protein levels and the impact of cellular senescence [153, 154]. Studies have shown that levels of key complement components, such as C3 and C4 are elevated with age [155], while higher C5a levels are correlated with disease severity in AD patients [156], implicating a role of complement factors in contributing to an enhanced chronic inflammatory state. BM-derived B cells from lupus-prone mice expressing the human complement receptor 2 (hCR2), demonstrated increased susceptibility to apoptosis and decreased ANA [157]. The presence of C3 was associated with a down-regulation hCR2, suggesting that C3 may interfere with central tolerance mechanisms, enhancing the probability of positively selecting autoreactive B cells [157]. Beyond its ability to drive aging via broad crosstalk with hallmarks of aging, such as altered metabolism and mitochondrial functions [158], often overlooked is the potential role of complement activation in modulating the autoreactivity of GC B cells. Specifically, it has been shown that complement-opsonized immune complexes are delivered by follicular B cells shuttling from the marginal zone to the FDCs in GCs [159], where antigen-specific B cells then capture the FDC-processed antigens and present them to CD4 + T cells, acquiring survival signals [160]. During aging, a decline in the phagocytic efficiency of immune cells, such as macrophages, can result in the accumulation of complement-opsonized debris and apoptotic cells, potentially containing self-antigens [161]. The binding of these complement-coated immune complexes to self-antigen-specific B cells can eventually lower the activation threshold for these cells, allowing them to bypass normal tolerance mechanisms [162].
Effectors actions of antibody-dependent enhancement through FcR signaling
In parallel, the FcRs are also critical determinants in autoAbs effector function by modulating immune responses, such as inflammation and immune cell activation, which can either enhance or suppress autoimmunity. Increased inflammatory signaling by effector cell activation via FcRs is seen in the pathogenic immune response, termed antibody-dependent enhancement (ADE). FcR engagement with non-neutralizing or sub-neutralizing antibodies promote viral infection, intensifying diseases [163, 164]. A relevant component of ADE lies in the context of pre-existing immunity typically induced by prior vaccination or infection, leading to enhanced FcR signaling, driving the expression of proinflammatory cytokines [165]. It is therefore not surprising that FcR are being studied as a potential mechanism mediating autoimmune disease, where autoAbs can interact with a variety of FcRs leading to ADE [166, 167]. IgG autoAbs taken from patients with SLE were shown to opsonize late apoptotic Jurket cells, inhibiting their ability to be phagocytized in a FcγR-dependent mechanism [168]. While the consequences of increased apoptotic cells were not investigated, one could speculate this could be potentially deleterious and disease-escalating. Fukue and co-workers found that the disease-modifying anti-rheumatic drug, abatacept, downregulated FcγR1 expression on monocytes in patients with RA. The decrease in FcγR1 expression led to suppression of pro-inflammatory cytokine production in ACPA immune complex treated cells [169]. Excessive engagement of FcRs through autoAb-dependent mechanisms appear to promote chronic inflammation, which could be involved in aberrant immune responses seen during obesity and aging. FcγR -/- mice fed a HFD had less weight gain, decreased insulin resistance and lower expression of pro-inflammatory genes compared to wild type controls [170]. In aging, increased IgG hinged on FcRn mediated recycling by macrophages [171]. Ablation of FcRn lowered adipose tissue IgG, improved survival and glucose metabolism, and facilitated a beiging phenotype in eWAT (epididymal white adipose tissue) [171]. FcR signaling was shown to activate a proliferative response in microglial cells when mice were administered anti-myelin oligodendrocyte glycoprotein (MOG) antibodies [172]. While pathogenic or therapeutic outcomes were not studied, the induction of microgliosis was amplified in mice harboring a constitutively active, plasma membrane-bound Brutons tyrosine kinase (BTK) [172]. This is interesting because reports from our laboratory and others have shown BTK as one of the top autoantigens in serum [3] and stromal vascular fraction [71] in obese individuals. This may mean that a portion of autoAbs seen in obesity could be involved in a compensatory mechanism to mitigate the diseased state. These studies suggest that FcR signaling is an important mediator in autoreactivity. Understanding how immune complexes interact with FcR in obesity and aging will be of high importance for future research.
Natural killer cells augment inflammation-driven AutoAb effector actions
Interestingly, as an effector of antibody-dependent cellular cytotoxicity (ADCC), NK cells in aged murine BM show a 2-fold expansion, contributing to reduced B cell lymphopoiesis via TNF-α-mediated inhibition of E47, a transcriptional regulator of B lineage commitment [173]. NK cells also show subset-specific effects in mediating tissue inflammation and senescence. For instance, IgG levels in SS patients and seropositive patients with early RA (i.e. positive for ACPA and/or RA) are correlated with the ratio of CD56bright to CD56dim NK cells, where the reduction in the circulating CD56dim NK cells contributes to accumulation of these cells into glands (and joints in the case of RA), enhancing focal immune injury and eventually systemic inflammation through secretion of pro-inflammatory cytokines (e.g. IFN-γ and TNF-α) [174,175,176]. While chronic inflammation driven by autoAbs plays a contributing role in the induction of immunosenescence, more work is needed to determine the relative contributions of obesity- or aging-associated autoAbs linked to NK cell-mediated ADCC.
Autoantigen targeting in obesity and aging
Immunogenicity of autoantigens in obese humans and mice
The immune responses mediated by autoAbs rely on the immunogenicity of autoantigens, which can be generated through obesity-associated events such as PTMs [177], dysregulated lipid metabolism [178], cellular stress, and perturbed proteostasis [179] (Fig. 1A). One of the top IgG antibody targets in obese human males with impaired insulin sensitivity, and even in studies of type 1 and 2 diabetes, is glial fibrillary acidic protein (GFAP), an intermediate filament protein expressed primarily in astrocytes and non-myelinating Schwann cells [3, 180,181,182]. Increased astrogliosis and GFAP immunoreactivity increased in the hypothalamus in mice fed a HFD, which formed distinct patterns around microvessels [183]. The close proximity to the vasculature may indicate a release of GFAP by activated astrocytes during obesity-associated central nervous system (CNS) inflammation. Previous reports confirmed that GFAP can be recognized by autoreactive IgG antibodies that can induce abnormal glucose metabolism [3, 184]. Given its correlation with the onset of T2D, GFAP has been used as a T2D biomarker in both humans and mice [180]. However, due to its location in immune privileged sites like the brain, targets like GFAP likely require an inflammatory milieu to break tolerance. In fact, many autoantigens that have been discovered linked to insulin resistance, which precedes T2D, are intracellular antigens present across many tissues (Table 1), and would likely need the correct inflammatory milieu to unmask their antigenicity to break tolerance [3]. How such inflammatory milieu is formed and how additional autoAb-autoantigen interactions contribute to age-related pathologies in the CNS and other tissues will be detailed in the later section.
In addition, products related to altered metabolism can also contribute to the immunogenicity of autoantigens during obesity. For example, elevated levels of advanced glycated end product (AGE)-modified proteins, such as serum albumin, are observed in circulating immune complexes of hyperglycemic plasma from patients with diabetes [185]. This observation is consistent with the findings that increased levels of circulating antibodies against AGE-modified proteins are found in patients with coronary atherosclerosis [186]. Notably, RA patients with T2D also exhibit high serum levels of autoAbs against AGE-modified IgG, exacerbating the disease severity [187]. Furthermore, obesity-associated dyslipidemia enriches the repertoire of self-lipid antigens, including various glycosphingolipids (GSLs), recognized by CD1d invariant natural killer T cells (iNKT) [187]. These iNKT cells are capable of promoting B cell production of autoAbs via cognate or non-cognate manners [71], as seen in a murine model of human lupus with heightened levels of anti-lipid IgG antibodies [188].
In addition to products of metabolic dysregulation, self-proteins from obesity-associated chronic inflammation are also candidates for autoantigens, as they are released from injured tissues during chronic inflammation. Antigens resulting from obesity-associated inflammation, including MDA and adipose-derived antigens also induce IgG autoAb production, interfering with the normal production of neutralizing antibodies upon viral challenges in obese SARS-CoV-2 patients [67]. Antigenic targets from chronically inflamed VAT will be discussed in more detail below. Notably, antigens from preexisting autoimmune conditions show an amplified antigenicity during obesity. For example, HFD feeding in MRL-lpr (lymphoproliferation) mice is accompanied by increased plasma levels of anti-dsDNA and ANA IgG antibodies, leading to an exacerbated SLE phenotype [189].
Taken together, these findings show that obesity may not only induce metabolic reprogramming in B cells and other immune cells but also facilitate structural and functional changes in autoAbs and autoantigens, which may also increase during low-level tissue damage with chronic inflammation. These events can worsen metabolic outcomes and favor the induction of autoimmune responses. In return, these changes can accelerate immunosenescence, weakening the immune system, and increasing susceptibility to infections and autoimmune diseases.
Immunogenicity of autoantigens in aging humans and mice
The aging process is driven by interconnected cellular, molecular, and systemic events, termed the hallmarks of aging [190], that ultimately lead to the breakdown of homeostasis and mortality [191]. In 2023, the hallmarks of aging were updated to include 12 interdependent features of cellular alterations that contribute to the decline in organismal function [190]. Our laboratory recently explored the hallmarks of aging through the view of immunity, by adding the contribution of chronic inflammation to each hallmark, integrating the immune system with all the features of aging [5]. Autoantigens found in aging populations are associated with pathways involved in the hallmarks of aging. Here we further explore the immune system’s participation in the hallmarks of aging with a particular emphasis on autoAbs and autoantigens.
During aging and ARDs, the ribosomal protein S6 kinase (RPS6KA6) has been reported as a top autoantigen associated with serum IgG autoAbs [136]. Additionally, eukaryotic translation initiation elongation factor 3 (eIF3) and ribosomal P proteins were autoantigen targets of autoAbs in autoimmune cerebellar ataxia, an age-associated autoimmune disease, and in patients with SLE [192, 193]. Such reductions in translational capacity and ribosome biogenesis have been shown to occur during aging, potentially leading to protein aggregates and perturbed protein translation [194, 195]. Dysregulated proteostasis also arises when there is dysregulated autophagy. While there is limited evidence that autoAb target autophagy-related proteins during aging, there are several links between autophagy and autoimmune diseases [196]. Individuals with aged-related macular degeneration (AMD) have several circulatory autoantigens related to autophagy that are targets of autoAbs [197]. In another study, among patients with wet or dry AMD, the top autoantigens found in their blood included alpha-synuclein, annexin V, heat shock protein 10 (HSP10), and GAPDH [198]. The range of antigens found in this study suggests that autoAbs are targeting proteins involved in protein folding, metabolism, and apoptosis. Mice lacking the immunity-related GTPase, IRGM1 gene, manifested an aberrant immune response characterized by type 1 interferonopathy, similar to the autoimmune disease Sjorgen’s syndrome [199]. The accumulation of type 1 IFN was due to impaired mitophagy, another feature of aging, which increased cytosolic mtDNA and mtRNA initiating an autoimmune response [199]. Although additional research is required to identify the autoantigen targets associated with aging, these studies underscore the interaction of autoAbs and their role in autophagy and age-related processes.
Increased fibrosis is associated with aging and is manifested in many ARDs, such as chronic kidney disease, fatty liver disease, idiopathic pulmonary fibrosis (IPF) and heart failure [200]. Analysis of lung tissues taken from older patients (65 ± 10 years of age) with IPF showed increased senescent biomarkers compared to controls. Additionally, treating fibroblasts with culture media taken from senescent cells resulted in greater expression in pro-fibrotic genes [201], suggesting that senescence factors can promote fibrosis. IgG antibodies in adipose tissue increase linearly with age in both mice and humans and may instigate a pro-fibrotic environment [171]. Treating young mice with IgG from old mice to mimic aging increased the expression of genes related to tissue fibrosis [171]. These authors then determined that IgG treatment led to higher levels of phosphorylated MEK, ERK1/2, and RAS GTP, which upregulates TGF-β in BM-derived monocytes and promotes fibrosis [171]. Mice deficient in B cells were protected from IgG-induced adipose tissue fibrosis, suggesting B cells can communicate with other immune cells to drive tissue remodeling [171]. In aging patients with different subtypes of pulmonary fibrosis, shorter leukocyte telomere lengths were observed compared to healthy age-matched controls [202]. Idiopathic pulmonary fibrosis was found to have one of the highest rates of telomerase mutations [203]. These studies provide evidence that pro-fibrotic environments are associated with telomere attrition.
In an autoimmune fibrotic disease, such as systemic sclerosis, characterized by accelerated aging, patients exhibit several hallmarks of aging such as mitochondrial stress, chromosome instability, and shortened telomere length [204]. Furthermore, a subset of patients diagnosed with systemic sclerosis had higher autoAbs targeting the telomere/shelterin complex, including the telomeric repeat-binding factor 1 (TERF1) as the most common autoantigen [205]. Another study discovered 8 additional autoantigens, including TERF2, associated with telomeres or telomerase in systemic sclerosis patients [206]. Analysis of peripheral blood leukocytes in systemic sclerosis patients revealed a negative association between TERF1 autoAbs and telomere length. These authors screened an additional cohort of patients with idiopathic pulmonary fibrosis and discovered that 11 out of 152 patients had TERF1 autoAbs [205], suggesting a potential relationship between telomere dysfunction, fibrosis, and the immune system. Notably, TERF1 levels decrease during aging in mice and humans, and a study using gene therapy to transiently overexpress TERF1 in mice significantly prevented age-related decline in neuromuscular function, glucose tolerance, and cognitive function [207, 208]. While studies that characterize TERF1 autoAb levels in healthy aging individuals are lacking, the correlation between TERF1 autoAbs, telomere shortening, and potentially age-adjusted telomere decline highlights their possible role in aging and immunosenescence. This area may benefit from further research, as understanding these connections could enhance our knowledge of telomere biology in aging and ARDs. Finally, aged mice lacking the gene for keratin 8 displayed high titers of autoAbs against the mitochondrial proteins, including glutamate dehydrogenase, catalase, mitochondrial HMG-CoA synthase, and aldehyde dehydrogenase E2 [209]. Therefore, mitochondrial health appears to rely on the integrity of intermediate filaments like keratin, the loss of which may promote oxidative stress and mitochondrial dysfunction. These studies suggest that alterations to the cytoskeletal network are associated with elevated autoAbs in pathways that are connected to some of the hallmarks of aging. More work will be needed to determine if changes in environmental forces associated with fibrosis and aging also facilitate autoantibody mediated production [210].
Autoantigen targets during obesity and aging
Skeletal muscle
Skeletal muscle accounts for roughly 40% of total body weight and plays a vital role in movement, posture and metabolism [211]. Aging and obesity compromise the regeneration of skeletal muscle and are accompanied by the loss of muscle mass and function [212, 213]. Diseases such as diabetes impact multiple cell populations within the skeletal muscle niche, resulting in a decline in muscle regeneration [214]. Single-cell RNA sequencing of human skeletal muscle has shown that aging is characterized by increased B and T cell populations, while M2-like macrophages decline [215]. In another human study, using single nuclear RNA sequencing study, antigen presentation, IFNγ responses, and complement cascades were upregulated in immune cells or fibro-adipogenic progenitors in old muscle, raising the possibility of classical antibody-dependent complement fixation signatures [216]. Additionally, blunted muscle hypertrophy and higher expression of senescence markers were found in old mice after surgery-induced muscle hypertrophy [217]. These results suggest that aging muscle is met with remodeling of the immune system and age-induced senescence which could be a contributing factor to sarcopenia (Fig. 1B).
Specific autoantigens have been investigated in skeletal muscle in the context of autoimmune disease, like Myasthenia Gravis (MG) or myositis, but less so in the context of obesity or aging. In MG, almost 90% of patients have IgG1 acetylcholine receptor autoantibodies and about 10% of patients have IgG4 muscle-specific kinase autoantibodies [218]. Myosin heavy chain has also been shown to be an autoantigen in skeletal muscle, and mice immunized with purified skeletal muscle myosin developed experimental autoimmune myositis [219, 220]. Additional autoantigens found in myositis include Histidyl tRNA synthetase (HRS/Jo-1), Mi-2, U1-70kD, and the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) [221]. Interestingly, these autoantigens are also expressed during skeletal muscle regeneration, suggesting that different stages of skeletal muscle injury/recovery may pose greater risks for autoAb pathogenicity. Furthermore, regeneration is impaired in aged skeletal muscle, which may be in part a result of dysregulated autoAb accumulation. For example, IgG-mediated autoimmunity has been proposed to be involved in the pathogenesis of sarcopenia, including potential targeting of cardiac troponin T in skeletal muscle [222]. Specifically, an increased abundance of IgG1 and IgG4 in skeletal muscle was associated with declined physical performance in humans and mice [218]. IgG co-localized with markers of the complement pathway, necroptosis, and loss of dystrophin [218] further linking autoAbs and skeletal muscle pathology. Going forward, it will be interesting to investigate whether subsets of obesity, including patients with sarcopenic obesity, which shows overlap with muscular features of aging, has any overlap in autoantibody repertoires.
Cardiovascular tissue
The prevalence of cardiovascular disease (CVD) increases during aging [223], in association with other risk factors such as smoking, obesity, hypertension, diabetes, sedentary lifestyle, poor dietary choices, and socio-economic status [224]. A major cause of CVD is atherosclerosis, which is now considered an immune-mediated etiology [225]. Foamy macrophages found in early and advanced atherosclerotic lesions express senescence markers [226], suggesting a role for immunosenescence in the pathology of atherosclerosis. Additionally, several autoantigens involved in the development of atherosclerosis have been detected (Fig. 1B). Glucose-regulated protein 78 (GRP78), an endoplasmic reticulum (ER) chaperone, can localize to the cell surface under periods of ER stress. In the early stages of atherosclerosis development, cell surface localization of GRP78 and increased anti-GRP78 autoAbs are seen, leading to NF-κB signaling in endothelial cells, and accelerating atherosclerotic lesion growth [227]. Another autoantigen found in atherosclerosis is heat shock protein 60 (HSP60). Injecting apolipoprotein E (ApoE)-deficient mice with anti-HSP60 autoAbs led to increased lesions in the aortic sinus [228]. Lorenzo and co-workers determined autoAbs that were reactive against atherosclerotic plaques. Interestingly the reactivity patterns were diverse, indicating that the plaques contained multiple antigenic targets [229]. One particular autoantigen studied was aldehyde dehydrogenase 4 family member A1 (ALDH4A1). These authors showed that anti-ALDH4A1 autoAbs delayed atherosclerosis plaque formation when infused into low-density lipoprotein receptor-deficient (Ldlr-/-) mice [229]. Other commonly studied targets during atherosclerosis include potentially pathogenic IgG antibodies against oxidation specific epitopes, and atheroprotective natural IgM anti-phosphorylcholine types [230]. Finally, an emerging antigenic target of adaptive immunity during atherosclerosis includes ApoB, though current research has been skewed more towards T cell targeting of ApoB than B cell targeting [231]. In summary, during CVD, several autoantigens can exacerbate disease, while others become targets of autoAbs, potentially delaying disease.
Adipose tissue and liver
The adipose tissue has a remarkable capacity to store excess energy, while also functioning as a regulator of hormones, appetite, immune responses, and thermogenesis [232, 233]. During aging, brown (BAT) and subcutaneous adipose tissues (SAT) decrease with a concomitant accumulation in VAT and ectopic lipids [234]. In addition to adipocyte remodeling, immune infiltration into the VAT increases resulting in a pro-inflammatory state during aging and obesity [6]. Obesity suppresses the secretion of regulatory adipokines, such as adiponectin, further leading to whole-body metabolic impairments [235]. Increased senescent preadipocytes were observed in VAT isolated from aged donors [236]. The senescent preadipocytes were characterized as being pro-inflammatory and capable of inducing inflammation in neighboring cells [236]. Therefore, aging and obesity promote a detrimental remodeling of adipose tissue, which may enhance immunosenescence. Adipose tissue has been found to not only be a source of inflammation but also the production and secretion of autoAbs [71, 237] (Fig. 1B). Indeed, self-antigens may be released from VAT through additional mechanisms linked to aging and obesity, including hypoxia, cell cytotoxicity, metabolism, hormones and DNA damage [71] (Table 1). Patients with general acquired lipodystrophy had anti-Perilipin-1 IgG antibodies that were capable of inducing lipolysis in-vitro [238]. Treating VAT cultures from obese patients with IgG-ACPAs, commonly seen in RA, led to increased expression of pro-inflammatory markers, while suppressing the expression of genes involved in the insulin signaling pathway [239].
In addition, obesity is associated with metabolic liver disease, driven in part by lipotoxicity, immune-mediated inflammation, and glucotoxicity, inducing liver damage [240,241,242]. Recently, a high-fat high-fructose (HFHF) diet was shown to promote MHC-II presentation of epitopes from protein disulfide isomerase family A member 3 (PDIA3) [15]. Anti-PDIA3 AutoAbs showed isotype switching from IgM to IgG3, and transfer of PDIA3-specific antibodies could exacerbate hepatocyte death. Increased humoral responses to PDIA3 were also observed in patients across a diverse array of chronic inflammatory liver conditions, such as autoimmune hepatitis, primary biliary cholangitis, and T2D, further supporting PDIA3 as an important liver-derived autoantigen during liver inflammation. Intriguingly, a novel subset of adipose tissue macrophages, termed inflammatory and metabolically activated macrophages (iMAMs), was found in obese humans and mice and enriched in PDIA3 expression [243]. PDIA3, in turn, exerts redox control on RhoA activity, strengthening the pro-inflammatory and migratory capacity of iMAMs through RhoA-YAP signaling, contributing to worsened VAT inflammation and metabolic outcomes [243]. This finding highlights an underappreciated role of the mechanosensing immune cells [244] and PDIA3 self-antigens in mediating a mechano-redox control of inflammation, especially during obesity and aging, where the VAT and the liver exhibit increased tissue stiffness due to hypertrophy, extracellular matrix accumulation, and fibrosis [245,246,247,248].
Spleen
As a critical extramedullary hematopoietic immune organ, the spleen maintains the homeostasis of the immune system in cooperation with the BM. However, aging is associated with an altered immune landscape in the spleen as it houses autoAb-producing B cells and other autoreactive immune cells that favor a pro-inflammatory and pro-immunosenescent phenotype [249]. The ABCs, as introduced above, are autoAb-secreting B cells often found to accumulate in aged or autoimmune-prone mice [125, 128]. For example, ABCs expand in spleens of MRL/lpr mice and produce autoAbs, such as anti-nucleosome and anti-RNA IgG, upon stimulation of TLR7 and TLR9 [127]. Other autoAbs specific to self-antigens include RF and ds-DNA are linked to conditions like SS, SLE, and malaria anemia [126]. The expansion of ABCs and autoreactive B cells in general might be related to impaired regulatory B cell (Bregs) function in the spleen. Specifically, Bregs can secrete IL-10, IL-35, and TGF-β to suppress CD4 + T cell function (including the pathogenic Th17 cells in the spleen that can promote autoAb production [250] and Tfh cell differentiation [251, 252] in patients and mice with autoimmune diseases, potentially preventing autoAb formation [253]. However, whether Bregs can directly suppress ABC-induced inflammation during aging and obesity needs further investigation.
Obesity fosters a chronic inflammatory state that promotes the survival and activation of splenic B cells. In DIO mice, the spleen often contains a lower percentage of IgM + IgD − cells and shows reduced spontaneous IgM production but increased IgG secretion, indicating that HFD-induced obesity induces a systemic humoral immune response [3]. T-bet + CD11c + B cells are also found to increase in spleens of HFD-fed mice and humans with higher body mass index [72]. There is a marked correlation between the proportion of T-bet + CD11c + B cells in the spleen and serum levels of IgM and IgG2c in both WT and Cd1d1-/- mice, suggesting that the expansion of these B cells and increased antibody production in obesity are supported by iNKT cells, an inflammatory subset capable of producing IFNγ [72]. Taken together, these findings show that the spleen plays an important role in autoAb-driven inflammation, as seen in autoimmune diseases, aging, and obesity. Mechanistically, the extrafollicular B cell activation and reduced GC responses lead to heightened production of pro-inflammatory cytokines and autoAbs, which are risk factors for immunosenescence [254,255,256].
Gastrointestinal
An emerging area of research is the interaction between the microbiota and host antibody production. Specific gut microbes can be a source of antigenic mimicry, induce autoreactivity in immune cells, or secrete metabolites that control inflammation [257]. Aging and obesity are associated with dysbiosis and weakening of the gut barrier integrity, exposing the intestinal immune system to various microbial pathogens [258, 259], some of which may exhibit molecular mimicry of certain autoantigens of the host. For instance, commensal bacteria expressing orthologs of Ro60, an RNA-binding protein commonly targeted by autoAbs in SLE, can trigger autoimmunity in genetically susceptible individuals [260]. In ulcerative colitis and some CD patients, anti-neutrophil cytoplasmic antibodies (ANCA) are strongly correlated with neutrophil extracellular traps (NET) formation and increased gut permeability [261,262,263,264]. ANCA also exhibits cross-reactivity with the outer membrane porin C (OmpC) of Bacteroides caccae and Escherichia coli, further instigating the inflammatory bowel disease (IBD) [265]. Additional self-antigens and microbial antigens exhibiting cross-reactivity with self-antigens, including glycoprotein 2 (GP2) and family with sequence similarity 84 member A (FAM84A), have been reviewed [266].
In addition, increased gut permeability during aging and obesity also allows for increased exposure of pathogenic bacteria and LPS to GC B cells in the intestinal TLS, accelerating B cell inflammatory function, senescence-like changes, leading to less IgA beneficial for homeostasis, decreased IgA diversity, and increased IgA autoAbs [261, 267,268,269], further contributing to inflammaging and immunosenescence [263] (Fig. 1B). Future research will need to elucidate the relationship between autoAbs and the microbiome in the context of aging and obesity. Moreover, it is also worth studying how B cells traffic from the intestine can impact tissue-specific inflammation and autoAb production across other tissues, or how gut-related therapies, including microbial mimics and dietary interventions could be used to target gut inflammation and/or pathogenic antibody production during obesity or aging to curb these conditions [264, 270]. Altogether, gut-derived antibodies contribute to the persistence of inflammation by continuously stimulating the intestinal immune system, leading to tissue damage and chronic inflammation associated with immunosenescence, impacting the gut’s ability to respond to constant bacterial challenges and maintain immune tolerance.
Central nervous system
Anti-neuronal autoAbs have been found in the periphery and in several brain-associated regions including the cortex [271], in the meningeal dura mater [272], and cerebrospinal fluid [273]. Here, they are expected to drive pathology in cases of autoimmune disease. In age-related cognitive conditions like PD and AD, autoAbs can drive both protective and deleterious effects (Fig. 1B). AutoAbs and other antibodies have been best studied in AD. Anti-amyloid antibodies are found at differential levels in AD patients, partially representing the sequestration by plaques [274]. These autoAbs are expected to be protective, targeting plaques for clearance by the abundant microglia population in AD, even being developed into drug candidates [275]. Given the linkages between APOE and AD, antibodies against APOE isoforms are being developed [276] but there is little evidence of their natural occurrence and autoAbs in AD. AutoAbs against other amyloid family proteins also represent an interesting AD resilience factor. The pancreas-derived islet amyloid polypeptide (IAPP)-IgA autoAb was also significantly lower in APOE4 carriers [277]. The IgA isotype is generated in the mucosa upon sampling of antigens encountered in tissues like the gut and lung [278]. Conversely, several other autoAbs were found at higher levels that were not neuron-specific [279], also representing chronic inflammation. Features of insulin resistance have been linked to AD and neurodegeneration [280]. The protective IgA class of antibodies generated in the gut is reduced in obese mice [262]. Whether this leads to increased levels of protective autoAbs or specifically neuroinflammation is unclear. As mentioned above, GFAP may be another CNS target incited during obesity, and studies in rats have shown increased deposition of IgG in the brains of HFD rodents, correlating with infiltration of microglia [184].
In PD, autoAbs against alpha-synuclein were found in increasing quantities in blood and cerebrospinal fluid (CSF) of patients, with a bias in males [281]. These synuclein autoAbs are expected to target neurons, drive inflammation and promote PD pathology [282]. Also, GFAP autoAbs found in DIO target cells like astrocytes and expected to correlate with astrogliosis and PD [274]. Interestingly, renin-angiotensin system (RAS) autoantibodies were also increased in PD serum [283], likely representing systemic inflammation linked age-related breakdown in tolerance.
Potential therapeutics extending lifespan and healthspan
Given the critical role that autoAbs play in driving chronic inflammation during aging and obesity, the potential of therapies traditionally purposed for autoimmune diseases in resolving inflammaging and improving health/lifespan is worth exploring. However, although autoAbs can have significant clinical effects and provide valuable insights for drug development, the employment of therapies targeting autoAbs or autoreactive B cells to counteract inflammaging or obesity still represents an understudied niche. A major obstacle has been the absence of experimental tools capable of unbiased, high-throughput detection of autoAbs across the human population on a proteome-wide scale. However, technologies, such as rapid extracellular antigen profiling (REAP), antigenic arrays, and phage immunoprecipitation sequencing (PhIP-seq) [284, 285], will allow for “autoAb-wide association studies”, enabling the identification of disease-modifying autoAbs not only for autoimmune diseases but also for aging and metabolic diseases [13] (Fig. 2). Consistently, one study mapping over 730,000 human-derived peptides showed that each individual, regardless of disease states, has unique and complex finger-prints of autoAbs that were best targeted using therapies against plasma cell markers like B cell maturation antigen (BCMA), instead of anti-CD19 or anti-CD20 [286].
Potential therapies targeting autoimmune components during aging and obesity. With next-generation autoAb detection methods, such as PhiP-Seq, antigen arrays, and REAP, playing a central role in providing a more detailed atlas of autoAbs, therapies traditionally used for autoimmune diseases can be repurposed to conquer the autoAbs and autoantigens generated during obesity and aging. T cell-based therapies targeting autoreactive B cells, antibody-based therapies targeting pro-inflammatory factors, and tolerance-inducing vaccines based on anti-idiotypic antibodies and antigen-encoding mRNA vaccines, are emerging techniques that are gaining increasing research focus. PhiP-Seq = phage immunoprecipitation sequencing; REAP = rapid extracellular antigen profiling; CAR-T = Chimeric antigen receptor T cells; CAAR-T = Chimeric autoantibody receptor T cells; BiTEs = Bispecific T cell engagers; SAAs = senescence-associated antigens
However, recent advancements in treating autoimmune diseases have been made by targeting autoAb-secreting B cells, an integral component to be reset during the sequential immunotherapy of autoimmune diseases [287]. Specifically, chimeric antigen receptor (CAR) T-cells targeting CD19 have been shown to successfully treat a patient with severe myositis and systemic sclerosis [288], as well as severe, anti-acetylcholine receptor antibody (anti-AchR) positive generalized MG [289]. B cells producing specific autoAbs can also be targeted by the chimeric autoantibody receptor (CAAR) T cells, which are engineered to produce and present specific antigens on their surface, allowing for recognition by corresponding B cells, which are consequently eliminated. Successful application of CAAR T cells has been made in treating anti-NMDA receptor (NMDAR) autoAb-mediated NMDAR encephalitis, the most common autoimmune encephalitis [290], as well as in anti-desmoglein 3 autoAb-mediated pemphigus vulgaris [291]. The CAR-based therapies have also shown the potential in aging research, for example, by targeting senescent cells markers, like urokinase plasminogen activator receptor (uPAR), to ameliorate the metabolic function decline in aged mice or DIO mice [292, 293]. Meanwhile, various senolytics or clearance of senescent cells, such as p21high cells, have shown efficacies in improving physiological functions during aging [294,295,296]. This research implicates the possibility of targeting senescent cells with antibodies against senescence-associated peptides, such as those encoded by Tns3 and Tmed3 in mice, which have shown high immunogenicity [297]. However, CAR-based therapies targeting age-related autoAbs warrant further investigation. Remarkably, with the newly developed CAR-enhancer technology and other CAR therapy improvements, the therapeutic potency of canonical CAR-based therapies will be significantly amplified [298, 299]. As well, additional approaches, such as the bispecific T cell engagers (BiTE) (e.g. blinatumomab that targets autoreactive B cells in RA) [300], may further expand the future therapeutic options for ARDs.
Importantly, some autoAbs possess dualistic properties that are sometimes beneficial and are less appreciated. For instance, IgG autoAbs generated in the WAT of DIO mice are found to facilitate phagocytosis of apoptotic adipocytes by macrophages, suggesting a beneficial role by some autoAbs during obesity [301]. Additional examples include anti-IFN-I autoAbs that can reduce SLE severity [302], anti-human epidermal growth factor receptor 2 (HER2) autoAbs that increase breast cancer patients’ survival rate [303], and anti-amyloidogenic peptides autoAbs that protect against AD [304]. Monoclonoal antibody-based drugs have been developed based on the antibodies above, including Anifrolumab for SLE [305], Trastuzumab for breast cancer [306], and Aducanumab for AD [307], indicating that autoAbs exhibit promising therapeutic value depending on different pathological conditions.
In addition, vaccines targeting autoAbs and “tolerance-inducing vaccines” targeting autoantigens have been gaining focus recently [308]. To avoid global immunosuppression during the induction of immune tolerance, antigen-specific tolerance represents a future avenue. For example, conjugating the antigen with a polymer glycosylated with N-acetylgalactosamine (pGal) induced antigen-specific tolerance mediated by Tregs in mice with experimental autoimmune encephalomyelitis (EAE) [309]. More importantly, anti-idiotypic antibodies, which target the idiotype of other antibodies, have gained interest for their potential to counteract harmful autoAbs in autoimmune diseases [310]. These antibodies are integral to the immune system’s regulation, as they can neutralize and inhibit the secretion of autoAbs, offering a novel form of immunotherapy with minimal side effects and prolonged immunity [311, 312]. While their application in age-related and metabolic diseases is not yet well-studied, emerging techniques like antigenic arrays, REAP, and PhIP-Seq are expected to identify candidate autoAbs, paving the way for vaccines targeting autoABs in these conditions.
Summary and concluding remarks
Aging and obesity contribute to autoAb production through chronic inflammation, immune dysregulation, and metabolic disturbances [7, 19]. Inflammaging is associated with disrupted immune tolerance and increased abundance of ABCs and other autoreactive B cells, leading to the production of autoAbs, such as anti-dsDNA and ANA IgG linked to AD and cardiovascular diseases [108, 189]. Obesity promotes similar effects through metainflammation, activating B cells through metabolic reprogramming towards a hypermetabolic, autoAb-secreting state [84]. Structural changes in antibodies, such as altered glycosylation, increase self-reactivity [162], while autoantigens can be generated through PTMs and metabolic disturbances, like AGE-modified proteins and glycosphingolipids, further exacerbating autoimmune responses [186].
These changes to antibodies during obesity and aging occur in the setting of chronic low-grade inflammation. Chronic inflammation is an important hallmark of aging, not only as a pathological consequence of disturbed homeostasis during aging but for its ability to instigate further age-related dysfunction [5]. AutoAbs resulting from obesity and aging can drive a common feedforward loop that promotes metabolic diseases, immune-mediated conditions, and a decline in immune function [147] (Fig. 3). In metabolic tissues, the gut, and the CNS, autoAbs lead to sustained immune activation, tissue damage, and oxidative stress, perpetuating a cycle of immune dysregulation [313, 314]. The chronic state of inflammation accelerates the aging of the immune system, resulting in immunosenescence, which in turn can promote a positive feedback loop that drives metabolic diseases and impaired health.
A vicious cycle of immune dysregulation during obesity and aging mediated by autoantibodies. During obesity and aging, the generation of autoantibodies depends on various interconnecting mechanisms, including ones that directly contribute to alteration in B cell functions and antibody structures, as well as several hallmarks of aging that promote the formation of autoantigens. The autoantibodies then play a critical role in fueling chronic inflammation that eventually contributes to immunosenescence, which continues to drive age-related diseases and metabolic syndromes. While obesity-related autoantibodies can contribute to metabolic syndromes, and age-related autoantibodies can contribute to age-related diseases (denoted as solid arrows), metabolic syndromes and age-related diseases can potentially create niches for the generation of age-related and obesity-related autoantibodies, respectively (denoted as dashed arrows). As a result, a vicious cycle between metabolic syndromes and age-related diseases forms and can exacerbate each other when driven by immunosenescence and chronic inflammation associated with autoantibodies. AGE = Advanced glycation end product; DAMPs = Danger-associated molecular patterns; PAMPs = Pathogen-associated molecular patterns; HFD = High-fat diet; SHM = Somatic hypermutation
The role of autoAbs in obesity, aging, and immunosenescence is a complex, yet vital, research area. Fueled by chronic inflammation, autoAbs not only accelerate the progression of metabolic syndrome and ARDs but also perpetuate a cycle of immune dysregulation that drives immunosenescence and impairs the healthspan. The persistent immune activation and tissue damage across various organs underscore the extensive impact of autoAbs on ARD progression. The rise in autoimmune diseases among the elderly, despite declining immune competence, is caused by a breakdown of immune tolerance. The accumulation of age-related failures in both central and peripheral tolerance likely contributes to the emergence of autoimmunity. Yet, significant gaps in our knowledge persist, including the extent to which these defects are intrinsic to B cells and how they interact with the systemic immune system.
A primary challenge is deciphering the molecular components and signaling networks that lead to tolerance breaches and subsequent autoAb generation. The heterogeneity and temporal variability of autoAbs present a substantial barrier, complicating our understanding and necessitating large cohort studies and longitudinal analyses [13]. In addition, while the role of autoAbs is central, the influence of genetic and environmental factors, as well as lifestyle needs to be considered [110]. To address the age-related generation of autoAbs, future research must investigate the cellular and molecular mechanisms by which immune aging transforms immune cells into auto-aggressive effectors. This includes investigating mitochondrial dysfunction, lysosomal failure, and chronic ER stress and expansion in T cells and B cells, as well as their communication with the environment, including microbes and the environmental exposome [101]. In addition, given the PDIA3 enrichment in macrophages in tissues of altered stiffness due to obesity and chronic inflammation, changes in potentially mechanosensing self-antigen levels might represent a novel target of study to ameliorate aging and obesity-related conditions [210]. In conclusion, by defining the interplay among various risk factors, we can enhance our understanding of the intrinsic and extrinsic factors that shape immunity as we age, paving the way for promising therapeutic strategies to combat autoimmune conditions associated with obesity and ARDs.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- APRIL:
-
A prolifeting-inducing ligang
- AchR:
-
Acetylcholine receptor
- AID:
-
Activation-induced cytidine deaminase
- AGEs:
-
Advanced glycated end products
- ABC:
-
Age-associated B cell
- ARDs:
-
Age-related diseases
- AMD:
-
Aged-related macular degeneration
- ALDH4A1:
-
Aldehyde dehydrogenase 4 family member A1
- AD:
-
Alzheimer’s disease
- ACPA:
-
Anti-citrullinated protein antibodies
- ADCC:
-
Antibody-dependent cellular cytotoxicity
- ADE:
-
Antibody-dependent enhancement
- ASCs:
-
Antibody-secreting cells
- ANCA:
-
Anti-neutrophil cytoplasm antibody
- AAV:
-
Anti-neutrophil cytoplasm antibody-associated vasculitis
- ANA:
-
Antinuclear antibody
- E-APOE:
-
Apolipoprotein
- AIM:
-
Apoptosis inhibitor of Macrophages
- AutoAb:
-
Autoantibodies
- BAFF:
-
B cell activating factor
- BAFF-R:
-
B cell activating factor receptor
- BLIMP-1:
-
B lymphocyte-induced maturation protein 1
- BCR:
-
B cell receptor
- BiTE:
-
Bispecific T cell engager
- BM:
-
Bone marrow
- BAT:
-
Brown adipose tissue
- CVD:
-
Cardiovascular disease
- CNS:
-
Central nervous system
- CSF:
-
Cerebrospinal fluid
- CAR:
-
Chimeric Antigen Receptor
- CAAR:
-
Chimeric Autoantibody Receptor
- CSR:
-
Class switch recombination
- COVID-19:
-
Coronavirus disease 2019
- CD:
-
Crohn’s disease
- CXCL12:
-
C-X-C Motif Chemokine Ligand 12
- CXCR4:
-
C-X-C chemokine receptor type 4
- DAMPs:
-
Danger-associated molecular patterns
- DNA:
-
Deoxyribonucleic acid
- DCs:
-
Dendritic cells
- DIO:
-
Diet induced obesity
- DNA-PKcs:
-
DNA-dependent protein kinase
- DN2:
-
Double negative
- dsDNA:
-
Double stranded DNA
- DMD:
-
Dystrophin
- eWAT:
-
epididymal white adipose tissue
- EAE:
-
Experimental autoimmune encephalomyelitis
- eIF3:
-
Eukaryotic translation initiation elongation factor-3
- FDC:
-
Follicular dendritic cell
- Tfh:
-
Follicular helper T cells
- FcR:
-
Fragment crystallizable receptor
- FcRn:
-
Fragment crystallizable receptor neonatal
- GAPDH:
-
Glyceraldehyde 3-phosphate dehydrogenase
- GC:
-
Germinal Center
- GFAP:
-
Glial fibrillary acidic protein
- GRP78:
-
Glucose regulated protein 78
- GSL:
-
Glyosphingolipid
- GP2:
-
Glycoprotein 2
- HSP:
-
Heat shock protein
- HFD:
-
High fat diet
- HFHF:
-
High Fructose/High Fat Diet
- HRS/Jo-1:
-
Histidyl tRNA synthetase
- HER2:
-
Human epidermal growth factor receptor 2
- HIV:
-
Human immunodeficiency virus
- IFN:
-
Interferon
- IL:
-
Interleukin
- Ig:
-
Immunoglobulin
- ITAM:
-
Immunoreceptor tyrosine-based activation motif
- ITIM:
-
Immunoreceptor tyrosine-based inhibitory motif
- IRGM1:
-
Immunity-related GTPase family M member 1
- iNOS:
-
Inducible nitric oxide synthase
- IGF:
-
Insulin-like growth factor
- iNKT:
-
Invariant natural killer T cell
- IAPP:
-
Islet amyloid polypeptide
- KO:
-
Knock out
- LPS:
-
Lipopolysaccharide
- Ldlr:
-
Low density lipoprotein receptor
- MDA:
-
Malondialdehyde
- MetS:
-
Metabolic syndrome
- mtDNA:
-
Mitochondrial DNA
- mtRNA:
-
Mitochondrial RNA
- mTOR:
-
Mammalian target of rapamycin
- MYH:
-
Myosin heavy chain
- MALT:
-
Mucosa-associated lymphoid structure
- MS:
-
Multiple Sclerosis
- Mtb:
-
Mycobacterium tuberculosis
- GlcNAc:
-
N-acetylglucosamine
- NK cells:
-
Natural killer cells
- NMDAR:
-
N-methyl-D-aspartate receptor
- NLR:
-
Nod like receptors
- NET:
-
Neutrophil extracellular traps
- OmpC:
-
Outer membrane porin C
- OXPHOS:
-
Oxidative Phosphorylation
- PD:
-
Parkinson’s disease
- PI3Kγ:
-
Phosphoinositide 3-kinase-gamma
- PhIP-seq:
-
Phage immunoprecipitation sequencing
- pGal:
-
Polymer glycosylated with N-acetylgalactosamine
- PTM:
-
Post-translational modification
- REAP:
-
Rapid extracellular antigen profiling
- Bregs:
-
Regulatory B cells
- Tregs:
-
Regulatory T cells
- RAS:
-
Renin-angiotensin system
- RA:
-
Rheumatoid Arthritis
- RNA:
-
Ribonucleic acid
- RPS6KA6:
-
Ribosomal protein S6 kinase
- SAAs:
-
Senescence-associated antigens
- SARS-CoV-2:
-
Severe acute respiratory syndrome coronavirus 2
- SHM:
-
Somatic hypermutation
- SLO:
-
Secondary lymphoid organs
- SASP:
-
Senescence-associated secretory phenotype
- SS:
-
Sjögren’s syndrome
- SAT:
-
Subcutaneous adipose tissue
- SLE:
-
Systemic lupus erythematosus
- TERF:
-
Telomeric repeat-binding factor
- TLS:
-
Tertiary lymphoid structures
- TLR:
-
Toll-like receptor
- TGF:
-
β-Transforming growth factor beta
- TNF:
-
Tumor necrosis factor
- T2D:
-
Type 2 diabetes
- VAT:
-
Visceral adipose tissue
- WT:
-
Wild Type
References
Fairweather D, Frisancho-Kiss S, Rose NR. Sex differences in autoimmune disease from a pathological perspective. Am J Pathol. 2008;173:600–9.
National Academies of Sciences, Engineering, and, Health M, and Medicine Division; Board on Population Health and Public Health Practice; Committee for the Assessment of NIH Research on Autoimmune Diseases. Enhancing NIH Research on Autoimmune Disease [Internet]. Washington (DC): National Academies Press (US); 2022 [cited 2024 Sep 28]. http://www.ncbi.nlm.nih.gov/books/NBK580299/
Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med. 2011;17:610–7.
Graham RC. Treatment of bacterial infections with specific antisera: a venerable idea born again. J Lab Clin Med. 1986;108:172–3.
Baechle JJ, Chen N, Makhijani P, Winer S, Furman D, Winer DA. Chronic inflammation and the hallmarks of aging. Mol Metab. 2023;74:101755.
Khan S, Chan YT, Revelo XS, Winer DA. The Immune Landscape of visceral adipose tissue during obesity and aging. Front Endocrinol (Lausanne). 2020;11:267.
Frasca D. Several areas of overlap between obesity and aging indicate obesity as a biomarker of accelerated aging of human B cell function and antibody responses. Immun Ageing. 2022;19:48.
Frasca D, Ferracci F, Diaz A, Romero M, Lechner S, Blomberg BB. Obesity decreases B cell responses in young and elderly individuals. Obes (Silver Spring). 2016;24:615–25.
Clemente DBP, Maitre L, Bustamante M, Chatzi L, Roumeliotaki T, Fossati S, et al. Obesity is associated with shorter telomeres in 8 year-old children. Sci Rep. 2019;9:18739.
Tzanetakou IP, Katsilambros NL, Benetos A, Mikhailidis DP, Perrea DN. Is obesity linked to aging? Adipose tissue and the role of telomeres. Ageing Res Rev. 2012;11:220–9.
Pérez LM, Pareja-Galeano H, Sanchis-Gomar F, Emanuele E, Lucia A, Gálvez BG. Adipaging: ageing and obesity share biological hallmarks related to a dysfunctional adipose tissue. J Physiol. 2016;594:3187–207.
Ghanemi A, Yoshioka M, St-Amand J. Ageing and obesity Shared patterns: from Molecular Pathogenesis to Epigenetics. Diseases. 2021;9:87.
Jaycox JR, Dai Y, Ring AM. Decoding the autoantibody reactome. Science. 2024;383:705–7.
Fonseca DLM, Filgueiras IS, Marques AHC, Vojdani E, Halpert G, Ostrinski Y, et al. Severe COVID-19 patients exhibit elevated levels of autoantibodies targeting cardiolipin and platelet glycoprotein with age: a systems biology approach. NPJ Aging. 2023;9:21.
Clement CC, Osan J, Buque A, Nanaware PP, Chang Y-C, Perino G, et al. PDIA3 epitope-driven immune autoreactivity contributes to hepatic damage in type 2 diabetes. Sci Immunol. 2022;7:eabl3795.
Frasca D, Diaz A, Romero M, Thaller S, Blomberg BB. Metabolic requirements of human pro-inflammatory B cells in aging and obesity. PLoS ONE. 2019;14:e0219545.
Crescioli S, Correa I, Ng J, Willsmore ZN, Laddach R, Chenoweth A, et al. B cell profiles, antibody repertoire and reactivity reveal dysregulated responses with autoimmune features in melanoma. Nat Commun. 2023;14:3378.
Shiwaku H, Katayama S, Gao M, Kondo K, Nakano Y, Motokawa Y, et al. Analyzing schizophrenia-related phenotypes in mice caused by autoantibodies against NRXN1α in schizophrenia. Brain Behav Immun. 2023;111:32–45.
Frasca D, Romero M, Diaz A, Blomberg BB. Obesity accelerates age defects in B cells, and weight loss improves B cell function. Immun Ageing. 2023;20:35.
Mazor RD, Nathan N, Gilboa A, Stoler-Barak L, Moss L, Solomonov I, et al. Tumor-reactive antibodies evolve from non-binding and autoreactive precursors. Cell. 2022;185:1208–e122221.
von Stemann JH, Rigas AS, Thørner LW, Rasmussen DGK, Pedersen OB, Rostgaard K, et al. Prevalence and correlation of cytokine-specific autoantibodies with epidemiological factors and C-reactive protein in 8,972 healthy individuals: results from the Danish blood Donor Study. PLoS ONE. 2017;12:e0179981.
Hagadorn KA, Peterson ME, Kole H, Scott B, Skinner J, Diouf A, et al. Autoantibodies inhibit Plasmodium Falciparum growth and are associated with protection from clinical malaria. Immunity. 2024;57:1769–e17794.
Meier HCS, Sandler DP, Simonsick EM, Weng N-P, Parks CG. Sex differences in the association between antinuclear antibody positivity with diabetes and multimorbidity in older adults: results from the Baltimore Longitudinal Study of Aging. Exp Gerontol. 2020;135:110906.
Shome M, Chung Y, Chavan R, Park JG, Qiu J, LaBaer J. Serum autoantibodyome reveals that healthy individuals share common autoantibodies. Cell Rep. 2022;39:110873.
Rubin SJS, Bloom MS, Robinson WH. B cell checkpoints in autoimmune rheumatic diseases. Nat Rev Rheumatol. 2019;15:303–15.
Ioannidou A, Goulielmaki E, Garinis GA. DNA damage: from chronic inflammation to age-related deterioration. Front Genet. 2016;7:187.
Wardemann H, Yurasov S, Schaefer A, Young JW, Meffre E, Nussenzweig MC. Predominant autoantibody production by early human B cell precursors. Science. 2003;301:1374–7.
Labrie JE, Borghesi L, Gerstein RM. Bone marrow microenvironmental changes in aged mice compromise V(D)J recombinase activity and B cell generation. Semin Immunol. 2005;17:347–55.
Rivera-Munoz P, Malivert L, Derdouch S, Azerrad C, Abramowski V, Revy P, et al. DNA repair and the immune system: from V(D)J recombination to aging lymphocytes. Eur J Immunol. 2007;37(Suppl 1):S71–82.
Liu Z, Davidson A. BAFF and selection of autoreactive B cells. Trends Immunol. 2011;32:388–94.
Alves da Costa T, Peterson JN, Lang J, Shulman J, Liang X, Freed BM, et al. Central human B cell tolerance manifests with a distinctive cell phenotype and is enforced via CXCR4 signaling in Hu-mice. Proc Natl Acad Sci U S A. 2021;118:e2021570118.
Carbone LD, Bůžková P, Fink HA, Robbins JA, Bethel M, Hamrick MW, et al. Association of Plasma SDF-1 with bone Mineral density, body composition, and hip fractures in older adults: the Cardiovascular Health Study. Calcif Tissue Int. 2017;100:599–608.
Bonasia CG, Abdulahad WH, Rutgers A, Heeringa P, Bos NA. B cell activation and escape of Tolerance checkpoints: recent insights from studying Autoreactive B cells. Cells. 2021;10:1190.
Vossenkämper A, Blair PA, Safinia N, Fraser LD, Das L, Sanders TJ, et al. A role for gut-associated lymphoid tissue in shaping the human B cell repertoire. J Exp Med. 2013;210:1665–74.
Weinstein JS, Herman EI, Lainez B, Licona-Limón P, Esplugues E, Flavell R, et al. TFH cells progressively differentiate to regulate the germinal center response. Nat Immunol. 2016;17:1197–205.
Cyster JG, Goodnow CC. Antigen-induced exclusion from follicles and anergy are separate and complementary processes that influence peripheral B cell fate. Immunity. 1995;3:691–701.
Brink R, Phan TG, Self-Reactive B. Cells in the Germinal Center reaction. Annu Rev Immunol. 2018;36:339–57.
Pritchard NR, Smith KGC. B cell inhibitory receptors and autoimmunity. Immunology. 2003;108:263–73.
Yong PFK, Salzer U, Grimbacher B. The role of costimulation in antibody deficiencies: ICOS and common variable immunodeficiency. Immunol Rev. 2009;229:101–13.
Luo W, Weisel F, Shlomchik MJ. B cell receptor and CD40 Signaling are rewired for synergistic induction of the c-Myc transcription factor in Germinal Center B cells. Immunity. 2018;48:313–e3265.
Ozaki K, Spolski R, Feng CG, Qi C-F, Cheng J, Sher A, et al. A critical role for IL-21 in regulating immunoglobulin production. Science. 2002;298:1630–4.
Ettinger R, Sims GP, Fairhurst A-M, Robbins R, da Silva YS, Spolski R, et al. IL-21 induces differentiation of human naive and memory B cells into antibody-secreting plasma cells. J Immunol. 2005;175:7867–79.
Petersone L, Walker LSK. T-cell help in the germinal center: homing in on the role of IL-21. Int Immunol. 2024;36:89–98.
Kim Y, Manara F, Grassmann S, Belcheva KT, Reyes K, Kim H et al. IL-21 shapes the B cell response in a context-dependent manner. bioRxiv. 2024;2024.07.13.600808.
Sato Y, Silina K, van den Broek M, Hirahara K, Yanagita M. The roles of tertiary lymphoid structures in chronic diseases. Nat Rev Nephrol. 2023;19:525–37.
Ligon MM, Wang C, DeJong EN, Schulz C, Bowdish DME, Mysorekar IU. Single cell and tissue-transcriptomic analysis of murine bladders reveals age- and TNFα-dependent but microbiota-independent tertiary lymphoid tissue formation. Mucosal Immunol. 2020;13:908–18.
Singh P, Coskun ZZ, Goode C, Dean A, Thompson-Snipes L, Darlington G. Lymphoid neogenesis and immune infiltration in aged liver. Hepatology. 2008;47:1680–90.
Sato Y, Oguchi A, Fukushima Y, Masuda K, Toriu N, Taniguchi K, et al. CD153/CD30 signaling promotes age-dependent tertiary lymphoid tissue expansion and kidney injury. J Clin Invest. 2022;132:e146071.
Allman D, Wilmore JR, Gaudette BT. The continuing story of T-cell independent antibodies. Immunol Rev. 2019;288:128–35.
Walker LSK. The link between circulating follicular helper T cells and autoimmunity. Nat Rev Immunol. 2022;22:567–75.
Sogkas G, Atschekzei F, Adriawan IR, Dubrowinskaja N, Witte T, Schmidt RE. Cellular and molecular mechanisms breaking immune tolerance in inborn errors of immunity. Cell Mol Immunol. 2021;18:1122–40.
Shirafkan F, Hensel L, Rattay K. Immune tolerance and the prevention of autoimmune diseases essentially depend on thymic tissue homeostasis. Front Immunol. 2024;15:1339714.
Almanan M, Raynor J, Ogunsulire I, Malyshkina A, Mukherjee S, Hummel SA, et al. IL-10-producing tfh cells accumulate with age and link inflammation with age-related immune suppression. Sci Adv. 2020;6:eabb0806.
Liu Z, Liu S, Zhang Y, Zeng W, Wang S, Ji P, et al. Distinct roles of ICOS and CD40L in human T-B cell adhesion and antibody production. Cell Immunol. 2021;368:104420.
Li K, Romero M, Cañardo M, Garcia D, Diaz A, Blomberg BB, et al. B cells from old mice induce the generation of inflammatory T cells through metabolic pathways. Mech Ageing Dev. 2023;209:111742.
Makhijani P, Basso PJ, Chan YT, Chen N, Baechle J, Khan S, et al. Regulation of the immune system by the insulin receptor in health and disease. Front Endocrinol (Lausanne). 2023;14:1128622.
Ersching J, Efeyan A, Mesin L, Jacobsen JT, Pasqual G, Grabiner BC, et al. Germinal Center Selection and Affinity Maturation require dynamic regulation of mTORC1 kinase. Immunity. 2017;46:1045–e10586.
Perl A. mTOR activation is a biomarker and a central pathway to autoimmune disorders, cancer, obesity, and aging. Ann N Y Acad Sci. 2015;1346:33–44.
Frasca D, Romero M, Diaz A, Alter-Wolf S, Ratliff M, Landin AM, et al. A molecular mechanism for TNF-α-mediated downregulation of B cell responses. J Immunol. 2012;188:279–86.
Frasca D, Diaz A, Romero M, Landin AM, Blomberg BB. High TNF-α levels in resting B cells negatively correlate with their response. Exp Gerontol. 2014;54:116–22.
Yang S, Zhao M, Jia S, Macrophage. Key player in the pathogenesis of autoimmune diseases. Front Immunol. 2023;14:1080310.
Li H, Wu Q, Li J, Yang P, Zhu Z, Luo B, et al. Cutting Edge: defective follicular exclusion of apoptotic antigens due to marginal zone macrophage defects in autoimmune BXD2 mice. J Immunol. 2013;190:4465–9.
Moss CE, Johnston SA, Kimble JV, Clements M, Codd V, Hamby S, et al. Aging-related defects in macrophage function are driven by MYC and USF1 transcriptional programs. Cell Rep. 2024;43:114073.
Hu H, Cheng X, Li F, Guan Z, Xu J, Wu D, et al. Defective efferocytosis by aged macrophages promotes STING signaling mediated inflammatory liver injury. Cell Death Discov. 2023;9:236.
Soler Palacios B, Estrada-Capetillo L, Izquierdo E, Criado G, Nieto C, Municio C, et al. Macrophages from the synovium of active rheumatoid arthritis exhibit an activin A-dependent pro-inflammatory profile. J Pathol. 2015;235:515–26.
Tsigalou C, Vallianou N, Dalamaga M. Autoantibody production in obesity: is there evidence for a link between obesity and autoimmunity? Curr Obes Rep. 2020;9:245–54.
Frasca D, Reidy L, Romero M, Diaz A, Cray C, Kahl K, et al. The majority of SARS-CoV-2-specific antibodies in COVID-19 patients with obesity are autoimmune and not neutralizing. Int J Obes (Lond). 2022;46:427–32.
Chauvin C, Retnakumar SV, Bayry J. Obesity negatively impacts maintenance of antibody response to COVID-19 vaccines. Cell Rep Med. 2023;4:101117.
Tedeschi SK, Cui J, Arkema EV, Robinson WH, Sokolove J, Lingampalli N, et al. Elevated BMI and antibodies to citrullinated proteins interact to increase rheumatoid arthritis risk and shorten time to diagnosis: a nested case-control study of women in the nurses’ Health studies. Semin Arthritis Rheum. 2017;46:692–8.
Li L, Feng D, Zeng J, Ye P, Chen Y, Wei D. Association between rheumatoid factor and metabolic syndrome in general population. Diabetol Metab Syndr. 2022;14:165.
Frasca D, Diaz A, Romero M, Garcia D, Jayram D, Thaller S, et al. Identification and characterization of adipose tissue-derived human antibodies with anti-self specificity. Front Immunol. 2020;11:392.
Hägglöf T, Vanz C, Kumagai A, Dudley E, Ortega V, Siller M, et al. T-bet + B cells accumulate in adipose tissue and exacerbate metabolic disorder during obesity. Cell Metab. 2022;34:1121–e11366.
Kirichenko TV, Markina YV, Bogatyreva AI, Tolstik TV, Varaeva YR, Starodubova AV. The role of Adipokines in inflammatory mechanisms of obesity. Int J Mol Sci. 2022;23:14982.
Revelo XS, Ghazarian M, Chng MHY, Luck H, Kim JH, Zeng K, et al. Nucleic acid-targeting pathways promote inflammation in obesity-related insulin resistance. Cell Rep. 2016;16:717–30.
Suurmond J, Diamond B. Autoantibodies in systemic autoimmune diseases: specificity and pathogenicity. J Clin Invest. 2015;125:2194–202.
Wen L, Zhang B, Wu X, Liu R, Fan H, Han L, et al. Toll-like receptors 7 and 9 regulate the proliferation and differentiation of B cells in systemic lupus erythematosus. Front Immunol. 2023;14:1093208.
Desnues B, Macedo AB, Roussel-Queval A, Bonnardel J, Henri S, Demaria O, et al. TLR8 on dendritic cells and TLR9 on B cells restrain TLR7-mediated spontaneous autoimmunity in C57BL/6 mice. Proc Natl Acad Sci U S A. 2014;111:1497–502.
Hanna Kazazian N, Wang Y, Roussel-Queval A, Marcadet L, Chasson L, Laprie C, et al. Lupus Autoimmunity and metabolic parameters are exacerbated upon high Fat Diet-Induced obesity due to TLR7 Signaling. Front Immunol. 2019;10:2015.
Kakalij RM, Dsouza DL, Ha L, Boesen EI. TLR7 activation by imiquimod worsens glycemic control in female FVB/N mice consuming a high-fat diet. Physiol Rep. 2024;12:e15949.
Cas MD, Roda G, Li F, Secundo F. Functional lipids in Autoimmune Inflammatory diseases. Int J Mol Sci. 2020;21:3074.
Matarese G. The link between obesity and autoimmunity. Science. 2023;379:1298–300.
Pal A, Lin C-T, Boykov I, Benson E, Kidd G, Fisher-Wellman KH, et al. High Fat Diet-Induced obesity dysregulates splenic B cell mitochondrial activity. Nutrients. 2023;15:4807.
Vivas-García Y, Efeyan A. The metabolic plasticity of B cells. Front Mol Biosci. 2022;9:991188.
Frasca D, Romero M, Diaz A, Garcia D, Thaller S, Blomberg BB. B cells with a senescent-Associated Secretory phenotype Accumulate in the adipose tissue of individuals with obesity. Int J Mol Sci. 2021;22:1839.
Pająk B, Zieliński R, Priebe W. The impact of Glycolysis and its inhibitors on the Immune response to inflammation and autoimmunity. Molecules. 2024;29:1298.
Li J, Zhao M, Luo W, Huang J, Zhao B, Zhou Z. B cell metabolism in autoimmune diseases: signaling pathways and interventions. Front Immunol. 2023;14:1232820.
Shaikh SR, Beck MA, Alwarawrah Y, MacIver NJ. Emerging mechanisms of obesity-associated immune dysfunction. Nat Rev Endocrinol. 2024;20:136–48.
Kim D-H, Do M-S. BAFF knockout improves systemic inflammation via regulating adipose tissue distribution in high-fat diet-induced obesity. Exp Mol Med. 2015;47:e129.
Tada F, Abe M, Kawasaki K, Miyake T, Shiyi C, Hiasa Y, et al. B cell activating factor in obesity is regulated by oxidative stress in adipocytes. J Clin Biochem Nutr. 2013;52:120–7.
Sánchez DCV, Castellanos SG, Sandoval MEV, García AG. B-Cell activating factor increases related to adiposity, insulin resistance, and endothelial dysfunction in overweight and obese subjects. Life (Basel). 2022;12:634.
Caro-Maldonado A, Wang R, Nichols AG, Kuraoka M, Milasta S, Sun LD, et al. Metabolic reprogramming is required for antibody production that is suppressed in anergic but exaggerated in chronically BAFF-exposed B cells. J Immunol. 2014;192:3626–36.
Thien M, Phan TG, Gardam S, Amesbury M, Basten A, Mackay F, et al. Excess BAFF rescues self-reactive B cells from peripheral deletion and allows them to enter forbidden follicular and marginal zone niches. Immunity. 2004;20:785–98.
Beyze A, Larroque C, Le Quintrec M. The role of antibody glycosylation in autoimmune and alloimmune kidney diseases. Nat Rev Nephrol. 2024;20:672–89.
Deriš H, Tominac P, Vučković F, Briški N, Astrup A, Blaak EE, et al. Effects of low-calorie and different weight-maintenance diets on IgG glycome composition. Front Immunol. 2022;13:995186.
Liu D, Li Q, Dong J, Li D, Xu X, Xing W, et al. The Association between normal BMI with Central Adiposity and Proinflammatory potential immunoglobulin G N-Glycosylation. Diabetes Metab Syndr Obes. 2019;12:2373–85.
Tanigaki K, Sacharidou A, Peng J, Chambliss KL, Yuhanna IS, Ghosh D, et al. Hyposialylated IgG activates endothelial IgG receptor FcγRIIB to promote obesity-induced insulin resistance. J Clin Invest. 2018;128:309–22.
Greto VL, Cvetko A, Štambuk T, Dempster NJ, Kifer D, Deriš H, et al. Extensive weight loss reduces glycan age by altering IgG N-glycosylation. Int J Obes (Lond). 2021;45:1521–31.
Arai S, Maehara N, Iwamura Y, Honda S, Nakashima K, Kai T, et al. Obesity-associated autoantibody production requires AIM to retain the immunoglobulin M immune complex on follicular dendritic cells. Cell Rep. 2013;3:1187–98.
Pham TD, Chng MHY, Roskin KM, Jackson KJL, Nguyen KD, Glanville J, et al. High-fat diet induces systemic B-cell repertoire changes associated with insulin resistance. Mucosal Immunol. 2017;10:1468–79.
Weyand CM, Goronzy JJ. Aging of the Immune System. Mechanisms and therapeutic targets. Ann Am Thorac Soc. 2016;13(Suppl 5):S422–8.
Zheng Y, Liu Q, Goronzy JJ, Weyand CM. Immune aging - a mechanism in autoimmune disease. Semin Immunol. 2023;69:101814.
Wang Y, Dong C, Han Y, Gu Z, Sun C. Immunosenescence, aging and successful aging. Front Immunol. 2022;13:942796.
Accardi G, Caruso C. Immune-inflammatory responses in the elderly: an update. Immun Ageing. 2018;15:11.
Frasca D, Diaz A, Romero M, Blomberg BB. Metformin enhances B cell function and antibody responses of Elderly individuals with Type-2 diabetes Mellitus. Front Aging. 2021;2:715981.
Zinecker H, Simon AK. Autophagy takes it all - autophagy inducers target immune aging. Dis Model Mech. 2022;15:dmm049345.
Iborra-Pernichi M, Ruiz García J, Velasco de la Esperanza M, Estrada BS, Bovolenta ER, Cifuentes C, et al. Defective mitochondria remodelling in B cells leads to an aged immune response. Nat Commun. 2024;15:2569.
Lanahan SM, Yang L, Jones KM, Qi Z, Cabrera EC, Cominsky LY, et al. PI3Kγ in B cells promotes antibody responses and generation of antibody-secreting cells. Nat Immunol. 2024;25:1422–31.
Khan S, Chakraborty M, Wu F, Chen N, Wang T, Chan YT et al. B cells promote T cell immunosenescence and mammalian aging parameters. bioRxiv. 2023;2023.09.12.556363.
Arvey A, Rowe M, Legutki JB, An G, Gollapudi A, Lei A, et al. Age-associated changes in the circulating human antibody repertoire are upregulated in autoimmunity. Immun Ageing. 2020;17:28.
Kumar SJ, Shukla S, Kumar S, Mishra P. Immunosenescence and Inflamm-Aging: clinical interventions and the potential for reversal of aging. Cureus. 2024;16:e53297.
Frasca D, Romero M, Garcia D, Diaz A, Blomberg BB. Hyper-metabolic B cells in the spleens of old mice make antibodies with autoimmune specificities. Immun Ageing. 2021;18:9.
Ma S, Ji Z, Zhang B, Geng L, Cai Y, Nie C et al. Spatial transcriptomic landscape unveils immunoglobin-associated senescence as a hallmark of aging. Cell. 2024;S0092-8674(24)01201-7.
Gibson KL, Wu Y-C, Barnett Y, Duggan O, Vaughan R, Kondeatis E, et al. B-cell diversity decreases in old age and is correlated with poor health status. Aging Cell. 2009;8:18–25.
Weksler ME. Changes in the B-cell repertoire with age. Vaccine. 2000;18:1624–8.
Rodriguez-Zhurbenko N, Quach TD, Hopkins TJ, Rothstein TL, Hernandez AM. Human B-1 cells and B-1 cell antibodies change with advancing age. Front Immunol. 2019;10:483.
Van der Put E, Frasca D, King AM, Blomberg BB, Riley RL. Decreased E47 in senescent B cell precursors is stage specific and regulated posttranslationally by protein turnover. J Immunol. 2004;173:818–27.
Frasca D, Landin AM, Lechner SC, Ryan JG, Schwartz R, Riley RL, et al. Aging down-regulates the transcription factor E2A, activation-induced cytidine deaminase, and ig class switch in human B cells. J Immunol. 2008;180:5283–90.
Collier DA, Ferreira IATM, Kotagiri P, Datir RP, Lim EY, Touizer E, et al. Age-related immune response heterogeneity to SARS-CoV-2 vaccine BNT162b2. Nature. 2021;596:417–22.
Bastard P, Gervais A, Le Voyer T, Rosain J, Philippot Q, Manry J, et al. Autoantibodies neutralizing type I IFNs are present in ~ 4% of uninfected individuals over 70 years old and account for ~ 20% of COVID-19 deaths. Sci Immunol. 2021;6:eabl4340.
Johnson SA, Rozzo SJ, Cambier JC. Aging-dependent exclusion of antigen-inexperienced cells from the peripheral B cell repertoire. J Immunol. 2002;168:5014–23.
Ahmed R, Omidian Z, Donner T, Hamad ARA. Hiding in plain sight: time to unlock autoimmune clues in human CD5 + B cells by using nextgen technology. Discov Med. 2018;26:79–83.
Anaya J-M, Shoenfeld Y, Rojas-Villarraga A, Levy RA, Cervera R, editors. Autoimmunity: From Bench to Bedside [Internet]. Bogota (Colombia): El Rosario University Press; 2013 [cited 2024 Sep 28]. http://www.ncbi.nlm.nih.gov/books/NBK459447/
Ruan P, Wang S, Yang M, Wu H. The ABC-associated Immunosenescence and Lifestyle interventions in Autoimmune Disease. Rheumatol Immunol Res. 2022;3:128–35.
Colonna-Romano G, Bulati M, Aquino A, Pellicanò M, Vitello S, Lio D, et al. A double-negative (IgD-CD27-) B cell population is increased in the peripheral blood of elderly people. Mech Ageing Dev. 2009;130:681–90.
Hao Y, O’Neill P, Naradikian MS, Scholz JL, Cancro MP. A B-cell subset uniquely responsive to innate stimuli accumulates in aged mice. Blood. 2011;118:1294–304.
Mouat IC, Goldberg E, Horwitz MS. Age-associated B cells in autoimmune diseases. Cell Mol Life Sci. 2022;79:402.
Nickerson KM, Smita S, Hoehn KB, Marinov AD, Thomas KB, Kos JT, et al. Age-associated B cells are heterogeneous and dynamic drivers of autoimmunity in mice. J Exp Med. 2023;220:e20221346.
Rubtsov AV, Rubtsova K, Kappler JW, Marrack P. TLR7 drives accumulation of ABCs and autoantibody production in autoimmune-prone mice. Immunol Res. 2013;55:210–6.
Dai D, Gu S, Han X, Ding H, Jiang Y, Zhang X, et al. The transcription factor ZEB2 drives the formation of age-associated B cells. Science. 2024;383:413–21.
Frasca D, Diaz A, Romero M, Garcia D, Blomberg BB. B Cell Immunosenescence. Annu Rev Cell Dev Biol. 2020;36:551–74.
Vidal-Pedrola G, Naamane N, Cameron JA, Pratt AG, Mellor AL, Isaacs JD, et al. Characterization of age-associated B cells in early drug-naïve rheumatoid arthritis patients. Immunology. 2023;168:640–53.
Wang S, Wang J, Kumar V, Karnell JL, Naiman B, Gross PS, et al. IL-21 drives expansion and plasma cell differentiation of autoreactive CD11chiT-bet + B cells in SLE. Nat Commun. 2018;9:1758.
Riley RL, Khomtchouk K, Blomberg BB. Age-associated B cells (ABC) inhibit B lymphopoiesis and alter antibody repertoires in old age. Cell Immunol. 2017;321:61–7.
Bagavant H, Durslewicz J, Pyclik M, Makuch M, Papinska JA, Deshmukh US. Age-associated B cell infiltration in salivary glands represents a hallmark of Sjögren’s-like disease in aging mice. Geroscience. 2024.
Nagele EP, Han M, Acharya NK, DeMarshall C, Kosciuk MC, Nagele RG. Natural IgG autoantibodies are abundant and ubiquitous in human sera, and their number is influenced by age, gender, and disease. PLoS ONE. 2013;8:e60726.
Yin J, Ibrahim S, Petersen F, Yu X. Autoimmunomic signatures of aging and age-related neurodegenerative diseases are Associated with brain function and ribosomal proteins. Front Aging Neurosci. 2021;13:679688.
Fernbach S, Mair NK, Abela IA, Groen K, Kuratli R, Lork M et al. Longitudinal analysis over decades reveals the Development and Immune implications of Type I Interferon autoantibodies in an Aging Population. medRxiv. 2024;2024.02.27.24303363.
Shen X, Wang C, Zhou X, Zhou W, Hornburg D, Wu S et al. Nonlinear dynamics of multi-omics profiles during human aging. Nat Aging. 2024.
Lehallier B, Gate D, Schaum N, Nanasi T, Lee SE, Yousef H, et al. Undulating changes in human plasma proteome profiles across the lifespan. Nat Med. 2019;25:1843–50.
Nie C, Li Y, Li R, Yan Y, Zhang D, Li T, et al. Distinct biological ages of organs and systems identified from a multi-omics study. Cell Rep. 2022;38:110459.
Olecka M, van Bömmel A, Best L, Haase M, Foerste S, Riege K, et al. Nonlinear DNA methylation trajectories in aging male mice. Nat Commun. 2024;15:3074.
Xia Y, Kellems RE. Receptor-activating autoantibodies and disease: preeclampsia and beyond. Expert Rev Clin Immunol. 2011;7:659–74.
Wu A-L, Kolumam G, Stawicki S, Chen Y, Li J, Zavala-Solorio J, et al. Amelioration of type 2 diabetes by antibody-mediated activation of fibroblast growth factor receptor 1. Sci Transl Med. 2011;3:113ra126.
Bauer ME. Accelerated immunosenescence in rheumatoid arthritis: impact on clinical progression. Immun Ageing. 2020;17:6.
van de Stadt LA, de Koning MHMT, van de Stadt RJ, Wolbink G, Dijkmans BAC, Hamann D, et al. Development of the anti-citrullinated protein antibody repertoire prior to the onset of rheumatoid arthritis. Arthritis Rheum. 2011;63:3226–33.
Wu C-Y, Yang H-Y, Lai J-H. Anti-citrullinated Protein Antibodies in patients with rheumatoid arthritis: Biological effects and mechanisms of Immunopathogenesis. Int J Mol Sci. 2020;21:4015.
Xiang Y, Zhang M, Jiang D, Su Q, Shi J. The role of inflammation in autoimmune disease: a therapeutic target. Front Immunol. 2023;14:1267091.
Malmström V, Catrina AI, Klareskog L. The immunopathogenesis of seropositive rheumatoid arthritis: from triggering to targeting. Nat Rev Immunol. 2017;17:60–75.
Visvabharathy L, Zhu C, Orban ZS, Yarnoff K, Palacio N, Jimenez M et al. Autoantibody production is enhanced after mild SARS-CoV-2 infection despite vaccination in individuals with and without long COVID. medRxiv. 2023;2023.04.07.23288243.
Acosta-Ampudia Y, Monsalve DM, Rojas M, Rodríguez Y, Zapata E, Ramírez-Santana C, et al. Persistent autoimmune activation and Proinflammatory State in Post-coronavirus Disease 2019 Syndrome. J Infect Dis. 2022;225:2155–62.
Cervia-Hasler C, Brüningk SC, Hoch T, Fan B, Muzio G, Thompson RC, et al. Persistent complement dysregulation with signs of thromboinflammation in active long covid. Science. 2024;383:eadg7942.
Yin K, Peluso MJ, Luo X, Thomas R, Shin M-G, Neidleman J, et al. Long COVID manifests with T cell dysregulation, inflammation and an uncoordinated adaptive immune response to SARS-CoV-2. Nat Immunol. 2024;25:218–25.
Giannoula Y, Kroemer G, Pietrocola F. Cellular senescence and the host immune system in aging and age-related disorders. Biomed J. 2023;46:100581.
Tanaka T, Biancotto A, Moaddel R, Moore AZ, Gonzalez-Freire M, Aon MA, et al. Plasma proteomic signature of age in healthy humans. Aging Cell. 2018;17:e12799.
Cao W, Zheng D, Wang G, Zhang J, Ge S, Singh M, et al. Modelling biological age based on plasma peptides in Han Chinese adults. Aging. 2020;12:10676–86.
Li Z, Wu H, Luo Y, Tan X. Correlation of serum complement factor 5a level with inflammatory response and cognitive function in patients with Alzheimer’s disease of different severity. BMC Neurol. 2023;23:319.
Pappworth IY, Kulik L, Haluszczak C, Reuter JW, Holers VM, Marchbank KJ. Increased B cell deletion and significantly reduced auto-antibody titre due to premature expression of human complement receptor 2 (CR2, CD21). Mol Immunol. 2009;46:1042–9.
Zheng R, Zhang Y, Zhang K, Yuan Y, Jia S, Liu J. The complement system, aging, and aging-related diseases. Int J Mol Sci. 2022;23:8689.
Phan TG, Grigorova I, Okada T, Cyster JG. Subcapsular encounter and complement-dependent transport of immune complexes by lymph node B cells. Nat Immunol. 2007;8:992–1000.
McCloskey ML, Curotto de Lafaille MA, Carroll MC, Erlebacher A. Acquisition and presentation of follicular dendritic cell-bound antigen by lymph node-resident dendritic cells. J Exp Med. 2011;208:135–48.
Hearps AC, Martin GE, Angelovich TA, Cheng W-J, Maisa A, Landay AL, et al. Aging is associated with chronic innate immune activation and dysregulation of monocyte phenotype and function. Aging Cell. 2012;11:867–75.
Lu LL, Suscovich TJ, Fortune SM, Alter G. Beyond binding: antibody effector functions in infectious diseases. Nat Rev Immunol. 2018;18:46–61.
Yang X, Zhang X, Zhao X, Yuan M, Zhang K, Dai J, et al. Antibody-dependent enhancement: ″Evil″ antibodies favorable for viral infections. Viruses. 2022;14:1739.
Sawant J, Patil A, Kurle S. A review: understanding molecular mechanisms of antibody-dependent enhancement in viral infections. Vaccines (Basel). 2023;11:1240.
Bournazos S, Gupta A, Ravetch JV. The role of IgG fc receptors in antibody-dependent enhancement. Nat Rev Immunol. 2020;20:633–43.
Chalayer E, Gramont B, Zekre F, Goguyer-Deschaumes R, Waeckel L, Grange L, et al. Fc receptors gone wrong: a comprehensive review of their roles in autoimmune and inflammatory diseases. Autoimmun Rev. 2022;21:103016.
Yu X, Lazarus AH. Targeting FcγRs to treat antibody-dependent autoimmunity. Autoimmun Rev. 2016;15:510–2.
Reefman E, Horst G, Nijk MT, Limburg PC, Kallenberg CGM, Bijl M. Opsonization of late apoptotic cells by systemic lupus erythematosus autoantibodies inhibits their uptake via an fcgamma receptor-dependent mechanism. Arthritis Rheum. 2007;56:3399–411.
Fukue R, Okazaki Y, Gono T, Kuwana M. Abatacept downregulates Fcγ receptor I on circulating monocytes: a potential therapeutic mechanism in patients with rheumatoid arthritis. Arthritis Res Ther. 2022;24:194.
van Beek L, Vroegrijk IOCM, Katiraei S, Heemskerk MM, van Dam AD, Kooijman S, et al. FcRγ-chain deficiency reduces the development of diet-induced obesity. Obes (Silver Spring). 2015;23:2435–44.
Yu L, Wan Q, Liu Q, Fan Y, Zhou Q, Skowronski AA, et al. IgG is an aging factor that drives adipose tissue fibrosis and metabolic decline. Cell Metab. 2024;36:793–e8075.
Pellerin K, Rubino SJ, Burns JC, Smith BA, McCarl C-A, Zhu J, et al. MOG autoantibodies trigger a tightly-controlled FcR and BTK-driven microglia proliferative response. Brain. 2021;144:2361–74.
King AM, Keating P, Prabhu A, Blomberg BB, Riley RL. NK cells in the CD19- B220 + bone marrow fraction are increased in senescence and reduce E2A and surrogate light chain proteins in B cell precursors. Mech Ageing Dev. 2009;130:384–92.
Ming B, Wu T, Cai S, Hu P, Tang J, Zheng F, et al. The increased ratio of blood CD56bright NK to CD56dim NK is a distinguishing feature of primary Sjögren’s syndrome. J Immunol Res. 2020;2020:7523914.
Chalan P, Bijzet J, Kroesen B-J, Boots AMH, Brouwer E. Altered natural killer cell subsets in Seropositive Arthralgia and Early Rheumatoid Arthritis are Associated with Autoantibody Status. J Rheumatol. 2016;43:1008–16.
Liu M, Liang S, Zhang C. NK Cells in Autoimmune diseases: protective or pathogenic? Front Immunol. 2021;12:624687.
Lastwika KJ, Lampe PD. Breaking tolerance: autoantibodies can target protein posttranslational modifications. Curr Opin Biotechnol. 2024;85:103056.
van Eijkeren RJ, Krabbe O, Boes M, Schipper HS, Kalkhoven E. Endogenous lipid antigens for invariant natural killer T cells hold the reins in adipose tissue homeostasis. Immunology. 2018;153:179–89.
Corona-Sanchez EG, Martínez-García EA, Lujano-Benítez AV, Pizano-Martinez O, Guerra-Durán IA, Chavarria-Avila E, et al. Autoantibodies in the pathogenesis of idiopathic inflammatory myopathies: does the endoplasmic reticulum stress response have a role? Front Immunol. 2022;13:940122.
Pang Z, Kushiyama A, Sun J, Kikuchi T, Yamazaki H, Iwamoto Y, et al. Glial fibrillary acidic protein (GFAP) is a novel biomarker for the prediction of autoimmune diabetes. FASEB J. 2017;31:4053–63.
Mancardi GL, Cadoni A, Tabaton M, Schenone A, Zicca A, De Martini I, et al. Schwann cell GFAP expression increases in axonal neuropathies. J Neurol Sci. 1991;102:177–83.
Yang Z, Wang KKW. Glial fibrillary acidic protein: from intermediate filament assembly and gliosis to neurobiomarker. Trends Neurosci. 2015;38:364–74.
Buckman LB, Thompson MM, Moreno HN, Ellacott KLJ. Regional astrogliosis in the mouse hypothalamus in response to obesity. J Comp Neurol. 2013;521:1322–33.
Yi C-X, Tschöp MH, Woods SC, Hofmann SM. High-fat-diet exposure induces IgG accumulation in hypothalamic microglia. Dis Model Mech. 2012;5:686–90.
Bhat S, Jagadeeshaprasad MG, Patil YR, Shaikh ML, Regin BS, Mohan V, et al. Proteomic insight reveals elevated levels of Albumin in circulating Immune complexes in Diabetic plasma. Mol Cell Proteom. 2016;15:2011–20.
Korça E, Piskovatska V, Börgermann J, Navarrete Santos A, Simm A. Circulating antibodies against age-modified proteins in patients with coronary atherosclerosis. Sci Rep. 2020;10:17105.
Dowds CM, Kornell S-C, Blumberg RS, Zeissig S. Lipid antigens in immunity. Biol Chem. 2014;395:61–81.
Wong-Baeza C, Reséndiz-Mora A, Donis-Maturano L, Wong-Baeza I, Zárate-Neira L, Yam-Puc JC, et al. Anti-lipid IgG antibodies are produced via Germinal centers in a murine model resembling human lupus. Front Immunol. 2016;7:396.
Zhang X, Meng J, Shi X, Quinet RJ, Davis W, Zakem J, et al. Lupus pathogenesis and autoimmunity are exacerbated by high fat diet-induced obesity in MRL/lpr mice. Lupus Sci Med. 2023;10:e000898.
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186:243–78.
Moqri M, Herzog C, Poganik JR, Biomarkers of Aging Consortium, Justice J, Belsky DW, et al. Biomarkers of aging for the identification and evaluation of longevity interventions. Cell. 2023;186:3758–75.
Guo L, Ren H, Fan S, Chao X, Liu M, Guan H, et al. Autoantibodies against eukaryotic translation elongation factor 1 delta in two patients with autoimmune cerebellar ataxia. Front Immunol. 2023;14:1289175.
Moadab F, Wang X, Najjar R, Ukadike KC, Hu S, Hulett T, et al. Argonaute, Vault, and ribosomal proteins targeted by autoantibodies in systemic Lupus Erythematosus. J Rheumatol. 2023;50:1136–44.
Stein KC, Morales-Polanco F, van der Lienden J, Rainbolt TK, Frydman J. Ageing exacerbates ribosome pausing to disrupt cotranslational proteostasis. Nature. 2022;601:637–42.
Turi Z, Lacey M, Mistrik M, Moudry P. Impaired ribosome biogenesis: mechanisms and relevance to cancer and aging. Aging. 2019;11:2512–40.
Lyu J, Zhang H, Wang C, Pan M. New insight in treating autoimmune diseases by targeting autophagy. Autoimmunity. 2024;57:2351872.
Iannaccone A, Giorgianni F, New DD, Hollingsworth TJ, Umfress A, Alhatem AH, et al. Circulating autoantibodies in Age-Related Macular Degeneration Recognize Human Macular Tissue Antigens Implicated in Autophagy, Immunomodulation, and Protection from oxidative stress and apoptosis. PLoS ONE. 2015;10:e0145323.
Korb CA, Beck S, Wolters D, Lorenz K, Pfeiffer N, Grus FH. Serum autoantibodies in patients with dry and wet age-related Macular Degeneration. J Clin Med. 2023;12:1590.
Rai P, Janardhan KS, Meacham J, Madenspacher JH, Lin W-C, Karmaus PWF, et al. IRGM1 links mitochondrial quality control to autoimmunity. Nat Immunol. 2021;22:312–21.
Selman M, Pardo A, Fibroageing. An ageing pathological feature driven by dysregulated extracellular matrix-cell mechanobiology. Ageing Res Rev. 2021;70:101393.
Schafer MJ, White TA, Iijima K, Haak AJ, Ligresti G, Atkinson EJ, et al. Cellular senescence mediates fibrotic pulmonary disease. Nat Commun. 2017;8:14532.
Adegunsoye A, Newton CA, Oldham JM, Ley B, Lee CT, Linderholm AL, et al. Telomere length associates with chronological age and mortality across racially diverse pulmonary fibrosis cohorts. Nat Commun. 2023;14:1489.
Alder JK, Armanios M. Telomere-mediated lung disease. Physiol Rev. 2022;102:1703–20.
Shen C-Y, Lu C-H, Wu C-H, Li K-J, Kuo Y-M, Hsieh S-C, et al. Molecular basis of Accelerated Aging with Immune dysfunction-mediated inflammation (Inflamm-Aging) in patients with systemic sclerosis. Cells. 2021;10:3402.
Adler BL, Boin F, Wolters PJ, Bingham CO, Shah AA, Greider C, et al. Autoantibodies targeting telomere-associated proteins in systemic sclerosis. Ann Rheum Dis. 2021;80:912–9.
Vulsteke J-B, Smith V, Bonroy C, Derua R, Blockmans D, De Haes P, et al. Identification of new telomere- and telomerase-associated autoantigens in systemic sclerosis. J Autoimmun. 2023;135:102988.
Derevyanko A, Whittemore K, Schneider RP, Jiménez V, Bosch F, Blasco MA. Gene therapy with the TRF1 telomere gene rescues decreased TRF1 levels with aging and prolongs mouse health span. Aging Cell. 2017;16:1353–68.
Hohensinner PJ, Kaun C, Buchberger E, Ebenbauer B, Demyanets S, Huk I, et al. Age intrinsic loss of telomere protection via TRF1 reduction in endothelial cells. Biochim Biophys Acta. 2016;1863:360–7.
Toivola DM, Habtezion A, Misiorek JO, Zhang L, Nyström JH, Sharpe O, et al. Absence of keratin 8 or 18 promotes antimitochondrial autoantibody formation in aging male mice. FASEB J. 2015;29:5081–9.
Du H, Bartleson JM, Butenko S, Alonso V, Liu WF, Winer DA, et al. Tuning immunity through tissue mechanotransduction. Nat Rev Immunol. 2023;23:174–88.
Frontera WR, Ochala J. Skeletal muscle: a brief review of structure and function. Calcif Tissue Int. 2015;96:183–95.
Heo J-W, Yoo S-Z, No M-H, Park D-H, Kang J-H, Kim T-W, et al. Exercise Training attenuates obesity-Induced skeletal muscle remodeling and mitochondria-mediated apoptosis in the skeletal muscle. Int J Environ Res Public Health. 2018;15:2301.
Morgan PT, Smeuninx B, Breen L. Exploring the impact of obesity on skeletal muscle function in older age. Front Nutr. 2020;7:569904.
Espino-Gonzalez E, Dalbram E, Mounier R, Gondin J, Farup J, Jessen N, et al. Impaired skeletal muscle regeneration in diabetes: from cellular and molecular mechanisms to novel treatments. Cell Metab. 2024;36:1204–36.
Kedlian VR, Wang Y, Liu T, Chen X, Bolt L, Tudor C, et al. Human skeletal muscle aging atlas. Nat Aging. 2024;4:727–44.
Perez K, Ciotlos S, McGirr J, Limbad C, Doi R, Nederveen JP, et al. Single nuclei profiling identifies cell specific markers of skeletal muscle aging, frailty, and senescence. Aging. 2022;14:9393–422.
Dungan CM, Figueiredo VC, Wen Y, VonLehmden GL, Zdunek CJ, Thomas NT, et al. Senolytic treatment rescues blunted muscle hypertrophy in old mice. Geroscience. 2022;44:1925–40.
Zhang T, Feng X, Dong J, Xu Z, Feng B, Haas KM, et al. Cardiac troponin T and autoimmunity in skeletal muscle aging. Geroscience. 2022;44:2025–45.
Wada K, Ueno S, Hazama T, Ogasahara S, Kang J, Takahashi M, et al. Radioimmunoassay for antibodies to human skeletal muscle myosin in serum from patients with polymyositis. Clin Exp Immunol. 1983;52:297–304.
Nemoto H, Bhopale MK, Constantinescu CS, Schotland D, Rostami A. Skeletal muscle myosin is the autoantigen for experimental autoimmune myositis. Exp Mol Pathol. 2003;74:238–43.
Casciola-Rosen L, Nagaraju K, Plotz P, Wang K, Levine S, Gabrielson E, et al. Enhanced autoantigen expression in regenerating muscle cells in idiopathic inflammatory myopathy. J Exp Med. 2005;201:591–601.
Zhang T. Autoimmunity as a novel mechanism underlying Sarcopenia. Aging. 2023;15:3221–2.
North BJ, Sinclair DA. The intersection between aging and cardiovascular disease. Circ Res. 2012;110:1097–108.
Şahin B, İlgün G. Risk factors of deaths related to cardiovascular diseases in World Health Organization (WHO) member countries. Health Soc Care Community. 2022;30:73–80.
Sharma H, Mossman K, Austin RC. Fatal attractions that trigger inflammation and drive atherosclerotic disease. Eur J Clin Invest. 2024;54:e14169.
Childs BG, Baker DJ, Wijshake T, Conover CA, Campisi J, van Deursen JM. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science. 2016;354:472–7.
Crane ED, Al-Hashimi AA, Chen J, Lynn EG, Won KD, Lhoták Š, et al. Anti-GRP78 autoantibodies induce endothelial cell activation and accelerate the development of atherosclerotic lesions. JCI Insight. 2018;3:e99363.
Foteinos G, Afzal AR, Mandal K, Jahangiri M, Xu Q. Anti-heat shock protein 60 autoantibodies induce atherosclerosis in apolipoprotein E-deficient mice via endothelial damage. Circulation. 2005;112:1206–13.
Lorenzo C, Delgado P, Busse CE, Sanz-Bravo A, Martos-Folgado I, Bonzon-Kulichenko E, et al. ALDH4A1 is an atherosclerosis auto-antigen targeted by protective antibodies. Nature. 2021;589:287–92.
Harrison J, Newland SA, Jiang W, Giakomidi D, Zhao X, Clement M, et al. Marginal zone B cells produce natural atheroprotective IgM antibodies in a T cell-dependent manner. Cardiovasc Res. 2024;120:318–28.
Khan A, Roy P, Ley K. Breaking tolerance: the autoimmune aspect of atherosclerosis. Nat Rev Immunol. 2024;24:670–9.
Sakers A, De Siqueira MK, Seale P, Villanueva CJ. Adipose-tissue plasticity in health and disease. Cell. 2022;185:419–46.
Scheja L, Heeren J. The endocrine function of adipose tissues in health and cardiometabolic disease. Nat Rev Endocrinol. 2019;15:507–24.
Nguyen TT, Corvera S. Adipose tissue as a linchpin of organismal ageing. Nat Metab. 2024;6:793–807.
Man K, Kallies A, Vasanthakumar A. Resident and migratory adipose immune cells control systemic metabolism and thermogenesis. Cell Mol Immunol. 2022;19:421–31.
Xu M, Tchkonia T, Ding H, Ogrodnik M, Lubbers ER, Pirtskhalava T, et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc Natl Acad Sci U S A. 2015;112:E6301–6310.
Frasca D, Diaz A, Romero M, Thaller S, Blomberg BB. Secretion of autoimmune antibodies in the human subcutaneous adipose tissue. PLoS ONE. 2018;13:e0197472.
Corvillo F, Aparicio V, López-Lera A, Garrido S, Araújo-Vilar D, de Miguel MP, et al. Autoantibodies against Perilipin 1 as a cause of Acquired Generalized Lipodystrophy. Front Immunol. 2018;9:2142.
Arias-de la Rosa I, Escudero-Contreras A, Ruiz-Ponce M, Cuesta-López L, Román-Rodríguez C, Pérez-Sánchez C, et al. Pathogenic mechanisms involving the interplay between adipose tissue and auto-antibodies in rheumatoid arthritis. iScience. 2022;25:104893.
Barrow F, Khan S, Fredrickson G, Wang H, Dietsche K, Parthiban P, et al. Microbiota-Driven activation of Intrahepatic B cells aggravates NASH through Innate and Adaptive Signaling. Hepatology. 2021;74:704–22.
Ghazarian M, Revelo XS, Nøhr MK, Luck H, Zeng K, Lei H, et al. Type I Interferon responses drive intrahepatic T cells to promote metabolic syndrome. Sci Immunol. 2017;2:eaai7616.
Huby T, Gautier EL. Immune cell-mediated features of non-alcoholic steatohepatitis. Nat Rev Immunol. 2022;22:429–43.
Luo J-H, Wang F-X, Zhao J-W, Yang C-L, Rong S-J, Lu W-Y et al. PDIA3 defines a novel subset of adipose macrophages to exacerbate the development of obesity and metabolic disorders. Cell Metab. 2024;S1550-4131(24)00361-9.
Lee M, Du H, Winer DA, Clemente-Casares X, Tsai S. Mechanosensing in macrophages and dendritic cells in steady-state and disease. Front Cell Dev Biol. 2022;10:1044729.
Naftaly A, Kislev N, Izgilov R, Adler R, Silber M, Shalgi R, et al. Nutrition alters the stiffness of adipose tissue and cell signaling. Int J Mol Sci. 2022;23:15237.
Abdennour M, Reggio S, Le Naour G, Liu Y, Poitou C, Aron-Wisnewsky J, et al. Association of adipose tissue and liver fibrosis with tissue stiffness in morbid obesity: links with diabetes and BMI loss after gastric bypass. J Clin Endocrinol Metab. 2014;99:898–907.
Abuhattum S, Kotzbeck P, Schlüßler R, Harger A, Ariza de Schellenberger A, Kim K, et al. Adipose cells and tissues soften with lipid accumulation while in diabetes adipose tissue stiffens. Sci Rep. 2022;12:10325.
Zhang Y-X, Ou M-Y, Yang Z-H, Sun Y, Li Q-F, Zhou S-B. Adipose tissue aging is regulated by an altered immune system. Front Immunol. 2023;14:1125395.
Lv J, Zhang C, Liu X, Gu C, Liu Y, Gao Y, et al. An aging-related immune landscape in the hematopoietic immune system. Immun Ageing. 2024;21:3.
Schnell A, Littman DR, Kuchroo VK. TH17 cell heterogeneity and its role in tissue inflammation. Nat Immunol. 2023;24:19–29.
Lin W, Jin L, Chen H, Wu Q, Fei Y, Zheng W, et al. B cell subsets and dysfunction of regulatory B cells in IgG4-related diseases and primary Sjögren’s syndrome: the similarities and differences. Arthritis Res Ther. 2014;16:R118.
Lin X, Wang X, Xiao F, Ma K, Liu L, Wang X, et al. IL-10-producing regulatory B cells restrain the T follicular helper cell response in primary Sjögren’s syndrome. Cell Mol Immunol. 2019;16:921–31.
Zhu Q, Rui K, Wang S, Tian J. Advances of Regulatory B cells in Autoimmune diseases. Front Immunol. 2021;12:592914.
Zhu DY-D, Maurer DP, Castrillon C, Deng Y, Mohamed FAN, Ma M et al. Lupus-associated innate receptors drive extrafollicular evolution of autoreactive B cells. bioRxiv. 2024;2024.01.09.574739.
Oleinika K, Slisere B, Catalán D, Rosser EC. B cell contribution to immunometabolic dysfunction and impaired immune responses in obesity. Clin Exp Immunol. 2022;210:263–72.
Lee JL, Fra-Bido SC, Burton AR, Innocentin S, Hill DL, Linterman MA. B cell-intrinsic changes with age do not impact antibody-secreting cell formation but delay B cell participation in the germinal centre reaction. Aging Cell. 2022;21:e13692.
Miyauchi E, Shimokawa C, Steimle A, Desai MS, Ohno H. The impact of the gut microbiome on extra-intestinal autoimmune diseases. Nat Rev Immunol. 2023;23:9–23.
Molinero N, Antón-Fernández A, Hernández F, Ávila J, Bartolomé B, Moreno-Arribas MV. Gut microbiota, an additional Hallmark of Human Aging and Neurodegeneration. Neuroscience. 2023;518:141–61.
Khan S, Luck H, Winer S, Winer DA. Emerging concepts in intestinal immune control of obesity-related metabolic disease. Nat Commun. 2021;12:2598.
Greiling TM, Dehner C, Chen X, Hughes K, Iñiguez AJ, Boccitto M, et al. Commensal orthologs of the human autoantigen Ro60 as triggers of autoimmunity in lupus. Sci Transl Med. 2018;10:eaan2306.
Shemtov SJ, Emani R, Bielska O, Covarrubias AJ, Verdin E, Andersen JK, et al. The intestinal immune system and gut barrier function in obesity and ageing. FEBS J. 2023;290:4163–86.
Luck H, Tsai S, Chung J, Clemente-Casares X, Ghazarian M, Revelo XS, et al. Regulation of obesity-related insulin resistance with gut anti-inflammatory agents. Cell Metab. 2015;21:527–42.
Hohman LS, Osborne LC. A gut-centric view of aging: do intestinal epithelial cells contribute to age-associated microbiota changes, inflammaging, and immunosenescence? Aging Cell. 2022;21:e13700.
Rojas OL, Pröbstel A-K, Porfilio EA, Wang AA, Charabati M, Sun T, et al. Recirculating intestinal IgA-Producing cells regulate Neuroinflammation via IL-10. Cell. 2019;176:610–e62418.
Cohavy O, Bruckner D, Gordon LK, Misra R, Wei B, Eggena ME, et al. Colonic bacteria express an ulcerative colitis pANCA-related protein epitope. Infect Immun. 2000;68:1542–8.
Garabatos N, Santamaria P. Gut microbial antigenic mimicry in autoimmunity. Front Immunol. 2022;13:873607.
Kawamoto S, Hara E. Crosstalk between gut microbiota and cellular senescence: a vicious cycle leading to aging gut. Trends Cell Biol. 2024;34:626–35.
Moll JM, Myers PN, Zhang C, Eriksen C, Wolf J, Appelberg KS, et al. Gut microbiota perturbation in IgA Deficiency is influenced by IgA-Autoantibody status. Gastroenterology. 2021;160:2423–e24345.
Luck H, Khan S, Kim JH, Copeland JK, Revelo XS, Tsai S, et al. Gut-associated IgA + immune cells regulate obesity-related insulin resistance. Nat Commun. 2019;10:3650.
Lin Y-H, Luck H, Khan S, Schneeberger PHH, Tsai S, Clemente-Casares X, et al. Aryl hydrocarbon receptor agonist indigo protects against obesity-related insulin resistance through modulation of intestinal and metabolic tissue immunity. Int J Obes (Lond). 2019;43:2407–21.
Gschmack E, Monoranu C-M, Marouf H, Meyer S, Lessel L, Idris R, et al. Plasma autoantibodies to glial fibrillary acidic protein (GFAP) react with brain areas according to Braak staging of Parkinson’s disease. J Neural Transm (Vienna). 2022;129:545–55.
Ahn JJ, Abu-Rub M, Miller RH. B cells in Neuroinflammation: New perspectives and mechanistic insights. Cells. 2021;10:1605.
Carmona-Iragui M, Fernández-Arcos A, Alcolea D, Piazza F, Morenas-Rodriguez E, Antón-Aguirre S, et al. Cerebrospinal fluid Anti-Amyloid-β autoantibodies and amyloid PET in cerebral amyloid angiopathy-related inflammation. J Alzheimers Dis. 2016;50:1–7.
Kellner A, Matschke J, Bernreuther C, Moch H, Ferrer I, Glatzel M. Autoantibodies against beta-amyloid are common in Alzheimer’s disease and help control plaque burden. Ann Neurol. 2009;65:24–31.
Mullard A. FDA approves third anti-amyloid antibody for Alzheimer disease. Nat Rev Drug Discov. 2024;23:571.
Ohgita T, Sakai K, Fukui N, Namba N, Nakano M, Kiguchi Y, et al. Generation of novel anti-apoe monoclonal antibodies that selectively recognize apoE isoforms. FEBS Lett. 2024;598:902–14.
Pocevičiūtė D, Roth B, Schultz N, Nuñez-Diaz C, Janelidze S et al. The Netherlands Brain Bank null,. Plasma IAPP-Autoantibody Levels in Alzheimer’s Disease Patients Are Affected by APOE4 Status. Int J Mol Sci. 2023;24:3776.
Pu A, Lee DSW, Isho B, Naouar I, Gommerman JL. The impact of IgA and the Microbiota on CNS disease. Front Immunol. 2021;12:742173.
Shim S-M, Koh YH, Kim J-H, Jeon J-P. A combination of multiple autoantibodies is associated with the risk of Alzheimer’s disease and cognitive impairment. Sci Rep. 2022;12:1312.
de la Monte SM. Insulin Resistance and neurodegeneration: Progress towards the development of new therapeutics for Alzheimer’s Disease. Drugs. 2017;77:47–65.
Akhtar RS, Licata JP, Luk KC, Shaw LM, Trojanowski JQ, Lee VM-Y. Measurements of auto-antibodies to α-synuclein in the serum and cerebral spinal fluids of patients with Parkinson’s disease. J Neurochem. 2018;145:489–503.
Bach J-P, Falkenburger BH. What autoantibodies tell us about the pathogenesis of Parkinson’s disease: an Editorial for measurements of auto-antibodies to α-synuclein in the serum and cerebral spinal fluids of patients with Parkinson’s disease on page 489. J Neurochem. 2018;145:433–5.
Labandeira CM, Pedrosa MA, Quijano A, Valenzuela R, Garrido-Gil P, Sanchez-Andrade M, et al. Angiotensin type-1 receptor and ACE2 autoantibodies in Parkinson´s disease. NPJ Parkinsons Dis. 2022;8:76.
Wang EY, Dai Y, Rosen CE, Schmitt MM, Dong MX, Ferré EMN, et al. High-throughput identification of autoantibodies that target the human exoproteome. Cell Rep Methods. 2022;2:100172.
Larman HB, Zhao Z, Laserson U, Li MZ, Ciccia A, Gakidis MAM, et al. Autoantigen discovery with a synthetic human peptidome. Nat Biotechnol. 2011;29:535–41.
Bodansky A, Yu DJ, Rallistan A, Kalaycioglu M, Boonyaratanakornkit J, Green DJ, et al. Unveiling the proteome-wide autoreactome enables enhanced evaluation of emerging CAR T cell therapies in autoimmunity. J Clin Invest. 2024;134:e180012.
Ramírez-Valle F, Maranville JC, Roy S, Plenge RM. Sequential immunotherapy: towards cures for autoimmunity. Nat Rev Drug Discov. 2024;23:501–24.
Wang X, Wu X, Tan B, Zhu L, Zhang Y, Lin L, et al. Allogeneic CD19-targeted CAR-T therapy in patients with severe myositis and systemic sclerosis. Cell. 2024;187:4890–e49049.
Haghikia A, Hegelmaier T, Wolleschak D, Böttcher M, Desel C, Borie D, et al. Anti-CD19 CAR T cells for refractory myasthenia gravis. Lancet Neurol. 2023;22:1104–5.
Reincke SM, von Wardenburg N, Homeyer MA, Kornau H-C, Spagni G, Li LY, et al. Chimeric autoantibody receptor T cells deplete NMDA receptor-specific B cells. Cell. 2023;186:5084–e509718.
Ellebrecht CT, Bhoj VG, Nace A, Choi EJ, Mao X, Cho MJ, et al. Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science. 2016;353:179–84.
Amor C, Feucht J, Leibold J, Ho Y-J, Zhu C, Alonso-Curbelo D, et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature. 2020;583:127–32.
Amor C, Fernández-Maestre I, Chowdhury S, Ho Y-J, Nadella S, Graham C, et al. Prophylactic and long-lasting efficacy of senolytic CAR T cells against age-related metabolic dysfunction. Nat Aging. 2024;4:336–49.
Wang B, Wang L, Gasek NS, Kuo C-L, Nie J, Kim T, et al. Intermittent clearance of p21-highly-expressing cells extends lifespan and confers sustained benefits to health and physical function. Cell Metab. 2024;36:1795–e18056.
Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018;24:1246–56.
Calabrò A, Accardi G, Aiello A, Caruso C, Galimberti D, Candore G. Senotherapeutics to counteract senescent cells are prominent topics in the context of anti-ageing strategies. Int J Mol Sci. 2024;25:1792.
Marin I, Boix O, Garcia-Garijo A, Sirois I, Caballe A, Zarzuela E, et al. Cellular Senescence is immunogenic and promotes Antitumor Immunity. Cancer Discov. 2023;13:410–31.
Rakhshandehroo T, Mantri SR, Moravej H, Louis BBV, Salehi Farid A, Munaretto L et al. A CAR enhancer increases the activity and persistence of CAR T cells. Nat Biotechnol. 2024.
Kagoya Y, Tanaka S, Guo T, Anczurowski M, Wang C-H, Saso K, et al. A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects. Nat Med. 2018;24:352–9.
Bucci L, Hagen M, Rothe T, Raimondo MG, Fagni F, Tur C, et al. Bispecific T cell engager therapy for refractory rheumatoid arthritis. Nat Med. 2024;30:1593–601.
Lempicki MD, Gray JA, Abuna G, Murata RM, Divanovic S, McNamara CA, et al. BAFF neutralization impairs the autoantibody-mediated clearance of dead adipocytes and aggravates obesity-induced insulin resistance. Front Immunol. 2024;15:1436900.
Fillatreau S. Antibodies against type I IFN: the bad guys self-restrain in systemic lupus erythematosus. Cell Rep Med. 2023;4:100903.
Tabuchi Y, Shimoda M, Kagara N, Naoi Y, Tanei T, Shimomura A, et al. Protective effect of naturally occurring anti-HER2 autoantibodies on breast cancer. Breast Cancer Res Treat. 2016;157:55–63.
Zhang Y, Chen H, Li R, Sterling K, Song W. Amyloid β-based therapy for Alzheimer’s disease: challenges, successes and future. Signal Transduct Target Ther. 2023;8:248.
Morand EF, Furie R, Tanaka Y, Bruce IN, Askanase AD, Richez C, et al. Trial of Anifrolumab in active systemic Lupus Erythematosus. N Engl J Med. 2020;382:211–21.
Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659–72.
Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, Maier M, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature. 2016;537:50–6.
Arnold C. Inverse vaccines could treat autoimmune disease - from multiple sclerosis to celiac disease. Nat Med. 2024;30:1218–9.
Tremain AC, Wallace RP, Lorentz KM, Thornley TB, Antane JT, Raczy MR, et al. Synthetically glycosylated antigens for the antigen-specific suppression of established immune responses. Nat Biomed Eng. 2023;7:1142–55.
Niazi SK. Anti-idiotypic mRNA vaccine to treat Autoimmune disorders. Vaccines (Basel). 2023;12:9.
Hampe CS. Protective role of anti-idiotypic antibodies in autoimmunity–lessons for type 1 diabetes. Autoimmunity. 2012;45:320–31.
Pan SY, Chia YC, Yee HR, Fang Cheng AY, Anjum CE, Kenisi Y, et al. Immunomodulatory potential of anti-idiotypic antibodies for the treatment of autoimmune diseases. Future Sci OA. 2020;7:FSO648.
Ramani S, Pathak A, Dalal V, Paul A, Biswas S. Oxidative stress in Autoimmune diseases: an under dealt malice. Curr Protein Pept Sci. 2020;21:611–21.
Zeng L, Yu G, Yang K, Li J, Hao W, Chen H. The efficacy of Antioxidative Stress Therapy on oxidative stress levels in rheumatoid arthritis: a systematic review and Meta-analysis of Randomized controlled trials. Oxid Med Cell Longev. 2021;2021:3302886.
Bastard P, Rosen LB, Zhang Q, Michailidis E, Hoffmann H-H, Zhang Y, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020;370:eabd4585.
Khan MWA, Al Otaibi A, Sherwani S, Khan WA, Alshammari EM, Al-Zahrani SA, et al. Glycation and oxidative stress increase autoantibodies in the Elderly. Molecules. 2020;25:3675.
Manoussakis MN, Tzioufas AG, Silis MP, Pange PJ, Goudevenos J, Moutsopoulos HM. High prevalence of anti-cardiolipin and other autoantibodies in a healthy elderly population. Clin Exp Immunol. 1987;69:557–65.
Falorni A, Gambelunghe G, Forini F, Kassi G, Cosentino A, Candeloro P, et al. Autoantibody recognition of COOH-terminal epitopes of GAD65 marks the risk for insulin requirement in adult-onset diabetes mellitus. J Clin Endocrinol Metab. 2000;85:309–16.
Zimering MB, Pan Z. Autoantibodies in type 2 diabetes induce stress fiber formation and apoptosis in endothelial cells. J Clin Endocrinol Metab. 2009;94:2171–7.
Davydova TV, Voskresenskaya NI, Fomina VG, Vetrile LA, Doronina OA. Induction of autoantibodies to glutamate in patients with Alzheimer’s disease. Bull Exp Biol Med. 2007;143:182–3.
Britschgi M, Olin CE, Johns HT, Takeda-Uchimura Y, LeMieux MC, Rufibach K, et al. Neuroprotective natural antibodies to assemblies of amyloidogenic peptides decrease with normal aging and advancing Alzheimer’s disease. Proc Natl Acad Sci U S A. 2009;106:12145–50.
Attanasio R, Brasky KM, Robbins SH, Jayashankar L, Nash RJ, Butler TM. Age-related autoantibody production in a nonhuman primate model. Clin Exp Immunol. 2001;123:361–5.
Acknowledgements
This work was funded, in part, through funds derived from the National Institutes of Health grant R01DK128435, and Canadian Institutes of Health Research grants PJT-186165, PJT-195795. T.R.V. was funded by a T32 NIH fellowship grant (NIA T32 AG000266). Figures 1, 2 and 3 were drawn using BioRender (www.Biorender.com).
Author information
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Valentino, T.R., Chen, N., Makhijani, P. et al. The role of autoantibodies in bridging obesity, aging, and immunosenescence. Immun Ageing 21, 85 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12979-024-00489-2
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12979-024-00489-2