Elsevier

Seminars in Immunology

Volume 20, Issue 1, February 2008, Pages 67-82
Seminars in Immunology

Review
Phenotypic and functional heterogeneity of human memory B cells

https://doi.org/10.1016/j.smim.2007.12.006Get rights and content

Abstract

Memory B cells are more heterogeneous than previously thought. Given that B cells play powerful antibody-independent effector functions, it seems reasonable to assume division of labor between distinct memory B cells subpopulations in both protective and pathogenic immune responses. Here we review the information emerging regarding the heterogeneity of human memory B cells. A better understanding of this topic should greatly improve our ability to target specific B cell subsets either in vaccine responses or in autoimmune diseases and organ rejection among other pathological conditions where B cells play central pathogenic roles.

Introduction

Immunological memory represents a highly effective mechanism to ensure quick protection against prevalent infections. B cell memory is generally viewed as supported by two cellular compartments: plasma cells, responsible for the production of antibodies (effector memory) and memory B cells which would represent precursors (central memory) capable of generating and replenishing the plasma cell compartment through a combination still incompletely understood of antigen-dependent and -independent mechanisms [1], [2], [3], [4]. This view is certainly valid when B cell effector functions are limited to the production of antibody. Yet, it is also incomplete as it fails to incorporate important effector and regulatory functions that may be played by “central” memory B cells (antigen-presentation, T cell and dendritic cell regulation and cytokine and chemokine production) [5], [6], [7]. When these critical B cell functions are considered, it can be postulated that effector memory is mediated by both plasma cells (through the production of antibodies) and by memory B cells (mostly through the production of cytokines). In turn, central memory B cells would be responsible not only for the generation and replenishment of plasma cells but also for the generation of distinct subsets of effector B cells. It is also possible that central memory B cells could play distinct regulatory roles.

Of note, the same properties of memory cells that are desirable for protective immune responses (long lifespan, prompt and enhanced responses to activation and ability to stimulate T cells) may be deleterious when it comes to avoiding chronic autoimmunity. Thus, once tolerance is broken in the B cell compartment, autoimmune memory would tend to persist (facilitated by availability of self antigens), could be easily reactivated and could break tolerance in the T cell compartment thereby providing a mechanism for further diversification and amplification [8], [9]. Given that substantial autoreactivity can be detected even in the IgG memory compartment of healthy subjects [10], it seems obvious that protective (anti-microbial) and potentially pathogenic (autoreactive) memory B cells must have different properties and be controlled by distinct regulatory mechanisms.

With regard to surface phenotype, substantial heterogeneity amongst human memory B cells has been documented by many studies including our own. Indeed, despite initial descriptions of CD27 as a universal marker of human memory cells, we and others have described memory populations that lack expression of CD27 and may be substantial in SLE and in some infections such as RSV [11], [12], [13], [14]. As discussed in this review, additional heterogeneity of memory B cells can also be demonstrated on the basis of the expression of CD38, CD21, CD24, CD19, B220, FcRH4 and CD25 [12], [15], [16], [17].

In addition to surface phenotype, the heterogeneity of human memory B cells can also be documented by: (1) differential regulation in autoimmune diseases and infections; (2) different repopulation kinetics after B cell depletion therapy (BCDT); (3) differential impact of other biological therapies; (4) different in vivo doubling times and half-life as documented by heavy water labeling (our own preliminary data indicating that CD27 memory cells have a much longer doubling time than CD27+ memory cells).

Importantly however, the actual functions of memory B cell subsets remains to be understood in terms of their relative participation in protective and autoimmune responses and in terms of their specific effectors and regulatory functions including cytokine production, antigen presentation and ability to differentiate into plasma cells. Also currently unknown is whether specific memory B cell subsets have different participation in central memory (understood as precursors of effector B cells and plasma cells) versus effector memory (defined as cytokine production). It is worth noting however that there is significant literature to support the ability of memory cells to produce abundant cytokines (TNF and lymphotoxin) with patterns different from naïve B cells (IL-10). Different studies also indicate that cytokine production by B cells may be relevant to human autoimmune diseases including SLE and MS. Finally, preliminary evidence indicates that excess production of pro-inflammatory cytokines by B cells may be reversed by BCDT in patients with MS [18].

Section snippets

Memory B cell heterogeneity

As recently discussed by others [19], our understanding of the diversity of memory B cell populations is hampered by pre-conceived notions of their surface phenotype, function and cellular and anatomical origin. Thus, the strict definition of a memory cell as one expressing isotype-switched, somatically mutated antibodies and generated through a T cell-dependent germinal center reaction would exclude important memory responses mediated by unswitched and/or unmutated B cells that at least in

Current schemes of classification of human memory B cell populations

By and large, the analysis of human B cell populations by flow cytometry has thus far relied on the expression of four major surface markers: CD19, IgD, CD38 and D27. With this four-color approach, two major classification schemes can be produced depending on the relative expression of either IgD and CD38 or IgD and CD27 (Fig. 1). Thus IgD/CD38 staining provides the so-called Bm1–Bm5 classification and can be used to identify multiple subsets in the human tonsil including: virgin naïve cells

Substantial additional complexity of human memory cells as revealed by multi-chromatic flow cytometry

In addition to the aforementioned classification schemes, human B cell subsets can also be defined by the surface expression levels of CD24 and CD38 (naïve, memory, GC, transitional B cells and plasmablasts). However, how the subsets defined by one classification scheme relate to those subsets defined by other schemes is not often clear. Hence, we have developed six different 10-color flow cytometry panels which provide a comprehensive phenotypic analysis of human B cell subsets and reveal

Anatomical distribution of human memory B cell subsets: The conundrum of the human spleen marginal zone and its equivalents

One of the main challenges in the study of human memory B cells is to understand their localization, phenotype and function of different subsets in the locales where they are generated in the course of orchestrated immune responses, where they reside as selected resting memory cells (in both cases presumably in the secondary lymphoid tissues although the contribution of the bone marrow needs to be more thoroughly assessed), and where they are recruited or generated as activated effector cells

Kinetics and homeostasis of memory B cell subsets: lessons from clinical situations and BCDT

The importance of elucidating human memory B cell heterogeneity is highlighted by our findings and those of others that certain memory subsets are preferentially expanded in particular clinical situations. Moreover, such studies have provided insight into the potential origin of diverse subsets and their dynamic generation in peripheral lymphoid tissue.

As shown by different groups, SLE patients display an expansion of switched memory in the peripheral blood which as previously discussed, is

Immuno-modulatory and effector functions of human memory B cells

While usually overshadowed by the production of antibodies, the ability of B cells to play important antibody-independent functions is well documented [5], [6], [7], [115]. These antibody-independent functions prominently include cytokine and chemokine production and antigen presentation. Through these functions B cells can profoundly influence the formation and organization of secondary lymphoid tissues and T cell development, activation and function [116], [117], [118], [119], [120], [121].

Summary

Experimental evidence continues to accumulate indicating that B cell memory is rather complex and includes T-cell dependent and independent memory, isotyope-switched and unswitched, mutated and unmutated memory B cells. It may also include a “natural” memory at least in part provided by conventional MZ B cells, several adaptive memory populations and possibly adaptive memory B cells that acquire marginal zone-like properties. Functionally, effective and appropriate (i.e., protective and

Acknowledgements

The work in this paper is supported in part by grants: R01 AI049660-01A1, U19 Autoimmunity Center of Excellence AI56390, and Center for Biodefense of Immuno-compromised Populations N01-AI50029 (IS); NIH-NIAMS K08AR048303, the Lupus Foundation of America, and the Alliance for Lupus Research (JA); NIH-NIAID K23A167501-01A1 (FEL).

References (139)

  • T. Lopes-Carvalho et al.

    Marginal zone B cells in lymphocyte activation and regulation

    Curr Opin Immunol

    (2005)
  • R.C. Lindsley et al.

    Generation of peripheral B cells occurs via two spatially and temporally distinct pathways

    Blood

    (2007)
  • B. Steiniger et al.

    The perifollicular and marginal zones of the human splenic white pulp: do fibroblasts guide lymphocyte immigration?

    Am J Pathol

    (2001)
  • P.L. Amlot et al.

    Impaired human antibody response to the thymus-independent antigen, DNP-Ficoll, after splenectomy. Implications for post-splenectomy infections

    Lancet

    (1985)
  • A. Tierens et al.

    Marginal-zone B cells in the human lymph node and spleen show somatic hypermutations and display clonal expansion

    Blood

    (1999)
  • B. Falini et al.

    Expression of the IRTA1 receptor identifies intraepithelial and subepithelial marginal zone B cells of the mucosa-associated lymphoid tissue (MALT)

    Blood

    (2003)
  • T. Marafioti et al.

    Phenotype and genotype of interfollicular large B cells, a subpopulation of lymphocytes often with dendritic morphology

    Blood

    (2003)
  • W.J. Hofmann et al.

    Thymic medullary cells expressing B lymphocyte antigens

    Hum Pathol

    (1988)
  • F. Sallusto et al.

    Central memory and effector memory T cell subsets: function, generation, and maintenance

    Annu Rev Immunol

    (2004)
  • A.F. Ochsenbein et al.

    Protective long-term antibody memory by antigen-driven and T help-dependent differentiation of long-lived memory B cells to short-lived plasma cells independent of secondary lymphoid organs

    PNAS

    (2000)
  • T. Dorner et al.

    Selecting B cells and plasma cells to memory

    J Exp Med

    (2005)
  • F. Martin et al.

    B cell immunobiology in disease: evolving concepts from the clinic

    Annu Rev Immunol

    (2006)
  • I. Sanz et al.

    B cell depletion therapy in autoimmune diseases

    Front Biosci

    (2007)
  • D.P. Harris et al.

    Reciprocal regulation of polarized cytokine production by effector B and T cells

    Nat Immunol

    (2000)
  • M.J. Mamula

    Epitope spreading: the role of self peptides and autoantigen processing by B lymphocytes

    Immunol Rev

    (1998)
  • H. Noorchashm et al.

    I-Ag7-mediated antigen presentation by B lymphocytes is critical in overcoming a checkpoint in T cell tolerance to islet {beta} cells of nonobese diabetic mice

    J Immunol

    (1999)
  • J.F. Fecteau et al.

    A new memory CD27-IgG+ B cell population in peripheral blood expressing VH genes with low frequency of somatic mutation

    J Immunol

    (2006)
  • C. Wei et al.

    A new population of cells lacking expression of CD27 represents a notable component of the B cell memory compartment in systemic lupus erythematosus

    J Immunol

    (2007)
  • S. Wirths et al.

    ABCB1 transporter discriminates human resting naive B cells from cycling transitional and memory B cells

    Eur J Immunol

    (2005)
  • G.R.A. Ehrhardt et al.

    Expression of the immunoregulatory molecule FcRH4 defines a distinctive tissue-based population of memory B cells

    J Exp Med

    (2005)
  • D. Culton et al.

    Similar CD19 dysregulation in two autoantibody-associated autoimmune diseases suggests a shared mechanism of B-cell tolerance loss

    J Clin Immunol

    (2007)
  • A.J. Cappione et al.

    Lupus IgG VH4.34 antibodies bind to a 220-kDa glycoform of CD45/B220 on the surface of human B lymphocytes

    J Immunol

    (2004)
  • S. Amu et al.

    The human immunomodulatory CD25+ B cell population belongs to the memory B cell pool

    Scand J Immunol

    (2007)
  • M. Duddy et al.

    Distinct effector cytokine profiles of memory and naive human B cell subsets and implication in multiple sclerosis

    J Immunol

    (2007)
  • S.M. Anderson et al.

    Intrinsic properties of human and murine memory B cells

    Immunol Rev

    (2006)
  • S. Weller et al.

    CD40-CD40L independent Ig gene hypermutation suggests a second B cell diversification pathway in humans

    Proc Natl Acad Sci USA

    (2001)
  • Y. Harada et al.

    Unmutated immunoglobulin M can protect mice from death by influenza virus infection

    J Exp Med

    (2003)
  • S. Kruetzmann et al.

    Human immunoglobulin M memory B cells controlling Streptococcus pneumoniae infections are generated in the spleen

    J Exp Med

    (2003)
  • S. Weller et al.

    Human blood IgM “memory” B cells are circulating splenic marginal zone B cells harboring a pre-diversified immunoglobulin repertoire

    Blood

    (2004)
  • A. Di Sabatino et al.

    Depletion of immunoglobulin M memory B cells is associated with splenic hypofunction in inflammatory bowel disease

    Am J Gastroenterol

    (2005)
  • Y. Shi et al.

    Regulation of aged humoral immune defense against pneumococcal bacteria by IgM memory B cell

    J Immunol

    (2005)
  • C.S. Ma et al.

    Selective generation of functional somatically mutated IgM+CD27+, but not Ig isotype-switched, memory B cells in X-linked lymphoproliferative disease

    J Clin Invest

    (2006)
  • S.M. Anderson et al.

    New markers for murine memory B cells that define mutated and unmutated subsets

    J Exp Med

    (2007)
  • J.M. Robertson et al.

    Not all CD4+ memory T cells are long lived

    Immunol Rev

    (2006)
  • K.K. McKinstry et al.

    Rapid default transition of CD4 T cell effectors to functional memory cells

    J Exp Med

    (2007)
  • M.-C. Hsu et al.

    B cell clones that sustain long-term plasmablast growth in T-independent extrafollicular antibody responses

    PNAS

    (2006)
  • J. Zhang et al.

    B cell memory to thymus-independent antigens type 1 and type 2: the role of lipopolysaccharide in B memory induction

    Eur J Immunol

    (1988)
  • T.V. Obukhanych et al.

    T-independent type II immune responses generate memory B cells

    J Exp Med

    (2006)
  • Y.J. Liu et al.

    Memory B cells in T cell-dependent antibody responses colonize the splenic marginal zones

    Eur J Immunol

    (1988)
  • U. Klein et al.

    Human immunoglobulin (Ig)M+IgD+ peripheral blood B cells expressing the CD27 cell surface antigen carry somatically mutated variable region genes: CD27 as a general marker for somatically mutated (memory) B cells

    J Exp Med

    (1998)
  • Cited by (0)

    View full text