Elsevier

Experimental Gerontology

Volume 138, September 2020, 110985
Experimental Gerontology

Review
Cell signaling and the aging of B cells

https://doi.org/10.1016/j.exger.2020.110985Get rights and content

Abstract

The uniqueness of each B cell lies in the structural diversity of the B-cell antigen receptor allowing the virtually limitless recognition of antigens, a necessity to protect individuals against a range of challenges. B-cell development and response to stimulation are exquisitely regulated by a group of cell surface receptors modulating various signaling cascades and their associated genetic programs. The effects of these signaling pathways in optimal antibody-mediated immunity or the aberrant promotion of immune pathologies have been intensely researched in the past in young individuals. In contrast, we are only beginning to understand the contribution of these pathways to the changes in B cells of old organisms. Thus, critical transcription factors such as E2A and STAT5 show differential expression or activity between young and old B cells. As a result, B-cell physiology appears altered, and antibody production is impaired. Here, we discuss selected phenotypic changes during B-cell aging and attempt to relate them to alterations of molecular mechanisms.

Introduction

As mammals age, their tissues undergo physiological transformations paralleled by the appearance of pathologies like rheumatoid arthritis (RA) (Bektas et al., 2017; Franceschi et al., 2018). The consequences of aging diverge extensively between individuals as underscored by the range of health status present in the human elderly population, from vigorous to frail (Hagen and Derudder, 2019; Pansarasa et al., 2019). The variety of age-related phenotypes observed in cells and tissues results from the combined effects of an array of influences, including environmental factors (e.g., pollution, nutrition, chronic infection), intrinsic homeostasis (e.g., inflammation, senescence) and epigenetic/genetic inheritance (e.g., single nucleotide polymorphisms) (Bulati et al., 2017; Campisi et al., 2019; Ferrucci and Fabbri, 2018; Franceschi et al., 2016; Melzer et al., 2019; Pinti et al., 2016; Singh et al., 2019).

One such tissue affected by aging is the immune system. Immunity preserves the integrity of the body by eradicating or neutralizing challenging threats, such as infection or cancer. The immune system comprises a collection of cooperating cell-types, broadly categorized into the myeloid (e.g., neutrophils and dendritic cells) and lymphoid (e.g., B and T cells) lineages. These cells derive from uncommitted hematopoietic stem and progenitor cells localized in the bone marrow of adult humans and mice (Beerman et al., 2010; Northrup and Allman, 2008). Aging has been reported to extensively reshape the generation and function of immune cells. Thus, in old humans and mice the hematopoietic stem cell pool appears biased toward myeloid cell production (Beerman et al., 2010; Pang et al., 2016), and elderly individuals accumulate a pro-inflammatory terminally-differentiated effector-type of T cells (Pinti et al., 2016). B cells serve a unique role in immunity as the sole producers of antibodies upon differentiation into plasma cells. In mice and humans, aging has profound effects on B-cell physiology, culminating in an altered distribution of mature B-cell subsets as well as compromised activation upon stimulation (Bulati et al., 2017; Hagen and Derudder, 2019; Kogut et al., 2012; Pinti et al., 2016).

One consequence of the age-dependent remodeling of the immune system is the deteriorated quality of antibody responses in vulnerable elderly and old mice (Bulati et al., 2017; Linterman, 2013; Nikolich-Žugich, 2017; Pinti et al., 2016; Weinberger, 2018). Of note, chronic inflammation is a reported trigger of the declining immunity in aging, when unrestrained by anti-inflammatory molecules (Franceschi et al., 2000; Franceschi et al., 2006; Hagen and Derudder, 2019; Pinti et al., 2016). However, the exact contribution of various pro-inflammatory cytokines or the existence of effective (local) concentrations remain to be established (Nikolich-Žugich, 2017).

Here, we discuss changes in signaling pathways at the molecular level in relation to remarkable alterations in the physiology of old mouse and human B cells. In addition, this review focuses on the impact of aging on B2 cells, the main lineage of B cells in adult animals. The situation in B1 cells, primarily generated early during life, will not be considered.

Section snippets

Young adults

In mice, the influence of aging on B2 cells, thereafter referred to as B cells, has been characterized by comparison to the situation in young adults (Boothby et al., 2019; Clark et al., 2013; Northrup and Allman, 2008). The B-lymphoid lineage is specified in uncommitted hematopoietic progenitor cells in the bone marrow upon expression of the cooperating transcription factors, E2A, EBF1 and Pax5 (Northrup and Allman, 2008). Pax5 expression ultimately marks B cell identity. Developing B cells

Aging and signaling in developing B cells

Longstanding investigations in young adult mice have shown that B-cell progression through the pro-B and pre-B stages depends on signals from the pre-BCR, IL-7R and CXCR4 (Fig. 1) (Clark et al., 2013; Fistonich et al., 2018; Herzog et al., 2009; Mandal et al., 2019). These receptors deliver positive and/or negative signals controlling processes critical to early B-cell development, i.e., survival, proliferation, positioning and differentiation. Several shared and distinct signaling pathways are

Persistence of mature B cells

While B cell development in the bone marrow is impaired, the size of the mature B compartment appears preserved over time in the spleen of mice (Kogut et al., 2012). This stability could be related to the reported decline of mature splenic B-cell turnover with age in mice (Johnson et al., 2002; Kline et al., 1999). Two major receptors modulate resting mature B-cell maintenance, the BCR and BAFFR (Boothby et al., 2019). However, the contribution of signals from these receptors to mature B-cell

Activated B cells

The impaired antibody responses to challenges in old organisms can be connected to several mechanistic alterations observed in activated B cells from old mice and humans, notably resulting in reduced AID expression and CSR (Bulati et al., 2017; Frasca et al., 2010a, Frasca et al., 2008, Frasca et al., 2004a; Pinti et al., 2016).

Final remarks

The age-associated reshaping of the B cell compartment is likely a combined result of imbalanced fate choices within the hematopoietic progenitor population, B cell autonomous alterations and extrinsic signals. Notably, chronic low-grade inflammation, commonly observed in old individuals, may precipitate the decline of humoral immunity (Franceschi et al., 2000; Hagen and Derudder, 2019; Henry et al., 2015). Overall, old mice and the elderly experience comparable alterations of B cell physiology

Acknowledgement

The authors are thankful for support from the Ingrid Shaker Nessmann Cancer Research Association (D-182400-020-012), the Tyrolean Science Fund (UNI-0404/1696), the Austrian Cancer Aid (KH15017) and the Austrian Science Fund (P32755) to V.L., and from the University Innsbruck Nachwuchsförderung (2017/BIO-5) and the Tyrolean Science Fund (UNI-0404/2310) to E.D. Figures were created with BioRender.com.

References (141)

  • D. Frasca et al.

    Obesity induces pro-inflammatory B cells and impairs B cell function in old mice

    Mech. Ageing Dev.

    (2017)
  • Y. Hao et al.

    A B-cell subset uniquely responsive to innate stimuli accumulates in aged mice

    Blood

    (2011)
  • D.L. Hodge et al.

    IFN-gamma AU-rich element removal promotes chronic IFN-gamma expression and autoimmunity in mice

    J. Autoimmun.

    (2014)
  • J. Jellusova et al.

    Context-specific BAFF-R signaling by the NF-κB and PI3K pathways

    Cell Rep.

    (2013)
  • Q. Jing et al.

    Involvement of microRNA in AU-rich element-mediated mRNA instability

    Cell

    (2005)
  • Z. Keren et al.

    B-cell depletion reactivates B lymphopoiesis in the BM and rejuvenates the B lineage in aging

    Blood

    (2011)
  • A.M. King et al.

    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)
  • I. Kogut et al.

    B cell maintenance and function in aging

    Semin. Immunol.

    (2012)
  • M. Kuraoka et al.

    The first B-cell tolerance checkpoint in mice and humans: control by AID

    Adv. Immunol.

    (2018)
  • K. Kwon et al.

    Instructive role of the transcription factor E2A in early B lymphopoiesis and germinal center B cell development

    Immunity

    (2008)
  • H. Li et al.

    Repression of Id2 expression by Gfi-1 is required for B-cell and myeloid development

    Blood

    (2010)
  • C. López-Otín et al.

    The hallmarks of aging

    Cell

    (2013)
  • R.W. McKenna

    Immunophenotypic analysis of hematogones (B-lymphocyte precursors) in 662 consecutive bone marrow specimens by 4-color flow cytometry

    Blood

    (2001)
  • J.P. Miller et al.

    B cells and aging: balancing the homeostatic equation

    Exp. Gerontol.

    (2007)
  • U.K. Mukhopadhyay et al.

    STAT5A is regulated by DNA damage via the tumor suppressor p53

    Cytokine

    (2016)
  • A.J. Nipper et al.

    Diminished antibody response to influenza vaccination is characterized by expansion of an age-associated B-cell population with low PAX5

    Clin. Immunol.

    (2018)
  • T. Ogawa et al.

    Age-related changes of human bone marrow: a histometric estimation of proliferative cells, apoptotic cells, T cells, B cells and macrophages

    Mech. Ageing Dev.

    (2000)
  • H. Pan et al.

    Key proteins and pathways that regulate lifespan

    J. Biol. Chem.

    (2017)
  • O. Pansarasa et al.

    Altered immune system in frailty: genetics and diet may influence inflammation

    Ageing Res. Rev.

    (2019)
  • S. Alivernini et al.

    MicroRNA-155 influences B-cell function through PU.1 in rheumatoid arthritis

    Nat. Commun.

    (2016)
  • I. Avivi et al.

    Depletion of B cells rejuvenates the peripheral B-cell compartment but is insufficient to restore immune competence in aging

    Aging Cell

    (2019)
  • G.V. Baracho et al.

    PDK1 regulates B cell differentiation and homeostasis

    Proc. Natl. Acad. Sci. U. S. A.

    (2014)
  • J.T. Barata et al.

    Flip the coin: IL-7 and IL-7R in health and disease

    Nat. Immunol.

    (2019)
  • A. Bektas et al.

    Aging, inflammation and the environment

    Exp. Gerontol.

    (2017)
  • M.R. Boothby et al.

    Molecular regulation of peripheral B cells and their progeny in immunity

    Genes Dev.

    (2019)
  • G.M. Borchert et al.

    Repression of human activation induced cytidine deaminase by miR-93 and miR-155

    BMC Cancer

    (2011)
  • C.F.A. de Bourcy et al.

    Phylogenetic analysis of the human antibody repertoire reveals quantitative signatures of immune senescence and aging

    Proc. Natl. Acad. Sci. U. S. A.

    (2017)
  • J. Campisi et al.

    From discoveries in ageing research to therapeutics for healthy ageing

    Nature

    (2019)
  • M.R. Clark et al.

    Orchestrating B cell lymphopoiesis through interplay of IL-7 receptor and pre-B cell receptor signalling

    Nat. Rev. Immunol.

    (2013)
  • M.E. Conley et al.

    Agammaglobulinemia and absent B lineage cells in a patient lacking the p85α subunit of PI3K

    J. Exp. Med.

    (2012)
  • S.W. Du et al.

    Generation of functional murine CD11c+ age-associated B cells in the absence of B cell T-bet expression

    Eur. J. Immunol.

    (2018)
  • S.W. Du et al.

    Functional characterization of CD11c+ age-associated B cells as memory B cells

    J. Immunol.

    (2019)
  • N.A. Duggal et al.

    An age-related numerical and functional deficit in CD19(+) CD24(hi) CD38(hi) B cells is associated with an increase in systemic autoimmunity

    Aging Cell

    (2013)
  • S.P. Fahl et al.

    Rpl22 loss impairs the development of B lymphocytes by activating a p53-dependent checkpoint

    J. Immunol.

    (2014)
  • L. Ferrucci et al.

    Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty

    Nat. Rev. Cardiol.

    (2018)
  • C. Fistonich et al.

    Cell circuits between B cell progenitors and IL-7+ mesenchymal progenitor cells control B cell development

    J. Exp. Med.

    (2018)
  • C. Franceschi et al.

    Inflamm-aging. An evolutionary perspective on immunosenescence

    Ann. N. Y. Acad. Sci.

    (2000)
  • C. Franceschi et al.

    Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans

    Mech. Ageing Dev.

    (2006)
  • C. Franceschi et al.

    Inflammaging and ‘Garb-aging’

    Trends Endocrinol. Metab.

    (2016)
  • C. Franceschi et al.

    The continuum of aging and age-related diseases: common mechanisms but different rates

    Front. Med. (Lausanne)

    (2018)
  • Cited by (0)

    View full text