You are on page 1of 7

Cutting Edge: Myosin 18A Is a Novel

Checkpoint Regulator in B Cell


Differentiation and Antibody-Mediated
Immunity
This information is current as
of November 14, 2021. Michael B. Cheung, Gospel Enyindah-Asonye, Ken Matsui,
Ivan Kosik, Nina Dvorina, William M. Baldwin III, Jonathan

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
W. Yewdell and Neetu Gupta
J Immunol 2021; 206:2521-2526; Prepublished online 17
May 2021;
doi: 10.4049/jimmunol.2100084
http://www.jimmunol.org/content/206/11/2521

Supplementary http://www.jimmunol.org/content/suppl/2021/05/17/jimmunol.210008
Material 4.DCSupplemental
References This article cites 33 articles, 11 of which you can access for free at:
http://www.jimmunol.org/content/206/11/2521.full#ref-list-1

Why The JI? Submit online.


• Rapid Reviews! 30 days* from submission to initial decision
• No Triage! Every submission reviewed by practicing scientists
• Fast Publication! 4 weeks from acceptance to publication
*average

Subscription Information about subscribing to The Journal of Immunology is online at:


http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2021 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Cutting Edge: Myosin 18A Is a Novel Checkpoint
Regulator in B Cell Differentiation and Antibody-
Mediated Immunity
Michael B. Cheung,* Gospel Enyindah-Asonye,* Ken Matsui,* Ivan Kosik,†
Nina Dvorina,* William M. Baldwin, III,* Jonathan W. Yewdell,† and Neetu Gupta*
We investigated the function of the newly discovered my- the proteins PSD95, Dlg1, and Zo-1), and an extended coiled-
osin family protein myosin 18A (Myo18A) in Ab-mediat- coil domain, the latter enabling homodimerization and interac-

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
ed immunity by generating B cellconditional Myo18A- tion with Myo2A bipolar filaments (12, 14). We previously
deficient mice. Myo18A deficiency led to expansion of reported that Myo18A is expressed in both precursor and mature
bone marrow progenitor B cells and mature B cells in B cells, and interacts with ezrin, Myo2A, and tyrosine-phosphor-
secondary lymphoid organs. Myo18A-deficient mice dis- ylated proteins (15), suggesting that it may regulate physiological
played serum IgM hyperglobulinemia and increased functions of B cells. In this work, we show that Myo18A is a
splenic IgM-secreting cells, with older mice switching to novel regulator that not only limits naive B cell and Ig levels, but
IgG1 hyperglobulinemia and autoantibody development. also restricts Ag-induced humoral immunity by restricting B cell
Immunization of Myo18A-deficient mice with inactivated differentiation into Ab-secreting cells (ASCs).
influenza virus led to development of more potent neu-
tralizing Abs against the major Ag hemagglutinin, associ-
ated with persistent accumulation of Ag-specific germinal Materials and Methods
center B cells and more Ag-specific bone marrow plasma Mice
cells. In vitro stimulation with TLR7 and BCR ligands Heterozygous Myo18A knockout first, conditional-ready mice (My-
revealed a greater ability of Myo18A-deficient B cells to o18AFRT/1) were developed with KOMP (University of California,
Davis) (MGI:4419827). Mice with floxed Myo18A alleles
differentiate into Ab-secreting cells, associated with higher (Myo18AFL/FL) were generated in house by crossing Myo18AFRT/1
AID and Blimp-1 expression. Overall, our study demon- with ACTB/FLPe mice (Jackson Laboratory) (16). B cellspecific con-
strates that Myo18A is a novel negative regulator of B ditional knockout mice (Myo18A BKO) were generated by crossing
cell homeostasis, differentiation, and humoral immunity. Myo18AFL/FL mice with Mb1Cre/1 mice (17), and have the genotype
The Journal of Immunology, 2021, 206: 25212526. Myo18AFL/FL Mb1Cre/1. Mb1Cre/1 mice, in which one Iga allele is
replaced with the gene for Cre-recombinase, served as controls in all
experiments. Experimental and control animals were not littermates or

T
cohoused in the same cage. Male and female mice aged 23 mo were
he B cell Ab response is tightly regulated to facilitate used for flow cytometry and immunization experiments and 6- to 8-
pathogen-specific immunity and prevent self-reactivi- mo-old mice for autoreactivity studies. All animal experiments were
ty. Membrane and actin cytoskeleton dynamics play approved by the Cleveland Clinic Institutional Animal Care and Use
Committee.
an important role in regulating B cell activation following
crosslinking of the BCR by Ag (14). Ezrin, a membrane-cy- Virus, immunization, and neutralization assay
toskeleton crosslinker, and myosin 2A (Myo2A), the only
Influenza A/Puerto Rico/8/1934 (PR8) (Mt. Sinai strain; H1N1) (18)
conventional class II myosin expressed in lymphocytes, regu-
was inactivated prior to immunization by exposure to UV. Mice were
late BCR clustering (4), signal transduction (57), B cell Ag immunized i.p. with 2,500 hemagglutination activating units of UV-
extraction, plasma cell (PC) differentiation, and humoral inactivated PR8 (UV-PR8) (18). Blood was collected from the tail
immune response (810). vein prior to and weekly following immunization, and blood, spleen,
The recently described class XVIII unconventional myosins and bone marrow were collected for analysis. Virus neutralization assay
are most closely related to Myo2A, and regulate important cellu- was performed as described (19). The frequency of infected cells was
normalized to a virus only control, and a nonlinear regression curve
lar processes in nonlymphoid cells (1113). Myosin 18A was generated using the dose-response inhibition model to calculate
(Myo18A) contains protein-protein interacting KE (lysine-glu- the serum dilution factor leading to half maximal infection (50% neu-
tamic acid)-rich and PDZ domains (domain contained within tralization dilution [ND50]), using Prism7 software (GraphPad).

*Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Address correspondence and reprint requests to Dr. Neetu Gupta, Cleveland Clinic, 9500
Cleveland, OH; and †Laboratory of Viral Diseases, National Institute of Allergy and Euclid Avenue, NE40, Cleveland, OH 44195. E-mail address: guptan@ccf.org
Infectious Diseases, National Institutes of Health, Bethesda, MD
The online version of this article contains supplemental material.
ORCIDs: 0000-0002-0021-3802 (M.B.C.) and 0000-0002-3826-1906 (J.W.Y.).
Abbreviations used in this article: ASC, Ab-secreting cell; GC, germinal center; LN, lymph
Received for publication January 28, 2021. Accepted for publication April 4, 2021. node; Myo18A, myosin 18A; Myo2A, myosin 2A; ND50, 50% neutralization dilution; PC,
plasma cell; PR8, influenza A/Puerto Rico/8/1934; UV-PR8, UV-inactivated PR8.
This work was supported by a National Institute of Allergy and Infectious Diseases, National
Institutes of Health grant (R21 AI117350) to N.G. Copyright © 2021 by The American Association of Immunologists, Inc. 0022-1767/21/$37.50

www.jimmunol.org/cgi/doi/10.4049/jimmunol.2100084
2522 CUTTING EDGE: Myo18A NEGATIVELY REGULATES B CELL IMMUNITY

Flow cytometry and Western blotting specific F(ab9)2 (Jackson ImmunoResearch Laboratories) for 48 h, and
Purified B and T cells were isolated using MACS Purification Kits (Mil- IgM1 ASCs quantified by ELISPOT assay. RNA was isolated from
tenyi Biotec) (Supplemental Table I). Western blotting for Myo18A naive and stimulated B cells, followed by cDNA synthesis, and quanti-
was performed as previously described (15). B cell progenitors and sub- tative RT-PCR performed using the PowerUp SYBR Green Master
sets were identified from single-cell suspensions of bone marrow, spleen, Mix (Applied Biosystems) on a QuantStudio 5 Real-Time PCR system
and draining lymph nodes (LNs) by staining with indicated Abs and (Applied Biosystems). Gene expression was analyzed using forward
LIVE/DEAD stain (Supplemental Table I). Activation markers were an- and reverse primers (Supplemental Table I) for Aicda (AID), Prdm1
alyzed on splenic CD191 B cells using specific Abs (Supplemental (Blimp-1), and Actb (b-actin) (Integrated DNA Technologies). Ex-
Table I). Cells were acquired with a BD LSR Fortessa Flow Cytometer pression was calculated by normalizing target gene expression to the
(BD Biosciences) and analyzed using FlowJo software (TreeStar) using housekeeping gene b-actin (DCT), and calculating the relative fold in-
established gating strategies (Supplemental Fig. 1AC). HA-specific B duction of stimulated cells versus naive B cells (2DDCt).
cells were identified using a PE-conjugated PR8 HA probe gifted by Dr.
Troy Randall (University of Alabama, Birmingham) (20). Statistical analysis
Statistical significance was determined by calculating p values by two-
ELISA and ELISPOT sided unpaired t test unless otherwise stated. Significant differences be-
ELISA was performed to quantify total IgM, IgG, and IgG subclass tween Mb1Cre/1 and Myo18A BKO mice are indicated as follows: *p <

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
(IgM, IgG, IgG1, IgG2b, IgG2c, or IgG3) using indicated reagents 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001. For all figures,
(Supplemental Table I) as previously described (7). HA-specific IgM mean ± SEM is shown, and each symbol indicates an individual biologi-
and IgG were measured using PR8 HA-coated ELISA plates (18). The cal replicate. In Fig. 4, statistically significant differences between mock,
area under the curve was calculated for each serum sample using Prism day 7, and day 28 UV-PR8immunized mice are indicated with a single
7 (GraphPad) software. IgM and IgG ASCs were quantified by ELI- dagger (†) for Mb1Cre/1 mice and a pound sign (#) for Myo18A BKO
SPOT as previously reported (7). mice as follows: †/#p < 0.05, ††/##p < 0.01, and †††/###p < 0.001. Data
were plotted and analyzed using Prism 7 software (GraphPad).
Autoantibody array
Serum samples from 6- to 8-mo-old animals were analyzed at the Uni-
versity of Texas Southwestern Medical Center Microarray Core using Results and Discussion
autoantigen microarray panel 1, profiling 95 autoantigens and eight in- Myo18A regulates B cell development
ternal controls. To quantify serum reactivity to autoantigens, the net
fluorescent intensity (NFI) of each Ag was calculated by subtracting the To investigate the function of Myo18A in B cells we developed
local background and negative control. The signal-to-noise ratio (SNR) Myo18AFRT/1 mice with one gene-trapped Myo18A allele (Fig.
was calculated by subtracting the median background from the median 1A). Breeding of heterozygous Myo18AFRT/1 mice revealed a
foreground and dividing by the SD of the background for each Ag. The sub-Mendelian ratio of approximately one-third fully wild-type
autoantibody score (Ab-score), calculated as log2([NFI  SNR] 1 1), is
depicted for both genotypes.
and two-third heterozygotes (Supplemental Table II). Genotyp-
ing of embryos showed that systemic genetic deletion of
In vitro differentiation and quantitative RT-PCR Myo18A was lethal by embryonic day 9.5 (Supplemental Table
Purified B cells were stimulated with 1 mg/ml Resiquimod (R848; In- II), consistent with reports in Drosophila (21, 22) and zebra fish
vivoGen) alone, or with 10 mg/ml goat anti-mouse IgM, m-chain (23, 24) in which Myo18A is required to maintain myofiber

FIGURE 1. B cellspecific deletion of Myo18A leads to expansion of bone marrow B cell progenitors and peripheral B cell subsets. (A) Strategy for develop-
ment of B cellspecific Myo18A conditional knockout (Myo18A BKO). (B) Lysates from Mb1Cre/1 and Myo18A BKO splenocytes, B cells, and T cells probed
for Myo18A and b-actin. (C) Bone marrow progenitor (Pro-Pre; B220loIgM), immature (Imm; B220loIgM1), and recirculating mature (Mat; B220hiIgM1) B
cell populations in 2- to 3-mo-old Mb1Cre/1 and Myo18A BKO mice. (DG) Number of splenocytes (D), CD191 B cells and CD31 T cells (E), splenic Fo
(CD191 CD93CD231CD21int), marginal zone (MZ; CD191CD93CD23CD21hi) and B1 B cells (CD191 CD93CD23CD21) (F), and CD51 B1a and
CD5 B1b cells (G) in naive 2- to 3-mo old Mb1Cre/1 and Myo18A BKO mice. (H and I) Number of LN cells (H) and B and T cells (I). n = 4 per genotype for
(C), and 5 per genotype for (D)(I). Representative data are shown from two to four experiments. *p < 0.05, **p < 0.01, ***p < 0.001.
The Journal of Immunology 2523

integrity during development. Myo18A BKO mice were devel- Mb1Cre/1 (Fig. 2A) but IgG was not (Fig. 2B). To investigate
oped as described in the Materials and Methods section (Fig. if higher IgM was due to altered splenic Ab secretion capacity
1A). B cell specificity of the Myo18A deletion was confirmed by in Myo18A BKO mice, we quantified Ab-secreting PCs by
immunoblotting of splenocytes, and purified B and T cells with flow cytometry and ELISPOT assay in naive 2- to 3-mo-old
an Ab specific to Myo18A (Fig. 1B). In humans, the 39 untrans- Mb1Cre/1 and Myo18A BKO mice. Myo18A BKO mice had
lated region of the MYO18A gene overlaps with the TIAF1 gene more PCs (Fig. 2C) and twice as many IgM-secreting cells in
(25). Although a homolog of TIAF1 has not been annotated in their spleens compared with Mb1Cre/1 mice (Fig. 2D). The
the mouse genome, it is important to note that our knockout number of IgG-secreting ASCs also trended higher in Myo18A
leaves the corresponding portion of the Myo18A gene intact. BKO mice compared with Mb1Cre/1 mice (Fig. 2E). In con-
Therefore, the effects described in this work are because of trast, bone marrow PCs (Fig. 2F) and IgM (Fig. 2G) and IgG
Myo18A deletion alone. Nonetheless, there is a potential for dif- (Fig. 2H) ASCs were similar in Mb1Cre/1 and Myo18A BKO
ferences between the regulation of and by Myo18A in mouse mice. These data demonstrate a higher overall Ab secretion ca-
and human B cells. As we have previously shown that B cell pro- pacity in Myo18A BKO mice, indicating that Myo18A regu-
genitors express Myo18A (15), we performed flow cytometry lates B cell differentiation into PCs in the spleens.

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
analysis of bone marrow cells from Mb1Cre/1 and Myo18A
BKO mice to investigate whether Myo18A deficiency affects B
cell development. Myo18A BKO mice had increased bone mar-
row propre-B cells, and immature B cells compared with
Mb1Cre/1 mice (Fig. 1C), whereas mature recirculating B cells
were not altered (Fig. 1C). The expansion of B cell progenitors
in the bone marrow suggests that Myo18A controls the earliest
stages of B cell development in a B cellintrinsic manner.
Others have reported a link between increased Myo18A expres-
sion in bone marrow stromal cells and enhanced hematopoietic
support (12). However, our data suggest a unique regulatory
role for Myo18A in B cell progenitors that is distinct from the
function attributed to it in bone marrow stromal cells. Cytokine
signals from the microenvironment, such as IL-7 and thymic
stromal lymphopoietin, and B cellintrinsic signals from the
pre-BCR and BCR are all important in B cell development (26)
and may be altered in Myo18A-deficient progenitors.

Myo18A regulates peripheral B cell homeostasis


We next evaluated the effect of Myo18A deletion on peripheral
B cell and other immune cell populations. Myo18A BKO mice
had a significant increase in the number of splenocytes com-
pared with Mb1Cre/1 controls (Fig. 1D) concomitant with ele-
vated B cells and no change in T cell numbers (Fig. 1E,
Supplemental Fig. 1D). The follicular (Fo) B cell subset was
significantly expanded in Myo18A BKO mice compared with
Mb1Cre/1 controls, whereas marginal zone B and total B1 B
cells were not affected (Fig. 1F). Within the B1 B cell subset,
however, the CD51 B1a population was expanded in Myo18A
BKO mice (Fig. 1G). Splenic B cells from Myo18A BKO mice
had similar levels of surface Igs (Supplemental Fig. 1E) and ac-
tivation markers MHC class II, CD69, CD44, and CD80
(Supplemental Fig. 1F). Myo18A BKO mice also had more
cells in their LNs compared with Mb1Cre/1 mice (Fig. 1H),
and the number of both B and T cells was increased in LNs of
Myo18A BKO mice (Fig. 1I). Elevated B cell numbers in both
spleens and LNs of Myo18A BKO mice suggests that Myo18A
expression in B cells regulates peripheral homeostasis, via higher
FIGURE 2. Deletion of Myo18A results in hyperglobulinemia. (A and B)
bone marrow B cell output or increased B cell entry or reten-
Serum concentration of total IgM (A) and IgG (B) in naive 2- to 3-mo-old
tion in secondary lymphoid organs. Mb1Cre/1 and Myo18A BKO mice. (C) Number of CD19B220loCD1381
splenic PCs. (D and E) ELISPOT assay for IgM1 (D) and IgG1 (E) ASCs
Hyperglobulinemia in Myo18A-deficient mice quantified as ASCs per million splenocytes. (F) Number of
To address if dysregulated numbers of peripheral B cells affect CD19B220loCD1381 bone marrow PCs. (G and H) ELISPOT assay for
IgM1 (G) and IgG1 (H) ASCs quantified as ASCs per million bone mar-
Ab production, we quantified serum Igs in Mb1Cre/1 and
row cells. n = 8 per genotype for (A) and (B), and 5 per genotype for
Myo18A BKO mice. Serum IgM was significantly elevated in (C)(H). Representative data are shown from two to five experiments. *p
naive 2- to 3-mo-old Myo18A BKO mice compared with < 0.05, **p < 0.01.
2524 CUTTING EDGE: Myo18A NEGATIVELY REGULATES B CELL IMMUNITY

Myo18A-deficiency increases the generation of autoantibodies autoantigen were increased (31, 32). Overt clinical signs of on-
We next examined if the hyperglobulinemia observed in 2- to going autoimmune or inflammatory disease, such as weight
3-mo-old mice persisted or worsened upon aging. Six-month- loss, increased dermatitis, or swelling of joints and limbs, were
old Myo18A BKO mice did not show a difference in serum not evident in the Myo18A BKO mice. Other pathological
IgM concentrations (Fig. 3A) but had a significant increase in symptoms of autoimmunity, such as glomerular inflammation
serum IgG compared with age-matched Mb1Cre/1 mice (Fig. and deposition of IgG were also not observed in the Myo18A
3B). Quantification of IgG subclasses by ELISA showed higher BKO mice. These data indicate that although the deletion of
IgG1 levels, with unaltered IgG2b, IgG2c, and IgG3 levels Myo18A leads to increased expansion and differentiation of
(Fig. 3C). Because naive Myo18A BKO mice have elevated T B1a B cells and increase in certain autoantibodies, the progres-
cells in their LNs, persistent B-T interactions may induce acti- sion to full blown autoimmune disease may require additional
vation signals that lead to class switching. An accumulation of hits in the Fo B cell and T cell compartments.
isotype switched B cells and IgG-secreting PCs may explain the
Myo18A deficiency elicits a more robust Ag-specific B cell Ab response
elevation in IgG levels in 6-mo-old Myo18A BKO mice. As el-
evated B cell numbers and serum IgG levels sometimes corre- Because the induction of Ab responses by Fo B cells are impor-

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
late with presence of autoreactive Abs, we employed an tant for neutralization and clearance of viruses, we examined the
autoantigen array to quantify Abs against 95 common self-anti- effect of Myo18A deficiency on Ag-specific immunity by immu-
gens in the sera of 6-mo-old Mb1Cre/1 and Myo18A BKO nizing Mb1Cre/1 and Myo18A BKO mice with UV-PR8. Com-
mice. Overall, Myo18A BKO mice had significantly higher pared with Mb1Cre/1, Myo18A BKO mice developed higher
concentrations of autoreactive Abs of both IgM (Supplemental serum anti-HA IgM (Fig. 4A), and anti-HA IgG (Fig. 4B). Day
Fig. 2A) and IgG (Supplemental Fig. 2B) isotype. IgM and 28 sera from Myo18A BKO mice blocked in vitro infection of
IgG Abs against a number of nuclear Ags including histone MDCK cells with live PR8 virus at higher serum dilutions than
H2B, citrullinated histones, Smith Ag D2 (Sm D2), nucleolin, sera from Mb1Cre/1, resulting in a significantly higher ND50 val-
ribosomal phosphoprotein P1, and proliferating-cell nuclear Ag ue (Fig. 4C). These data show that the deletion of Myo18A in B
were increased in Myo18A BKO mice (Fig. 3D, E). Autoreac- cells enables a stronger Ag-specific neutralizing Ab response to
tive Abs against the extracellular matrix components fibrinogen immunization. The greater neutralization capacity antiserum
S, citrullinated fibrinogen, and aggrecan were also significantly from immunized Myo18A BKO mice may result from a quanti-
higher in Myo18A BKO mice (Fig. 3D, 3E). B1a B cells not tative increase in HA-specific IgG, an increase in the affinity of
only make much of the natural IgM (27), but also express au- such Abs, and/or an increase in neutralizing Abs against other flu
toreactive BCRs (2830). Furthermore, B1a B cells can switch Ags. We next examined the cellular basis for the stronger anti-
from IgM- to IgG-expressing cells and differentiate into HA Ab response in Myo18A BKO mice by performing flow cy-
IgG-secreting cells (30, 31). Therefore, it is conceivable that ex- tometry analysis of spleen cells from mock-immunized mice that
pansion of CD51 B1a B cells in Myo18A BKO mice supports received PBS, or mice immunized with UV-PR8 for 7 or 28 d.
both, increase in serum IgM levels as well as IgM and IgG auto- In all three groups, Myo18A BKO mice had more splenocytes
antibodies to self-antigens. Although we did not observe higher than Mb1Cre/1 mice (Fig. 4D). HA-specific B cells were elevated
autoantibodies to chromatin and dsDNA in Myo18A BKO in UV-PR8immunized Myo18A BKO mice relative to
mice, circulating Abs to the lupus-associated Sm D2 Mb1Cre/1 mice on day 7, with further increase on day 28 (Fig.

FIGURE 3. Myo18A-deficiency leads to


development of self-reactive Abs. (AC) Se-
rum concentration of IgM (A), IgG (B),
and IgG subclasses IgG1, IgG2b, IgG2c,
and IgG3 (C) in 6- to 8-mo-old Mb1Cre/1
and Myo18A BKO mice. (D and E) Heat-
maps of normalized Ab-score data for
IgM- (D) and IgG-reactive (E) autoantigens
in Mb1Cre/1 and Myo18A BKO mice. n =
67 mice per genotype. Representative
data are shown from three experiments.
*p < 0.05, **p < 0.01, ***p < 0.001.
The Journal of Immunology 2525

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
FIGURE 4. Deletion of Myo18A promotes more robust flu virusspecific cellular and Ab response by enhancing B cell differentiation. Serum anti-PR8 HA
IgM (A) and IgG (B) in immunized Mb1Cre/1 and Myo18A BKO quantified as area under the curve. (C) Neutralization capacity of day 28 serum expressed as
ND50. (DG) Number of splenocytes (D), HA1CD191 B cells (E), HA1CD191GL71CD951 GC B cells (F), and HA1IgG1 ASCs (G) in Mb1Cre/1 and
Myo18A BKO mice immunized with UV-inactivated PR8. (HJ) Differentiation of B cells in vitro assessed by ELISPOT for IgM1 ASCs (H) and Blimp-1 (I)
and AID (J) gene expression, in 48-h cultures of R848 and anti-IgMstimulated B cells from Mb1Cre/1 and Myo18A BKO mice. n = 45 per genotype for
(A)(F), and 3 for (G)(J). Representative data are shown from one to three experiments. †p < 0.05, ††p < 0.01. ##p < 0.01, ###p < 0.001. *p < 0.05, **p <
0.01, **** p < 0.0001.

4E). Greater expansion of HA-specific germinal center (GC) B of B cells of both genotypes by R848; however, Myo18A-defi-
cells was observed in Myo18A BKO mice after 7 d compared cient B cells displayed greater fold induction relative to naive
with Mb1Cre/1 mice, and although Mb1Cre/1 GC B cells con- cells than did Mb1Cre/1 B cells (Fig. 4I). Simultaneous TLR7
tracted by 28 d, significantly higher numbers of HA1 GC B cells and BCR stimulation led to even greater expression of Blimp-1
persisted in Myo18A BKO spleens (Fig. 4F). Myo18A BKO but the expression was again significantly higher in Myo18A-
mice had significantly more HA-specific IgG1 ASCs in their deficient B cells (Fig. 4I). Similar results were obtained for the
bone marrow (Fig. 4G) than Mb1Cre/1 mice, indicating an im- GC differentiation transcription factor AID (Fig. 4J). These
proved long-lived PC response. Because naive Myo18A-deficient data demonstrate that Myo18A restricts B cell differentiation to
B cells do not exhibit pre-existing activation prior to immuniza- ASCs in a B cellintrinsic manner and that its deletion releases
tion (Supplemental Fig. 1E, 1F), the increase in Ag-specific GC B cells from this inhibition.
B cells suggests that either more B cells are activated postimmu- Unconventional myosin family proteins have a vast array of
nization and enter the GC reaction or there is greater expansion functions in a variety of immune and nonimmune cells (33). In
of B cells within the GC. Alternately, improved survival and per- this work, we report a novel role for the newest member of this
sistence of B cells during GC selection may explain the observed family, Myo18A in B cell development, homeostasis, and Ab-
increase in Ag-specific GC B cells in the later phase of the im- mediated immunity. Conditional genetic deletion of Myo18A in
mune response. B cells resulted in expansion of both developing and mature B
Myo18A-deficient B cells exhibit greater differentiation cells, hyperglobulinemia, autoantibody development, persistent
GC output, and stronger neutralizing Ab response to influenza A
To determine the mechanism underlying enhanced humoral virus. Taken together, our data demonstrate that Myo18A is a
immunity observed in UV-PR8immunized Myo18A BKO novel regulatory checkpoint protein whose absence leads to exag-
mice, we modeled the response of splenic Myo18A-deficient B gerated B cell differentiation and Ab responses.
cells to R848, a TLR7/8 agonist and influenza virus ssRNA
mimic, and anti-IgM, a surrogate for Ag in vitro. Increased
IgM1 ASCs were detected in B cells from Myo18A BKO mice Acknowledgments
compared with Mb1Cre/1 upon stimulation for 48 h with The authors acknowledge assistance from the University of Texas Southwestern
R848 alone as well as with R848 1 anti-IgM (Fig. 4H). To de- Microarray Core facility.
lineate the molecular basis of increased differentiation, we ana-
lyzed the expression of key transcription factors AID and
Blimp-1 in naive and R848 1 anti-IgM-stimulated B cells by Disclosures
quantitative RT-PCR. Blimp-1 was induced upon stimulation The authors have no financial conflicts of interest.
2526 CUTTING EDGE: Myo18A NEGATIVELY REGULATES B CELL IMMUNITY

18. Angeletti, D., J. S. Gibbs, M. Angel, I. Kosik, H. D. Hickman, G. M. Frank, S. R.


References Das, A. K. Wheatley, M. Prabhakaran, D. J. Leggat, et al. 2017. Defining B cell
1. Harwood, N. E., and F. D. Batista. 2010. Early events in B cell activation. Annu. immunodominance to viruses. Nat. Immunol. 18: 456463.
Rev. Immunol. 28: 185210. 19. Kosik, I., D. Angeletti, J. S. Gibbs, M. Angel, K. Takeda, M. Kosikova, V. Nair,
2. Treanor, B., D. Depoil, A. Gonzalez-Granja, P. Barral, M. Weber, O. Dushek, A. H. D. Hickman, H. Xie, C. B. Brooke, and J. W. Yewdell. 2019. Neuraminidase
Bruckbauer, and F. D. Batista. 2010. The membrane skeleton controls diffusion inhibition contributes to influenza A virus neutralization by anti-hemagglutinin
dynamics and signaling through the B cell receptor. Immunity 32: 187199. stem antibodies. J. Exp. Med. 216: 304316.
3. Tolar, P. 2017. Cytoskeletal control of B cell responses to antigens. Nat. Rev. Im- 20. Allie, S. R., J. E. Bradley, U. Mudunuru, M. D. Schultz, B. A. Graf, F. E. Lund,
munol. 17: 621634. and T. D. Randall. 2018. The establishment of resident memory B cells in the lung
4. Gupta, N., B. Wollscheid, J. D. Watts, B. Scheer, R. Aebersold, and A. L. DeFranco. requires local antigen encounter. Nat. Immunol. 20: 97108.
2006. Quantitative proteomic analysis of B cell lipid rafts reveals that ezrin regulates 21. Ivanov, A. I., A. C. Rovescalli, P. Pozzi, S. Yoo, B. Mozer, H. P. Li, S. H. Yu, H.
Higashida, V. Guo, M. Spencer, and M. Nirenberg. 2004. Genes required for Dro-
antigen receptor-mediated lipid raft dynamics. Nat. Immunol. 7: 625633.
5. Parameswaran, N., G. Enyindah-Asonye, N. Bagheri, N. B. Shah, and N. Gupta. sophila nervous system development identified by RNA interference. Proc. Natl.
Acad. Sci. USA 101: 1621616221.
2013. Spatial coupling of JNK activation to the B cell antigen receptor by tyrosine-
22. Bonn, B. R., A. Rudolf, C. Hornbruch-Freitag, G. Daum, J. Kuckwa, L. Kastl, D.
phosphorylated ezrin. J. Immunol. 190: 20172026.
Buttgereit, and R. Renkawitz-Pohl. 2013. Myosin heavy chain-like localizes at cell
6. Treanor, B., D. Depoil, A. Bruckbauer, and F. D. Batista. 2011. Dynamic cortical
contact sites during Drosophila myoblast fusion and interacts in vitro with Rolling
actin remodeling by ERM proteins controls BCR microcluster organization and in- pebbles 7. Exp. Cell Res. 319: 402416.
tegrity. J. Exp. Med. 208: 10551068. 23. Cao, J., S. Li, M. Shao, X. Cheng, Z. Xu, and D. Shi. 2014. The PDZ-containing
7. Pore, D., N. Parameswaran, K. Matsui, M. B. Stone, I. Saotome, A. I. McClatchey, unconventional myosin XVIIIA regulates embryonic muscle integrity in zebrafish.
S. L. Veatch, and N. Gupta. 2013. Ezrin tunes the magnitude of humoral immuni- J. Genet. Genomics 41: 417428.

Downloaded from http://www.jimmunol.org/ at Red de Bibliotecas del CSIC on November 14, 2021
ty. J. Immunol. 191: 40484058. 24. Cao, J. M., X. N. Cheng, S. Q. Li, S. Heller, Z. G. Xu, and D. L. Shi. 2016. Iden-
8. Vascotto, F., D. Lankar, G. Faure-Andre, P. Vargas, J. Diaz, D. Le Roux, M. I. Yuseff, tification of novel MYO18A interaction partners required for myoblast adhesion
J. B. Sibarita, M. Boes, G. Raposo, et al. 2007. The actin-based motor protein myosin and muscle integrity. Sci. Rep. 6: 36768.
II regulates MHC class II trafficking and BCR-driven antigen presentation. J. Cell Biol. 25. Chang, N. S., J. Mattison, H. Cao, N. Pratt, Y. Zhao, and C. Lee. 1998. Cloning
176: 10071019. and characterization of a novel transforming growth factor-beta1-induced TIAF1
9. Natkanski, E., W. Y. Lee, B. Mistry, A. Casal, J. E. Molloy, and P. Tolar. 2013. B cells protein that inhibits tumor necrosis factor cytotoxicity. Biochem. Biophys. Res. Com-
use mechanical energy to discriminate antigen affinities. Science 340: 15871590. mun. 253: 743749.
10. Hoogeboom, R., E. M. Natkanski, C. R. Nowosad, D. Malinova, R. P. Menon, A. 26. Carsetti, R. 2000. The development of B cells in the bone marrow is controlled by
Casal, and P. Tolar. 2018. Myosin IIa Promotes Antibody Responses by Regulating B the balance between cell-autonomous mechanisms and signals from the microenvi-
Cell Activation, Acquisition of Antigen, and Proliferation. Cell Rep. 23: 23422353. ronment. J. Exp. Med. 191: 58.
11. Berg, J. S., B. C. Powell, and R. E. Cheney. 2001. A millennial myosin census. 27. Lalor, P. A. 1991. An evolutionarily-conserved role for murine Ly-1 B cells in pro-
Mol. Biol. Cell 12: 780794. tection against bacterial infections. Autoimmunity 10: 7176.
12. Furusawa, T., S. Ikawa, N. Yanai, and M. Obinata. 2000. Isolation of a novel 28. Deng, J., X. Wang, Q. Chen, X. Sun, F. Xiao, K.-H. Ko, M. Zhang, and L. Lu.
PDZ-containing myosin from hematopoietic supportive bone marrow stromal cell 2016. B1a cells play a pathogenic role in the development of autoimmune arthritis.
lines. Biochem. Biophys. Res. Commun. 270: 6775. Oncotarget 7: 1929919311.
13. Mori, K., T. Furusawa, T. Okubo, T. Inoue, S. Ikawa, N. Yanai, K. J. Mori, and 29. Hoffmann, A., S. Kerr, J. Jellusova, J. Zhang, F. Weisel, U. Wellmann, T. H. Win-
M. Obinata. 2003. Genome structure and differential expression of two isoforms of kler, B. Kneitz, P. R. Crocker, and L. Nitschke. 2007. Siglec-G is a B1 cell-inhibitory
receptor that controls expansion and calcium signaling of the B1 cell population.
a novel PDZ-containing myosin (MysPDZ) (Myo18A). J. Biochem. 133: 405413.
Nat. Immunol. 8: 695704.
14. Billington, N., J. R. Beach, S. M. Heissler, K. Remmert, S. Guzik-Lendrum, A.
30. Diana, J., Y. Simoni, L. Furio, L. Beaudoin, B. Agerberth, F. Barrat, and A. Lehuen.
Nagy, Y. Takagi, L. Shao, D. Li, Y. Yang, et al. 2015. Myosin 18A coassembles
2012. Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells ini-
with nonmuscle myosin 2 to form mixed bipolar filaments. Curr. Biol. 25: tiates autoimmune diabetes. Nat. Med. 19: 6573.
942948. 31. Enghard, P., J. Y. Humrich, V. T. Chu, E. Grussie, F. Hiepe, G.-R. Burmester, A.
15. Matsui, K., N. Parameswaran, N. Bagheri, B. Willard, and N. Gupta. 2011. Prote- Radbruch, C. Berek, and G. Riemekasten. 2010. Class switching and consecutive
omics analysis of the ezrin interactome in B cells reveals a novel association with loss of dsDNA-reactive B1a B cells from the peritoneal cavity during murine lupus
Myo18aa. J. Proteome Res. 10: 39833992. development. Eur. J. Immunol. 40: 18091818.
16. Rodrıguez, C. I., F. Buchholz, J. Galloway, R. Sequerra, J. Kasper, R. Ayala, A. F. 32. McClain, M. T., P. A. Ramsland, K. M. Kaufman, and J. A. James. 2002. Anti-sm
Stewart, and S. M. Dymecki. 2000. High-efficiency deleter mice show that FLPe is autoantibodies in systemic lupus target highly basic surface structures of complexed
an alternative to Cre-loxP. Nat. Genet. 25: 139140. spliceosomal autoantigens. J. Immunol. 168: 20542062.
17. Hobeika, E., S. Thiemann, B. Storch, H. Jumaa, P. J. Nielsen, R. Pelanda, and M. 33. Maravillas-Montero, J. L., and L. Santos-Argumedo. 2011. The myosin family: un-
Reth. 2006. Testing gene function early in the B cell lineage in mb1-cre mice. Proc. conventional roles of actin-dependent molecular motors in immune cells. J. Leukoc.
Natl. Acad. Sci. USA 103: 1378913794. Biol. 91: 3546.

You might also like