ArticlePublisher preview available

Germinal Center B Cells Latently Infected with Epstein-Barr Virus Proliferate Extensively but Do Not Increase in Number

American Society for Microbiology
Journal of Virology
Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

In this study we show that in long-term persistent infection, Epstein-Barr virus (EBV)-infected cells undergoing a germinal center (GC) reaction in the tonsils are limited to the follicles and proliferate extensively. Despite this, the absolute number of infected cells per GC remains small (average of 3 to 4 cells per germinal center; range, 1 to 9 cells), and only about 38 to 55% (average, 45%) of all GCs carry infected cells. The data fit a model where, on average, cells in the GC divide approximately three times; however, only one progeny cell survives to undergo a further three divisions. Thus, a fraction of cells undergo multiple rounds of division without increasing in numbers; i.e., they die at the same rate that they are dividing. We conclude that EBV-infected cells in the GC undergo the extensive proliferation characteristic of GC cells but that the absolute number is limited either by the immune response or by the availability of an essential survival factor. We suggest that this behavior is a relic of the mechanism by which EBV establishes persistence during acute infection. Lastly, the expression of the viral latent protein LMP1 in GC B cells, unlike in vitro, does not correlate directly with the expression of bcl-2 or bcl-6. This emphasizes our claim that observations made regarding the functions of EBV proteins in cell lines or in transgenic mice should be treated with skepticism unless verified in vivo.
JOURNAL OF VIROLOGY, Jan. 2010, p. 1158–1168 Vol. 84, No. 2
0022-538X/10/$12.00 doi:10.1128/JVI.01780-09
Copyright © 2010, American Society for Microbiology. All Rights Reserved.
Germinal Center B Cells Latently Infected with Epstein-Barr Virus
Proliferate Extensively but Do Not Increase in Number
Jill E. Roughan,† Charles Torgbor,‡ and David A. Thorley-Lawson*
Department of Pathology, Tufts University School of Medicine, Jaharis Building, Boston, Massachusetts 02111
Received 22 August 2009/Accepted 22 October 2009
In this study we show that in long-term persistent infection, Epstein-Barr virus (EBV)-infected cells
undergoing a germinal center (GC) reaction in the tonsils are limited to the follicles and proliferate extensively.
Despite this, the absolute number of infected cells per GC remains small (average of 3 to 4 cells per germinal
center; range, 1 to 9 cells), and only about 38 to 55% (average, 45%) of all GCs carry infected cells. The data
fit a model where, on average, cells in the GC divide approximately three times; however, only one progeny cell
survives to undergo a further three divisions. Thus, a fraction of cells undergo multiple rounds of division
without increasing in numbers; i.e., they die at the same rate that they are dividing. We conclude that
EBV-infected cells in the GC undergo the extensive proliferation characteristic of GC cells but that the absolute
number is limited either by the immune response or by the availability of an essential survival factor. We
suggest that this behavior is a relic of the mechanism by which EBV establishes persistence during acute
infection. Lastly, the expression of the viral latent protein LMP1 in GC B cells, unlike in vitro, does not
correlate directly with the expression of bcl-2 or bcl-6. This emphasizes our claim that observations made
regarding the functions of EBV proteins in cell lines or in transgenic mice should be treated with skepticism
unless verified in vivo.
Epstein-Barr virus (EBV) is a human herpesvirus that es-
tablishes a lifetime persistent infection in 90% of all adults
(reviewed in references 21 and 51). The virus persists in rest-
ing, latently infected memory B cells that circulate in the pe-
riphery and lymph nodes (5; reviewed in references 53 and 55).
In these cells the virus is quiescent, at least at the level of viral
protein expression (23). This lack of viral proteins is presum-
ably a major reason why these cells are able to persist in the
face of a healthy immune response. The other property for
which EBV is well known is its ability to infect resting B cells
and drive them to become activated proliferating lymphoblasts
through the expression of nine latent proteins and several
untranslated RNAs (reviewed in reference 45). The latter in-
clude a large number of microRNAs (miRNAs) (7, 12, 17, 44).
The current model of Epstein-Barr virus persistence holds
that the virus drives resting B cells to become activated lym-
phoblasts so that they can differentiate through a germinal
center (GC) reaction to become resting memory B cells (55–
57), where the virus persists. The GC is the structure in the
follicles of secondary lymphoid organs where antigen-activated
B cells undergo a T-cell-dependent immune response (1, 35,
37). The production and maintenance of GCs are absolutely
dependent on the expression of the transcription factor bcl-6
(13, 58) and are initiated by the rapid proliferation of antigen-
specific B cells. During this expansion phase, B cells divide
approximately every 6 to 12 h (2, 37) so that 3 2 founder cells
can create a GC of approximately 10
5
cells in a few days (29,
36, 37). In the classical model of GC development (37), it was
proposed that in the proliferating state, the B cells (centro-
blasts) reside in the dark zone of the follicle and do not express
their antigen receptor while actively undergoing the processes
of somatic hypermutation and immunoglobulin class switching.
These processes are governed by the enzyme AID (activation-
induced cytidine deaminase), whose expression is also a defin-
ing feature of GC B cells (39). After several rounds of division,
the cells move to the light zone, rest, reexpress their surface
immunoglobulin (centrocytes), and compete for antigen bind-
ing and T-cell help. Survival requires that the B cell success-
fully competes for both signals. It should be noted, though, that
the clear-cut distinction between centroblasts and centrocytes
has been called into question (1).
While in the GC, B cells are highly apoptosis prone, and a
failure to receive the requisite survival signals leads rapidly to
death. If the survival signals are received, then there are three
possible fates for the GC cell. It can recapitulate the process by
once more becoming a centroblast, or it can upregulate pro-
survival molecules such as bcl-2 (29, 36) and leave the GC as a
plasma or memory B cell. Which fate the B cell follows de-
pends on the cytokine environment and the relative strength
and endurance of its interaction with antigen and T-helper
cells.
In a previous study we presented direct evidence that EBV-
infected cells transiting from activated lymphoblast to memory
B cells are found to physically reside in tonsil GCs and partic-
ipate in the GC reaction (46). This included the expression of
bcl-6 and AID, defining makers of the GC, and GC-specific
chemokine receptors that would cause them to be retained in
the GC. Moreover, the cells express markers associated with
* Corresponding author. Mailing address: Dept. of Pathology, Jaha-
ris Building, Tufts University School of Medicine, 150 Harrison Ave.,
Boston, MA 02111. Phone: (617) 636-2726. Fax: (617) 636-2990.
E-mail: david.thorley-lawson@tufts.edu.
Present address: Department of Immunology and Microbial Sci-
ence, The Scripps Research Institute, La Jolla, CA 92037.
Present address: Department of Biochemistry and Biotechnology,
Kwame Nkrumah University of Science and Technology, Kumasi,
Ghana, West Africa.
Published ahead of print on 4 November 2009.
1158
... In contrast, no transcripts were detected in the absence of EBV genome assessed by PCR. 29 Regarding the expression of LMP1 protein, it was reported that EBERsÀ cases by conventional ISH did not express LMP1 protein. 15 In contrast, we proved LMP1 protein expression in EBERs+ cases, and in EBERsÀ cases with LMP1 transcripts expression as well. ...
Article
Full-text available
In 2017, the World Health Organization (WHO) confirmed a new entity, Epstein Barr virus (EBV) + Diffuse large B cell lymphoma (DLBCL), not otherwise specified (NOS). Traces of EBV transcripts were described in lymphomas, including DLBCL, that were diagnosed as EBV negative by conventional methods. The aim of this study was to detect viral genome by qPCR, as well as LMP1 and EBNA2 transcripts, with a more sensitive method in DLBCL cases from Argentina. Fourteen cases originally considered as EBV negative expressed LMP1 and/or EBNA2 transcripts. In addition, LMP1 and/or EBNA2 transcripts were also observed in bystander cells. However, EBERs+ cells cases by conventional ISH showed higher numbers of cells with LMP1 transcripts and LMP1 protein. In the cases that were EBERS− in tumor cells but with expression of LMP1 and/or EBNA2 transcripts, the viral load was below the limit of detection. This study provides further evidence that EBV could be detected in tumor cells by more sensitive methods. However, higher expression of the most important oncogenic protein, LMP1, as well as increased viral load, are only observed in cases with EBERs+ cells by conventional ISH, suggesting that traces of EBV might not display a key role in DLBCL pathogenesis.
... The virus infects cell types that express the receptor CR2 or CD21, such as pharynx cells (Birkenbach et al. 1992;Jiang et al. 2008;Smith et al. 2020). The pharynx cells contain few unintegrated copies (episomes) of the virus genome, which are replicated every time B cell divides (Roughan et al. 2010). From these EBV producing plasma B-cells EBV can be transferred to local (oropharyngeal) epithelial cells where replication is increased and the virus is shed into saliva (Hadinoto et al. 2009;Tsang et al. 2014). ...
Article
Full-text available
Epstein-Barr Virus can cause various diseases, from acute inflammatory diseases such as fatal or chronic EBV infection, infectious mononucleosis as well as lymphoid and epithelial cancer, various autoimmune diseases, and also could interact with malaria. As EBV infects 95% of the world population, and more than 30% are infected with the protozoan parasite, with more than 500,000 deaths due to malaria cases. It is important to understand how EBV dysregulates the immune system, especially when the virus is interacting with other pathogens such as malaria parasites, causing more severe conditions in certain people like Burkitt Lymphoma. This review will be informative about the mechanism of how EBV interacts with malaria parasites and how it affects the immune system. Knowledge of various cytokines triggering the immune system which may provide links to control/minimize malaria disease severity.
... In total, there are 25 parameters, which we fixed to published values to obtain a reference fit of the model (Supplementary Table 1) (12)(13)(14). The latently infected memory B cell compartment, B m , was taken as a proxy for EBV VL. ...
Article
Full-text available
Epstein-Barr virus (EBV) establishes a lifelong latent infection in healthy humans, kept under immune control by cytotoxic T cells (CTLs). Following paediatric haematopoetic stem cell transplantation (HSCT), a loss of immune surveillance leads to opportunistic outgrowth of EBV-infected cells, resulting in EBV reactivation, which can ultimately progress to post-transplant lymphoproliferative disorder (PTLD). The aims of this study were to identify risk factors for EBV reactivation in children in the first 100 days post-HSCT and to assess the suitability of a previously reported mathematical model to mechanistically model EBV reactivation kinetics in this cohort. Retrospective electronic data were collected from 56 children who underwent HSCT at Great Ormond Street Hospital (GOSH) between 2005 and 2016. Using EBV viral load (VL) measurements from weekly quantitative PCR (qPCR) monitoring post-HSCT, a multivariable Cox proportional hazards (Cox-PH) model was developed to assess time to first EBV reactivation event in the first 100 days post-HSCT. Sensitivity analysis of a previously reported mathematical model was performed to identify key parameters affecting EBV VL. Cox-PH modelling revealed EBV seropositivity of the HSCT recipient and administration of anti-thymocyte globulin (ATG) pre-HSCT to be significantly associated with an increased risk of EBV reactivation in the first 100 days post-HSCT (adjusted hazard ratio (AHR) = 2.32, P = 0.02; AHR = 2.55, P = 0.04). Five parameters were found to affect EBV VL in sensitivity analysis of the previously reported mathematical model. In conclusion, we have assessed the effect of multiple covariates on EBV reactivation in the first 100 days post-HSCT in children and have identified key parameters in a previously reported mechanistic mathematical model that affect EBV VL. Future work will aim to fit this model to patient EBV VLs, develop the model to account for interindividual variability and model the effect of clinically relevant covariates such as rituximab therapy and ATG on EBV VL.
... Naive B cells migrate to the mantle zone of the follicles below the epithelium and remain there for several days, and then return to the circulation. Infection of new naive B cells occurs in the intraepithelial layer, and not in the mantle zone [11], that is, when an infected B cell enters the follicle, it is already a blast and cannot migrate to the mantle zone. EBV infects cells through the interaction of viral glycoproteins gp350/220 with CD21 and gp42/gH/gL with class II HLA in a B cell. ...
Chapter
Full-text available
Infection of Epstein-Barr virus (EBV) is about 90% among people over the age of 40. The EBV causes a chronic infection that is characterized by chronic or recurrent symptoms and persists for a long time. Recombinant interferon-gamma (IFN-γ) has high clinical and antiviral efficacy in the treatment of herpesvirus infections. 110 patients with chronic EBV infection were examined. The patients were divided into three groups for different treatment regimens: Group 1—IFN-γ therapy (15 injections of Ingaron i/m, 500,000 IU every other day); Group 2—valaciclovir (Valtrex 500 mg × 2 times/day, orally for 2 months); Group 3—valganciclovir (Valcyte 450 mg × 2 times/day, orally for 2 months) and IFN-γ (10–20 injections of Ingaron i/m, 500,000 IU every other day). The best results were obtained in group 3–73.07% negative PCR. In this group, the combination of valganciclovir + IFN-γ was different. We showed that the efficacy of therapy in patients with chronic EBV is determined by the duration of INF-γ administration. We also determined spontaneous and induced production of IFN-α and -γ cytokines in serum and in lymphocyte culture. We demonstrated that in patients with an initially low level of induced IFN-γ, the production of this cytokine significantly increased in three months after the end of therapy.
Article
Full-text available
Chromatin accessibility fundamentally governs gene expression and biological response programs that can be manipulated by pathogens. Here we capture dynamic chromatin landscapes of individual B cells during Epstein-Barr virus (EBV) infection. EBV+ cells that exhibit arrest via antiviral sensing and proliferation-linked DNA damage experience global accessibility reduction. Proliferative EBV+ cells develop expression-linked architectures and motif accessibility profiles resembling in vivo germinal center (GC) phenotypes. Remarkably, EBV elicits dark zone (DZ), light zone (LZ), and post-GC B cell chromatin features despite BCL6 downregulation. Integration of single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq), single-cell RNA sequencing (scRNA-seq), and chromatin immunoprecipitation sequencing (ChIP-seq) data enables genome-wide cis-regulatory predictions implicating EBV nuclear antigens (EBNAs) in phenotype-specific control of GC B cell activation, survival, and immune evasion. Knockouts validate bioinformatically identified regulators (MEF2C and NFE2L2) of EBV-induced GC phenotypes and EBNA-associated loci that regulate gene expression (CD274/PD-L1). These data and methods can inform high-resolution investigations of EBV-host interactions, B cell fates, and virus-mediated lymphomagenesis.
Article
Epstein-Barr virus (EBV) is an usually harmless virus whose oncogenic properties in vitro are related to its ability to transform lymphoid cells, and, in consequence, it can be associated with lymphomas. Since a few studies detected EBV presence in supposedly EBV-negative lymphomas, our aim was to evaluate EBV presence by sensitive gene expression assays in the tonsils from healthy pediatric donors from a region with high incidence of EBV-associated lymphomas. EBERs transcripts were detected by View RNA ISH in all cases, even in cases assessed negative by widely used in situ hybridization. The presence of LMP1 transcripts was proved in 93% of cases, co-expressed with EBNA2 in 30%. In this study, evidence for the expression of different latent and lytic viral genes in a population of young age of primary infection, detected with more sensitive methods, in particular at the germinal center, where most EBV-associated lymphomas originate, was provided.
Article
Introduction: Angioimmunoblastic T-cell lymphoma (AITL) is an aggressive T-cell lymphoma commonly associated with B-cell dysregulation. Correlations involving B-cell dysregulation and clinicopathological features remain unclear. Methods: We prospectively collected blood samples from 11 AITL patients and 17 healthy controls. The percentages of B-cell subpopulations and lymphocytes with IL-21 production were assessed using flow cytometry. Peripheral blood lymphocyte morphology was evaluated microscopically. Results: Six of 11 (54.5%) patients presented with polyclonal hypergammaglobulinemia. Three of 11 (27.3%) tumor biopsies showed monoclonal immunoglobulin gene rearrangement. The patients exhibited significantly lower levels of naive (p < 0.001) and class-switched (p < 0.001) B cells than controls. The percentages of IgD-CD27- B cells (p = 0.007) and antibody-secreting cells (ASCs) (p = 0.001) were increased. Blood smears revealed atypical lymphocytes and immature plasma cells with morphological diversity. In comparison to normal controls, IL-21 production significantly increased in CD4+ (p < 0.001) and CD8+ (p = 0.020) T cells. B-cell clonality, RHOA G17V mutation, and the presence of sheets of clear cells and immature/mature plasma cells in lymph nodes were significantly associated with percentages of class-switched B cells and ASCs. The patients with circulating EBV DNA had a lower percentage of naive B cells (p = 0.009). Conclusions: Our results demonstrated a wide spectrum of peripheral B-cell morphologies and immunophenotypes of peripheral B cells in AITL. These findings correspond to dysregulated B-cell immunity and heterogeneous clinicopathological features.
Article
Full-text available
Human B cells encompass functionally diverse lineages and phenotypic states that contribute to protective as well as pathogenic responses. Epstein-Barr virus (EBV) provides a unique lens for studying heterogeneous B cell responses, given its adaptation to manipulate intrinsic cell programming. EBV promotes the activation, proliferation, and eventual outgrowth of host B cells as immortalized lymphoblastoid cell lines (LCLs) in vitro, which provide a foundational model of viral latency and lymphomagenesis. Although cellular responses and outcomes of infection can vary significantly within populations, investigations that capture genome-wide perspectives of this variation at single-cell resolution are in nascent stages. We have recently used single-cell approaches to identify EBV-mediated B cell heterogeneity in de novo infection and within LCLs, underscoring the dynamic and complex qualities of latent infection rather than a singular, static infection state. Here, we expand upon these findings with functional characterizations of EBV-induced dynamic phenotypes that mimic B cell immune responses. We found that distinct subpopulations isolated from LCLs could completely reconstitute the full phenotypic spectrum of their parental lines. In conjunction with conserved patterns of cell state diversity identified within scRNA-seq data, these data support a model in which EBV continuously drives recurrent B cell entry, progression through, and egress from the Germinal Center (GC) reaction. This “perpetual GC” also generates tangent cell fate trajectories including terminal plasmablast differentiation, which constitutes a replicative cul-de-sac for EBV from which lytic reactivation provides escape. Furthermore, we found that both established EBV latency and de novo infection support the development of cells with features of atypical memory B cells, which have been broadly associated with autoimmune disorders. Treatment of LCLs with TLR7 agonist or IL-21 was sufficient to generate an increased frequency of IgD-/CD27-/CD23-/CD38+/CD138+ plasmablasts. Separately, de novo EBV infection led to the development of CXCR3+/CD11c+/FCRL4+ B cells within days, providing evidence for possible T cell-independent origins of a recently described EBV-associated neuroinvasive CXCR3+ B cell subset in patients with multiple sclerosis. Collectively, this work reveals unexpected virus-driven complexity across infected cell populations and highlights potential roles of EBV in mediating or priming foundational aspects of virus-associated immune cell dysfunction in disease.
Article
Full-text available
Infection of inbred mice with murine gammaherpesvirus 68 (MHV68) has proven to be a powerful tool to study gammaherpesvirus pathogenesis. However, one of the limitations of this system has been the inability to directly detect infected cells harvested from infected animals. To address this issue, we generated a transgenic virus that expresses the enhanced yellow fluorescent protein (YFP), driven by the human cytomegalovirus immediate-early promoter and enhancer, from a neutral locus within the viral genome. This virus, MHV68-YFP, replicated and established latency as efficiently as did the wild-type virus. During the early phase of viral latency, MHV68-YFP efficiently marked latently infected cells in the spleen after intranasal inoculation. Staining splenocytes for expression of various surface markers demonstrated the presence of MHV68 in distinct populations of splenic B cells harboring MHV68. Notably, these analyses also revealed that markers used to discriminate between newly formed, follicular and marginal zone B cells may not be reliable for phenotyping B cells harboring MHV68 since virus infection appears to modulate cell surface expression levels of CD21 and CD23. However, as expected, we observed that the overwhelming majority of latently infected B cells at the peak of latency exhibited a germinal center phenotype. These analyses also demonstrated that a significant percentage of MHV68-infected splenocytes at the peak of viral latency are plasma cells (ca. 15% at day 14 and ca. 8% at day 18). Notably, the frequency of virus-infected plasma cells correlated well with the frequency of splenocytes that spontaneously reactivate virus upon explant. Finally, we observed that the efficiency of marking latently infected B cells with the MHV68-YFP recombinant virus declined at later times postinfection, likely due to shut down of transgene expression, and indicating that the utility of this marking strategy is currently limited to the early stages of virus infection.
Article
Full-text available
The current model of Epstein-Barr virus (EBV) infection and persistence in vivo proposes that EBV uses the germinal center (the GC model) to establish a quiescent latent infection in otherwise-normal memory B cells. However, the evidence linking EBV-infected cells and the GC is only indirect and limited. Therefore, a key portion of the model, that EBV-infected cells physically reside and participate in GCs, has yet to be verified. Furthermore, recent experiments suggested that upon infection of GC cells the viral growth latency transcription program is dominant and GC functionality and phenotype are ablated, i.e., EBV infection is not consistent with GC function. In this study we show that in vivo, EBV-infected B cells in the tonsils retain expression of functional and phenotypic markers of GC cells, including bcl-6 and AID. Furthermore, these cells are physically located in the GC and express a restricted form of latency, the default latency program. Thus, the EBV default latency transcription program, unlike the growth latency program, is consistent with the retention of GC functionality in vivo. This work verifies key components of the GC model of EBV persistence and suggests that EBV and the GC can interact to produce the latently infected memory cells found in the periphery. Furthermore, it identifies latently infected GC B cells as a potential pathogenic nexus for the development of the EBV-positive, GC-associated lymphomas Hodgkin's disease and Burkitt's lymphoma.
Article
We have developed an approach to identify microRNAs ( miRNAs) that is based on bioinformatics and array-based technologies, without the use of cDNA cloning. The approach, designed for use on genomes of small size (< 2 Mb), was tested on cells infected by either of two lymphotropic herpesviruses, KSHV and EBV. The viral genomes were scanned computationally for pre-miRNAs using an algorithm (VMir) we have developed. Candidate hairpins suggested by this analysis were then synthesized as oligonucleotides on microarrays, and the arrays were hybridized with small RNAs from infected cells. Candidate miRNAs that scored positive on the arrays were then subjected to confirmatory Northern blot analysis. Using this approach, 10 of the known KSHV pre-miRNAs were identified, as well as a novel pre-miRNA that had earlier escaped detection. This method also led to the identification of seven new EBV-encoded pre-miRNAs; by using additional computational approaches, we identified a total of 18 new EBV pre-miRNAs that produce 22 mature miRNA molecules, thereby more than quadrupling the total number of hitherto known EBV miRNAs. The advantages and limitations of the approach are discussed.
Article
Nature Reviews Microbiology 6, 913–924 (2008), doi:10.1038/nrmicro2015 In the above article, the term E3 ubiquitin-protein ligase was incorrectly added after CDKN2A. On page 918, in Box 3, the following sentence is incorrect: When MYC is overexpressed, the tumour suppressor p53 is activated, predominantly through CDKN2A (E3 ubiquitin-protein ligase; also known as p14ARF), which in turn interacts with and antagonizes MDM2, the negative regulator of p53 (Ref.
Article
Germinal centers develop in the B cell follicles of secondary lymphoid tissues during T cell-dependent (TD) antibody responses. The B cells that give rise to germinal centers initially have to be activated outside follicles, in the T cell-rich zones in association with interdigitating cells and T cell help. After immunization with a single dose of protein-based antigen, the germinal centers formed are oligoclonal; on average three B blasts colonize each follicle. These blasts undergo massive clonal expansion and activate a site-directed hypermutation mechanism that acts on their immunoglobulin-variable (Ig-v)-region genes. Mature germinal centers are divided into dark and light zones. The proliferating blasts, centroblasts, occupy the dark zone and give rise to centrocytes that are not in cell cycle and fill the light zone. The light zone contains a rich network of follicular dendritic cells (FDC) that have the capacity to take up antigen and hold this on their surface for periods of more than a year. The antigen is held as an immune complex in a native unprocessed form; but the antigen may be taken up from FDC by B cells, which can process this and present it to T cells. Centrocytes appear to be selected by their ability to interact with antigen held on FDC. There is a high death rate among centrocytes in vivo, and when these cells are isolated in vitro, they undergo apoptosis within hours on culture. The onset of apoptosis can be delayed by crosslinking centrocytes' surface Ig, and long-term survival is achieved by signalling through their surface CD40. After activation through CD40 the centrocytes increase their surface Ig and acquire characteristics of memory B cells. These observations suggest centrocyte selection involves uptake and processing of antigen held on FDC and its presentation to T cells that can be induced to express CD40 ligand at the point of cognate interaction. Other signals that induce a proportion of germinal center cells to become plasma cells have also been described. Germinal centers persist for about 3 weeks following immunization, but after this, memory B blasts continue to proliferate in follicles throughout the months of T cell-dependent antibody responses. These cells are probably the source of plasma cells and memory cells required to maintain long-term antibody production and memory after the first 3 weeks of T cell-dependent antibody responses.
Article
Using a set of surface markers including IgD and CD38, human tonsillar B cells were classified into discrete subpopulations. Molecular and functional analysis allowed us to identify: i) two sets of naive B cells (Bm1 and Bm2); ii) germinal center founder cells (Bm2'); iii) an obscure population of germinal center B cells, displaying a high load of somatic mutations in IgV genes, C mu to C delta switch and preferential Ig lambda light chain usage: these cells may represent the precursors of normal and malignant IgD-secreting plasma cells; iv) the centroblasts (Bm3) in which somatic mutation machinery is activated; v) the centrocytes (Bm4) in which isotype switch occurs; vi) the memory B cells. The characterization of these subpopulations showed that: i) programmed cell death is set before somatic mutations, possibly providing an efficient way for affinity maturation; ii) only high affinity centrocytes are allowed to switch isotype; iii) CD40-ligation inhibits plasmacytic differentiation of mature B lymphocytes; iv) memory B cells preferentially differentiate into plasma cells; v) IgD isotype switch occurs in normal B cells; vi) receptor editing may be induced by somatic mutations in germinal centers. We also characterized two types of antigen-presenting cells in germinal centers: follicular dendritic cells that select high affinity B cells, and a new subset of germinal center dendritic cells that activate germinal center T cells.
Article
The production of a mouse monoclonal antibody, Ki-67, is described. The Ki-67 antibody recognized a nuclear antigen present in proliferating cells, but absent in resting cells. Immunostainings with Ki-67 revealed nuclear reactivity in cells of germinal centres of cortical follicles, cortical thymocytes, neck cells of gastrointestinal mucosa, un-differentiated spermatogonia and cells of a number of human cell lines. The Ki-67 antibody did not react with cells known to be in a resting stage, such as lymphocytes, monocytes, parietal cells and Paneth's cells of gastrointestinal mucosa, hepatocytes, renal cells, mature sperm cells, brain cells, etc. Expression of the antigen recognized by Ki-67 could be induced in peripheral blood lymphocytes after stimulation with phytohaemagglutinin, whereas it disappeared from HL-60 cells stimulated with phorbol esters to differentiate into mature macrophages in a resting stage. These findings suggest that Ki-67 is directed against a nuclear antigen associated with cell proliferation. A first series of immunostainings of tumour biopsies indicated that Ki-67 may be a potent tool for easy and quick evaluation of the proportion of proliferating cells in a tumour.
Article
B cells undergo selection within germinal centers on the basis of their capacity to be activated by antigen held on follicular dendritic cells. Isolated germinal center B cells in culture kill themselves by apoptosis but this is prevented if their receptors for antigen are cross‐linked. In this study it is confirmed that almost all germinal center B cells, unlike other B cells, do not express the 25‐kDa protein encoded by the bcl‐2 oncogene. Cross‐linking the surface Ig of isolated germinal center cells causes them to express bcl‐2 protein. Two other stimuli which inhibit the entry of germinal center cells to apoptosis result in the expression of bcl‐2 protein. These stimuli are: (a) CD40 antibody and (b) recombinant 25‐kDa fragment of the CD23 protein plus recombinant interleukin 1α. Respectively, these induce germinal center cells to differentiate to resting B cells or plasmablasts. Dual‐fluorescence studies on small lymphocytes confirm the presence of bcl‐2 protein in mitochondria but show that this is also present in other extra‐nuclear areas. Burkitt lymphoma cells have a phenotype which indicates that they are neoplastic cells of germinal center origin. The expression of bcl‐2 protein by Burkitt lymphoma lines was also studied. Burkitt lines which retain the phenotype of fresh Burkitt lymphoma cells can be induced to enter apoptosis on culture with the Ca ²⁺ ionophore ionomycin. These cells were found not to express bcl‐2 protein. By contrast, Burkitt lines which have drifted towards a lymphoblastoid cell line phenotype and are resistant to the induction of apoptosis express high levels of the bcl‐2 protein. The findings support the concept that the susceptibility of germinal center cells to entering apoptosis is associated with their lack of expression of bcl‐2 protein. Aberrant expression of bcl‐2 protein by some neoplastic germinal center cells may allow survival in situations where their normal counterparts die.