ArticlePDF Available

The efficiency of B cell receptor (BCR) editing is dependent on BCR light chain rearrangement status

Authors:
  • Israeli Knesset, Jerusalem, Israel

Abstract and Figures

Anti-DNA knock-in mice serve as models for studying B cell tolerance mechanisms to a ubiquitous antigen. We have constructed six strains of double transgenic (C57BL/6xBALB/c)F1 mice, each expressing an unmutated or somatically mutated anti-DNA heavy (H) chain, combined with one of three different light (L) chains, namely V(kappa)1-J(kappa)1, V(kappa)4-J(kappa)4 and V(kappa)8-J(kappa)5. In vitro analysis of the various Ig H/L chain combinations showed that all had a similar specificity for single-stranded DNA and double-stranded DNA, but that antibodies encoded by the mutated H chain had higher affinities for the autoantigen. None of the targeted mouse strains exhibited significant levels of serum anti-DNA activity. However, while B cells from mice carrying the V(kappa)1-J(kappa)1 transgenic L chains were tolerized almost exclusively by L chain receptor editing in an affinity-independent manner, the mice expressing V(kappa)8-J(kappa)5 L chains have utilized affinity-dependent clonal anergy as their sole mechanism of B cell tolerance. V(kappa)4-J(kappa)4 targeted mice exhibited an intermediate phenotype with respect to these two mechanisms of B cell tolerance. Our results suggest that receptor editing is the preferred mechanism of B cell tolerance and that the efficiency of L chain editing is directly related to the number of available J(kappa) segments on the expressed V(kappa) allele.
Content may be subject to copyright.
0014-2980/02/0404-1164$17.50+ .50/0 © WILEY-VCH Verlag GmbH, D-69451 Weinheim, 2002
The efficiency of B cell receptor (BCR) editing is
dependent on BCR light chain rearrangement
status
Nurit Yachimovich, Gustavo Mostoslavsky, Yuval Yarkoni, Inna Verbovetski and
Dan Eilat
Department of Medicine, Hadassah University Hospital, Faculty of Medicine, Hebrew University,
Jerusalem, Israel
Anti-DNA knock-in mice serve as models for studying B cell tolerance mechanisms to a
ubiquitous antigen. We have constructed six strains of double transgenic (C57BL/6×BALB/
c)F1 mice, each expressing an unmutated or somatically mutated anti-DNA heavy (H) chain,
combined with one of three different light (L) chains, namely V
1-J
1, V
4-J
4andV
8-J
5. In
vitro analysis of the various Ig H/L chain combinations showed that all had a similar specific-
ity for single-stranded DNA and double-stranded DNA, but that antibodies encoded by the
mutated H chain had higher affinities for the autoantigen. None of the targeted mouse strains
exhibited significant levels of serum anti-DNA activity. However, while B cells from mice car-
rying the V
1-J
1 transgenic L chains were tolerized almost exclusively by L chain receptor
editing in an affinity-independent manner, the mice expressing V
8-J
5 L chains have utilized
affinity-dependent clonal anergy as their sole mechanism of B cell tolerance. V
4-J
4tar-
geted mice exhibited an intermediate phenotype with respect to these two mechanisms of B
cell tolerance. Our results suggest that receptor editing is the preferred mechanism of B cell
tolerance and that the efficiency of L chain editing is directly related to the number of avail-
able J
segments on the expressed V
allele.
Key words: Tolerance / Receptor editing / Anergy / Lupus / Transgenic mice
Received 14/9/01
Revised 13/12/01
Accepted 22/1/02
[I 22467]
Abbreviations: BCR: B cell receptor ds: Double stranded
ss: Single stranded NZB: New Zealand Black NZW: New
Zealand White ES: Embryonic stem CFSE: Carboxyfluo-
rescein diacetate succinimidyl ester RAG: Recombinase
activating gene
1 Introduction
B cell receptor (BCR) editing by secondary rearrange-
ment of immunoglobulin (Ig) genes has been identified in
recent years as a major mechanism of B cell tolerance to
self antigens [1–3]. However, its relation to other well-
known tolerance mechanisms, such as clonal deletion
[4, 5] and clonal anergy [6, 7] has not been well estab-
lished. For example, it is not known whether there is a
temporal relationship between receptor editing and other
tolerance mechanisms. Nor is it clear how the specificity
or affinity of the tolerogenic antigen influences the choice
of the appropriate tolerance mechanism.
We have previously established two transgenic mouse
lines, in which germ-line-encoded or somatically mu-
tated, rearranged heavy chain variable region genes,
derived from an anti-DNA hybridoma (D42) were targeted
to the IgH-chain locus of the mouse [8]. On a non-
autoimmune (C57BL/6×BALB/c)F1 mouse genetic back-
ground, these H-chain-only knock-in mice were shown
to be tolerant in that they did not produce significant
amounts of anti-DNA antibodies in their serum [8]. How-
ever, when transferred to the lupus-prone NZB/NZW F1
genetic background, the targeted D42H chain gave rise
to very high titers of DNA-binding antibodies due to
selection of particular Ig heavy/light chain combinations
with high affinity for double-stranded (ds) DNA [9]. In tol-
erant mice, a quantitative analysis of B cell populations
inthebonemarrow(BM)aswellasofJ
utilization and
DNA binding of hybridoma Ab suggested that immature
B cell deletion and L chain editing were the major toler-
ance mechanisms. Surprisingly, these mechanisms were
less effective in targeted mice expressing the somatically
mutated anti-DNA H chain (D42H), than in mice express-
ing the germ-line-encoded H chain (glD42H), possibly
due to the greater abundance of high affinity, anti-DNA B
cells in mice with the mutated D42H. Autoreactive B cells
in the periphery of D42H targeted mice exhibited several
features of functional inactivation (clonal anergy) [8].
1164 N. Yachimovich et al. Eur. J. Immunol. 2002. 32: 1164–1174
Fig. 1. DNA specificity of Ig H/L chain combinations. DNA
specificity of secreted transfectoma IgM antibodies was
evaluated in competition experiments, in which increasing
amounts of M13 phage ssDNA (squares) or dsDNA (circles)
were used as competitive inhibitors of E. coli [14C]dsDNA
binding to the individual purified antibodies in the nitrocellu-
lose filter assay. The amount of each antibody was adjusted
tobind50percentof25ng(1,500cpm)input[
14C]dsDNA in
the absence of inhibitor.
To gain a better understanding of the inter-relationships
between different tolerance mechanisms and their de-
pendence on the specificity and affinity for the tolerogen,
we have now constructed several new knock-in (C57BL/
6×BALB/c)F1 mouse lines with discrete Ig H/L chain
combinations. Specifically, the D42H and glD42H trans-
genic mice were crossed with knock-in mice expressing
V
1- J
1, V
4- J
4 [10] and V
8- J
5 [11] L chains. None of
the ensuing six lines of H/L chain double transgenic mice
produced DNA binding antibodies in their serum. A
detailed analysis of the various mechanisms of B cell tol-
erance in these mouse lines shows a hierarchy in their
ability to edit their BCR and suggests that the L chain
rearrangement status, rather than BCR affinity for the self
antigen determines the efficiency of receptor editing. In
contrast, non-editing self-reactive B cells are subjected
to clonal anergy and the intensity of their functional inac-
tivation is dependent on their BCR affinity for the self
antigen.
2 Results
2.1 Characterization of anti-DNA Ig H/L chain
combinations
Non-secreting NSO myeloma cells were stably co-
transfected with linearized plasmid vectors expressing
various combinations of the anti-DNA
?
HchainsD42Hor
glD42H [12] and the
L chains V
1-J
1 targeting vector
(this report), V
4- J
4 [13] and V
8-J
5 [14]. G418-
selected, cloned transfectomas were grown in serum-
free medium and the secreted antibodies were purified
by column chromatography and evaluated for DNA
specificity. Fig. 1 shows that, within the limits of experi-
mental error, the different
?
/
chain combinations had
the same specificity for single-stranded (ss) DNA and
dsDNA. In all cases, both ssDNA and dsDNA with the
same nucleotide sequence (M13 phage DNA) competed
with transfectoma antibodies for binding to E. coli
dsDNA in solution. These binding measurements in solu-
tion eliminate possible orientation effects that are likely
to occur when DNA is bound to solid-phase supports
(e.g. in ELISA). The results shown in Fig. 1 are consistent
with the notion [8] that the affinity and specificity of
D42H/L chain anti-DNA antibodies are primarily depen-
dent on the D42 heavy chain. Additionally, the various
?
transfectoma antibodies showed a very similar speckled
pattern in the fluorescent anti-nuclear Ab (FANA) test
(data not shown; see also Fig. 5C in [9]).
The apparent affinities of the various Ig
?
/
chain combi-
nations for dsDNA were measured by the nitrocellulose
filter assay and calculated from Scatchard plots [12].
Table 1 shows that the six transfected H/L chain combi-
nations may be divided into a lower affinity group com-
posed of the three L chains paired with the germ-line-
encoded D42H chain and a higher affinity group, com-
posed of the same L chains paired with mutated D42H.
Within each group, the difference in apparent affinity is
1.4–2.3-fold, while the two groups differ by a factor of
2–8. A similar H chain influence on antibody affinity is
evident in the
?
/
transfectoma derived from the original
NZB/NZW anti-DNA hybridoma, where the D42H/V
D42
H/L pair has about 12-fold higher affinity for dsDNA than
its unmutated counterpart (Table 1 and [12]). However,
the D42H/V
D42 chain combination that is representa-
tive of anti-DNA antibodies produced by NZB/NZW H
chain-only transgenic B cells [9] has 50–90-fold higher
affinity for DNA than the combination of the same H
chain with the three transfected L chains. Thus, the H/L
pairs in this study represent low to medium affinity anti-
DNA antibodies that are typically produced by H chain-
only non-autoimmune mice [9].
2.2 Anti-DNA activity in the serum of Ig H/L
double-transgenic mice
Six lines of anti-DNA knock-in mice, heterozygous for
both targeted heavy and light chains were constructed
by crossing glD42H or D42H single transgenic C57BL/6
mice with BALB/c mice, targeted with V
1-J
1, V
4-J
4or
V
8-J
5 L chains. All of the double transgenic mice pro-
duced low serum titers of dsDNA binding antibodies
(Fig. 2) that did not increase with age and were not sig-
nificantly different from those of non-transgenic or D42H
chain-only transgenic (C57BL/6×BALB/c)F1 mice. For
Eur. J. Immunol. 2002. 32: 1164–1174 B cell receptor editing and clonal anergy in anti-DNA transgenic mice 1165
Fig. 2. Anti-DNA activity in the serum of Ig H/L chain tar-
geted mice. Undiluted or sequentially diluted in borate-
buffered saline, heat-inactivated (56°C, 30 min) serum sam-
ples (10
?
l) were tested for dsDNA binding in the nitro-
cellulose filter assay. Input E. coli [14C]dsDNA was 25 ng
(1,500 cpm). Background radioactivity (with no added
serum) was subtracted from all binding values. Average
binding values are indicated for each mouse strain.
comparison, D42H chain-only female NZB/NZW F1 mice
had serum anti-DNA titers that were several orders of
magnitude higher than those of all single- and double-
transgenic non-autoimmune mice (Fig. 2).
2.3 BCR editing in anti-DNA Ig H/L chain double-
transgenic mice
The very low titers of DNA binding antibodies in the sera
of H/L chain targeted mice could be due to the relatively
low affinity of the anti-DNA antibodies produced in these
mice (Table 1) or to the induction of tolerance mecha-
nisms that operate on higher affinity autoreactive Bcells.
To test the latter possibility, the mechanism of receptor
editing by secondary rearrangements in Ig L chains was
evaluated by direct sequencing of expressed L chains in
LPS hybridomas obtained from spleen cells of the vari-
ous double transgenic mouse lines. Fig. 3 shows the
rearrangement status of
light chains obtained from
hybridomas produced from the different double trans-
genic mice. The vast majority of both low-affinity
glD42H/V
1-J
1 and medium-affinity D42H/V
1-J
1B
cells (100% and 89%, respectively) were shown to edit
Table 1. DNA binding affinity of Ig H/L chain combinations
H/L combination Apparent affinity (M–1/bp)
glD42H/V
1-J
14.6×10
6
D42H/V
1-J
11.1×10
7
glD42H/V
4-J
42.0×10
6
D42H/V
4-J
48.7×10
6
glD42H/V
8-J
53.3×10
6
D42H/V
8-J
51.6×10
7
glD42H/V
D42-J
56.5×10
7
D42H/V
D42-J
57.8×10
8
their
L chains by secondary rearrangements to down-
stream J
minigenes, particularly to J
5 (87% and 56%
for glD42H/V
1 and D42H/V
1, respectively). These sec-
ondary rearrangements were not biased towards any
particular V
segment and both J
-proximal and J
-distal
V
genes were identified in L chain cDNA (data not
shown). PCR analysis of
alleles in glD42H/V
1-J
1
hybridomas showed that 10 of 32 edited hybridomas
(31%) retained a germ-line configuration on the untar-
geted
allele, suggesting that in most of these B cells, L
chain editing involved both the transgenic and endoge-
nous
alleles (data not shown). Some of these hybrid-
omas (4/19) demonstrated a PCR product that could
reflect an RS-dependent C
deletion [15] of the original
transgene (not shown). It should be pointed out that the
percentage of non-rearranged endogenous
alleles is a
maximum estimate, since rearrangement by inversion to
any J
segment except J
1 could retain the PCR product
of the germ-line configuration.
In a sharp contrast to the almost complete L chain
receptor editing in mice expressing D42H/V
1BCR,the
overwhelming majority of B cells expressing glD42H/
V
8-J
5 and D42H/V
8-J
5 retained their original Ig H/L
chain combination (Fig. 3). PCR analysis of
alleles in
glD42H/V
8-J
5 hybridoma DNA samples indicated that
all (18/18) had retained the germ-line configuration on
the untargeted
allele (data not shown). None of these
hybridomas showed an RS-dependent C
deletion. In
transgenic mice expressing glD42H/V
4orD42H/V
4
BCR, the level of L chain receptor editing was intermedi-
ate between the D42/V
1 and D42V
8 knock-in mice
(Fig. 3). Of the V
4-J
4-expressing hybridomas, 40–60%
retained their original H/L chain combination. The rest
have edited their L chains, mostly to J
5. The great
majority of these secondary rearrangements appeared to
involve both
alleles, since only 3 of 16 originally
1166 N. Yachimovich et al. Eur. J. Immunol. 2002. 32: 1164–1174
Fig. 3. J
usage in hybridoma cells derived from Ig H/L chain
targeted mice. J
usage was determined by direct sequenc-
ing of hybridoma RNA following reverse transcription-PCR
amplification. Empty bars represent expression of the origi-
nal V
-J
transgenic L chain.
Table 2.
Q
light chain and
?
aexpression in splenic B cells of Ig H/L chain targeted mice
Mouse B cells/spleen
10–7)
Q
-expressing cells
(% of B cells)
?
a-expressing cells
(% of B cells)
glD42H/V
1-J
12.5±0.9(n=9) 5.1±1.2(n=7) 7.3±1.3(n=6)
D42H/V
1-J
11.4±0.7(n= 11) 6.9 ± 0.8 (n=8) 19.8±6.5(n=5)
glD42H/V
4-J
43.2±0.5(n=4) 3.2±1.0(n=4) 2.9±0.4(n=4)
D42H/V
4-J
41.7±0.7(n=7) 7.6±1.4(n=7) 14.9±2.7(n=5)
glD42H/V
8-J
55.0 ± 1.9 (n=12) 0.4 ± 0.2 (n=7) 1.4 ± 0.9 (n=9)
D42H/V
8-J
53.7 ± 2.1 (n=7) 0.2 ± 0.1 (n=4) 2.0 ± 1.6 (n=9)
expressing V
4-J
4 hybridomas have retained the
unrearranged germ-line configuration. Secondary
rearrangements apparently occurred also in V
4-J
4
hybridomas that continued to express the targeted
transgene, since 6/9 of these hybridomas had a non-
germ-line configuration on the unexpressed allele. As in
the case of V
1-J
1 hybridomas, no bias of particular V
genes in L chain secondary rearrangements was evident
in V
4-J
4 hybridomas (data not shown).
To explore further the difference between the various Ig
H/L chain transgenic mice in their capacity to induce
BCR editing, the relative number of splenic B cells
expressing
Q
L chains was investigated by FACS analy-
sis. Table 2 shows that V
1-J
1 and V
4-J
4knock-in
mice had similar levels (3.2–7.6%) of
Q
-expressing
splenic B cells. If correction is made for the number of B
cells per spleen, and assuming that
Q
-expressing cells
are not deleted, then there is little difference in the levels
of
Q
expression between B cells harboring low-affinity
glD42H combinations with V
1-J
1orV
4-J
4 and their
higher affinity D42H/L chain counterparts. Strikingly,
however, the number of
Q
-expressing cells among B cells
from mice transgenic for glD42H and D42H chain combi-
nations with V
8-J
5 was extremely low (Table 2), further
indicating that these cells did not edit their BCR L chains.
In a previous study [12] we showed that the targeted, b-
allotype D42H chain could be inactivated by specific
rearrangements of DHor VH-DHinto the H chain leader
intron, thereby activating the non-transgenic, endoge-
nous H chain allele that expresses the a-allotype H
chains. Although these secondary rearrangements are
made possible due to the non-physiological retention of
the heavy chain D region on the targeted allele, they may
nevertheless represent an ongoing editing process.
Indeed, the relative number of
?
a-expressing B cells in
mice targeted with V
8-J
5 H/L chain combinations was
very low compared with mice having the V
1-J
1 and
V
4-J
4 H/L combinations (Table 2). This difference still
holds when one corrects for the number of B cells per
spleen and assumes that
?
a-expressing B cells are not
deleted. The same proportions of
?
aand
?
bchains were
seen in the various hybridoma populations derived from
the corresponding transgenic mice, suggesting that LPS
hybridomas truly represent the spectrum of B cell popu-
lations in vivo. The lack of H chain editing in V
8-J
5-
expressing B cells is compatible with the very low levels
of
and
Q
L chain editing in mice harboring these trans-
genic B cells.
Finally, RAG mRNA levels were measured in bone mar-
row cells derived from the various Ig H/L chain knock-in
mice. RAG levels in targeted bone marrow B cells should
be roughly proportional to the rate of ongoing BCR edit-
ing, since both H and L chains were prerearranged in B
cells of these double-transgenic animals. Total RNA was
prepared, and levels of RAG-2 were determined by a
Eur. J. Immunol. 2002. 32: 1164–1174 B cell receptor editing and clonal anergy in anti-DNA transgenic mice 1167
Fig. 4. RAG-2 expression in bone marrow cells from Ig H/L
chain targeted mice. RAG-2 mRNA was measured by a
semiquantitative reverse transcription-PCR, followed by blot
hybridization with CD19 and RAG-2 DNA probes. The ratio
of CD19 to RAG-2 signal was determined by densitometry
after subtracting the individual backgrounds.
semiquantitative reverse transcription-PCR. As a control
for the B cell number and gel loading, samples were ana-
lyzed in parallel for CD19 expression (Fig. 4). Quanti-
tation of the radioactive signal revealed that RAG-2
expression levels were similar in non-transgenic (C57BL/
6×BALB/c)F1 mice and transgenic D42H/ V
1-J
1 and
D42H/ V
4-J
4 mice. In contrast, RAG-2 levels in D42H/
V
8-J
5 knock-in mice were tenfold lower (Fig. 4). These
results are consistent with the low rate of H and L chain
editing in V
8-J
5-expressing mice and suggest that V
8-
targeted mice are not using receptor editing as a major
mechanism of B cell tolerance.
2.4 B cell clonal anergy in anti-DNA Ig H/L chain
double-transgenic mice
Since Ig H/L chain targeted mice carrying the V
8-J
5
transgenic L chain did not seem to edit their BCR, other
mechanisms of immunological tolerance should be
responsible for the very low anti-DNA levels in their
serum. We, therefore, looked at different parameters of
clonal anergy in these knock-in mice. In particular, BCR
density [7, 8, 16] as well as B cell proliferation [17] follow-
ing LPS [7, 8] or anti-CD40 [18] stimulation were exam-
ined in these mice. Fig. 5A shows that the mean fluores-
cence intensity (MFI) of IgMb, which is a measure of BCR
density on the surface of B cells from transgenic mice
carrying the V
8-J
5 L chain, was reduced compared
with the IgMbsurface density of non-transgenic C57BL/6
B cells. However, the induction of clonal anergy, as rep-
resented by BCR density, was dependent on BCR affini-
ty for antigen. The MFI of IgMbon D42H/V
8-J
5-
expressing B cells was drastically reduced (
˚
sevenfold),
while the MFI of IgMbon glD42H/ V
8-J
5wasonly
slightly reduced (
˚
twofold), as compared with non-
transgenic C57BL/6 mice (Fig. 5A). Presumably, a rela-
tively small decrease in receptor density is sufficient to
anergize B cells that express low-affinity BCR. This situa-
tion is different from that of L chain editing (Fig. 3), where
secondary rearrangements were independent of BCR
affinity for antigen within the range of affinities measured
for anti-DNA antibodies in this study. The BCR densities
of D42H/V
4-J
4 and D42H/V
1-J
1 transgenic B cells
were also reduced (
˚
fourfold), although to a lesser
extent than that of D42H/V
8-J
5 (Fig. 5A), probably
because some of their edited H/L chain combinations
(estimated at
˚
30%, [8]) are still capable of binding DNA,
due to the dominance of the targeted H chain (Indeed,
the BCR densities of the editing H/L combinations were
similar to that of D42H chain-only transgenic mice, data
not shown). However, the difference between BCR den-
sities of mutated and non-mutated H chains in combina-
tion with editing V
4-J
4andV
1-J
1 was narrowed as
compared with the difference between D42H/V
8-J
5
and glD42H/V
8-J
5 (Fig. 5A). This is apparently because
the latter, non-editing combinations are fixed in their
anti-DNA affinities, while most of the editing L chains (in
combination with either glD42H or D42H) have given rise
to similar, non-DNA binding H/L chain combinations.
To further demonstrate features of clonal anergy and
BCR affinity dependence of anergy induction, splenic B
cells from the H/L chain knock-in mice were stimulated
in vitro for 3–4 days with either 1
?
g/ml LPS or anti-CD40
antibody and measured for proliferation as compared
with non-transgenic C57BL/6 B cells. Fig. 5B shows by
CFSE dilution [17] that the highest inhibition of cell divi-
sion was apparent in LPS-stimulated D42H/V
8-J
5
transgenic B cells, but not in glD42H/V
8-J
5 B cells.
Substantial differences in B cell proliferation were also
noted between the mutated and non-mutated D42H
chain combinations with V
4-J
4andV
1-J
1. Virtually
identical results of B cell proliferation were obtained fol-
lowing anti-CD40 antibody stimulation in vitro (data not
shown). Collectively, these data indicate that the medium
affinity D42H/V
8-J
5-expressing B cells are tolerized by
functional silencing (clonal anergy), while the effect of
this tolerance mechanism on the lower affinity glD42H/
V
8-J
5ismuchsmaller,suggestingthatthestrengthof
the inactivation signal is critically dependent on BCR
affinity for the tolerizing antigen.
2.5 Bone marrow B cell compartments in anti-
DNA Ig H/L chain double-transgenic mice
Table 3 shows the relative sizes of the major develop-
mental B cell compartments in the bone marrow in the
various strains of Ig H/L chain knock-in mice. It is evident
that the pro-B cell compartment has about the same rel-
ative size in all the transgenic mice tested. However, the
mice expressing H/L chain combinations with V
8-J
5
targeted L chain have substantially different proportions
1168 N. Yachimovich et al. Eur. J. Immunol. 2002. 32: 1164–1174
Fig. 5. B cell anergy in Ig H/L chain targeted mice. Splenic B cells from mice transgenic for V
1-J
1, V
4-J
4orV
8- J
5 L chains,
in combination with glD42H (thin line) or D42H (thick line) H chains were measured for BCR density (A), using a heavy chain anti-
mouse
?
bantibody in flow cytometric analysis. For comparison, non-transgenic
?
b-expressing C57BL/6 B cells were also ana-
lyzed. Splenic (B220+) B cells from the H/L double-transgenic mice were also studied for LPS-induced proliferation, as analyzed
by CFSE dilution (B). The MFI of
?
bBCR density (A) and the MFI of R1-gated, dividing cells and their percentage of total trans-
genic B cells (B) are given in the accompanying table. The results represent several experiments in which the same relative MFI
were obtained.
Table 3. Bone marrow B cell compartments in Ig H/L chain targeted mice
Pro-B Pre-B Immature B Mature B
Mouse n%a) %a) %a) %a)
B220lo CD43+B220lo CD43B220lo CD43B220hi CD43
IgMIgMIgM+IgM+
Non-transgenic 10 3.2 ± 2.1 66.2 ± 7.0 19.1 ± 7.3 11.5 ± 4.3
glD42H/V
1-J
1 6 5.3 ± 2.6 71.1 ± 11.9 10.4 ± 4.3 13.1 ± 10.3
D42H/V
1-J
1 6 3.0 ± 1.9 75.0 ± 11.1 13.1 ± 4.3 8.7 ± 16.0
glD42H/V
4-J
4 5 12.2 ± 3.6 63.4 ± 11.9 7.8 ± 2.9 16.5 ± 8.2
D42H/V
4-J
4 6 2.6 ± 0.6 76.2 ± 13.9 12.0 ± 5.9 9.0 ± 5.1
glD42H/V
8-J
5 10 3.2 ± 1.2 43.0 ± 7.9 21.4 ± 8.9 32.4 ± 13.0
D42H/V
8-J
5 6 2.9 ± 0.7 39.4 ± 6.2 18.3 ± 9.2 39.3 ± 12.2
a) Percent of B cells
Eur. J. Immunol. 2002. 32: 1164–1174 B cell receptor editing and clonal anergy in anti-DNA transgenic mice 1169
of pre-B, immature B and mature B cell compartments in
the bone marrow. In V
1-J
1- and V
4-J
4-expressing B
cells, there is a very high ratio of IgM-“pre-B” cells to
IgM+immature/mature B cells. This probably reflects the
fact that most of the cells that initially express the tar-
geted autoreactive BCR are induced to modulate their
BCR as a result of contact with antigen and assume a
pre-B-like phenotype to edit their receptors [19, 20]. It is
also consistent with the finding that B cells undergoing
editing are specifically delayed in the small pre-BII cell
compartment [21]. In contrast, the ratio of immature/
mature B cells to pre-B cells is reversed in the bone mar-
row of Ig H/L chain knock-in mice that express the V
8-
J
5 transgenic L chain. These anergized B cells would
readily enter the immature and mature developmental
stage and leave the bone marrow to enter peripheral
organs such as the spleen. Indeed, the total number of
splenic B lymphocytes, known to consist of immature
and mature B cells, was found to be higher in V
8-J
5-
expressing H/L chain mice, than in the corresponding
V
1-J
1orV
4-J
4 transgenic mice (Table 2). This sug-
gests that many of the latter B cells are lost through non-
productive secondary rearrangements.
3 Discussion
This study extends our investigation of B cell tolerance
mechanisms to a ubiquitous antigen (DNA) in normal and
autoimmune-prone animals. The anti-DNA H-chain-only
mice [8, 9] have the advantage that their targeted H
chains were allowed to combine with the complete,
native L chain repertoire of the mouse, presumably giv-
ing rise to a wide spectrum of anti-DNA antibody affini-
ties. However, one has to make several assumptions to
interpret the experimental results. These include [8] (i)
that the targeted H chains will combine with a large vari-
ety of L chains to give DNA binding BCR, and (ii) that the
dominance of the H chain in determining affinity for DNA
can result in higher affinity anti-DNA BCR for most H/L
chain combinations containing the mutated D42 H chain,
as compared with those having the germ-line-encoded
H chain. None of these assumptions have to be made in
the present study, where the targeted H/L pairs were
compared for their affinity and anti-DNA specificity. The
limitation of this study, however, is that very high affinity
anti-DNA H/L combinations, that are typical of lupus-
prone NZB/NZW F1 mice (Table 1) and are relatively rare
for a given anti-DNA H chain [9], were not included. This
would explain why clonal deletion, a B cell tolerance
mechanism that is probably responsible for direct elimi-
nation of the very-high-affinity anti-DNA B cells in the
normal animal [4, 8] is not demonstrated in this study,
except as an indirect consequence of receptor editing. In
contrast, the low- and medium-affinity anti-DNA trans-
genic mice that we have generated permit us to study
several important characteristics of two other well-
documented B cell tolerance mechanisms, clonal anergy
and receptor editing.
The relationship between editing and anergy comes to
focus in this study, because of the similarities in affinity
and specificity and the differences in rearrangement sta-
tus within the different H/L-targeted mice. Most striking
is the difference in the choice of tolerance mechanism by
anti-DNA transgenic mice carrying the V
1-J
1 and V
8-
J
5 L chains. The V
1-J
1 mouse strains edit the majority
of their L chains in an affinity-independent manner within
the range of DNA binding affinities measured in this
study. Receptor editing has been previously found to be
exquisitely sensitive to ultralow affinity by Lang et al. [22],
who studied anti-class I transgenic mice. The L chain
editing demonstrated by the anti-DNA V
1-J
1trans-
genic B cells is not limited to the targeted
allele which
can obviously utilize its remaining available J
segments
for secondary rearrangements, but is also evident on the
second
allele as well as on
Q
alleles.
In contrast to V
1-J
1 H/L chain double-transgenic mice,
the V
8-J
5 B cells of anti-DNA knock-in mice do not
show any tendency to edit their L chains, either at the
non-transgenic
or the
Q
alleles. Instead, clonal anergy
is the tolerance mechanism of choice, and unlike recep-
tor editing, this mechanism is affinity-dependent, as
demonstrated by the difference in the anergy parameters
that were measured (Fig. 5), between the higher affinity
D42H/V
8-J
5 and lower affinity glD42H/V
8-J
5 trans-
genic B cells. These results confirm our earlier work with
anti-DNA H chain-only knock-in mice [8], where D42H
transgenic B cells had lower receptor densities and were
less responsive to mitogenic stimulation than glD42H B
cells. The difference in the fate of V
8-J
5 and V
1-J
1
targeted B cells in this study is also reflected in the level
of RAG expression in the bone marrow (Fig. 4), and in the
much higher ratio of bone marrow immature/mature cells
to pre-B cells in the former B cell population (Table 3),
resulting in higher number of B cells in the spleen of mice
expressing the V
8-J
5 transgenic L chain (Table 2). The
higher number of splenic B cells may also reflect previ-
ous reports indicating that anergized anti-DNA B cells
are long lived [16, 23].
Casellas et al. [21], who studied knock-in mice express-
ing the same V
8-J
5 L chain and a different anti-DNA H
chain (3H9), observed little editing, if any, of the targeted
L chain allele, in full agreement with the results reported
here. However, when the same V
8-J
5 L chain was
expressed in combination with the native repertoire of
the mouse H chains, about 18% of B cells replaced the
targeted L chain. This may reflect a strong selection for a
1170 N. Yachimovich et al. Eur. J. Immunol. 2002. 32: 1164–1174
dominant, endogenous H chain which results in an H/L
pair with very high-affinity autoreactive specificity.
The anti-DNA Ig H/L chain mice expressing the V
4-J
4
transgenic L chain were perfectly capable of inducing
Q
L
chain rearrangement (Table 2), but their LPS hybridomas
showed an intermediate
chain editing efficiency
between V
1-J
1 and V
8-J
5-expressing anti-DNA
knock-in mice. This is evident in Fig. 3, where 40% and
60% of the glD42H/V
4-J
4 and D42H/V
4-J
4, respec-
tively, expressed their original Ig H/L chain combination,
as compared with only 10% or less of the V
1-J
1-
expressing hybridomas. Moreover, the great majority
(13/16) of the editing V
4-J
4 transgenic hybridomas
seem to have edited both their transgenic and endoge-
nous
allele. Interestingly, of the “non-edited” V
4-J
4-
expressing hybridomas (Fig. 3), the majority (6/9) had a
non-germ-line configuration on the unexpressed allele,
suggesting that secondary rearrangements may occur in
editing B cells, while the original H/L combination is still
expressed (see below).
The clear distinction between the two routes of tolerance
in V
1-J
1- and V
8-J
5-expressing anti-DNA knock-in
mice raises the question of what determines the choice
of B cell tolerance mechanism. It is clear from Table 1
and Fig. 1 that there is no fundamental difference in the
DNA affinity or specificity of Ig D42H or glD42H combi-
nations comprising the two L chains. Therefore,the DNA
antigen is not likely to make the choice between anergy
and editing. On the other hand, the two L chains differ in
their V-gene family and rearrangement status (J
1vs.J
5
rearrangement). It is hard to see why the difference in
V-gene family would determine the route of tolerance,
unless a tolerizing antigen other than DNA reacts differ-
ently with the two H/L chain combinations (see below).
Other considerations that might, in principle, affect the
efficiency of receptor editing, such as the number, dis-
tance and orientation of potentially rearranging V
genes
are not applicable in the targeted locus. This is because
the targeting procedure was identical for the two trans-
genic L chains and the full repertoire of endogenous V
genes remains intact in both models. The targeting pro-
cedure is also not likely to play a role since conventional
(non-targeted) transgenic B cells carrying the same V
8-
J
5 L chain also become anergic [16, 23]. Additionally,
we have searched the V
1andV
8 genes for cryptic
”heptamer-like” sequences that could potentially facili-
tate L chain inactivation through V-gene replacement
[24, 25]. A higher number of heptamer-like sequences
were identified in V
8 than in V
1, suggesting that such
mechanism is not likely to account for the difference in
editing between the two L chains. Also, we have not
identified in our sequenced hybridomas any functional
“hybrid” V
sequences, and this putative replacement
mechanism has not been identified in mouse L chains
[10, 11].
Our data, therefore, suggest that the choice between
BCR editing and clonal anergy (or deletion) is determined
by the rearrangement status of the
L chain. The
sequence of events that is consistent with the data
described in this report suggests the following princi-
ples: (i) receptor editing is the preferred mechanism of
tolerance due to its greater sensitivity to low affinity tole-
rogens. Autoreactive B cells that are incapable of recep-
tor editing would likely resort to other mechanisms of tol-
erance, such as clonal anergy. (ii) Allelic and isotype
exclusion is fully maintained during the process of recep-
tor editing, at least under the conditions (i.e. range of
antigen affinities) described in this study. Consequently,
the tolerized cell would express an edited L chain allele
only if the original
-expressing gene has been replaced
by a productive secondary rearrangement. Alternatively,
the endogenous
or
Q
alleles would be activated when
secondary rearrangement on the active allele is non-
productive, or due to RS-dependent C
deletion [15, 26]
or somatic mutation [27]. (iii) Secondary rearrangements
on the active
allele is the rate-limiting step in L chain
editing. The efficiency of rearrangement within a given
“window of opportunity”, should be directly proportional
to the number of chromatin accessible recombination
signal sequences (RSS) [28]. This number is dependent
on the available J
segments on the active
allele. Thus,
editing V
1-J
1 L chain has the maximal opportunity to
rearrange or inactivate the targeted allele within a given
time frame, resulting in over 90% of edited BCR in these
mice. V
4-J
4-targeted mice can only utilize J
5forL
chain secondary rearrangement on the active allele; con-
sequently, only half of the V
4-J
4 B cells express an
edited BCR. In the case of V
8-J
5 L chain, only RS-
dependent C
deletion would permit the expression of
edited
(or
Q
) alleles and this may be a very slow pro-
cess.
As discussed above, allelic and isotype exclusion is
maintained during the editing process, although second-
ary rearrangements may occur on both alleles. Indeed,
we found no evidence for the expression of more than
one H and one
chain in all of our sequenced hybrid-
omas. Likewise, in FACS analysis, no
/
Q
double produc-
ing splenic B cells could be identified. Allelic exclusion
has been reported to be violated in other anti-DNA trans-
genic mouse models that include (i) transgenic mice in
which conventional rather than targeted Ig H/L chains
were employed [13], (ii) transgenic Ig chains that were
backcrossed onto an autoimmune genetic background
[29], (iii) transgenic Ig chains with very high affinity for
DNA (e.g. 3H9 56R; [30]).
Eur. J. Immunol. 2002. 32: 1164–1174 B cell receptor editing and clonal anergy in anti-DNA transgenic mice 1171
There may be alternative explanations for the relative
inefficiency of autoreactive B cells expressing V
8-J
5in
editing their Ig chains. These include: (i) the possibility
that a productive rearrangement to J
5 may slow down
editing by limiting accessibility of the other L chain
alleles to the rearrangement machinery and by down
regulation of RAG enzymes [31]. (ii) A V
-J
5 productive
rearrangement may accelerate the transition of B cells to
a mature phenotype that is not amenable to induction of
receptor editing [1, 3]. Indeed, conventional transgenic
3H9H/V
8-J
5 anergic B cells were shown to be pheno-
typically mature and long-lived [23]. (iii) One has to con-
sider the possibility that D42H/V
8-J
5 B cells are anerg-
ized first by the tolerizing antigen and anergic cells are
refractory to editing. This possibility seems unlikely,
however, because receptor editing has been shown in
this study to be more sensitive to low-affinity tolerogens
than clonal anergy. Also, it has been reported recently
that anergic B cells in hen egg lysozyme Ig (Ig-HEL)/
soluble HEL double-transgenic mice show evidence of
receptor editing in vivo [32].
One remaining possibility is that one or more of the self
antigens that induce immunological tolerance in vivo are
different from DNA. To be consistent with the results
reported here, this would require that V
1-J
1-expressing
B cells be tolerized by one cross-reacting antigen, V
8-
J
5-expressing cells by a second antigen, and V
4-J
4-
expressing cells which show features of editing and
anergy be tolerized by both antigens. This is a rather
unlikely scenario, because all three H/L chain combina-
tions were shown to bind DNA with comparable specific-
ities, and at least in the case of V
8-J
5-expressing B
cells, the degree of tolerance induction can be directly
related to their affinity for DNA. The weight of the evi-
dence, therefore, suggests that L chain receptor editing
is the preferred mechanism of B cell tolerance and its
efficiency is dependent on the rearrangement status of
the expressed L chain. It should be noted, however, that
although receptor editing is clearly the preferred mecha-
nism of tolerance in these anti-DNA transgenic mice, this
study focuses on B cells specific for one particular auto-
antigen and with a rather limited window of affinities.
Studies of B cells specific for other self antigens are
needed to support a general conclusion regarding the
preferred use of receptor editing for induction of B cell
tolerance.
4 Materials and methods
4.1 Knock-in mice
C57BL/6 transgenic mice, heterozygous for D42H and
glD42H targeted heavy chains, were constructed by homol-
ogous recombination in ES cells as described previously [8].
BALB/c mice, heterozygous for the targeted L chains V
4-
J
4andV
8-J
5wereproducedinasimilarway[10].Togen-
erate V
1-J
1-targeted mice, a 10.5-kb BamHI fragment
containing the rearranged gene was cloned into a targeting
vector designed to allow the insertion of rearranged
genes
upstream of the J
region ([10], and M. Shannon, S. Ibrahim,
E. Luning Prak and M. Weigert, unpublished data). E14–1 ES
cells were transfected with the SalI linearized targeting vec-
tor. ES cells were subjected to positive-negative selection
using G418 and gancyclovir, respectively, and tested for
homologous recombination by PCR [10]. Targeted ES cells
were injected into blastocysts obtained from C57BL/6 preg-
nant females and these blastocysts were implanted into
pseudopregnant females. Chimeric animals were identified
by coat color. These chimeric mice were then mated with
C57BL/6 mice and agouti-colored offspring were screened
for the L-chain replacement by V
1-J
1 and gene targeting
PCR assays. Analysis of LPS hybridomas from heterozy-
gous V
1-J
1-targeted mice demonstrated that 82% of the
hybridoma clones retained the V
1-J
1 knock-in, 9% deleted
the knock-in, and 9% inverted the knock-in. In the course of
this analysis, a duplicated germ-line
locus was identified
by PCR and Southern blotting in the V
1-J
1 knock-in mice,
in addition to the targeted allele. The additional
germ-line
gene (
G
20 kb) does not interfere with the properly targeted
V
1-J
1 allele with respect to allelic exclusion (95%) or
receptor editing (M. Shannon and M. Weigert, unpublished
results). The presence of D42H, V
1L, V
4L and V
8L trans-
genesintailDNAwastestedbyPCR,usingDNAprimers
described previously [8, 10]. All transgenic and control mice
were bred and maintained at the SPF animal facility of the
Hebrew University Faculty of Medicine.
4.2 Transfection of myeloma cells with Ig H/L chain
combinations and purification of IgM antibodies
Linearized expression vectors containing D42H or glD42H
?
heavy chains [12] and V
1-J
1, V
4-J
4[13]orV
8-J
5[14]
light chains were used in different combinations to transfect
non-producer NSO myeloma cells, essentially as described
previously [12]. Following selection in 1.5 mg/ml active G418
and cell cloning, IgM-secreting transfectomas were grown in
serum-free, protein-free medium (Sigma Chemical Co., St.
Louis, MO; cat. no. S2897) and antibodies were isolated by
elution from a DE-52 column (Whatman, Maidstone, GB)
with 0.5 M NaCl in 0.05 M phosphate buffer pH 8.
4.3 Measurement of antibody binding affinity and
specificity
Antibody binding to DNA was assayed in solution by the
nitrocellulose filter assay [12, 32]. This method also mea-
sured DNA binding affinity at equilibrium, using 32P-labeled
plasmid DNA [12]. The relative specificities of anti-DNA anti-
bodies to dsDNA and ssDNA antigens were evaluated
1172 N. Yachimovich et al. Eur. J. Immunol. 2002. 32: 1164–1174
by competition experiments, in which single-stranded or
double-stranded M13 mp18 circular DNA species (New
England Biolabs, Beverly, MA) were used as competitive
inhibitors of antibody binding to E. coli [14C]dsDNA in the
nitrocellulose filter assay [33].
4.4 Flow cytometric analysis
Single-cell suspensions from bone marrow or spleen of
transgenic mice were stained with monoclonal or polyclonal
antibodies and analyzed by FACScan (Becton Dickinson,
San Jose, CA) using the “Lysis II” program. The antibodies
used for staining were the same as described previously [8].
Additional antibodies included biotin-conjugated goat anti-
mouse
chain (clone R33–18–10.1, obtained from Dr. Klaus
Rajewsky, Cologne, Germany), and FITC-conjugated goat
anti-
Q
(clone Ls 136, Southern Biotechnology, Birmingham,
AL).
4.5 PCR and sequence analysis of hybridoma
antibodies
Hybridomas were produced by fusion of BALB/c NSO mye-
loma cells with spleen cells from wild-type or transgenic
mice following a 3-day incubation with 40
?
g/ml LPS
(Sigma). Secretion of IgMaand IgMbantibodies in hybridoma
supernatants was measured with allotype-specific anti-
bodies by ELISA [8]. cDNA preparation and direct sequenc-
ing of hybridoma DNA was carried out as described previ-
ously [8]. Germ-line configuration of the untargeted
chain
allele was assayed by PCR, using a sense strand primer
upstream of J
1, 5’-TGTGACATGCTTCTAAAGCAAAAGA-3’
and an antisense strand primer downstream of J
1, 5’-CAA-
GATCTACCCTGCTTCTTTGAGCAT-3’. RS1-RS-dependent
C
deletion was determined by PCR using primers b and c
in [26].
4.6 Measurement of B cell proliferation
B cell proliferation following LPS stimulation was evaluated
by CFSE (Molecular probes, Leiden, The Netherlands) dilu-
tion, measured by flow cytometry [17].
4.7 Measurement of RAG-2 expression in bone marrow
cells
Total RNA was extracted from bone marrow cells and
reverse transcribed to cDNA using random primers and M-
MLV reverse transcriptase (Promega). RAG-2 mRNA levels
were quantitated as described previously [34], using CD19
as an internal control.
Acknowledgements: This study was supported by the
United-States Israel Binational Science Foundation, by the
Israel Science Foundation administered by the Israeli Acad-
emy of Science and Humanities and by the German-Israeli
Foundation for Scientific Research and Development. We
thank Drs. Martin Weigert and Eline Luning Prak for provid-
ing the L chain targeted mice and for their critical reading of
the manuscript, and Dr. Doron Melamed for his helpful com-
ments and his procedure for measurement of RAG expres-
sion.
References
1Nemazee, D. and Weigert, M., Revising B cell receptors. J. Exp.
Med. 2000. 5: 1813–1817.
2Nemazee, D., ReceptorselectioninBandTlymphocytes.Annu.
Rev. Immunol. 2000. 18: 19–51.
3Meffre, E., Casellas, R. and Nussenzweig, M. C., Antibody reg-
ulation of B cell development. Nat. Immunol. 2000. 1: 379–385.
4Chen, C., Nagy, Z., Radic, M. Z., Hardy, R. R., Huszar, D.,
Camper,S.A.andWeigert,M.,Thesiteandstageofanti-DNA
B cell deletion. Nature 1995. 373: 252–255.
5Nemazee, D. and Buerki, K., Clonal deletion of B lymphocytes
in a transgenic mouse bearing anti-MHC class I antibody genes.
Nature 1989. 337: 562–566.
6Nossal,G.J.V.,Clonal anergy of B cells: a flexible, reversible,
and quantitative concept. J. Exp. Med. 1996. 183: 1953–1956.
7Goodnow, C. C., Crosbie, J., Adelstein, S., Lavoie, T. B.,
Smith-Gill, S. J., Brink, R. A., Pritchard-Briscoe, H., Wother-
spoon. J. S., Loblay, R. H., Raphael, K., Trent, R. J. and Bas-
ten, A., Altered immunoglobulin expression and functional silenc-
ing of self-reactive B lymphocytes in transgenic mice. Nature
1988. 334: 676–682.
8Pewzner-Jung, Y., Friedmann, D., Sonoda, E., Jung. S.,
Rajewsky. K. and Eilat, D., B cell deletion, anergy, and receptor
editing in “knock-in” mice targeted with a germline-encoded or
somatically mutated anti-DNA heavy chain. J. Immunol. 1998.
161: 4634–4645.
9Friedmann, D., Yachimovich, N., Mostoslavsky, G., Pewzner-
Jung, Y., Ben-Yehuda, A., Rajewsky, K. and Eilat, D., Produc-
tion of high affinity autoantibodies in autoimmune New Zealand
black/New Zealand white F1 mice targeted with an anti-DNA
heavy chain. J. Immunol. 1999. 162: 4406–4416.
10 Luning Prak, E. and Weigert, M., Light chain replacement: a
newmodelforantibodygenerearrangement.J. Exp. Med. 1995.
182: 541–548.
11 Chen, C., Luning Prak, E. and Weigert, M., Editing disease-
associated autoantibodies. Immunity 1997. 6: 97–105.
12 Pewzner-Jung, Y., Simon, T. and Eilat, D., Structural elements
controlling anti-DNA antibody affinity and their relationship
to anti-phosphorylcholine activity. J. Immunol. 1996. 156:
3065–3073.
13 Gay, D., Saunders, T., Camper, S. and Weigert, M., Receptor
editing: An approach by autoreactive B cells to escape tolerance.
J. Exp. Med. 1993.177: 999–1008.
14 Carmack, C. E., Camper, S. A., Mackle, J. J., Gerhard, W. U.
and Weigert, M., Influence of a V
8Lchaintransgeneonen-
dogenous rearrangements and the immune response to the HA
(SB) determinant on influenza virus. J. Immunol. 1991. 147:
2024–2033.
Eur. J. Immunol. 2002. 32: 1164–1174 B cell receptor editing and clonal anergy in anti-DNA transgenic mice 1173
15 Moore, M., Durdik, W. J., Persiani, D. M. and Selsing, E., Dele-
tion of
chain constant region genes in mouse
Q
chain-producing
B cells involve intrachromosomal DNA recombinations similar to
V-J joining. Proc. Natl. Acad. Sci. USA 1985. 82: 6211–6215.
16 Nguyen, K., Mandik, L., Bui, A., Kavaler, J., Norvell, A., Mon-
roe, J., Roark, J. and Erikson, J., Characterization of anti-
single-stranded DNA B cells in a non-autoimmune background.
J. Immunol. 1997. 159: 2633–2644.
17 Lyons, A. B. and Parish, C. R., Determination of lymphocyte
division by flow cytometry. J. Immunol. Methods 1994. 171:
131–137.
18 Rolink, A. G., Andersson, J. and Melchers, F., Characterization
of immature B cells by a novel monoclonal antibody, by turnover
and by mitogen reactivity. Eur. J. Immunol. 1998. 28: 3738–3748.
19 Pelanda, R., Schwers, S., Sonoda, E., Torres R. M., Nemazee,
D. and Rajewsky, K., Receptor editing in a transgenic mouse
model: site, efficiency and role in B cell tolerance and antibody
diversification. Immunity 1997. 7: 765–775.
20 Yamagami, T., Boekel, E. T., Andersson, J., Rolink, A. and
Melchers, F., Frequencies of multiple IgL chain gene rearrange-
ments in single normal or
L chain-deficient B lineage cells.
Immunity 1999.11: 317–327.
21 Casellas, R., Yang Shih, T.A., Kleinwietfeld, M., Rakonjac, J.,
Nemazee, D., Rajewsky, K. and Nussenzweig, M. C., Contri-
bution of receptor editing to the antibody repertoire. Science
2001. 291: 1541–1544.
22 Lang, J., Jackson, M., Teyton, L., Brunmark, A., Kane, K. and
Nemazee, D., B cells are exquisitely sensitive to central toler-
ance and receptor editing induced by ultralow affinity, membrane
bound antigen. J. Exp. Med. 1996. 184: 1685–1697.
23 Noorchashm, H., Bui, A., Li, H. L., Eaton, A., Mandik-Nayak,
L., Sokol, C., Potts, K. M., Pur ´e, E. and Erikson, J., Character-
ization of anergic anti-DNA B cells: B cell anergy is a T cell-
independent and potentially reversible process. Int. Immunol.
1999. 11: 765–776.
24 Chen, C., Nagy, Z., Luning Prak, E. and Weigert, M., Immuno-
globulin heavy chain gene replacement: A mechanism for recep-
tor editing. Immunity 1995. 3: 747–755.
25 Wilson, P. C., Wilson, K., Liu, Y-J., Banchereau, J., Pascual, V.
and Capra, J. D., Receptor revision of immunoglobulin heavy
chain variable region genes in normal human B lymphocytes.
J. Exp. Med. 2000. 191: 1881–1894.
26 Retter,M.W.andNemazee,D.,Receptor editing occurs fre-
quently during normal B cell development. J. Exp. Med. 1998.
188: 1231–1238.
27 Brard, F., Shanon, M., Luning Prak, E., Litwin, S. and Weigert,
M., Somatic mutation and light chain rearrangement generate
autoimmunity in anti-single-stranded DNA transgenic MRL/lpr
mice. J. Exp. Med. 1999. 190: 691–704.
28 Roth, D. B. and Roth, S. Y., Unequal access: regulating V(D)J
recombination through chromatin remodeling. Cell 2000. 133:
699–702.
29 Spatz, L., Saenko V., Iliev, A., Jones, L., Geskin, L. and Dia-
mond, B., Light chain usage in anti-double-stranded DNA B cell
subsets: role in cell fate determination. J. Exp. Med. 1997. 185:
1317–1326.
30 Li, Y., Li, H. and Weigert, M., Autoreactive B cells in the marginal
zone that express dual receptors. J. Exp. Med.. 2002. 195:
181–188.
31 Schlissel, M. S. and Stanhope-Baker, P., Accessibility and the
developmental regulation of V(D)J recombination. Semin.
Immunol. 1997. 9: 161–170.
32 Tze, L. E., Baness, E. A., Hippen, K. L. and Behrens, T. W., Ig
light chain receptor editing in anergic B cells. J. Immunol. 2000.
165: 6796–6802.
33 Eilat, D. and Fischel, R., Recurrent utilization of genetic ele-
ments in V regions of anti nucleic acid antibodies from autoim-
mune mice. J. Immunol. 1991. 147: 361–368.
34 Melamed, D., Kench, J. A., Grabstein, K., Rolink, A. and
Nemazee, D., A functional B cell receptor transgene allows effi-
cient IL-7-Independent maturation of B cell precursors. J. Immu-
nol. 1997. 159: 1233–1239.
Correspondence: Dan Eilat, Department of Medicine,
Hadassah University Hospital, P.O. Box 12000, Jerusalem
91120, Israel
Fax: +972-2-6437775
e-mail: eilatd
md2.huji.ac.il
1174 N. Yachimovich et al. Eur. J. Immunol. 2002. 32: 1164–1174
... To examine further the correlation between L chain allelic inclusion and receptor editing, single B cells from the three groups of mice were tested for Jk usage. Nonediting B cells mostly use Jk1 or Jk2 for k-chain rearrangement, whereas editing B cells often shift to Jk4 or Jk5 usage (22,23). Fig. 2C shows that normal mice and wild-type NZB/NZW mice had a similar profile of Jk usage, with a higher proportion of Jk1 + Jk2 compared with Jk4 + Jk5 usage and a low level of L chain allelic inclusion. ...
... This example may represent a special case, in which k receptor editing was induced following l rearrangement as a result of autoreactivity (9); however, this process could not eliminate DNA binding because l genes are incapable of secondary rearrangements (17). The final outcome of this special situation is the survival of isotypically included B cells that retain their autoreactivity, similarly to autoreactive transgenic B cells in which the Vk gene is rearranged to Jk5, and, therefore, receptor editing is inhibited (23) and the cells resort to clonal anergy (21). However, in the majority of allelically included B cells employing the two k alleles, no restriction on editing both alleles should exist and, therefore, most of these B cells would lose their autoreactivity. ...
Article
L chain allelic inclusion has been proposed as a B cell tolerance mechanism in addition to clonal deletion, clonal anergy, and receptor editing. It is said to rescue autoreactive B cells from elimination by diluting out the self-reactive BCR through the expression of a second innocuous L chain. In autoimmune animals, such as lupus-prone mice, allelically included B cells could be activated and produce pathogenic autoantibodies. We have previously shown that anti-DNA hybridomas from diseased New Zealand Black/New Zealand White F1 mice exhibit nearly perfect allelic exclusion. In the current study, we have analyzed single B cells from these and from nonautoimmune mice. In addition, we have cloned and expressed the Ig variable regions of several L chain-included B cells in cell culture. We find that although the number of L chain-included B cells increases as a result of receptor editing, the majority of such cells do not retain an autoreactive HxL chain combination and, therefore, allelic inclusion in itself does not serve as a B cell tolerance mechanism in these autoimmune mice.
... Because we were interested in B cell tolerance mechanisms, namely, clonal deletion (1), clonal anergy (19), and receptor editing (20), and their apparent failure in autoimmune mice, we further generated several H3L double-knock-in mice on normal and B/W genetic backgrounds (21,22). These included the D42H transgene, combined with Vk1-Jk1, Vk4-Jk4, or Vk8-Jk5. ...
... On a normal mouse background, (C57BL/6 3 BALB/c)F 1 , we found that the doubletransgenic mice edited their L chains extensively and that the extent of receptor editing depended greatly on the availability of the unrearranged Jk segments on the targeted L chain allele. Thus, D42H/Vk1-Jk1 B cells, having three remaining unrearranged Jk segments, edited their L chain in almost every recovered hybridoma of LPS-activated spleen cells; D42H/Vk4-Jk4 B cells with only one remaining Jk were only partially edited; and D42H/Vk8-Jk5 B cells remained essentially unedited (21). Surprisingly, when backcrossed to the B/W autoimmune background, the doubletransgenic mice edited their L chains to the same extent as the normal mice (22). ...
Article
Systemic lupus erythematosus is considered a prototype of systemic autoimmune diseases; however, despite considerable advances in recent years in the understanding of basic mechanisms in immunology, little progress has been made in elucidating the etiology and pathogenesis of this disease. This even holds for inbred mice, such as the lupus-prone New Zealand Black/New Zealand White F(1) mice, which are all genetically programmed to develop lupus at a predetermined age. This frustrating state of affairs calls for a fundamental change in our scientific thinking and the opening of new directions in lupus research. In this study, we suggest that intrinsic B cell tolerance mechanisms are not grossly impaired in lupus-prone mice, but that an unusually strong positive selection event recruits a small number of autoreactive B cells to the germinal centers. This event could be facilitated by nucleic acid-protein complexes that are created by somatic changes in the susceptible animal.
... The CDR-L3 region is similarly diverse, although without the contribution from a D gene, the degree of variability is less. However, light chains can also be important for the binding specificity of antibodies; light chains are swapped during receptor editing to change the specificity of the antibody (5,6). Hence, the contribution of light chains to the antigen-binding sites must not be overlooked. ...
Article
Full-text available
Antibody variable regions are composed of a heavy and a light chain, and in humans, there are two light chain isotypes: kappa and lambda. Despite their importance in receptor editing, the light chain is often overlooked in the antibody literature, with the focus being on the heavy chain complementarity-determining region (CDR)-H3 region. In this paper, we set out to investigate the physicochemical and structural differences between human kappa and lambda light chain CDR regions. We constructed a dataset containing over 29,000 light chain variable region sequences from IgM-transcribing, newly formed B cells isolated from human bone marrow and peripheral blood. We also used a published human naïve dataset to investigate the CDR-H3 properties of heavy chains paired with kappa and lambda light chains and probed the Protein Data Bank to investigate the structural differences between kappa and lambda antibody CDR regions. We found that kappa and lambda light chains have very different CDR physicochemical and structural properties, whereas the heavy chains with which they are paired do not differ significantly. We also observed that the mean CDR3 N nucleotide addition in the kappa, lambda, and heavy chain gene rearrangements are correlated within donors but can differ between donors. This indicates that terminal deoxynucleotidyl transferase may work with differing efficiencies between different people but the same efficiency in the different classes of immunoglobulin chain within one person. We have observed large differences in the physicochemical and structural properties of kappa and lambda light chain CDR regions. This may reflect different roles in the humoral immune response.
... The VH125Tg/Vκ125 SD model lacks all downstream Jκ which necessitates chromatin remodeling for rearrangement on the other allele, and should thus represent a relatively stringent barrier to receptor editing. This is suggested by the finding that nearly all anti-DNA B cells undergo receptor editing when downstream Jκ2-5 are present on the targeted allele, but virtually no anti-DNA B cells undergo receptor editing when downstream Jκ are absent (60). Competition for survival factors and follicular entry are also relaxed in the VH125Tg/Vκ125 SD model, as the vast majority of developing B cells recognize insulin (Fig. 1C). ...
Article
Autoreactive B lymphocytes that commonly arise in the developing repertoire can be salvaged by receptor editing, a central tolerance mechanism that alters BCR specificity through continued L chain rearrangement. It is unknown whether autoantigens with weak cross-linking potential, such as insulin, elicit receptor editing, or whether this process is dysregulated in related autoimmunity. To resolve these issues, we developed an editing-competent model in which anti-insulin Vκ125 was targeted to the Igκ locus and paired with anti-insulin VH125Tg. Physiologic, circulating insulin increased RAG-2 expression and was associated with BCR replacement that eliminated autoantigen recognition in a proportion of developing anti-insulin B lymphocytes. The proportion of anti-insulin B cells that underwent receptor editing was reduced in the type 1 diabetes-prone NOD strain relative to a nonautoimmune strain. Resistance to editing was associated with increased surface IgM expression on immature (but not transitional or mature) anti-insulin B cells in the NOD strain. The actions of mAb123 on central tolerance were also investigated, because selective targeting of insulin-occupied BCR by mAb123 eliminates anti-insulin B lymphocytes and prevents type 1 diabetes. Autoantigen targeting by mAb123 increased RAG-2 expression and dramatically enhanced BCR replacement in newly developed B lymphocytes. Administering F(ab')2123 induced IgM downregulation and reduced the frequency of anti-insulin B lymphocytes within the polyclonal repertoire of VH125Tg/NOD mice, suggesting enhanced central tolerance by direct BCR interaction. These findings indicate that weak or faulty checkpoints for central tolerance can be overcome by autoantigen-specific immunomodulatory therapy.
... Such secondary rearrangements replace (genotypic editing) or outcompete (phenotypic editing) the primary light-chain rearrangements, thereby modifying autoreactive BCR specificity. [2][3][4] Another process, BCR revision, occurs in mature B cells and involves the modification of self-reactive heavy-chain V-D-J rearrangements by IGHV gene replacement. [5][6][7][8] Two different types of replacement have been described. ...
Article
Full-text available
In B cells, BcR immunoglobulin revision is a common route for modifying unwanted antibody specificities via a mechanism called VH replacement. This in vivo process, mostly affecting heavy-chain rearrangement, involves the replacement of all or part of a previously rearranged IGHV gene with another germline IGHV gene located upstream. Two different mechanisms of IGHV replacement have been reported: type 1, involving the RAG complex and requiring a FR3 internal recombination signal; and type 2, involving an unidentified mechanism different from that of type 1. In the case of light chain loci, BcR immunoglobulin editing ensures that a second V-J rearrangement occurs. This helps to maintain tolerance, by generating a novel BcR with a new antigenic specificity. We report that human B cells can, surprisingly, undergo type 2 replacement associated with kappa light chain rearrangements. The de novo IGKV-IGKJ products result from the partial replacement of a previously rearranged IGKV gene by a new germline IGKV gene, in-frame and without deletion or addition of nucleotides. There are wrcy/rgyw motifs at the "IGKV donor-IGKV recipient chimera junction" as described for type 2 IGHV replacement, but AID expression was not detected. This unusual mechanism of homologous recombination seems to be a variant of gene conversion-like recombination, which does not require AID. The recombination phenomenon described here provides new insight into IG locus recombination and BcR immunoglobulin repertoire diversity. This article is protected by copyright. All rights reserved.
... To directly test whether B cells harboring anti-insulin VH125 undergo receptor editing, VH125Tg mice were intercrossed with mice harboring Igκ-targeted Vκ transgenes to generate three lines, VH125Tg/Vκ1ki, VH125Tg/Vκ4ki, and VH125Tg/Vκ8ki. Vκ1ki, Vκ4ki, and Vκ8ki Tgs are useful in this regard because their ability to undergo receptor editing when paired with an anti-DNA IgH has been well examined in elegant studies on anti-DNA specific BCRs (150,199). In addition, a prior study in NOD found that VH125 can pair with closely related Vκ1-110 (Vκ1ki) and Vκ4-81 (Vκ4ki) genes (197). ...
Thesis
Les maladies auto-immunes, qui touchent plus de 5% de la population, sont induites par une perte de la tolérance aux antigènes du Soi. Ces pathologies, généralement multifactorielles, résultent de l’effet combiné de plusieurs allèles de susceptibilité et de différents facteurs environnementaux. Les agents infectieux ont été tout particulièrement incriminés, mais les mécanismes en jeu restent encore mal élucidés. Les lymphocytes B, qui jouent un rôle central dans la pathogénie de nombreuses maladies auto-immunes, sont susceptibles d’être activés selon différents mécanismes au cours d’un processus infectieux et cette activation peut englober des cellules autoréactives. On ne sait cependant pas si cette activation peut entraîner la production d’auto-anticorps pathogènes de forte affinité et d’isotype IgG à partir du pool de cellules productrices d’auto-anticorps naturels de faible affinité, qui sont présentes de façon constitutive dans le répertoire B de l’individu sain. Nous avons mis au point un nouveau modèle murin knock-in pour des lymphocytes B présentant une affinité intermédiaire pour leur auto-antigène, la protéine HEL2x mutée (Hen-Egg Lysozyme). Ce modèle autoréactif d’affinité intermédiaire SWHEL X HEL2x, élaboré sur un fond génétique non auto-immun, permet de suivre le processus de maturation d’affinité des cellules B anti-HEL en présence de leur auto-antigène HEL2x au cours de l’infection chronique par la bactérie Borrelia burgdorferi. L’infection induit au niveau ganglionnaire une prolifération ainsi qu’une activation lymphocytaire B incluant des cellules anergiques. Certains clones autoréactifs sont capables de gagner les centres germinatifs ganglionnaires, de commuter vers l’isotype IgG et présentent des mutations somatiques au niveau de la région variable de la chaîne lourde de leur immunoglobuline, dans la zone d’interaction avec HEL2x, indiquant un processus de sélection par l’auto-antigène. Malgré un taux augmenté d’auto-anticorps d’isotype IgM, ces animaux ne produisent pas de plasmocytes capables de sécréter des auto-anticorps d’isotype IgG. Nos observations suggèrent l’existence de mécanismes de tolérance périphérique intrinsèques mis en place en particulier au niveau du centre germinatif. Un premier point de contrôle va éliminer les lymphocytes B autoréactifs ayant commuté de classe et présentant des mutations somatiques leur conférant une affinité augmentée pour l’auto-antigène tandis qu’un second point de contrôle va empêcher la différenciation en plasmocytes IgG+. Chez l’individu non prédisposé génétiquement, des mécanismes pourraient ainsi permettre de prévenir le développement d’une auto-immunité pathogène au cours d’un épisode infectieux.
Article
Full-text available
Germinal centers (GCs) are the primary site at which clonal expansion and affinity maturation of B cells occur. B cells encounter antigen and receive T cell help in the GC light zone (LZ) and then migrate to the dark zone where they proliferate and undergo somatic mutation before cycling back to the LZ for further rounds of selection. Tolerance to autoantigens is frequently lost de novo as GC B cells undergo class switching and somatic mutation. This loss of tolerance is regulated by a variety of mechanisms including cell death, failure to compete for T cell help, and failure to differentiate into effector cells. Systemic lupus erythematosus (SLE) is characterized by loss of tolerance to nucleic acid antigens. While defects in tolerance occur in the naïve repertoire of SLE patients, pathogenic autoantibodies also arise in the GC by somatic mutation from non-autoreactive precursors. Several B cell defects contribute to the loss of GC tolerance in SLE, including polymorphisms of genes encoded by the Sle1 locus, excess TLR7 signaling, defects in FcRIIB expression, or defects of B cell apoptosis. Extrinsic soluble factors, such as Type-1 IFN and B cell-activating factor, or an increased number of T follicular helper cells in the GC also alter B cell-negative selection. Finally, defects in clearance of apoptotic debris within the GC result in BCR-mediated internalization of nucleic acid containing material and stimulation of autoantibody production by endosomal TLR-driven mechanisms.
Article
Full-text available
BAFF inhibition is a new B cell-directed therapeutic strategy for autoimmune disease. Our purpose was to analyze the effect of BAFF/APRIL availability on the naive and Ag-activated B cell repertoires in systemic lupus erythematosus, using the autoreactive germline D42 H chain (glD42H) site-directed transgenic NZB/W mouse. In this article, we show that the naive Vκ repertoire in both young and diseased glD42H NZB/W mice is dominated by five L chains that confer no or low-affinity polyreactivity. In contrast, glD42H B cells expressing L chains that confer high-affinity autoreactivity are mostly deleted before the mature B cell stage, but are positively selected and expanded in the germinal centers (GCs) as the mice age. Of these, the most abundant is VκRF (Vκ16-104*01), which is expressed by almost all IgG anti-DNA hybridomas derived from the glD42H mouse. Competition with nonautoreactive B cells or BAFF/APRIL inhibition significantly inhibited selection of glD42H B cells at the late transitional stage, with only subtle effects on the glD42H-associated L chain repertoire. However, glD42H/VκRF-encoded B cells were still vastly overrepresented in the GC, and serum IgG anti-DNA Abs arose with only a slight delay. Thus, although BAFF/APRIL inhibition increases the stringency of negative selection of the naive autoreactive B cell repertoire in NZB/W mice, it does not correct the major breach in B cell tolerance that occurs at the GC checkpoint.
Article
Random assembly of antibody variable (V), diversity (D) and joining (J) gene segments creates a vast repertoire of antigen receptors, including autoreactive ones. Three ways that are known to reduce autoreactivity in the B-cell compartment include clonal deletion, functional inactivation and receptor editing, a mechanism involving a change in antigen receptor specificity through continued V(D)J recombination. New data suggest that editing can efficiently eliminate autoreactivity, yet, in an autoimmune context, secondary antibody gene rearrangements might also contribute to autoimmunity.
Article
Full-text available
Immunological tolerance has been demonstrated in double-transgenic mice expressing the genes for a neo-self antigen, hen egg lysozyme, and a high affinity anti-lysozyme antibody. The majority of anti-lysozyme B-cells did not undergo clonal deletion, but were no longer able to secrete anti-lysozyme antibody and displayed markedly reduced levels of surface IgM while continuing to express high levels of surface IgD. These findings indicate that self tolerance may result from mechanisms other than clonal deletion, and are consistent with the hypothesis that IgD may have a unique role in B-cell tolerance.
Article
Full-text available
We isolated and characterized the germ-line counterpart of a DNA segment designated RS (for recombining sequence), that is frequently recombined in mouse lambda light chain-producing B lymphocytes. Using Southern blot analyses of myelomas and mouse-Chinese hamster fusion cell lines, we found that RS DNA sequences are located on mouse chromosome 6, evidently more than 15 kilobases downstream of the kappa light-chain locus. We find that a typical recognition site for Ig gene recombination is situated within germ-line RS sequences near the recombination points observed in at least two lambda chain-producing cell lines. This represents a complete and functional Ig recognition site that is not directly associated with Ig genes. We also characterized a recombined RS segment isolated from the cell line BM18-4.13.9. This recombined segment has a variable region kappa light chain gene (V kappa) joined directly to RS sequences. Our results suggest that the deletion of the kappa light chain constant region (C kappa) exon in many lambda chain-producing B cells is the result of RS recombination and that C kappa deletion may be mediated by the same processes as antibody gene V-J joining (J = joining segment gene). We discuss the potential biological significance of RS DNA recombination in B-cell maturation.
Article
Immunological tolerance has been demonstrated in double-transgenic mice expressing the genes for a neo-self antigen, hen egg lysozyme, and a high affinity anti-lysozyme antibody. The majority of anti-lysozyme B-cells did not undergo clonal deletion, but were no longer able to secrete anti-lysozyme antibody and displayed markedly reduced levels of surface IgM while continuing to express high levels of surface IgD. These findings indicate that self tolerance may result from mechanisms other than clonal deletion, and are consistent with the hypothesis that IgD may have a unique role in B-cell tolerance.
Article
To determine the fate of anti-DNA antibody-bearing B cells in normal mice, we generated transgenic mice bearing the heavy (H) and light (L) chain genes of a well-characterized anti-double-stranded DNA antibody. This antibody was originally isolated from a diseased MRL/lpr mouse and has characteristics common to spontaneously arising anti-DNA antibodies. Results show that the H/L transgene (tg) immunoglobulin receptor is not expressed by animals bearing both tgs, although single tg animals (H or L) express their transgenes. Young H/L tg animals express few B cells, whereas adult H/L tg animals maintain almost normal B cell numbers. Analysis of the immunoglobulin receptors used by adult B cells shows that all contain the tg H chain in association with endogenous L chains. These B cells transcribe the L tg as well as the rearranged endogenous L chain gene, and loss of endogenous L chain gene transcription results in resurrection of the 3H9 H/L tg product. Examination of the endogenous L chains used by these cells shows that they represent a highly restricted subset of V genes. Taken together, these data suggest that autoreactive transgenic B cells can rearrange endogenous L chain genes to alter surface receptors. Those L chains that compete successfully with the L tg for H chain binding, and that create a nonautoreactive receptor, allow the B cell to escape deletion. We suggest that this receptor editing is a mechanism used by immature autoreactive B cells to escape tolerance.
Article
To assess the sensitivity of B cell tolerance with respect to receptor/autoantigen affinity, we identified low affinity ligands to the 3-83 (anti-major histocompatibility complex class I) antibody and tested the ability of these ligands to induce central and peripheral tolerance in 3-83 transgenic mice. Several class I protein alloforms, including Kbm3 and Dk, showed remarkably low, but detectable, affinity to 3-83. The 3-83 antibody bound Kb with K lambda approximately 2 x 10(5) M-1 and bound 10-fold more weakly to the Kbm3 (K lambda approximately 2 x 10(4) M-1) and Dk antigens. Breeding 3-83 immunoglobulin transgenic mice with mice expressing these ultralow affinity Kbm3 and Dk ligands resulted in virtually complete deletion of the autoreactive B cells from the peripheral lymphoid tissues. These low affinity antigens also induced receptor editing, as measured by elevated RAG mRNA levels in the bone marrow and excess levels of id- variant B cells bearing lambda light chains in the spleen. Reactive class I antigens were also able to mediate deletion of mature B cells when injected into the peritoneal cavity of 3-83 transgenic mice. Although the highest affinity ligand, Kk, was consistently able to induce elimination of the 3-83 peritoneal B cells, the lower affinity ligands were only partially effective. These results demonstrate the remarkable sensitivity of the deletion and receptor-editing mechanisms in immature B cells, and may suggest a higher affinity threshold for deletion of peripheral, mature B cells.
Article
Receptor editing, clonal deletion, and anergy are the mechanisms by which B cells maintain tolerance to self antigens. To determine the extent to which receptor editing shapes the normal antibody repertoire, we generated an immunoglobulin κ polymorphism that facilitates the detection of editing of immunoglobulin light chains in vivo. We found that B cells are targeted for editing during a 2-hour delay in development at the pre-BII cell stage, and that about 25% of all antibody molecules are produced by gene replacement. These results suggest that receptor editing represents a major force in shaping the antibody repertoire.
Article
Two IgG anti-DNA and two IgG anti-RNA autoantibodies derived from lupus prone NZB/NZW F1 mice have been analyzed for their Ag fine specificities and for their H and L chain V-region sequences. A remarkable similarity of VH gene sequences with previously sequenced antinucleic acid autoantibodies (Eilat, D., D. M. Webster and A. R. Rees. J. Immunol. 141:1745, 1988) was noted. This finding indicates that a small number of unique VH genes is involved in this autoimmune response and that the sequences of these genes are correlated with the different specificities for the autoantigen. The VK sequences appeared, by computer search, to be selected nonrandomly, but their use was not restricted to autoantibodies. An additional striking feature was evident in the construction of the D region elements, giving rise to CDR3 peptides that can interact with DNA and RNA. These constructs probably include D-D fusion products, which are relatively rare in Ig rearrangements.
Article
A rearranged murine V kappa 8/J kappa 5 L chain gene that codes for the L chain of most antibodies generated in the primary response of BALB/c mice to the antigenic site, Sb, of the hemagglutinin (HA) molecule of influenza virus A/PR/8/34 (PR8) has been cloned. Three transgenic lines were generated by microinjecting the gene. Lines Ga and L each contain a single copy of the transgene whereas line Gb contains three complete copies. Mice of the Ga lineage showed increased V kappa 8-specific mRNA levels only in spleen, but not in nonlymphoid organs and therefore displayed apparently normal lymphoid-specific regulation of the Ig transgene. B cell hybridomas generated from these mice were analyzed for rearrangements of endogenous V kappa genes. Greater than 90% of the C kappa alleles were retained in germ-line configuration in the Ga line, compared with only 0 to 18% in the L line. Thus, a wide variation in the frequency of endogenous rearrangements is seen among mice of different lineages using the same transgene construct. None of more than 150 hybridomas derived from LPS-stimulated splenic B cells of Ga mice exhibited HA-binding activity although they expressed the transgene and, in most cases, excluded endogenous V kappa rearrangements. In contrast, a large fraction of hybridomas isolated after primary immunization with PR8 were HA(Sb)-specific. This indicated that the transgene was functional but formed HA-specific antibodies with a more restricted set of H chains than previously hypothesized. The primary anti-HA response to immunization with PR8 was diminished in all lines compared with normal mice except for a slightly accelerated but transient burst of anti-HA antibody formation in two out of three lines (Ga and Gb). This early response in G lineage mice was largely specific for HA(Sb) and thus appeared to be composed of transgene-expressing antibodies. No differences in serum titers were observed in the secondary anti-HA responses to booster inoculation with PR8 between transgenic and normal mice.
Article
B lymphocytes can be rendered specifically unresponsive to antigen by experimental manipulation in vivo and in vitro, but it remains unclear whether or not natural tolerance involves B-cell tolerance because B cells are controlled by T lymphocytes, and in their absence respond poorly to antigen (reviewed in ref. 7). In addition, autoantibody-producing cells can be found in normal mice and their formation is enhanced by B-cell mitogens such as lipopolysaccharides. We have studied B-cell tolerance in transgenic mice using genes for IgM anti-H-2k MHC class I antibody. In H-2d transgenic mice about 25-50% of the splenic B cells bear membrane immunoglobulin of this specificity, and abundant serum IgM encoded by the transgenes is produced. In contrast, H-2k x H-2d (H-2-d/k) transgenic mice lack B cells bearing the anti-H-2k idiotype and contain no detectable serum anti-H-2k antibody, suggesting that very large numbers of autospecific B cells can be controlled by clonal deletion.