You are on page 1of 17

High-resolution structures of the IgM Fc domains

reveal principles of its hexamer formation


Roger Müllera,1, Melissa A. Gräwerta,b,1, Thomas Kerna,c,1, Tobias Madla,c,d, Jirka Pescheka, Michael Sattlera,c,2,
Michael Grolla,2, and Johannes Buchnera,2
a
Munich Center for Integrated Protein Science at the Department Chemie, Technische Universität München, 85748 Garching, Germany; bDeutsches
Elektronen Synchrotron, European Molecular Biology Laboratory, 22603 Hamburg, Germany; cInstitute of Structural Biology, Helmholtz Zentrum München,
85764 Neuherberg, Germany; and dInstitute of Chemistry, Karl-Franzens Universität Graz, 8010 Graz, Austria

Edited by Robert Huber, Max Planck Institute of Biochemistry, Planegg-Martinsried, Germany, and approved May 6, 2013 (received for review
January 10, 2013)

IgM is the first antibody produced during the humoral immune with three domains in the Fc part (11). Fluorescence data (12)
response. Despite its fundamental role in the immune system, IgM and the subsequently determined crystal structure (11) of IgE Fc
is structurally only poorly described. In this work we used X-ray revealed that the IgE Fc is sharply bent. Interestingly, the latest
crystallography and NMR spectroscopy to determine the atomic IgM model suggests a similar Fc region with the Cμ4 domains
structures of the constant IgM Fc domains (Cμ2, Cμ3, and Cμ4) protruding out of the plane defined by Cμ2, Cμ3, and the Fab
and to address their roles in IgM oligomerization. Although the domains (13).
isolated domains share the typical Ig fold, they differ substantially Owing to its flexibility, crystallization trials of the IgM oligomer
in dimerization properties and quaternary contacts. Unexpectedly, have not been successful to date. In this study, we applied X-ray
the Cμ4 domain and its C-terminal tail piece are responsible and crystallography and NMR spectroscopy to determine the atomic
sufficient for the specific polymerization of Cμ4 dimers into cova- details of all individual mouse IgM Fc domains (Cμ2, Cμ3, and
lently linked hexamers of dimers. Based on small angle X-ray scat- Cμ4). Analysis of their quaternary arrangements in solution re-
tering data, we present a model of the ring-shaped Cμ4 structure, vealed unexpectedly that Cμ4 forms defined ring-like hexamers of
which reveals the principles of IgM oligomerization. dimers. SAXS data of the hexameric domain assembly together
with modeling suggest a model for the IgM Fc structure.
antibody oligomerization | hybrid approach | dimer interfaces
Results

A ntibodies, also referred to as Igs, defend against infection


by the inactivation of viruses or bacteria and by recruiting
downstream effectors such as the complement system or cells
Characterization of the IgM Fc Domains. To gain insight into the
elusive structure of the IgM Fc segment, we produced the in-
dividual Fc domains recombinantly in Escherichia coli and char-
specialized to kill invading microorganisms (1, 2). IgM is the first acterized their properties after purification. Investigation of the
antibody to be produced during the humoral immune response influence of intersubunit disulfide bonds on oligomerization was
(3). Early IgM antibodies are secreted before B cells have un- addressed by serine mutations of the corresponding cysteine res-
dergone somatic hypermutations and therefore tend to be of low idues in Cμ2 (C337), Cμ3 (C414), and Cμ4tp (C575). During
affinity. To compensate for the reduced binding efficiency of the thermal denaturation, all domains unfold cooperatively with
monomers, IgM forms oligomers (Fig. 1) whose multiple antigen- transition midpoints ranging from 59–65 °C (Table 1 and Fig. S1).
binding sites confer high overall avidity (4). Moreover, the com- The lack of thermodynamic stability parameters is caused by the
plex structure of IgM makes it especially effective in activating the irreversibility of the transitions. However, guanidium chloride
complement system (5). (GdmCl)-induced equilibrium unfolding (Fig. S1) resulted in re-
Whereas the antigen-binding fragment (Fab) of different Ig versible transitions of all domains, and thermodynamic parame-
classes follow a common topology, the constant fragment (Fc) ters are given in Table 1. All domains behave like two-state folders,
parts differ substantially in domain composition and architec- not significantly populating any equilibrium intermediates.
ture. In contrast to IgG, the Fc part of IgM is composed of three To investigate the quaternary structure of the isolated domains,
Ig domains (Cμ2, Cμ3, and Cμ4) and an additional C-terminal we first performed size-exclusion chromatography (SEC) experi-
tail piece (tp). The IgM polymer is composed of subunits in ments (Fig. S2). The Cμ2 domain was purified as a disulfide-linked
which two heavy chains (μ) are covalently paired with two light dimer, as determined by nonreducing SDS/PAGE, which is in
chains (L). IgM is present either as pentamers (μ2L2)5J in the agreement with the elution profile of the SEC column (Fig. S2A,
presence of the J chain, or as hexamers (μ2L2)6 in the absence of dotted line). Moreover, our data demonstrate that dimer forma-
the J chain, a small protein that is involved in IgM assembly and tion was independent of the protein concentration. However, when
secretion (6–8). Upon assembly, the heavy chains are covalently
linked in the Cμ2, Cμ3, and Cμ4 (tp) domains by interchain
BIOCHEMISTRY

disulfide bridges (Fig. 1). Author contributions: R.M., M.S., M.G., and J.B. designed research; R.M., M.A.G., T.K., T.M.,
The Fc region of IgM is of outstanding interest because its and J.P. performed research; R.M., M.A.G., T.K., T.M., J.P., M.S., M.G., and J.B. analyzed data;
and R.M., M.S., M.G., and J.B. wrote the paper.
structure, oligomerization, and effector protein binding clearly
differs from other Ig Fc regions. Our current understanding of The authors declare no conflict of interest.

the IgM structure largely originates from negative-stain EM (9), This article is a PNAS Direct Submission.

which identified the pentamer as a planar, star-shaped oligomer Data deposition: The atomic coordinates and structure factors have been deposited in the
Protein Data Bank, www.pdb.org [PDB ID codes 4JVU (Cμ2 domain), 4BA8 (Cμ3 domain),
with the Fab fragments pointing away from the inner core com-
4JVW (Cμ4 domain), and 4BLE (C-alpha coordinates of the Cμ4tp hexamer of dimers small
posed of the Fc regions. Early structural models of complement angle X-ray scattering model).
activation were based on small angle X-ray scattering (SAXS) 1
R.M., M.A.G., and T.K. contributed equally to this work.
data and modeling (10) and suggest a planar Fc disk containing a 2
To whom correspondence may be addressed. E-mail: sattler@helmholtz-muenchen.de,
central Cμ4 ring. The Cμ3 and Cμ2 domains are attached in a star- michael.groll@ch.tum.de, or johannes.buchner@tum.de.
shaped manner. The latest model makes use of the high similarity This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.
of IgM and IgE. IgM shares the basic IgE domain architecture 1073/pnas.1300547110/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1300547110 PNAS | June 18, 2013 | vol. 110 | no. 25 | 10183–10188


interaction in the SEC and aUC experiments. Taken together,
these experiments conclude that the Cμ2 and Cμ4 domains form
dimers in solution, whereas the Cμ3 domain is monomeric. Next,
we performed structural analysis of the respective Cμ domains.

Crystal Structure of the Cμ2 Domain. Crystals of the Cμ2 domain


were obtained in the space group C2, with the unit cell dimensions
a = 92.4 Å, b = 44.8 Å, and c = 54.6 Å, and two molecules in
the asymmetric unit accounting for 37% solvent content. The phase
problem of the IgM Cμ2 domain was determined by Patterson
search calculations using the IgE Ce2 (1O0V) (11) as a starting
model and the structure was refined to an Rfree of 18.1% at
1.3 Å resolution (Table S1). Overall the model is well defined in
its electron density map, yet some loops (gap between chain-A
Glu-293 to Thr-298 and chain-B Phe-248 to Lys-255 and Thr-293
Fig. 1. Schematic view of hexameric IgM (Left) and a single subunit (Right). to Gln-300) were structurally disordered and have therefore been
Heavy chains are depicted in light blue and light chains in orange. Red lines omitted. The Cμ2 domain crystallized as a dimer, and the con-
represent intersubunit disulfide bridges and the green hexagons show formation of Cμ2 shows a typical Ig fold with a two-layer sandwich
glycosylation sites. The cysteine residues that form interdomain disulfide of seven antiparallel β-strands arranged in two β-sheets with
bridges between the Cμ2 domains (C337) and covalently link the IgM hex- a Greek key topology (Fig. 2A). Structural superposition of the
amer in Cμ3 (Cys414) and the C-terminal tp (C575) are depicted. Cα-atoms of both chains yields a rms deviation of 0.6 Å. Both
chains possess an Ig fold typical internal disulfide bond between
Cys-260 and Cys-320. The electron density map shows high oc-
the interchain disulfide bond was removed by mutating Cys337 to cupancy and low Debye–Waller factors in the disulfide bridges,
Ser, the elution time became concentration-dependent. The single which are oriented perpendicular to the β-sheets. A common
peak is indicative of a fast monomer/dimer equilibrium (Fig. S2A). feature of many members of the Ig superfamily is the presence
Both the wild type and the C414S mutant of the Cμ3 domain eluted of at least one cis proline residue in the native structure. Both
predominantly as monomers and did not show any peak-shift (Fig. domains harbor this cis peptide bond between Thr-266 and Pro-
S2 B and C). However, in the case of wild-type Cμ3, small amounts 267. However, in chain A there is an additional cis peptide bond
of covalent dimers were observed. The Cμ4 domain behaved involving Pro-253, whereas the corresponding loop in chain B is
similar to the Cμ2C337S mutant. It also exhibits a fast monomer/ not resolved.
dimer equilibrium, although with lower affinity as estimated from To analyze the biological assembly state of the structure, the
the smaller shift at higher protein concentrations (Fig. S2D). When PISA server (15) was used and confirmed that Cμ2 forms a phys-
the C-terminal tp with the penultimate C575 was attached to the iological dimer comprising a buried area of ∼1,660 Å2. The Cμ2
Cμ4 domain, at least one additional prominent species with sub- dimer shares the mode of domain pairing seen in IgE Ce2 with
stantially increased molecular weight appeared (Fig. S2E). This its less extensive interaction surface and interchain disulfide bonds
peak could be minimized by mutating Cys575 to Ser (Fig. S2F), (11). However, unlike IgE Ce2, the interface is dominated by
indicating that this species seems to correspond to a covalently a central hydrophobic core (Fig. 2A) as typically seen in other Ig
linked Cμ4tp dimer or higher-order oligomers (discussed below). domain structures. The dimer is further stabilized by six hydrogen
To gain further insight into Cμ domain association, we per- bonds, one salt bridge (between chain A Glu-261 OE2 and chain
formed sedimentation equilibrium (SE) and sedimentation ve- B Lys-257 NZ, 2.77 Å) as well as an additional disulfide bridge
locity (SV) analytical ultracentrifugation (aUC). Data from SE between the C-terminal Cys-337 residues of chain A and B (Fig. 2A).
runs performed at different rotor speeds and protein concen- The hydrophobic contact interface also accounts for the observed
trations were fitted with a model for self-association (14) to cal- dimer formation in the absence of the C-terminal disulfide bridge.
culate dissociation constants for the monomer/dimer equilibrium
and were confirmed by SV-aUC (Table 1 and Fig. S3). For the Solution Structure of the Cμ3 Domain. Because crystallization trials
Cμ2C337S mutant without the interdomain disulfide bridge, a of the Cμ3C414S domain failed, this structure was elucidated
Kd of 2.1 ± 0.1 μM was assigned. The weaker dimerization of the by heteronuclear NMR spectroscopy (Table S2). We determined
Cμ4 domain deduced from the SEC experiment was verified by the overall tumbling correlation time of Cμ3C414S to be 6.0 ± 0.4 ns
aUC, resulting in a Kd of 86 ± 3 μM. The presence of the C- from 15N NMR relaxation data (Fig. S4). Comparison with an
terminal tp (Cμ4tpC575S) decreased the affinity of the Cμ4 do- expected value of 7.5 ns for the monomer and 14.4 ns for the
main dimer to a Kd of 224 ± 7 μM. This finding might be the dimer confirms that Cμ3C414S is monomeric, in agreement with
consequence of a not-fully-unstructured tp that is interacting with the sedimentation data described above. The solution structure of
the Cμ4 domain. In contrast, the Cμ3C414S domain showed no Cμ3C414S consists of the typical Ig fold with a β-sheet sandwich

Table 1. Oligomeric state and conformational stability of the IgM Fc domains


Domain Mass, kDa Kd, μM ΔGunfolding, kJ·mol−1 meq, kJ·mol−1·M−1 Tmelt, °C

Cμ2 24.6 — 36.2 18.6 64.5


Cμ2C337S 23.6 2.1 ± 0.1 26.0 16.6 60.0
Cμ3C414S 11.3 — 15.8 10.2 57.9
Cμ4 17.8* 86 ± 3 15.8 14.4 58.9
Cμ4tpC575S 15.4* 224 ± 7 21.1 17.5 60.9

Molar masses and Kd values for domain dimerization were calculated from SV and SE aUC runs, respectively.
*For Cμ4 and Cμ4tpC575S, monomeric and dimeric species could not be separated due to fast exchange rates.
ΔGunfolding and the cooperativity parameter (meq) originate from GdmCl denaturation experiments and melting
temperature (Tmelt) from thermal denaturation experiments.

10184 | www.pnas.org/cgi/doi/10.1073/pnas.1300547110 Müller et al.


Fig. 2. Structures of the individual IgM Fc domains.
(A) Cartoon representation of the crystal structure of
the Cμ2 domain (Upper, Protein Data Bank ID code
4JVU), the arrangement of the two molecules in the
crystallographic asymmetric unit forming a stable di-
mer. Inter- and intramolecular disulfide bonds are
shown in stick representation. The surface of one Cμ2
domain (chain A, yellow), showing the contact area
with the second Cμ2 domain (chain B, red), is repre-
sented as Cα-trace (Lower). (B) Solution structure of the
Cμ3 domain (Protein Data Bank ID code 4BA8). Super-
position of the 10 lowest-energy NMR structures are
shown as cartoon (Upper). The highly flexible DE loop is
labeled. Chemical shift perturbations in the Cμ3 do-
main, when bound to the Cμ4 domain, are shown
(Lower). The color scale is as follows: yellow, 0.025–0.05
ppm; orange, 0.05–0.1 ppm; and red, >0.1 ppm. The
positions for the interdomain disulfide bridge (Cys414)
and the glycosylation site (Asn402) are labeled and
shown in stick representation. (C) Cartoon represen-
tation of the crystal structure the Cμ4 domain (Upper,
Protein Data Bank ID code 4JVW). Intramolecular disul-
fide bonds are shown in stick representation. The surface of one Cμ4 domain (chain A, blue), showing the contact area with the second Cμ4 domain (chain B, red), is
represented as Cα-trace (Lower). Color coding for the interfaces: gray, hydrophilic contact residues; green, hydrophobic contact residues.

that includes an intramolecular disulfide bridge between Cys367 hydrophobic protein interactions resulting in a stable dimer of
and Cys426 (Fig. 2B). NMR signals of residues in the loop between Cμ4 (Fig. 2C). Interestingly, compared with the dimerization
strands D and E connecting the two β-sheets are line-broadened behavior of other Ig folds, the two molecules are arranged in
owing to chemical exchange, further confirmed by comparison of a parallel manner (Fig. 2C). Particularly, the N termini are
the transverse R2 15N relaxation rates to offset-corrected R1ρ rates located on the same side of the dimer, and the C termini on the
(Fig. S4). The increased dynamics of this loop had been already opposite end, which is important for the oligomer formation. A
observed in the corresponding IgG Cγ2 domain in the absence of summary of the different homo–dimer association modes of
glycosylation in the DE loop (16), suggesting that the DE loop in Ig Fc domains is shown in Fig. S5.
Cμ3C414S may be stabilized by glycosylation of Asn402. Chemical
shift perturbations (CSP) of Cμ3C414S and Cμ3C414S–Cμ4 do- Cμ4 Oligomerization. As shown above, the Cμ4tp domain forms
main constructs (Fig. S4) have been obtained by comparing the weak noncovalent dimers in the absence of Cys575 in the tp. In
chemical shifts of their 1HN, 15N resonance from the backbone the presence of Cys575, we also observed noncovalently linked
assignments. The CSPs have been calculated according to Eq. S2. dimers in dilute solutions. At higher concentrations, however,
In the spectra of the tandem construct the region between those dimers form disulfide-linked oligomers. The analysis of the
Phe354–Phe358 has been broadened out completely owing to the native Cμ4tp by SEC revealed a concentration-dependent equi-
interaction in the interface, and therefore no exact CSP could librium between dimers and oligomers with masses of 31 kDa
be assigned. Residues Thr374–Leu378 and Val396–Ser399 have and 198 kDa applying multiangle light scattering (MALS) (Fig.
been excluded from the analysis, because their 1HN, 15N resonances 3A). Interestingly, the latter species corresponds well to a hex-
could not be assigned owing to line broadening in the single domain amer of Cμ4 dimers, as observed for native IgM in the absence of
construct. This allowed mapping of the binding interface of the Cμ3 the J chain (6, 17, 18). Thus, the isolated Cμ4tp domain is suf-
domain with Cμ4 (Fig. 2B). Interestingly, this interface on Cμ3 ficient for the assembly of defined hexamers of dimers.
comprises predominantly the two helices at the C-terminal side of Further support of oligomer formation was received by SV
Cμ3 and is very similar to the interactions seen between the cor- aUC runs at different protein concentrations (1–20 μM) (Fig.
responding domains in other Ig classes. The mapped interface on 3B), revealing a major species that sediments with 8.8 S and a
Cμ3 was then used for modeling experiments, eventually yielding minor one with 2.6 S. Both the sedimentation coefficients and
the overall domain arrangements below. the fitted frictional coefficient f/f0 of 1.34 calculated the masses
for the two species to be 190 kDa and 30 kDa, respectively.
Crystal Structure of the Cμ4 Domain. The Cμ4 domain crystallized These values validate the dimer/hexamer of dimer equilibrium
in the space group C2 with the cell dimensions a = 169.2 Å, b = of Cμ4tp dimers. Furthermore, also the relative ratios could
41.2 Å, and c = 67.1 Å. Four molecules are located in the asym- be shifted toward the hexamer of dimers in a concentration-
metric unit leading to 40% solvent content. Using the structure of dependent manner (Fig. 3B, Inset).
BIOCHEMISTRY

the Cμ2 domain as a starting model, the IgM Cμ4 could be de-
termined by molecular replacement methods and refined to a Rfree Model for the Cμ4 Hexamer Structure. SAXS experiments were
of 23.4% at 2.0 Å resolution (Table S1). The conformations of all used to further characterize the hexameric structure of Cμ4tp
four IgM Cμ4 monomers show a typical Ig topology that is stabi- dimers (Materials and Methods gives details). The molecular
lized by a disulfide bond between Cys474 and Cys536 (Fig. 2C) and mass of Cμ4tp of 162 ± 4 kDa, determined based on the I(0), fits
a cis peptide bond involving Pro481 is observed. well to the expected molecular mass of 176 kDa for a hexamer.
The analysis of the biological assembly using the Protein Inter- The radius of gyration (Rg) of 41.6 ± 0.5 and a maximum dimen-
faces, Surfaces and Assemblies (PISA) server predicts that the sion of 114 Å also support hexamer of dimer formation (Fig. S6A).
monomers A and D as well as B and C form stable dimers. The The SAXS data together with the crystal structure of the Cμ4
conformation of both dimers in the asymmetric unit match to domain allowed us to model the hexameric structure of Cμ4tp
each other with an rmsd on the Cα-atoms of 0.3 Å and 0.4 Å, dimers. Note that the additional Cμ4–Cμ4 interface observed in
respectively. Approximately 1,900 Å2, which corresponds to 17% the crystal structure cannot be used to construct the hexamer
of the surface area, are buried in the interface that comprises (Fig. S6E). Therefore, no additional restraints have been used

Müller et al. PNAS | June 18, 2013 | vol. 110 | no. 25 | 10185
we present a hybrid approach using X-ray crystallography, NMR,
and SAXS to solve the structures of the individual domains
of the mouse IgM Fc (Cμ2, Cμ3, and Cμ4) and to reconstruct the
Fc oligomer. Aside from the expected domain topologies with
their characteristic β-sheet sandwich and two short α-helices
(20), we observed unexpected domain associations that were not
considered in previous models. Based on electron microscopy,
X-ray scattering, and mutagenesis studies, we know that the IgM
polymer is assembled through interactions of identical domains,
that is, Cμ2–Cμ2, Cμ3–Cμ3, and Cμ4–Cμ4, and that the corre-
sponding cysteine residues (C337, C414, and C575) form the
interchain disulfide bonds (21–23). A detailed mutagenesis study
showed that the cysteine at position 575 is essential for efficient
assembly, whereas C337 and C414 are not needed for polymer
formation (24). The latest IgM model is based on the IgE
structure (13).
To obtain a better understanding of the elusive structure of
the IgM Fc, we first characterized the isolated Fc domains. The
Cμ2 domain, the most N-terminal domain of the IgM Fc, replaces
the hinge region found in IgG (25). It forms a disulfide-linked
dimer with a unique interface dominated by hydrophobic inter-
actions. In the absence of the disulfide bridge, the Kd is around
2 μM. This weak interaction needs to be further stabilized by the
covalent linkage (Cys337). The IgE Ce2 domain is functionally
equivalent to Cμ2 and it also bears some structural resemblance.
Both have a unique association mode with a small interface
area, compared with other Ig constant domains, and a distinct
rotation angle (110° for Cμ2 and 105° for Ce2) between the
domains (Fig. S5A). However, there are important differ-
ences. These include the arrangement of the disulfide bridges
in Ce2 (11) and the interface that is polar and less pronounced
in Ce2, leading to a monomeric protein in the absence of the
disulfide bonds (26).
The Cμ3 domain does not make any stable dimer contacts. This
Fig. 3. Characterization of the Cμ4tp oligomer. (A) SEC MALS profile of the is similar to the corresponding IgG Cγ2 and IgE Ce3 domains,
Cμ4tp oligomer. UV absorbance is shown in black, and calculated masses in which do not have any direct contacts either. In contrast to IgG and
red. (B) Cμ4tp oligomer characterized by aUC SV runs at 20 μM (black), 5 μM IgE, IgM forms larger oligomers. During assembly, the Cμ3 do-
(red), and 1 μM (cyan). The histogram shows the percentages of hexamer of
mains come into close proximity and the weak Cμ3–Cμ3 inter-
dimers (dark gray) and dimers (light gray) of the total protein in solution.
actions are stabilized via disulfide bridges (Cys414) only in the
complete IgM oligomer. The position of the Cys414 at the edge of
in the SAXS modeling. All structures share a characteristic to- the Cμ3 domain and its mainly polar environment supports such
roidal assembly (Fig. 4B). The generated structures fit well to the a weak contact that must be stabilized by a disulfide bridge.
experimental data (χ = 1.98–3.34) (Fig. S6G). Interestingly, in The mode of dimerization elucidated here for Cμ4 has so far
the oligomer the Cμ4tp dimers contact each other involving the not been observed among Igs (Fig. 2 and Fig. S5A): Whereas all
surface of the β-sheets and several loop regions with a surface other C-terminal antibody domain dimers such as IgG Cγ3, IgA
Cα3, and IgE Ce4 are arranged in an antiparallel fashion, with
area of ∼1,000 Å2 (Dataset S1). The N termini of Cμ4tp are
the N and C termini on opposing sides, the IgM Cμ4 topology
pointing outward of the hexameric ring, whereas the C termini,
results in the location of the N termini and C termini on the
which are structurally constrained by the disulfide bond, are
same side. This is explained by the rotation angles between the
facing its interior (Fig. 4B). domains. Whereas IgG Cγ3, IgA Cα3, and IgE Ce4 have rota-
In conclusion, the various structural and functional data tion angles of 132°, 138°, and 142°, respectively, the Cμ4 of IgM
described above allowed us to generate a model of the entire harbors a rotation angle of 70°, which is important for the self-
Cμ2–Cμ3–Cμ4tp hexamer (Fig. 4A) using (i) the structures of the assembly of the dimers into pentamers or hexamers of dimers.
individual subunits, (ii) the SAXS-derived structure of the hex- Interestingly, IgA Cα3 and IgE Ce4 share up to 50% sequence
americ Cμ4tp dimers, (iii) the distances for the inter-Cμ3 disulfide identity with Cμ4. Whereas the dimer interfaces of IgM Cμ4, IgA
bonds, and (iv) NMR chemical shift mapping of the Cμ3–Cμ4 Cα3, and IgE Ce4 share a conserved, mostly hydrophobic, core,
binding interface (Fig. 2C and Fig. S4). We conclude that the most residues lying at the outer part of the Cμ4 interface show
Cμ2–Cμ3 subunits in IgM point apart from the hexameric ring little identity or homology to IgA or IgE (Fig. S5B). These dif-
formed by Cμ4tp dimers (Fig. 4C). The domain interactions, in- ferences must lead to the different dimer association of Cμ4.
cluding the short linkers between the Cμ2, Cμ3, and Cμ4 domains, Hence, their different oligomeric states are caused by the unique
sterically restrict the accessible conformational space (Fig. 4C). amino acid composition at the interface that defines the con-
These findings demonstrate that the Cμ4 domain orchestrates the figuration of the dimer.
assembly of the oligomeric IgM, whereas the covalent linkages in A hallmark of the current work is the identification of the
the tail piece and the Cμ3 domain stabilize this complex. minimum requirements for IgM oligomerization. Attaching the
C-terminal tp to the Cμ4 domain is already sufficient for oligomer
Discussion formation, including the intermolecular disulfide bridge involving
Whereas the structure and assembly of IgG is well-studied (19), Cys575, the penultimate amino acid in the tp. Our results therefore
for IgM we largely lack detailed structural information. Here show that the Cμ4 domain possesses two dimerization interfaces

10186 | www.pnas.org/cgi/doi/10.1073/pnas.1300547110 Müller et al.


Fig. 4. SAXS analysis of Cμ4tp and hexameric IgM Fc modeling. (A) Scheme of the IgM Fc hexamer with the information used for model building is in-
dicated. (B) Structural model of the Cμ4tp hexamer of dimers (Protein Data Bank ID code 4BLE). The hexameric subunits and N/C termini of the Cμ4 dimer
structure are annotated. One single Cμ4tp dimer is shown in blue. (C ) Cartoon representation of the IgM Fc hexamer model (Left). A side view of the Fc
hexamer is given (Right), showing that the inner core (Cμ4) is protruding out of the plane defined by the Cμ2 and Cμ3 domains. Cμ2 dimers are shown in
yellow, Cμ3 in gray, and the Cμ4 core ring in blue. The termini and linker residues are represented as Cα dummy atoms by the program CORAL and are
shown as spheres.

consisting of the four β-strands as well as a weak interaction site of six or more subunits are secreted (30, 31). Furthermore, the J
distinct from the first one that is stabilized by the interdimer chain plays an important role in the selective assembly and it is
disulfide bridge. Thus, our data reveal the existence of a non- expected that its major function is a place holder for one IgM
covalently associated Cμ4 dimer that is covalently surrounded by subunit in the pentamer compared with the hexamer (32). This is
two adjacent Cμ4 dimers. The noncovalent Cμ4 oligomer does not supported by the hexameric ERp44/ERGIC53 assembly platform
seem especially stable (Fig. 3) at protein concentrations usually that is involved in the formation of pentameric and hexameric
found in plasma (ca. 1 μM). However, the oligomer is further IgM assemblies. Therefore, the hexameric Cμ4 ring presented here
stabilized by the Cμ2 dimer interface and the inter-HC disulfide might be identical to a pentameric Cμ4/J-chain ring with one Cμ4
bridge at the C terminus of the Cμ2 domain (23). dimer subunit exchanged by one J-chain protein. Notably, hex-
Based on the ring-like Cμ4 oligomer and the data from the americ IgM that lacks the J chain activates the complement system
single domains, this study describes a model for the hexameric 15- to 20-fold more efficiently than pentameric IgM (33, 34).
IgM Fc, composed of the Cμ4 inner core and the Cμ3 and Cμ2 In summary, our study provides a comprehensive structural
domains that build a flexible star-shaped arrangement around analysis of the IgM Fc domains as well as of the oligomerization
the inner core. This is similar to previous models based on
of IgM subparticles. Despite the high structural similarity of in-
electron microscopy (9) and SAXS (10) data. However, the
dividual Ig domains from various classes, interactions between
model presented here is not planar and the central region (Cμ4)
the domains are different and adapted for the desired function.
is projecting out of the plane defined by the Cμ2 and Cμ3
domains (Fig. 4C). The dimensions of this structure are in good Notably, it is the Cμ4 domain of IgM together with its C-terminal
agreement with previous estimations based on cryo-atomic force extension that, apart from any antibody domain characterized
microscopy (AFM) data (13). In addition to the nonplanar to date, confers the intrinsic ability to oligomerize into defined
shape, also the size of the central circular region, composed of hexamers of dimer. Because the sequences of mouse and human
the Cμ3 and Cμ4 domains, with a diameter of ∼180 Å, fits well to IgM Fc are conserved (68% identity), we assume that the model
BIOCHEMISTRY

the core region dimension of 190 ± 20 Å seen in the cryo- presented here is also true for human IgM Fc.
AFM experiments.
In conclusion, the Cμ4tp domain is responsible and sufficient Materials and Methods
Protein Production and Purification. Proteins were expressed as inclusion
for the specific IgM polymerization. In vivo the situation seems
bodies in E. coli, oxidatively refolded, and purified as described in SI Mate-
more complicated. In the crowded environment of an antibody-
rials and Methods.
producing cell, a sophisticated quality control system is present
(27). In the case of IgM, the formation of pentamer/hexamer Analytical Gel Filtration. Analytical SEC measurements were performed in PBS
complexes is stringently controlled by the ERp44/ERGIC53 as outlined in SI Materials and Methods. For the Cμ4tp domain detection and
assembly platform including a thiol retention mechanism via mass calculations of the observed species a Dawn Heleos MALS detector
Cys575 (28, 29). In addition, it was shown that carbohydrates was used.
have an influence on oligomerization (30), particularly the glycan
linked to Asn563 in the tp of Cμ4 that interacts with ERp44. In Analytical Ultracentrifugation. Domain association and determination of
its absence, pentamers cannot be formed in vivo and polymers dissociation constants was assessed with analytical ultracentrifugation as

Müller et al. PNAS | June 18, 2013 | vol. 110 | no. 25 | 10187
described in SI Materials and Methods. For dimerizing domains, data were correlation acquired on a 10% 13C-labeled sample as previously described
fitted to a self-association model (14). (37, 38). Details are provided in SI Materials and Methods.

Optical Spectroscopy. Fluorescence and CD measurements were carried out in SAXS Measurements and Modeling. SAXS data for solutions of Cμ4tp were
PBS as described in SI Materials and Methods. To obtain thermodynamic measured for several solute concentrations in the range from 1 to 10 mg/
parameters from the GdmCl-induced unfolding and refolding transitions, a mL. The structure of the Cμ4tp hexamer was modeled using the program
two-state model was applied (35). CORAL (39) as described in SI Materials and Methods. The model of the IgM
Fc hexamer was generated in CORAL using the structures of (i) Cμ2, (ii) Cμ3,
Crystallization and Structure Determination. Crystals of Cμ2 and Cμ4 were (iii) the Cμ4 hexameric ring as determined based on SAXS data, (iv) the
grown at 20 °C using the sitting drop vapor diffusion method. Protein so- disulfide linkages (C414 and C575), and (v) the Cμ3–Cμ4 interface derived
lution (10 mg/mL) in 10 mM Tris·HCl, pH 7.5, was mixed with an equal vol- from NMR chemical shift titrations as input. Details are provided in SI Materials
ume of buffer C2 [1.0 M lithium chloride, 0.1 M MES (pH 5.4), and 23% PEG and Methods.
6000] for the Cμ2 domain and buffer C4 [15% PEG 8000 and 0.1M Hepes (pH
7.2)] for the Cμ4 domain, respectively. Diffraction datasets were collected ACKNOWLEDGMENTS. The mouse IgM cDNA was a kind gift from Roberto
using synchrotron radiation at the X06SA beamline at the Swiss Lightsource Sitia (San Raffaele Scientific Institute, Milan). We thank the staff of the Swiss
Lightsource for their assistance in X-ray data collection. We are grateful to
(Cμ2) or on a Bruker Microstar/X8 Proteum (Bruker AXS Inc.) with a Cu ro-
Matthias Feige and Moritz Marcinowski for helpful discussion and techni-
tating anode (λ = 1.54 Å; Cμ4 domain). For structure determination, mo- cal support and Thomas Kriehuber for assistance during MALS measure-
lecular replacement was performed in Phaser (36) using the coordinates of ments. We thank the Bavarian NMR Centre for NMR measurement time and
the Ce2 domain of human IgE (1O0V) for Cμ2 and the coordinates of the Anton Paar GmbH for access to the SAXSess mc2 in-house SAXS instrument.
here determined structure of the Cμ2 domain for Cμ4, respectively. Details This work was supported by grants from the Swiss National Foundation (to
are provided in SI Materials and Methods. R.M.), DFG (to M.S., M.G., and J.B.), the Emmy Noether Program Grant MA
5703/1-1 (to T.M.), the Helmholtz Association, the Bavarian Ministry of Sci-
ences, Research and the Arts, Bavarian Molecular Biosystems Research Net-
NMR Spectroscopy. NMR data were acquired at 25 °C on a Bruker Avance III work (to T.M.), and the Austrian Academy of Sciences (Austrian Programme
600 MHz spectrometer with a 300-μM uniformly 15N, 13C-labeled Cμ3C414S for Advanced Research and Technology Fellowship, to T.M.). M.A.G. was
sample except for the stereospecific assignment of valine and leucine supported by the Peter und Traudl Engelhornstiftung and J.P. by the Stud-
side chains, which was based on a 1H-13C heteronuclear multiple quantum ienstiftung des deutschen Volkes.

1. Wannemuehler MJ, Galvin JE (1994) Bacterial immunogens and protective immunity 21. Beale D, Feinstein A (1969) Studies on the reduction of a human 19S immunoglobulin
in swine. Vet Immunol Immunopathol 43(1-3):117–126. M. Biochem J 112(2):187–194.
2. Ohanian SH, Schlager SI (1981) Humoral immune killing of nucleated cells: mecha- 22. Milstein CP, Richardson NE, Dieverson EV, Feinstein A (1975) Interchain disulphide
nisms of complement-mediated attack and target cell defense. Crit Rev Immunol 1(3): bridges of mouse immunoglobulin M. Biochem J 151(3):615–624.
165–209. 23. Davis AC, Roux KH, Pursey J, Shulman MJ (1989) Intermolecular disulfide bonding
3. Boes M (2000) Role of natural and immune IgM antibodies in immune responses. Mol in IgM: effects of replacing cysteine residues in the mu heavy chain. EMBO J 8(9):
Immunol 37(18):1141–1149. 2519–2526.
4. Thomas HI, Morgan-Capner P (1990) The avidity of specific IgM detected in primary 24. Wiersma EJ, Shulman MJ (1995) Assembly of IgM. Role of disulfide bonding and
rubella and reinfection. Epidemiol Infect 104(3):489–497.
noncovalent interactions. J Immunol 154(10):5265–5272.
5. Taylor B, et al. (1994) C1q binding properties of monomer and polymer forms
25. Saphire EO, et al. (2001) Crystal structure of a neutralizing human IGG against HIV-1:
of mouse IgM mu-chain variants. Pro544Gly and Pro434Ala. J Immunol 153(11):
A template for vaccine design. Science 293(5532):1155–1159.
5303–5313.
26. McDonnell JM, et al. (2001) The structure of the IgE Cepsilon2 domain and its role in
6. Brewer JW, Corley RB (1997) Late events in assembly determine the polymeric
stabilizing the complex with its high-affinity receptor FcepsilonRIalpha. Nat Struct
structure and biological activity of secretory IgM. Mol Immunol 34(4):323–331.
7. Sidman C (1981) B lymphocyte differentiation and the control of IgM mu chain ex- Biol 8(5):437–441.
pression. Cell 23(2):379–389. 27. Sitia R, Braakman I (2003) Quality control in the endoplasmic reticulum protein fac-
8. Sitia R, et al. (1990) Developmental regulation of IgM secretion: The role of the tory. Nature 426(6968):891–894.
carboxy-terminal cysteine. Cell 60(5):781–790. 28. Anelli T, et al. (2007) Sequential steps and checkpoints in the early exocytic com-
9. Feinstein A, Munn EA (1969) Conformation of the free and antigen-bound IgM an- partment during secretory IgM biogenesis. EMBO J 26(19):4177–4188.
tibody molecules. Nature 224(5226):1307–1309. 29. Wang L, et al. (2008) Crystal structure of human ERp44 shows a dynamic functional
10. Perkins SJ, Nealis AS, Sutton BJ, Feinstein A (1991) Solution structure of human and modulation by its carboxy-terminal tail. EMBO Rep 9(7):642–647.
mouse immunoglobulin M by synchrotron X-ray scattering and molecular graphics 30. de Lalla C, Fagioli C, Cessi FS, Smilovich D, Sitia R (1998) Biogenesis and function of
modelling. A possible mechanism for complement activation. J Mol Biol 221(4): IgM: the role of the conserved mu-chain tailpiece glycans. Mol Immunol 35(13):
1345–1366. 837–845.
11. Wan T, et al. (2002) The crystal structure of IgE Fc reveals an asymmetrically bent 31. Cortini M, Sitia R (2010) ERp44 and ERGIC-53 synergize in coupling efficiency and
conformation. Nat Immunol 3(7):681–686. fidelity of IgM polymerization and secretion. Traffic 11(5):651–659.
12. Zheng Y, Shopes B, Holowka D, Baird B (1991) Conformations of IgE bound to its 32. Randall TD, Brewer JW, Corley RB (1992) Direct evidence that J chain regulates
receptor Fc epsilon RI and in solution. Biochemistry 30(38):9125–9132. the polymeric structure of IgM in antibody-secreting B cells. J Biol Chem 267(25):
13. Czajkowsky DM, Shao Z (2009) The human IgM pentamer is a mushroom-shaped 18002–18007.
molecule with a flexural bias. Proc Natl Acad Sci USA 106(35):14960–14965. 33. Davis AC, Roux KH, Shulman MJ (1988) On the structure of polymeric IgM. Eur J Im-
14. Lebowitz J, Lewis MS, Schuck P (2002) Modern analytical ultracentrifugation in pro-
munol 18(7):1001–1008.
tein science: a tutorial review. Protein Sci 11(9):2067–2079.
34. Randall TD, King LB, Corley RB (1990) The biological effects of IgM hexamer forma-
15. Krissinel E, Henrick K (2007) Inference of macromolecular assemblies from crystalline
tion. Eur J Immunol 20(9):1971–1979.
state. J Mol Biol 372(3):774–797.
35. Bolen DW, Santoro MM (1988) Unfolding free energy changes determined by the
16. Borrok MJ, Jung ST, Kang TH, Monzingo AF, Georgiou G (2012) Revisiting the role of
linear extrapolation method. 2. Incorporation of delta G degrees N-U values in a
glycosylation in the structure of human IgG Fc. ACS Chem Biol 7(9):1596–1602.
17. Brewer JW, Randall TD, Parkhouse RME, Corley RB (1994) IgM hexamers? Immunol thermodynamic cycle. Biochemistry 27(21):8069–8074.
Today 15(4):165–168. 36. McCoy AJ, et al. (2007) Phaser crystallographic software. J Appl Cryst 40(Pt 4):658–674.
18. Davis AC, Shulman MJ (1989) IgM—molecular requirements for its assembly and 37. Szyperski T, Neri D, Leiting B, Otting G, Wüthrich K (1992) Support of 1H NMR as-
function. Immunol Today 10(4):118–122, 127–128. signments in proteins by biosynthetically directed fractional 13C-labeling. J Biomol
19. Feige MJ, Hendershot LM, Buchner J (2010) How antibodies fold. Trends Biochem Sci NMR 2(4):323–334.
35(4):189–198. 38. Coggins BE, et al. (2003) Structure of the LpxC deacetylase with a bound substrate-
20. Feige MJ, et al. (2008) The structure of a folding intermediate provides insight into analog inhibitor. Nat Struct Biol 10(8):645–651.
differences in immunoglobulin amyloidogenicity. Proc Natl Acad Sci USA 105(36): 39. Petoukhov MV, Svergun DI (2005) Global rigid body modeling of macromolecular
13373–13378. complexes against small-angle scattering data. Biophys J 89(2):1237–1250.

10188 | www.pnas.org/cgi/doi/10.1073/pnas.1300547110 Müller et al.


Supporting Information
Müller et al. 10.1073/pnas.1300547110
SI Materials and Methods 1–20 μM at 35,000 rpm. Data analysis was carried out by the C(s)
Cloning, Mutagenesis, Expression, and Purification of the Constant method with TI (time invariant) and RI (radial invariant) noise
Fragment Domains. The Cμ2 (Ile225–Ala339), Cμ3 (Asp344– fitting using SEDFIT version 14.1 software (2).
Lys443), Cμ4 (Glu446–Lys558), and Cμ4tp (Glu446–Tyr576) genes For Kd measurements, sedimentation equilibrium runs with
were obtained by PCR amplification using the mouse IgM heavy- protein samples of different concentrations were performed.
chain cDNA as template. Amino acid numbering is according to Sedimentation was detected at 230, 280, or 300 nm, depending
the UniProt entry P01872 (IGHM_MOUSE) with the VH (122 on protein concentration. Data from various concentrations and
amino acids) added. The genes were cloned into the pET28a ex- rotor speeds were fitted globally to a self-association model (3)
pression vector (Merck/Novagene) via the NcoI and HindIII re- in Origin (OriginLab) according to Eq. S1:
striction sites. The C337S, C414S, and C575S mutations were  
introduced using the QuikChange site-directed mutagenesis strat- ω2  2 2 
aðrÞ = c1 ðr0 Þ«1 d exp Mb r − r0
egy (Stratagene/Agilent). Expression was performed in Escherichia 2RT
coli BL21 cells at 37 °C. At an OD600 of 0.6–0.8, expression was   [S1]
ω2  2 2 
induced with 1 mM isopropylthio-β-galactoside. Cells were har- + K12 c1 ðr0 Þ2 2«1 d exp 2Mb r − r0 :
vested after overnight growth and inclusion bodies were prepared 2RT
as previously described (1). Inclusion bodies were solubilized in
50 mM Tris·HCl (pH 7.5), 10 mM EDTA, 8 M urea, and 2 mM The c1(r0) value denotes the molar concentration at a reference
β-mercaptoethanol at 4 °C for 2 h. Insoluble components were position r0; Mb, e1, and d designate the buoyant mass, molar ex-
removed by centrifugation (46,000 × g, 25 min, 4 °C). The su- tinction coefficient, and the optical path length, respectively. K12
pernatant was applied to a Q-Sepharose column equilibrated with is the association constant for the monomer/dimer equilibrium.
50 mM Tris (pH 7.5), 10 mM EDTA, and 5 M urea. The protein
Fluorescence and CD Measurements. CD measurements were carried
was present in the flow-through, and refolding was carried out
via dialysis into 250 mM Tris·HCl (pH 8.0), 100 mM L-arginine, out at 25 °C in a Jasco J-720 spectropolarimeter. Far UV (FUV)-CD
10 mM EDTA, 1 mM oxidized glutathione, and 0.5 mM reduced spectra were recorded from 195–260 nm at a protein concentra-
glutathione at 4 °C overnight. To remove misfolded protein and tion of 10 μM in 1 mm quartz cuvettes. Near UV (NUV)-CD
remaining impurities, the protein was applied to a HiLoad Su- spectra were collected form 260–340 nm at a protein concentration
perdex75 26/60-pg gel-filtration column (GE Healthcare) equil- of 40 μM in 2-mm quartz cuvettes. All spectra were accumulated
ibrated in PBS buffer. For NMR (chemical shift perturbation 16 times and buffer-corrected. For temperature-induced equi-
analysis) and SAXS measurements, a Cμ3C414S–Cμ4 construct librium unfolding, the change in ellipticity at 205 nm was recorded
was cloned, which included the cysteine at position 337 (Cys337– and the rates for heating and cooling were set to 10 °C h−1.
Lys558) and the protein was expressed, refolded, and purified as Fluorescence measurements were performed in a Spex Fluo-
disulfide linked dimer. Isotope-labeled proteins were expressed roMax-4 spectrofluorimeter (Instruments SA). To determine the
in M9 minimal medium containing 15N ammonium chloride as the maximum difference between native and guanidium chloride
only nitrogen source and in addition13C glucose as the only carbon (GdmCl)-induced unfolded protein, intrinsic tryptophan fluo-
source. Partially deuterated protein was expressed in 70% D2O- rescence spectra were obtained from 310–450 nm, with exitation
containing medium. All constructs were sequenced and the mass at 280 nm and excitation and emission slit width set to 1 nm and
of the purified proteins was verified using matrix-assisted laser 6 nm, respectively. Measurements were carried out using a pro-
desorption ionization time-of-flight mass spectrometry. tein concentration of 1 μM in 1-cm quartz cuvettes.
GdmCl-induced equilibrium unfolding was monitored by fluo-
Analytical Gel Filtration. The oligomeric state of the domains were rescence and CD spectroscopy. For fluorescence measurements,
determined by analytical size-exclusion chromatography (SEC) the excitation wavelength was set to 280 nm and the emission was
measurements using a Shimadzu HPLC System and an analytical determined at 360 nm with slit width of 1.5 nm and 6 nm, re-
Superdex75 10/300 GL column (GE Healthcare) equilibrated spectively. For CD measurements, the change in ellipticity at 212
with PBS (8.09 mM Na2HPO4, 1.76 mM KH2PO4, 137 mM NaCl, nm (Cμ2), 213 nm (Cμ3), 220 nm (Cμ4), or 221 nm (Cμ4tp) was
and 2.65 mM KCl, pH 7.4) at 20 °C. All experiments were per- monitored. All measurements were performed at a protein con-
formed with a flow rate of 0.5 mL/min. Fifty-microliter samples centration of 10 μM. For GdmCl-induced unfolding transitions,
were injected at concentrations of 1, 10, and 100 μM, and the the samples were incubated over night at 25 °C at different
elution profiles were detected by absorbance at 280 nm or by GdmHCl concentrations before measurements.
fluorescence emission at 350 nm. For the Cμ4tp domain, two Data were evaluated using Origin (OriginLab); for GdmCl-
Superdex200 5/150 GL columns (GE Healthcare) in series and a induced unfolding transitions, a two-state model was applied (4).
Dawn Heleos multiangle light scattering detector (Wyatt) were
used in addition. Crystallization and Structure Determination. Crystallization of the
Cμ2 and Cμ4 domains of IgM was performed by the sitting drop
Analytical Ultracentrifugation. Analytical ultracentrifugation was vapor diffusion method at 20 °C. Ten milligrams per milliliter
carried out with a ProteomLab XL-I (Beckman-Coulter) equip- (800 μM) of protein solution in 10 mM Tris·HCl (pH 7.5) were
ped with absorbance optics. All experiments were performed mixed with an equal volume of buffer C2 [1.0 M lithium chloride,
using PBS at 20 °C. For the determination of the masses of the 0.1 M MES (pH 5.4), and 23% PEG 6000] for the Cμ2 domain
individual domains and mutants, sedimentation velocity experi- and buffer C4 [15% PEG 8000 and 0.1M Hepes (pH 7.2)] for the
ments were performed at a rotor speed of 50,000 rpm. Sedi- Cμ4 domain. In both cases, crystals grew to a final size of ∼50 ×
mentation was detected at 230, 280, or 300 nm, depending on 50 × 25 μm3 within 48 h. Crystals were either soaked in 2 M
protein concentration. For the Cμ4tp oligomer, sedimentation lithium sulfate at pH 5.4 (Cμ2 domain) or buffer C4 with addi-
velocity runs were performed with protein concentrations of tional 25% PEG 200 (Cμ4 domain) before being cooled in a

Müller et al. www.pnas.org/cgi/content/short/1300547110 1 of 11


stream of liquid nitrogen at 100 K. Native diffraction datasets were experiment. Distance restraints were extracted from 15N and 13C
collected using synchrotron radiation at the X06SA-beamline edited NOESY spectra with mixing times of 70 ms (17, 18). Fi-
(Swiss Lightsource) (Cμ2 domain at 1.3 Å resolution, Table S1) nally, 15N R1 and R2 relaxation rates, as well as {1H}-15N het-
or on a Bruker Microstar/X8 Proteum (Bruker AXS Inc.) with eronuclear NOE values, were measured at 600 MHz proton
a Cu rotating anode (λ = 1.54 Å; Cμ4 domain at 1.9 Å reso- Larmor frequency at 298 K (19). All data were acquired with a
lution, Table S1). 300-μM uniformly 15N,13C-labeled Cμ3C414S sample, except for
For structure determination of the Cμ2 domain, data were the stereospecific assignment of valine and leucine side chains,
processed using the program package XDS (5). The self-rotation which was based on a 1H-13C heteronuclear multiple quantum
function, calculated with MOLREP (6), revealed noncrystallo- correlation acquired on a 10% 13C-labeled sample as previously
graphic twofold symmetries indicating two Cμ2 domain mole- described (20, 21).
cules in the asymmetric unit cell. Determination of the crystal NOE cross-peaks were assigned automatically by the software
structure was performed by molecular replacement using the CYANA 3.0 (22) and then inspected manually for assignment and
program PHASER (7). Coordinates of the corresponding do- completeness. The distance restraints extracted from the CYANA
main of the human IgE (1O0V, residues 228–328) were applied calculation as well as torsion angles derived by TALOS+ (23) were
as starting model for the ligand structure. After rigid body used in a water refinement calculation in Aria 2.2 (24). Finally, the
and preliminary positional refinement with REFMAC (8) as quality of the structure ensemble was validated using WHATIF
well as noncrystallographic symmetry averaging using the soft- (25) and PROCHECK (26). The data of the Cμ3 domain have been
ware package MAIN (9), both Cμ2 domains were traced. The deposited in the Protein Data bank (ID code 4BA8).
refined model resulted in improved density and allowed final- To map the Cμ3/Cμ4 interface, chemical shift perturbation
ization of the last missing secondary structures and loop con- analysis was performed, following chemical shifts in the HNCA
nections. The structures were completed in successive rounds spectra of the 15N-labeled dimeric Cμ3C414S–Cμ4 construct
using the interactive 3D graphic programs COOT (10) and an- compared with the labeled Cμ3C414S and plotted for each res-
isotropically refined with REFMAC5 (8). Water molecules were idue in Cμ3C414S. The chemical shift perturbations have been
located either by using ARP/wARP (11) within the CCP4i GUI calculated according to Eq. S2:
(12) or by using COOT (10). All solvent molecules were verified sffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi

manually in COOT (10). Temperature factors were corrected with  
δ15N 2
restraints between bonded atoms and between noncrystallo- CSPðppmÞ = + δ1H 2 : [S2]
graphic symmetry related atoms using translation/libration/ 10
screw (TLS) parameters with current crystallographic values of
Rwork = 0.138 and Rfree = 0.168 (13). Data collection and re- The chemical shift perturbations of the different nuclei have been
finement statistics are summarized in Table S1. Coordinates normalized using their gyromagnetic ratio.
were confirmed to have good stereochemistry from the Ram-
achandran plot using PROCHECK (14), which displays 99.0% of Small Angle X-Ray Scattering Measurements. Small angle X-ray
residues in the most favored region and 1.0% of residues in the scattering (SAXS) data for solutions of Cμ4tp were recorded
additionally allowed regions. The asymmetric unit cell contains on an in-house SAXS instrument (SAXSess mc2; Anton Paar
two Cμ2 domains (with the N-terminal Met and the last two Ala GmbH) equipped with a Kratky camera, a sealed X-ray tube
residues at the C terminus being structurally disordered) and 291 source, and a one-dimensional reverse-biased silicon diode array
water molecules (Table S1). The data of the Cμ2 domains have detector (Mythen 1K; Dectris). The scattering patterns were
been deposited in the Protein Data bank (PDB ID code 4JVU). measured with a 30-min exposure time (three frames, each 10 min)
For structure determination of the Cμ4 domain, data were for several solute concentrations in the range from 188 to 751 μM.
processed with the Proteum 2 software suite (version 2006; Radiation damage was excluded based on a comparison of in-
Bruker AXS Inc.). MOLREP revealed twofold symmetries sug- dividual frames of the 30 min exposures, where no changes were
gesting four Cμ4 domains in the asymmetric unit cell. Initial detected. A range of momentum transfer of 0.01 < s < 0.7 Å−1
phases were received by Patterson search calculations applying was covered (s = 4π sin(θ)/λ, where 2θ is the scattering angle and
PHASER and using the coordinates of the previously determined λ = 1.5 Å is the X-ray wavelength).
model structure of the Cμ2 domain as starting model. Similarly SAXS data for solutions of Cμ4tp and Cμ3C414S–Cμ4 in the
as described for the structure elucidation of the Cμ2 domain, presence of 5 mM DTT were recorded at the X33 beamline of
cycles of model building, fourfold noncrystallographic symmetry the European Molecular Biology Laboratory at Deutsches Elek-
averaging for electron density map improvement and TLS tronen Synchrotron using a MAR345 image plate detector. The
refinement yielded the final Cμ4 model. Data collection and scattering patterns were measured with a 2-min exposure time
refinement statistics are summarized in Table S1. The stereo- (eight frames, each 15 s) for several solute concentrations in the
chemical quality of the Cμ4 domains were assessed with PRO- range from 110 to 578 μM. Radiation damage was excluded based
CHECK, displaying in total 97.9% of residues in the most on a comparison of individual frames of the 2-min exposures,
favored region and 2.1% of residues in the additionally allowed where no changes were detected. Using the sample–detector
regions. The asymmetric unit cell contains four Cμ4 structures distance of 2.7 m, a range of momentum transfer of 0.01 < s < 0.6
(with the N-terminal five amino acids being structurally disor- Å-1 was covered (s = 4π sin(θ)/λ, where 2θ is the scattering angle
dered) and 428 water molecules (Table S1). The data of the Cμ4 and λ = 1.5 Å is the X-ray wavelength).
domains have been deposited in the Protein Data bank (PDB All SAXS data were analyzed with the package ATSAS. The
ID code 4JVW). Molecular illustrations were prepared using data were processed with the SAXSQuant software (version 3.9)
the PyMOL Molecular Graphics System (DeLano Scientific). and desmeared using the program GNOM (27). The forward
scattering, I(0), the radius of gyration, Rg, the maximum di-
NMR Spectroscopy and Structure Calculation. NMR data were ac- mension, Dmax, and the interatomic distance distribution func-
quired at 298 K on a Bruker Avance III 600-MHz spectrometer tions, (P(R)), were computed with the program GNOM (27).
equipped with a TCI CryoProbe head. Processing was done with The masses of the solutes were evaluated by comparison of the
NMRPipe (15) and analysis was carried out using Sparky (16). forward scattering intensity with that of a human serum albumin
The backbone resonances were assigned manually based on reference solution (molecular mass 69 kDa). The structure of the
HNCACB, HN(CO)CACB, HNCO, and HN(CA)CO experi- Cμ4tp hexamer was modeled using the program CORAL (28).
ments (17). Side chains were assigned using an (H)CCH-TOCSY The crystal structure of Cμ4 was taken as input, a C6 symmetry

Müller et al. www.pnas.org/cgi/content/short/1300547110 2 of 11


was defined, and the flexible N- and C-terminal regions (residues calculation in which none of the Cμ4 interfaces in the hexamer
E446–H448 and Gly557–Tyr576) were randomized. No con- was restrained (Fig. S6). C-alpha coordinates of the Cμ4tp hex-
nections between the Cμ4 dimers have been defined owing to amer of dimers SAXS model have been deposited in the Protein
lack of experimental data defining the interface. A total of 50 Data Bank (ID code 4BLE).
structures were calculated, and the best scoring structure (based To model the structure of the entire IgM, the Cμ4tp hexamer
on the solvation free energy gain upon formation of the in- modeled based on SAXS data was taken as input for its exten-
terface, ΔiG) was selected to model the flexible C terminus. To sion with Cμ2, Cμ3, and the flexible linkers/termini (Cμ2–Cμ3,
test whether the additional Cμ4 interface observed in the crystal Cμ3–Cμ4, and the tail piece extending Cμ4). The following input
structures can be used to construct the structure of the hexamer and data were used in the rigid body docking program CORAL:
we calculated an ensemble of structures using the crystal struc- the atomic resolution structures of (i) Cμ2, (ii) Cμ3, (iii) the
ture of the Cμ4 dimer of dimers as input. The calculations were SAXS model of the Cμ4 hexameric ring, (iv) the disulfide link-
carried out using the program CORAL (28) with the same set- ages (C414 and C575), and (v) the Cμ3–Cμ4 interface derived
tings as described above, but defining C3 symmetry and distance from NMR data as input. The connections of Cμ4 to the Cμ3
restraints between the dimers of dimers according to the addi- domains has been made based on the NMR chemical shift per-
tional interface observed in the crystal structure. Because the turbations observed for the free Cμ3C414S domain compared
Cμ4 dimers of dimers might adopt orientations different from with the Cμ3C414S–Cμ4 dimer (Fig. S4). Based on that, am-
those in the hexamer, we used all possible combinations of dis- biguous distance restraints defining that any proton in Cμ3 res-
tance restraints (i.e., contacts of chain A with chain B/C of the idues F354 and F358 has to be within a distance closer than 6 Å
next Cμ4tp dimer of dimers, chain A–chain C/B, chain D–chain to any proton of Cμ4 have been defined. A total of 50 structures
B/C, and chain D–chain C/B). The resulting structures are in were calculated, and the structure with the best fit to the ex-
worse agreement with the experimental data compared with the perimental data was selected to prepare Fig. 4C.

1. Mayer M, Buchner J (2004) Refolding of inclusion body proteins. Methods Mol Med 16. Goddard TD, Kneller D (2007) SPARKY3 (Univ of California, San Francisco).
94:239–254. 17. Sattler M, Schleucher J, Griesinger C (1999) Prog Nucl Magn Reson Spectrosc 34:
2. Schuck P (2000) Size-distribution analysis of macromolecules by sedimentation 93–158.
velocity ultracentrifugation and lamm equation modeling. Biophys J 78(3):1606–1619. 18. Otting G, Wüthrich K (1990) Heteronuclear filters in two-dimensional [1H,1H]-NMR
3. Lebowitz J, Lewis MS, Schuck P (2002) Modern analytical ultracentrifugation in spectroscopy: Combined use with isotope labelling for studies of macromolecular
protein science: A tutorial review. Protein Sci 11(9):2067–2079. conformation and intermolecular interactions. Q Rev Biophys 23(1):39–96.
4. Bolen DW, Santoro MM (1988) Unfolding free energy changes determined by the 19. Farrow NA, et al. (1994) Backbone dynamics of a free and phosphopeptide-
linear extrapolation method. 2. Incorporation of delta G degrees N-U values in a complexed Src homology 2 domain studied by 15N NMR relaxation. Biochemistry
thermodynamic cycle. Biochemistry 27(21):8069–8074. 33(19):5984–6003.
5. Kabsch W (1993) Automatic processing of rotation diffraction data from crystals of 20. Szyperski T, Neri D, Leiting B, Otting G, Wüthrich K (1992) Support of 1H NMR
initially unknown symmetry and cell constants. J Appl Cryst 26:795–800. assignments in proteins by biosynthetically directed fractional 13C-labeling. J Biomol
6. Vagin A, Teplyakov A (2010) Molecular replacement with MOLREP. Acta Crystallogr NMR 2(4):323–334.
D Biol Crystallogr 66(Pt 1):22–25. 21. Coggins BE, et al. (2003) Structure of the LpxC deacetylase with a bound substrate-
7. McCoy AJ, et al. (2007) Phaser crystallographic software. J Appl Cryst 40(Pt 4): analog inhibitor. Nat Struct Biol 10(8):645–651.
658–674. 22. Güntert P (2009) Automated structure determination from NMR spectra. Eur Biophys
8. Murshudov GN, Vagin AA, Dodson EJ (1997) Refinement of macromolecular J 38(2):129–143.
structures by the maximum-likelihood method. Acta Crystallogr D Biol Crystallogr 23. Shen Y, Delaglio F, Cornilescu G, Bax A (2009) TALOS+: A hybrid method for
53(Pt 3):240–255. predicting protein backbone torsion angles from NMR chemical shifts. J Biomol NMR
9. Turk D (1992) Improvement of a programme for molecular graphics and manipulation 44(4):213–223.
of electron densities and its application for protein structure determination. PhD 24. Linge JP, Williams MA, Spronk CA, Bonvin AM, Nilges M (2003) Refinement of protein
thesis (Technische Universität München). structures in explicit solvent. Proteins 50(3):496–506.
10. Emsley P, Cowtan K (2004) Coot: Model-building tools for molecular graphics. Acta 25. Vriend G (1990) WHAT IF: A molecular modeling and drug design program. J Mol
Crystallogr D Biol Crystallogr 60(Pt 12 Pt 1):2126–2132. Graph. 8(1):52–56, 29.
11. Perrakis A, Morris R, Lamzin VS (1999) Automated protein model building combined 26. Laskowski RA, Rullmannn JA, MacArthur MW, Kaptein R, Thornton JM (1996) AQUA
with iterative structure refinement. Nat Struct Biol 6(5):458–463. and PROCHECK-NMR: Programs for checking the quality of protein structures solved
12. Winn MD, et al. (2011) Overview of the CCP4 suite and current developments. Acta by NMR. J Biomol NMR 8(4):477–486.
Crystallogr D Biol Crystallogr 67(Pt 4):235–242. 27. Svergun DI (1992) Determination of the regularization parameter in indirect-
13. Brünger AT (1992) Free R value: A novel statistical quantity for assessing the accuracy transform methods using perceptual criteria. J Appl Cryst 25:495–503.
of crystal structures. Nature 355(6359):472–475. 28. Petoukhov MV, Svergun DI (2005) Global rigid body modeling of macro-
14. Laskowski RA, MacArthur MW, Moss DS, Thornton JM (1993) PROCHECK: A program molecular complexes against small-angle scattering data. Biophys J 89(2):
to check the stereochemical quality of protein structures. J Appl Cryst 26:283–291. 1237–1250.
15. Delaglio F, et al. (1995) NMRPipe: A multidimensional spectral processing system
based on UNIX pipes. J Biomol NMR 6(3):277–293.

Müller et al. www.pnas.org/cgi/content/short/1300547110 3 of 11


Fig. S1. Stability against thermal and chemical unfolding. (A) Temperature denaturation of the Cμ2 (black), Cμ3C414S (red), Cμ4 (blue), and Cμ4tpC575S
(green) domains, followed by the far-UV CD signal at 205 nm at a heating rate of 20 °C/h. Data were fitted by a Boltzmann function. GdmCl-induced equi-
librium unfolding (filled circles) and refolding (open circles) of the (B) Cμ2, (C) Cμ3C414S, (D) Cμ4, and (E) Cμ4tpC575S domains. GdmCl-induced unfolding/
refolding transitions were monitored by intrinsic tryptophan fluorescence. Data were fitted by a two-state unfolding model.

Müller et al. www.pnas.org/cgi/content/short/1300547110 4 of 11


Fig. S2. Characterization of domain oligomerization. SEC profiles of (A) Cμ2 (dotted line) and Cμ2C337S domain, (B) Cμ3 domain, (C) Cμ3C414S domain, (D)
Cμ4, (E) Cμ4tp domain, and (F) Cμ4tpC575S domain. The proteins were loaded with 1 μM (black line), 10 μM (red line), and 100 μM (blue line). The fluorescence
signal was normalized to the highest peak intensity.

Müller et al. www.pnas.org/cgi/content/short/1300547110 5 of 11


Fig. S3. Sedimentation velocity analytical ultracentrifugation runs of (A) Cμ2, (B) Cμ2C337S, (C) Cμ3C337S, (D) Cμ4, and (E) Cμ4tpC575S. Runs were performed
at 50,000 rpm in PBS buffer. Representative analytical ultracentrifugation sedimentation equilibrium runs of (F) Cμ2C337S (10 μM), (G) Cμ3C414S (25 μM), (H)
Cμ4 (5 μM), and (I) Cμ4tpC575S (20 μM). Runs were performed at 34,000 rpm (black triangles), 38,000 rpm (red circles), and 42,000 rpm (blue squares) in PBS
buffer. Data were fitted globally to a self-association monomer–dimer equilibrium model (solid lines).

Müller et al. www.pnas.org/cgi/content/short/1300547110 6 of 11


Fig. S4. Experimental 15N backbone amide relaxation parameters and chemical shift perturbation (CSP) of the free Cμ3C414S domain compared with the
Cμ3C414S–Cμ4 dimer. (A) hetNOE values, (B) longitudinal (R1) rates, and (C) comparison of transverse relaxation rates R2, measured directly (black) and derived
from R1ρ (red). All experiments have been recorded at a Larmor frequency of 600 MHz at 298 K. Correlation times values were determined from 15N relaxation
data as described in SI Materials and Methods. (D) Chemical shifts in free form (Cμ3C414S) and the tandem construct (Cμ3C414S–CH4) have been assigned using
3D HNCA spectra. The secondary structure is pictured above (α-helixes in red, β-sheets in blue).

Müller et al. www.pnas.org/cgi/content/short/1300547110 7 of 11


Fig. S5. Comparison and alignments of Ig constant domains. (A) Comparison of the various Ig dimerization modes (cartoon representation). (Top) Display of
the respective chain A. (Middle) Display of the dimer form. (Bottom) Surface representation of the dimers. (B) Structure and sequence alignments of IgM Cμ4
(pink), IgA Cα3 (blue), and IgE Ce4 (green). The amino acids involved in the dimer interface are colored in green. Interface residues that are not conserved are
shown in the alignment in red and highlighted in the structure by red arrows. The following coordinates were used (IgA, 1OW0; IgE,1O0V; IgG, 3HKF).

Müller et al. www.pnas.org/cgi/content/short/1300547110 8 of 11


Fig. S6. Summary of the SAXS data and modeling. (A) Guinier plots of unmerged Cμ4tp scattering data collected at different concentrations. The Rgs and
calculated molecular weights (based on BSA standard) are shown. (B–D) SAXS data showing a comparison of the experimental radial density distributions of
Cμ4tp (B) and Cμ4tp (C) and Cμ3C414S–Cμ4 (D) in the presence of 5 mM DTT at different concentrations. (E) Modeling of the hexamer structure using the
program CORAL and defining C3 symmetry. Different combinations of the additional Cμ4 interface observed in the crystal structures were used as restraints
(SI Materials and Methods gives details). The fit (χ) between the experimental and back-calculated data are shown. (F) SAXS data showing a comparison of the
experimental radial density distribution of Cμ4tp (blue) and the Cμ4 dimer (red) [back-calculated from the crystal structure determined here, using the program
Crysol (1)]. (G) Comparison of experimental Cμ4tp SAXS data with SAXS data back-calculated from the CORAL model of the Cμ4tp hexamer. Both the s and I(s)
axes are shown in a logarithmic representation. The angular ranges from 0.0014–0.6 nm−1 are compared.

1. Svergun DI, Barberato C, Koch MHJ (1995) CRYSOL – A program to evaluate X-ray solution scattering of biological macromolecules from atomic coordinates. J Appl Cryst 28:768–773.

Müller et al. www.pnas.org/cgi/content/short/1300547110 9 of 11


Table S1. Crystallographic statistics
Parameters Cμ2 domain Cμ4 domain

Crystal parameters
Space group C2 C2
Cell constants a = 92.44 Å, b = 44.78 Å, c = 54.57 Å a = 169.16 Å, b = 41.21 Å, c = 67.1 Å
α = γ = 90°; β =118° α = γ = 90°; β = 92.3°
Data collection
Beamline X06SA, SLS CuKα, rotating anode
Wavelength, Å 1.0 1.5418
Resolution range, Å* 40–1.30 (1.4–1.30) 40–2.0 (2.1–2.0)
No. of observations 15,5963 15,4481
No. of unique reflections† 48,207 31,721
Completeness, %* 99.3 (99.6) 100 (100)
Rmerge, %*,‡ 3.8 (30.9) 4.6 (35.4)
I/σ (I)* 16.1 (5.2) 19.0 (3.1)
Refinement (REFMAC5)
Resolution range, Å 15–1.3 15–2.0
No. of reflections working set 45,796 28,566
No. of reflections test set 2,414 1,537
No. of nonhydrogen 1,615 3,356
No. of solvent water 291 428
Rwork/Rfree, %§ 13.8/16.8 19.7/23.4
Rmsd bond, Å{/angles, ° 0.019/1.857 0.007/1.256
Average B-factor, Å2 20.0 26.1
Ramachandran plot, %k 99.0/1.0/0.0 97.9/2.1/0.0
PDB ID code 4JVU 4JVW

*The values in parentheses of resolution range, completeness, Rmerge, and I/σ (I) correspond to the last resolution shell.

Friedel pairs were treated as different reflections.
‡ P P P
Rmerge ðIÞ = hkl j ½IðhklÞj -IðhklÞ =½ hkl Ihkl , where I(hkl)j is the jth measurement of the intensity of reflection hkl and <I(hkl)> is the
average intensity.
P P
§
R = hkl kFobs j − jFcalc k= hkl jFobs j; where Rfree is calculated without a sigma cutoff for a randomly chosen 5% of reflections, which were
not used for structure refinement, and Rwork is calculated for the remaining reflections.
{
Deviations from ideal bond lengths/angles.
k
Number of residues in favored region/allowed region/outlier region.

Table S2. NMR and refinement statistics for Cμ3C414S domain


Parameters For all residues For residues 3-29,38-51,66-100

NOE-based distance restraints


Intraresidual, sequential 772
Medium range (2 ≤ ji-jj ≤ 4) 158
Medium range (ji-jj ≥ 5) 563
Total 1,493
Violated 0
Other restraints
ϕ + ψ dihedral restraints 100
Coordinate precision
Backbone, A 0.49 ± 0.10
Heavy atom, A 0.94 ± 0.11
Common Interface for NMR structure Generation
Red, % 16 11
Orange, % 18 14
Green, % 67 67
Whatcheck
First-generation packing quality 2.918 ± 1.251
Second-generation packing quality 4.617 ± 1.785
Ramachandran plot appearance 0.886 ± 0.227
χ1/χ2 rotamer normality −3.471 ± 0.486
Backbone conformation 1.647 ± 0.186
Ramachandran plot, %
Most favored regions 91.9
Allowed regions 7.0
Generously allowed regions 0.9
Disallowed regions 0.3
PDB ID code 4BA8

Müller et al. www.pnas.org/cgi/content/short/1300547110 10 of 11


Other Supporting Information Files

Dataset S1 (TXT)

Müller et al. www.pnas.org/cgi/content/short/1300547110 11 of 11

You might also like