You are on page 1of 12

http://informahealthcare.

com/mbc
ISSN: 0968-7688 (print), 1464-5203 (electronic)

Mol Membr Biol, 2014; 31(7–8): 250–261


! 2014 Informa UK Ltd. DOI: 10.3109/09687688.2014.990998

ORIGINAL ARTICLE

Purification and interaction analyses of two human lysosomal vitamin


B12 transporters: LMBD1 and ABCD4
Justin C. Deme1, Mark A. Hancock2, Xiaobing Xia3, Chitra A. Shintre3, Maria Plesa1,4, Jaeseung C. Kim1,4,
Elisabeth P. Carpenter3, David S. Rosenblatt4, and James W. Coulton1
1
Department of Microbiology and Immunology, McGill University, Montreal, Canada, 2McGill University Surface Plasmon Resonance (SPR) Facility,
Montreal, Canada, 3Structural Genomics Consortium, University of Oxford, Oxford, UK, and 4Department of Human Genetics, McGill University,
Montreal, Canada
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15

Abstract Keywords
Mutations in human LMBRD1 and ABCD4 prevent lysosomal export of vitamin B12 to the Cobalamin, inborn errors of metabolism,
cytoplasm, impairing the vitamin B12-dependent enzymes methionine synthase and membrane proteins, protein–protein
methylmalonyl-CoA mutase. The gene products of LMBRD1 and ABCD4 are implicated in interactions, vitamin B12 metabolism
vitamin B12 transport at the lysosomal membrane and are proposed to act in complex.
To address the mechanism for lysosomal vitamin B12 transport, we report the novel History
recombinant production of LMBD1 and ABCD4 for detailed biophysical analyses. Using blue
native PAGE, chemical crosslinking, and size exclusion chromatography coupled to multi-angle Received 5 August 2014
light scattering (SEC-MALS), we show that both detergent-solubilized LMBD1 and detergent- Revised 18 October 2014
solubilized ABCD4 form homodimers. To examine the functional binding properties of these Accepted 8 November 2014
For personal use only.

proteins, label-free surface plasmon resonance (SPR) provides direct in vitro evidence that: Published online 23 December 2014
(i) LMBD1 and ABCD4 interact with low nanomolar affinity; and (ii) the cytoplasmic vitamin
B12-processing protein MMACHC also interacts with LMBD1 and ABCD4 with low nanomolar
affinity. Accordingly, we propose a model whereby membrane-bound LMBD1 and ABCD4
facilitate the vectorial delivery of lysosomal vitamin B12 to cytoplasmic MMACHC, thus
preventing cofactor dilution to the cytoplasmic milieu and protecting against inactivating side
reactions.

Introduction generated from homocysteine by cytoplasmic MS and


MeCbl. AdoCbl is used by mitochondrial MCM for the
Dubbed nature’s most beautiful cofactor (Stubbe, 1994),
isomerization of methylmalonyl-CoA to succinyl-CoA for
cobalamin (Cbl or vitamin B12) is the largest of all vitamins
entry into the tricarboxylic acid (TCA) cycle.
and the most structurally complex (Hodgkin et al., 1956). It is
Nine inherited defects of intracellular Cbl metabolism,
composed of a central cobalt atom that is coordinated by a
designated cblA to cblG, cblJ and mut, have been discovered
tetrapyrrole framework termed the corrin ring and by two axial
through somatic complementation assays (Coelho et al., 2012;
ligands. The lower axial ligand is in a ‘‘base-on’’ state when the
Froese & Gravel, 2010). Defects in this pathway result in MCM
pendant dimethylbenzimidazole (DMB) base, connected to the
and MS substrate accumulation; the resultant methylmalonic
corrin ring, coordinates cobalt. Protonation of DMB or
aciduria and/or homocystinuria can give rise to severe
histidine substitution results in the ‘‘base-off’’ or ‘‘base-off/
hematological and neurological conditions. Patients within
His-on’’ forms of Cbl, respectively. Cbl diversity is provided
the cblF (Rosenblatt et al., 1985; Watkins & Rosenblatt, 1986)
by its upper axial ligand that covalently attaches to the central
or cblJ (Coelho et al., 2012; Kim et al., 2012) groups have
cobalt. Methyl, 50 -deoxyadenosyl, hydroxyl, or cyano groups
similar clinical phenotypes: hypotonia, lethargy, poor feeding,
can serve as upper axial ligand, giving rise to several forms of
bone marrow suppression, macrocytic anemia and heart
Cbl: methylcobalamin (MeCbl), 50 -adenosylcobalamin
defects. Cultured fibroblasts from cblF patients accumulate
(AdoCbl), hydroxocobalamin (OHCbl), and cyanocobalamin
nonprotein-bound Cbl in lysosomes (Rosenblatt et al., 1985;
(CNCbl). Methylmalonyl-CoA mutase (MCM; EC 5.4.99.2)
Vassiliadis et al., 1991). The accumulation of unprocessed,
and methionine synthase (MS; EC 2.1.1.13) use Cbl as cofactor
nonprotein-bound Cbl within the cell has also been reported for
to catalyze chemically unique reactions. Methionine is
cblJ patients (Coelho et al., 2012; Kim et al., 2012) who mimic
the cblF phenotype (Rosenblatt et al., 1985). Mutations in
Correspondence: Prof. James W. Coulton, Department of Microbiology LMBRD1 (Rutsch et al., 2009) and ABCD4 (Coelho et al.,
and Immunology, McGill University, Lyman Duff Medical Building,
Room 403, 3775 University Street, Montreal, Canada H3A 2B4. Tel: +1 2012) were recently identified as the cause of the cblF and cblJ
514 398 3929. Fax: +1 514 398 7052. E-mail: james.coulton@mcgill.ca defects, respectively. Transfection of LMBRD1 into cblF
DOI: 10.3109/09687688.2014.990998 Interactions: LMBD1, ABCD4, MMACHC 251

fibroblasts or ABCD4 into cblJ fibroblasts rescues AdoCbl and chromatography coupled to multi-angle light scattering (SEC-
MeCbl synthesis and restores MCM and MS function, MALS), we determined that detergent-solubilized LMBD1
implicating these gene products in the lysosomal export of and ABCD4 formed homodimers in solution. Adopting label-
Cbl (Coelho et al., 2012; Rutsch et al., 2009). free, real-time surface plasmon resonance (SPR), LMBD1 and
LMBRD1 encodes LMBD1, a 61.4 kDa membrane protein ABCD4 interacted with low nanomolar affinity. As predicted
of 540 residues and six predicted lysosomal N-glycosylation by phage display, SPR also confirmed that MMACHC can
sites (Rutsch et al., 2009). LMBD1 localizes primarily to the interact with both LMBD1 and ABCD4 with low nanomolar
lysosome (Chapel et al., 2013; Rutsch et al., 2009) but also affinities. Our results are consistent with a model that
appears in the plasma membrane (Tseng et al., 2013) and implicates LMBD1 and ABCD4 in the vectorial delivery of
perinuclear region (Rutsch et al., 2009). Based on topology lysosomal Cbl to cytoplasmic MMACHC. Our direct evi-
modeling programs, predicted glycosylation pattern and dence for protein interactions involved in egress of lysosomal
freeze-fracture replica immunolabeling experiments (Rutsch Cbl advances basic understanding of early intracellular Cbl
et al., 2009), LMBD1 spans the lysosomal membrane via nine metabolism that, to date, has largely been limited to studies in
transmembrane (TM) helices with a lysosomal N-terminus and cultured cells.
cytoplasmic C-terminus. LMBD1 demonstrates homology to
the lipocalin-1 interacting membrane receptor (LIMR) Methods
involved in the internalization of lipocalins that bind small
Protein purification
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15

hydrophobic molecules like steroids and lipids (Rutsch et al.,


2009, 2011). cDNA corresponding to LMBRD1 or ABCD4 were cloned into
ABCD4 encodes ABCD4, a non-glycosylated protein of 606 the pFB-CT10HF-LIC transfer vector that encodes down-
residues with molecular mass of 68.6 kDa (Holzinger et al., stream sequences in the following order: a TEV cleavage site
1997; Kashiwayama et al., 2009). While ABCD4 was origin- (ENLYFQS); a decahistidine tag (His10); and a FLAG
ally identified as peroxisomal (Shani et al., 1997), recent (DYKDDDDK) tag. Baculoviruses were constructed accord-
studies have suggested its localization to the ER (Kashiwayama ing to the Bac-to-Bac system (Life Technologies). Spodoptera
et al., 2009) or within the lysosome of human beings (Coelho frugiperda (Sf9) insect cells were grown in shaker flasks in
et al., 2012) and rats (Chapel et al., 2013). ABCD4 consists of I-MAX (Wisent) or Sf-900 II SFM media (Life Technologies)
two domains: an N-terminal TM domain and a C-terminal at 27  C and infected with amplified recombinant baculovirus
For personal use only.

nucleotide-binding domain (NBD) (Coelho et al., 2012). The (MOI of 0.25–0.5) for 72 h. Following infection, cells were
TM domain has six predicted TM helices (Coelho et al., 2012; collected by centrifugation (1000 g for 10 min), washed in
Kashiwayama et al., 2009) whereas the NBD contains highly PBS, flash frozen in liquid nitrogen, and stored at 80  C.
conserved motifs involved in ATP and Mg2+ binding, Thawed, infected cells were resuspended (25 ml/L of cells)
including Walker A and B motifs plus a modified ATP-binding in cold lysis buffer (50 mM HEPES pH 7.5 plus a Complete
cassette (ABC) signature motif (Coelho et al., 2012). Based on EDTA-free protease inhibitor cocktail tablet [Roche]) and
bioinformatics predictions and loop-directed glycosylation lysed by three passes through an EmulsiFlex-C5 homogenizer
experiments, the topology of ABCD4 has both the N- and (Avestin) at 15 000 psi. Supernatants collected following
C-termini exposed to the cytoplasm (Coelho et al., 2012; centrifugation (10 000 g at 4  C for 10 min) were supplemented
Kashiwayama et al., 2009). ABCD4 is a member of the D with NaCl to a final concentration of 1 M. Membranes were
subfamily of ABC half transporters that dimerize to form recovered by ultracentrifugation (100 000 g at 4  C for 75 min)
active, full transporters (Hillebrand et al., 2007; Liu et al., and resuspended in lysis buffer plus 1 M NaCl using a Dounce
1999). ABCD4 has significant amino acid identity (25–27%) homogenizer. After an additional round of ultracentrifugation
with these other members, but lacks a hydrophilic (100 000 g at 4  C for 75 min), membranes expressing LMBD1
N-terminal tail that mediates localization to the peroxisome were resuspended in extraction buffer [50 mM HEPES pH 7.5,
(Kashiwayama et al., 2009). 200 mM NaCl, 5% glycerol, 1% (w/v) n-dodecyl-b-D-malto-
The functional interplay between LMBD1 and ABCD4 was side (DDM) and 0.1% (w/v) cholesteryl hemisuccinate (CHS)].
originally proposed by a fluorescence microscopy study that Membranes expressing ABCD4 were treated with extraction
demonstrated colocalization of both proteins to the lysosomal buffer plus 0.5 mM tris(2-carboxyethyl)phosphine (TCEP) and
membrane (Coelho et al., 2012). Further supported by partial 1.0 mM MgCl2 for 16 h at 4  C by slow mixing on a rotating
functional rescue by transfection of ABCD4 cDNA into mixer.
cultured cblF fibroblasts (Coelho et al., 2012), it is suggested Detergent-extracted membranes collected after ultracentri-
that ABCD4 and LMBD1 form a complex that is critical to the fugation (100 000 g at 4  C, 45 min) were supplemented
export of lysosomal Cbl to the cytoplasm. These lysosomal with 10 mM imidazole. Ni-NTA Superflow (Qiagen), pre-
membrane proteins are also proposed to mediate the direc- equilibrated in purification buffer (50 mM HEPES pH 7.5,
tional transfer of Cbl to the downstream cytoplasmic 200 mM NaCl, 5% glycerol, 0.03% DDM, 0.003% CHS, plus
Cbl-processing protein MMACHC (Froese & Gravel, 2010; 0.5 mM TCEP and 1 mM MgCl2 for ABCD4 preparations)
Rutsch et al., 2011). with 10 mM imidazole, was added to detergent-extracted
This study provides detailed characterizations of the membranes, followed by gentle mixing for 1 h at 4  C. Resin
protein interactions involved in Cbl egress from the lysosome. was captured through a gravity column and washed with 15
To our knowledge, this is the first description of recombinant column volumes (CV) of purification buffer plus 20 mM
LMBD1 and ABCD4 production. Exploiting blue native imidazole, followed by stringent washing using 15 CV of
PAGE (BN-PAGE), chemical crosslinking, and size exclusion purification buffer supplemented with 40 mM imidazole.
252 J. C. Deme et al. Mol Membr Biol, 2014; 31(7–8): 250–261

Protein was eluted by addition of purification buffer containing filtration buffer. For LMBD1 crosslinking, ultrapure EM grade
300 mM imidazole and exchanged into gel filtration buffer glutaraldehyde (EM Sciences) was mixed with samples to a
(20 mM HEPES pH 7.5, 200 mM NaCl, 0.02% DDM, 0.002% final concentration of 0.025% (w/w). Aliquots of LMBD1 were
CHS, plus 0.5 mM TCEP and 1.0 mM MgCl2 for ABCD4), removed from the reaction mixture following incubation for
using a PD Minitrap G-25 desalting column (GE Healthcare 10, 30, and 60 min at 25  C. To ensure specific crosslinking
Life Sciences). TEV protease was added to each sample in a 1:2 with glutaraldehyde, a control reaction of LMBD1 preincu-
mass ratio of TEV:protein, and Peptide-N-Glycosidase bated with 1% SDS followed by glutaraldehyde treatment
F (PNGase F) was concurrently added to LMBD1 samples (0.025%) for 1 h was performed concurrently. For ABCD4,
in a 1:30 mass ratio of PNGaseF:LMBD1 and incubated for formaldehyde (Pierce) was added to a final concentration of
72 h at 4  C. 0.075% (w/v) to initiate crosslinking; aliquots were removed
Cleaved LMBD1 and ABCD4 were separated from His6- following incubation for 30 min, 1 h, and 4 h at 25  C. To
tagged TEV protease and PNGase F by incubation for 1 h terminate crosslinking for all reactions, aliquots were treated
with Ni-NTA Superflow resin at 4  C. Resin was collected in a with SDS-PAGE sample buffer for 10 min prior to loading onto
gravity column and flow through was pooled and concentrated a 4–12% polyacrylamide gel prepared in-house. Following
to 1 mg/ml using a centrifugal filter (Millipore; Amicon electrophoresis, crosslinked samples were visualized by silver
100 kDa MWCO). MMACHC and MMADHC were cloned, staining.
expressed, and purified as previously described (Deme et al.,
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15

2012; Plesa et al., 2011). Protein concentrations were


Size exclusion chromatography – multi-angle light
derived from A280 measurements using the theoretical extinc-
scattering (SEC-MALS)
tion coefficients and molecular masses for LMBD1
(106 230 M1 cm1; 62.2 kDa), ABCD4 (92 250 M1 cm1; For molecular mass determination of protein-detergent com-
69.4 kDa), MMACHC (47 900 M1 cm1; 31.9 kDa), and plexes (PDCs), size exclusion chromatography was coupled to
MMADHC (32 430 M1 cm1; 31.9 kDa). multi-angle light scattering, refractive index, and UV detec-
Purity of LMBD1 and ABCD4 was assessed by SDS- tion. Both LMBD1 and ABCD4 were applied to a Superdex
PAGE using 8% polyacrylamide gels followed by silver 200 10/300 GL column (GE Healthcare Life Sciences)
staining. Apparent protein molecular masses derived from connected to a Waters 2695 HPLC system at 20  C at a flow
SDS-PAGE were estimated using molecular mass standards. rate of 0.25 ml/min using gel filtration buffer. Elution was
For personal use only.

Monodispersity of LMBD1 and ABCD4 protein-detergent monitored inline by absorption at 280 nm (Waters 2489 UV/
complexes (PDCs) were assessed using analytical size Vis detector), by refractive index at 658.0 nm using an Optilab
exclusion chromatography (aSEC). Briefly, samples (50 mg) rEX detector (Wyatt Technology), and by multi-angle light
were applied onto a Superdex 200 10/300 GL column (GE scattering at 656.0 nm using the three detectors (49 , 90 , 131 )
Healthcare Life Sciences) equilibrated in gel filtration buffer of the miniDAWN TREOS (Wyatt Technology). Data were
and apparent molecular masses were derived from soluble analyzed using ASTRA 5.3.4.16 software (Wyatt Technology)
protein standards (Bio-Rad). Purified LMBD1 and ABCD4 using a standard template for determination of PDC molecular
were subjected to Western blotting using anti-FLAG and anti- masses and the ‘‘protein conjugate’’ module for protein and
His6 antibodies and to mass spectrometry (Structural detergent contributions to the PDC, according to Slotboom
Genomics Consortium, University of Oxford, UK) to confirm et al. (2008). Rabbit muscle aldolase (GE Healthcare Life
primary sequence fidelity. Sciences; 158 kDa) was used as internal standard for detector
normalization. A (dn/dc)protein value of 0.192 ml/mg was used
Blue native PAGE (BN-PAGE) for LMBD1 and ABCD4 based on SEDFIT estimations
(Schuck, 2000). The (dn/dc)detergent of DDM-CHS, 0.185 ml/
Blue native PAGE was performed using 4–20% polyacryl- mg, was calculated offline in batch mode using a standard
amide gradient gels generated in-house. The buffering curve of known detergent concentrations in gel filtration
components of the gel included 50 mM Bis-tris and 500 mM buffer. The extinction coefficient of LMBD1 at 280 nm
aminocaproic acid, pH 7.0. Protein samples (8 mg) were (1.707 ml mg1 cm1) was calculated from primary sequence
incubated in the presence or absence of SDS (1%) for 10 min whereas the contribution of DDM-CHS at 280 nm was
prior to loading. Samples and unstained protein standards considered negligible (Slotboom et al., 2008).
(NativeMark; Life Technologies) were run in cold anode
(50 mM Bis-tris pH 7.0) and deep blue cathode buffers
(50 mM Tricine, 15 mM Bis-tris, 0.02% Coomassie G-250, Surface plasmon resonance (SPR)
pH 7.0) for 1 h at 100 V, whereupon the cathode buffer was
Interactions between ABCD4 (310 kDa), LMBD1 (220 kDa),
replaced with slightly blue cathode buffer (50 mM Tricine,
MMACHC (32 kDa), and/or MMADHC (32 kDa) were
15 mM Bis-tris, 0.002% Coomassie G-250, pH 7.0) and
examined using label-free, real-time BIACORE 3000 instru-
electrophoresis continued for 2–3 h at 180 V. Following
mentation (GE Healthcare Bio-Sciences AB, Uppsala,
electrophoresis, gels were stained with Coomassie brilliant
Sweden). Experiments were performed on research-grade
blue R-250.
CM4 sensor chips at 25  C using filtered (0.2 mm) and degassed
HBS-ED running buffer (10 mM HEPES pH 7.4, 150 mM
Chemical crosslinking
NaCl, 3.4 mM EDTA, 0.02% (w/v) DDM). Fatty acid free
Prior to crosslinking, LMBD1 was diluted to 2.5 mM (0.15 mg/ bovine serum albumin (BSA, 66 kDa) was from Sigma
ml) and ABCD4 was diluted to 1.2 mM (0.08 mg/ml) in gel (#A8806), Pierce Gentle Elution (PGE) was from Thermo
DOI: 10.3109/09687688.2014.990998 Interactions: LMBD1, ABCD4, MMACHC 253

Scientific (#21027), and Anatrace detergents were from Results


Affymetrix (DDM #D310 and Empigen #D350); maltose-
Purification of recombinant LMBD1 and ABCD4
binding protein (MBP, 42 kDa) was purified in-house and all
other chemicals were reagent-grade quality. Extensive construct and detergent screening for both LMBD1
Low- (100 RU), medium- (400 RU), and high-density and ABCD4 was performed in collaboration with the Integral
(1000 RU) ABCD4-immobilized surfaces were prepared Membrane Proteins (IMP) group of the Structural Genomics
using the Biacore Amine Coupling kit (10 mg/ml ABCD4 in Consortium Oxford. Briefly, N- and C-terminal truncation
10 mM sodium acetate pH 5.0 containing 0.02% (w/v) DDM); constructs of LMBD1 and ABCD4 (20 each) were cloned into
corresponding reference surfaces were prepared in the expression vectors with either a His6-tag at the N-terminus or
absence of any ABCD4 protein. To examine binding affinity, a His10/FLAG-tag at the C-terminus. Selection of the deletion
LMBD1 (0–100 nM, 2-fold serial dilutions) was titrated to constructs was based on secondary structure, disorder,
steady-state plateaus using low flow rates (10 ml/min  10 min sequence alignments, and topology predictions. Baculovirus
association + 20 min dissociation). To cross-validate our for all LMBD1 and ABCD4 constructs were generated and
results in the reversed ‘‘ligand-analyte’’ orientation, amine- used to infect small-scale Sf9 cultures for purification
coupled LMBD1-immobilized surfaces were prepared and screening. This involved extraction of all LMBD1 and
tested in the similar manner. MMACHC and MMADHC were ABCD4 constructs from infected Sf9 membranes in the
also amine-coupled (200–700 RU each) in complementary standard detergent n-dodecyl-b-D-maltoside (DDM) followed
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15

experiments to test for their interaction with ABCD4 by immobilized metal affinity chromatography and SDS-
and LMBD1, as well as solution-phase MMACHC or PAGE. While several constructs of LMBD1 and ABCD4 were
MMADHC (positive controls). In all cases, sensor chip identified as soluble using this technique, we selected the
surfaces were regenerated between sample injections at 50 ml/ more physiologically-relevant full-length (not truncated) C-
min using two 30-sec pulses of solution I (PGE containing terminal His10/FLAG-tag constructs of LMBD1 and ABCD4
0.05% (v/v) Empigen), solution II (1 M imidazole containing for detergent screening assays. These constructs were purified
0.05% (v/v) Empigen), solution III (PGE containing in 12 detergent conditions and tested for monodispersity using
33 mM H3PO4, 1.5 M NaCl, and 0.05% (v/v) Empigen), analytical size exclusion chromatography (aSEC). LMBD1
solution IV (50 mM NaOH containing 1 M NaCl and 0.05% was monodisperse by aSEC in DDM and mixed DDM-
(v/v) Empigen), and solution V (HBS-ED) followed by cholesteryl hemisuccinate (CHS) micelles but decylmaltoside
For personal use only.

routine washing procedures (i.e. ‘‘extraclean’’ and ‘‘wash’’ (DM), undecylmaltoside (UDM), and decyl maltose neopen-
options). tyl glycol (DMNG) necessitated supplementation with
SPR data were doubled-referenced (Myszka, 1999) and are cholesteryl hemisuccinate for monodispersity (data not
representative of triplicate injections acquired from at least shown). For ABCD4, solubilization in mixed DDM-choles-
three independent trials. For each replicate series, a buffer teryl hemisuccinate (CHS) micelles yielded non-aggregated
blank was injected first, the highest titrant concentration protein by aSEC but was polydisperse (described below).
second, and serial dilutions followed (from the lowest to the To obtain sufficient material for biophysical studies, full-
highest concentration repeated); comparing responses length constructs of LMBD1 and ABCD4 (incorporating
between the two highest titrant injections verified consistent tandem C-terminal affinity tags, His10-FLAG) were expressed
immobilized surface activity throughout each assay. Apparent and purified from large-scale Sf9 cultures (4 l each) using
equilibrium dissociation constants (KD) were determined by baculovirus generated from small-scale screening. LMBD1
global fitting of the data (averaged responses at the end of and ABCD4 were extracted from Sf9 membranes by solubil-
each association phase [Req] plotted versus concentration) to a ization in DDM plus CHS (DDM-CHS) and subjected to Ni-
‘‘steady-state affinity’’ model in the BIAevaluation software NTA affinity chromatography (Figure 1A). Affinity-purified
(v 4.1). In all cases, theoretical binding maxima were LMBD1 (495% pure) exhibited smearing by SDS-PAGE,
predicted using the following equation: indicative of the six N-glycosylation sites that are predicted
(Rutsch et al., 2009). Smearing was eliminated by adding
Rmax ¼ ðMA =ML ÞðRL ÞðnÞ
Peptide-N-Glycosidase F (PNGase F) to samples prior to
where Rmax is the maximal binding response (RU) at electrophoresis. Following affinity tag removal and extensive
saturation; MA is the molecular mass (kDa) of the injected deglycosylation, LMBD1 migrated as two distinct species
analyte; ML is the molecular mass (kDa) of the immobilized (55 kDa and 57 kDa). Continued SDS-PAGE heterogeneity
ligand; RL is the amount (RU) of immobilized ligand; and n is was attributed to the incomplete deglycosylation of LMBD1
the predicted binding stoichiometry (e.g. 1:1). by PNGase F; only the 55 kDa band was visualized following
exogenous PNGase F addition. These results indicated that
PNGase F activity in SDS is maintained and yielded complete
Phage display
deglycosylation only when LMBD1 was denatured, i.e.
Phage display was performed for MMACHC as previously deglycosylated LMBD1 migrated to an apparent molecular
described (Plesa et al., 2011). Pair-wise alignments of weight of 55 kDa, whereas partially deglycosylated LMBD1
affinity-selected peptides against MMACHC were performed migrated to 57 kDa. Based on the marginal difference in mass,
against LMBD1 and ABCD4 using the REceptor LIgand we propose that partially deglycosylated LMBD1 is detected
Contacts (RELIC) server MATCH (Mandava et al., 2004) and because of a single N-glycosylation site that is inaccessible to
the MatchScan program (Deme et al., 2012; Plesa et al., PNGase F under native conditions; resistance to PNGase F
2011). treatment is a common outcome for detergent-solubilized
254 J. C. Deme et al. Mol Membr Biol, 2014; 31(7–8): 250–261

calculations, consistent with membrane proteins’ binding SDS


to a greater degree than soluble proteins (Rath et al., 2009).
Purified LMBD1 and ABCD4 in DDM-CHS were sub-
jected to aSEC to assess hydrodynamic properties of protein-
detergent complexes (PDCs) (Figure 1B). LMBD1 was
monodisperse in solution, yielding a peak corresponding to
an apparent PDC molecular mass of 230 kDa based on
molecular mass standards. ABCD4 yielded a polydisperse
profile with three apparent peaks: 600 kDa minor leading
peak, 350 kDa major peak, and 200–250 kDa minor trailing
peak. Purification of an ABCD4 construct with an N-terminal
affinity tag demonstrated increased susceptibility to TEV
protease but yielded a similar polydisperse aSEC profile (data
not shown), indicating that the heterogeneity of recombinant
ABCD4 is not influenced by the location of the affinity tag but
may be an intrinsic property of the membrane protein itself.
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15

Oligomerization of LMBD1 and ABCD4


Due to inherent difficulties in elucidating membrane protein
oligomerization by aSEC, alternative techniques were adopted.
Blue native PAGE (BN-PAGE) assesses membrane protein
complexes by native electrophoresis such that negatively
Figure 1. Purified, detergent-solubilized LMBD1 and ABCD4. (A) Left, charged Coomassie G-250 exchanges with the protein-bound
SDS-PAGE of recombinant LMBD1; lane M, molecular mass standards;
lane 1, affinity-purified LMBD1; lane 2, affinity-purified LMBD1 plus
detergent; the membrane protein thus migrates through the
PNGase F; lane 3, LMBD1 following proteolysis by TEV protease and electric field. This technique has widely been utilized in
deglycosylation by PNGase F; lane 4, cleaved LMBD1 following reverse combination with SDS-mediated complex dissociation to
For personal use only.

affinity purification; lane 5, cleaved LMBD1 following reverse affinity analyze purified components of bacterial secretion systems
purification plus exogenous PNGase F. Right, SDS-PAGE of recombin-
ant ABCD4; lane M, molecular mass standards; lane 1, affinity-purified (Duong, 2003; Maillard et al., 2007; van der Does et al., 2003).
ABCD4; lane 2, ABCD4 following TEV proteolysis; lane 3, ABCD4 LMBD1 subjected to BN-PAGE yielded a homogeneous band
following reverse affinity purification; lane 4, ABCD4 following reverse with apparent molecular mass of 180–200 kDa when compared
purification plus exogenous PNGase F. (B) aSEC chromatogram of to a molecular weight ladder derived from soluble proteins
LMBD1 (black), ABCD4 (grey), and molecular weight standards
(dashed; scaled to LMBD1 axis); T, thyroglobulin (670 kDa); (Figure 2A). Treatment of LMBD1 with SDS prior to
G, g-globulin (158 kDa); O, ovalbumin (44 kDa). electrophoresis resulted in a shift to an apparent molecular
mass of 90–100 kDa. While elucidation of the molecular
masses of membrane protein complexes using soluble stand-
ards is inherently difficult due to the differential Coomassie-
membrane proteins (Boulter & Wang, 2001). Extending binding properties of soluble and membrane proteins, our
PNGase F incubation times, screening other buffer conditions, results suggest that subunit dissociation through SDS treatment
and testing the activity of alternate deglycosylating enzymes yields monomeric LMBD1 and that native LMBD1 is dimeric.
(such as endoglycosidase H) did not alleviate this heterogen- Similar to its aSEC profiles, BN-PAGE profiles of ABCD4
eity. Regardless of the observed glycosylation heterogeneity, were heterogeneous, with visible bands corresponding to
LMBD1 was homogeneous and monodisperse based on molecular masses of 300 and 600 kDa (Figure 2A). SDS-
aSEC (Figure 1B). treated ABCD4 migrated to a single species with apparent
ABCD4 was purified using a similarly optimized work- molecular mass of 90 kDa, again suggesting that ABCD4 exists
flow. Following preliminary Ni-NTA purification, ABCD4 as a higher-order oligomeric complex.
migrated at a molecular mass of 62 kDa and was 495% pure To further assess oligomerization, chemical crosslinking
by SDS-PAGE (Figure 1A). Following removal of the affinity was performed on purified LMBD1 or ABCD4 followed by
tag, subsequent purification yielded two bands (62 kDa and SDS-PAGE and visualization with silver staining. Untreated
60 kDa). ABCD4 is not predicted to be glycosylated and, LMBD1 migrated to a molecular mass of 55 kDa, whereas 1 h
accordingly, we demonstrate that treatment with PNGase F treatment of LMBD1 with glutaraldehyde (0.025%) resulted
does not shift its molecular mass by SDS-PAGE (Figure 1A). in the formation of a protein adduct with molecular mass of
Alternatively, immunoblotting (anti-His10 or anti-FLAG) 110 kDa, consistent with dimeric LMBD1 (Figure 2B). To
revealed that the 62 kDa band retained its affinity tag, ascertain that this adduct did not arise from non-specific
whereas the 60 kDa did not, indicative of incomplete TEV crosslinking, LMBD1 was treated with both SDS and
proteolysis (data not shown). Extending TEV protease glutaraldehyde, effectively preventing dimer formation. For
incubation times or altering the TEV:ABCD4 ratio did not ABCD4, treatment with formaldehyde (0.1%) over 4 h
reduce the presence of the 62 kDa band. The apparent resulted in the formation of complexes consistent in mass
molecular masses derived by SDS-PAGE for both LMBD1 with dimeric ABCD4 (120–150 kDa) and a higher molecular
and ABCD4 deviated from the theoretical primary sequence weight oligomer, compared to monomeric molecular masses
DOI: 10.3109/09687688.2014.990998 Interactions: LMBD1, ABCD4, MMACHC 255
Figure 2. Self-association of LMBD1 and
ABCD4. (A) BN-PAGE of purified LMBD1
(left) and ABCD4 (right). Lanes 1,
DDMCHS-solubilized; lanes 2, SDS-solubi-
lized (1%); lanes M, native molecular mass
markers. (B) Chemical crosslinking of
LMBD1 with glutaraldehyde (0.025%) at
25  C for 10 min (lane 3), 30 min (lane 4), and
1 h (lane 5). Untreated LMBD1 (lane 1) or a
1 h glutaraldehyde-treated LMBD1 sample
containing 1% SDS (lane 2) were used as
controls. (C) Chemical crosslinking of
ABCD4 with formaldehyde (0.1%) at 25  C
for 30 min (lane 2), 1 h (lane 3) and 4 h (lane
4). Untreated ABCD4 (lane 1) used as
control. (D) SEC-MALS analysis of LMBD1
(solid) and ABCD4 (dashed), with absorb-
ance (280 nm) profiles displayed and plotted
against molar masses. The three major peaks
of ABCD4, corresponding to PDCs, are
indicated as red dots. For LMBD1, molecular
mass contributions of protein (blue) and
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15

detergent (yellow) were deduced from the


molecular mass of the PDC (green) using the
ASTRA method (Slotboom et al., 2008).
For personal use only.

of 60 and 62 kDa in untreated samples (Figure 2C). The PDC would be too substantial to permit ABCD4 solubilization
dimeric form predominated at 30 min and 1 h, with the higher based upon the minor amount of bound detergent (30 kDa).
molecular weight oligomer occurring only after extended Overall, our results suggest that the leading shoulder
incubation (4 h). (500–600 kDa) is tetrameric ABCD4, and the trailing shoulder
Building upon our BN-PAGE and crosslinking analyses, we (220–240 kDa) is monomeric ABCD4.
then employed size exclusion chromatography with multi-
angle light scattering (SEC-MALS). SEC-MALS allows for
LMBD1 and ABCD4 interact by surface plasmon
direct molecular mass determination of PDCs (including
resonance (SPR)
contributions arising from the individual protein or detergent
components) and does not require a calibration curve Surface plasmon resonance was employed to test for the
composed of molecular mass standards (Slotboom et al., interaction between both membrane proteins. Initially, single-
2008). Analysis of LMBD1 revealed a molecular mass of cycle titrations detected specific, dose-dependent binding
220 kDa for the PDC (Figure 2D), consistent with the 230 kDa between LMBD1 (0–100 nM) and amine-coupled ABCD4
estimation from our aSEC analyses (Figure 1B); the LMBD1 (Supplementary Figure 1A, available online); in comparison,
protein component contributed 135 kDa, whereas the protein- little or no signal changes were observed with significantly
bound detergent (DDM-CHS) contributed 85 kDa. Given the higher concentrations of MBP (0–400 nM) or BSA (0–600 nM)
theoretical molecular mass of deglycosylated LMBD1 is titrated under similar conditions. While saturable LMBD1-
62.2 kDa, our SEC-MALS analyses indicated that LMBD1 ABCD4 binding was also detectable using multi-cycle titra-
was dimeric. While the heterogeneity of ABCD4 precluded tions in the reversed ‘‘ligand-analyte’’ orientation
estimates of individual protein/detergent contributions to the (Supplementary Figure 1B–C), it was easier to regenerate
PDC, we confirmed molecular masses of the three prominent sensor chip surfaces when LMBD1 was flowed over immobi-
PDCs observed in our aSEC profiles: a minor leading peak lized ABCD4. Hence, this orientation was adopted for
with molecular mass of 510 kDa, a minor trailing peak of subsequent experiments.
240 kDa and the prominent peak corresponding to 310 kDa To further analyze the kinetics of the LMBD1-ABCD4
(Figure 2D). Based on the theoretical molecular mass of interaction, LMBD1 was titrated over low-, medium-, and
ABCD4 (69.5 kDa) and our chemical crosslinking results, we high-density amine-coupled ABCD4 surfaces (Figures 3A, C,
propose that this major 310 kDa peak represents homodimeric and E, respectively). Immobilized ABCD4 was functional in
ABCD4. Our findings are consistent with literature reports all cases (i.e. surface activities480%) and the saturable, dose-
indicating that ABC half transporters dimerize to reconstitute dependent LMBD1 binding responses correlated well with a
functional transporters (Hillebrand et al., 2007; Liu et al., 1:1 PDC:PDC stoichiometry (based upon the PDC molecular
1999). While ABC half transporters have also been shown to masses calculated by SEC-MALS, i.e. 220 kDa for LMBD1
tetramerize (Xu et al., 2004), we contend that the protein and 310 kDa for ABCD4). Slow dissociation rates (510%) did
contribution of tetrameric ABCD4 (280 kDa) to the 310 kDa not permit fitting of the data using the simple 1:1 kinetics
256 J. C. Deme et al. Mol Membr Biol, 2014; 31(7–8): 250–261
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15
For personal use only.

Figure 3. Representative SPR kinetics of LMBD1 binding to amine-coupled ABCD4 at variable surface densities. Kinetics of LMBD1 (0–100 nM, 2-
fold serial) binding to low (A, 125 RU), medium (C, 350 RU), and high (E, 1150 RU) density ABCD4 surfaces in multi-cycle mode (25 ml/min  10 min
association + 20 min dissociation). Corresponding isotherms (B, D, F) in which steady-state binding responses were plotted versus analyte
concentrations (circles); fits to the ‘‘steady-state’’ affinity model in BIAevaluation (lines) predict low nanomolar KD for LMBD1-ABCD4 interaction
with 1:1 stoichiometry overall.

model. Alternatively, the multi-density titrations were ana- kinetics) and high-affinity (i.e. apparent equilibrium dissoci-
lyzed according to the steady-state amounts of LMBD1 bound ation constant (KD) of 10 nM; n ¼ 7).
at the end of each association phase (Figure 3B, D, and F,
respectively); importantly, the acquired binding isotherms and
Identification of potential MMACHC-binding sites on
low nanomolar KD values were similar in either ‘‘analyte-
LMBD1 and on ABCD4 by phage display
ligand’’ orientation (see Supplementary Figure 1C for com-
parison). Independent of mass transport effects, steady-state Phage display was used to predict regions on LMBD1 that
analyses for LMBD1 over all ABCD4 surfaces were may interact with MMACHC. Ensembles of 129 Ph.D.-C7C
unequivocal in terms of: (i) specific, dose-dependent binding peptides and 149 Ph.D.-12 peptides previously isolated
characterized by slow dissociation kinetics; and (ii) low from MMACHC-binding phages (Plesa et al., 2011) were
nanomolar binding affinities. Taken together, our SPR data aligned against the primary sequence of LMBD1, using
indicate that the specific, dose-dependent interaction between RELIC/MATCH and MatchScan. Thirty-four MMACHC-
LMBD1 and ABCD4 is stable (i.e. slow dissociation rate affinity selected peptides aligned to cytoplasmic loops
DOI: 10.3109/09687688.2014.990998 Interactions: LMBD1, ABCD4, MMACHC 257
Figure 4. Phage display identifies putative
MMACHC binding sites on LMBD1. (A)
MMACHC affinity-selected peptides aligned
to three regions within cytoplasmic loops of
LMBD1 (I–III). Exact sequence matches are
highlighted in black; conservative matches
are highlighted in grey. (B) Topology dia-
gram of TM helices 3–6 of LMBD1 using
boundaries from UniProt (Q9NUN5) and
visualization with TOPO2. MMACHC-bind-
ing regions displayed in blue and denoted by
roman numerals.
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15
For personal use only.

(Supplementary Table 1, available online) based on the (32 kDa each) were immobilized, thereby maximizing the
proposed topology of LMBD1 (UniProt: Q9NUN5), produ- signal range with the significantly larger LMBD1 (220 kDa)
cing three distinct clusters (Figure 4). Eight peptides aligned and ABCD4 (310 kDa) membrane proteins. Fixed concentra-
to residues 135–145 (region I), the loop between TM helices 3 tion injections of LMBD1 and ABCD4 (100 nM each) yielded
and 4. The largest cluster consisted of 20 aligned peptides specific binding responses (1500–2000 RU) against immobi-
(region II), aligning to residues 215–225 within the largest lized MMACHC as compared to BSA (50 RU; Figure 6A);
cytoplasmic segment of LMBD1 between TM5 and TM6. The even with normalization for the 3- to 5-fold difference in
remaining six peptides mapped within this same loop but to molecular mass between the PDCs and BSA, the binding
residues 260–267 (region III). responses with LMBD1 and ABCD4 were still significantly
The same procedure was used to predict regions on ABCD4 larger compared to BSA (e.g. 50 RU observed, as compared to
that may interact with MMACHC. Affinity-selected peptides 4-fold scale up ¼ 200 RU). In contrast, LMBD1 and
against MMACHC (Plesa et al., 2011) were mapped to the ABCD4 elicited only minor binding responses (20–30 RU)
primary sequence of ABCD4. Forty-three affinity-selected against immobilized MMADHC as compared to BSA (5 RU;
peptides aligned to ABCD4 and formed distinct clusters 4-fold scale up ¼ 20 RU; Figure 6B). Even with normaliza-
(Figure 5; Supplementary Table 2). All four predicted tion for the 3.5-fold difference in the amine-coupled densities
MMACHC-binding regions (I–IV) localized to the proposed between MMACHC (700 RU) and MMADHC (200 RU), the
cytoplasmic C-terminal NBD of ABCD4 (UniProt: O14678). LMBD1 and ABCD4 binding responses were still extremely
Region I (residues 392–404) was N-terminal to the proposed low with MMADHC when compared to the theoretical Rmax
Walker A motif (residues 421–428). Regions II (residues 456– values (signal range of 1300–1900 RU anticipated, as readily
464) and III (residues 501–511) mapped between the Walker A achieved with MMACHC). While these results are not
motif and proposed ABC signature (residues 524–530), Walker suggestive of a role for MMADHC interacting with LMBD1
B motifs (544–549), and D-loop (548–555). The final region and ABCD4, we cross-validated the functional integrity of our
(IV; residues 584–592) mapped to the C-terminus of the NBD. cytoplasmic protein preparations by titrating MMACHC with
MMADHC (Supplementary Figure 2A); the acquired binding
profiles were consistent with our previous publications (thiol-
Binary interactions of LMBD1 or ABCD4 with MMACHC
coupled MMACHC, Plesa et al., 2011; amine-coupled
or MMADHC by SPR
MMADHC, Deme et al., 2012).
To corroborate our phage display predictions, additional SPR To examine their binding specificity with MMACHC
analyses were performed in which MMACHC and MMADHC in greater detail, detergent-solubilized ABCD4 (Figures 6C
258 J. C. Deme et al. Mol Membr Biol, 2014; 31(7–8): 250–261

Figure 5. Phage display identifies putative


MMACHC binding sites on ABCD4. (A)
MMACHC affinity-selected peptides aligned
to four regions within the C-terminal NBD of
ABCD4 (I–IV). Exact sequence matches are
highlighted in black; conservative matches
are highlighted in grey. (B) Linear represen-
tation of the NBD of ABCD4 with proposed
catalytic motifs shown in orange: A, Walker
A; S, ABC signature; B + D, Walker B and
D-loop. MMACHC-binding regions are
annotated as roman numerals and displayed
in green.
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15
For personal use only.

and D) and LMBD1 (Figure 6E and F) were titrated over 2001), in contrast to LMBD1 which demonstrates only minor
amine-coupled MMACHC surfaces. ABCD4 and LMBD1 localization to the plasma membrane (Tseng et al., 2013).
exhibited saturable, dose-dependent binding to MMACHC Similar to LMBD1, LIMR contains nine putative TM helices.
and similar affinities were detected (KD of 65  10 nM versus LIMR binds and internalizes lipocalins via receptor-mediated
14  2 nM, respectively; n ¼ 3 for each). As a positive control, endocytosis. Recombinant production of human LIMR in a
we also titrated MMACHC over the same immobilized Drosophila expression system was recently demonstrated by
MMACHC surfaces (Supplementary Figure 2B); similar to Hesselink and colleagues (Hesselink & Findlay, 2013).
our previous publication (Deme et al., 2012), the association Congruent with our LMBD1 data, the authors demonstrated
phase deviated from simple 1:1 kinetics (i.e. multi-phasic that DDM-solubilized LIMR forms dimers in solution. Our
MMACHC-MMACHC interaction). BN-PAGE, crosslinking, and SEC-MALS results for LMBD1
are consistent with those for LIMR, in that LMBD1 predom-
Discussion
inantly forms dimers in solution.
The molecular bases of inborn cblF and cblJ defects in vitamin Recombinant ABCD4 demonstrates heterogeneity in its
B12 metabolism were established when LMBRD1 and ABCD4 oligomeric state, but our results indicate that dimeric ABCD4
were identified to encode putative lysosomal exporters of Cbl is the predominant form when solubilized in mixed DDM and
(Coelho et al., 2012; Rutsch et al., 2009). Mutations in their CHS micelles. ABCD4 belongs to the D subfamily of
encoded gene products, LMBD1 and ABCD4 respectively, peroxisomal ABC half transporters and shares homology
prevent the lysosomal release of Cbl to the cytoplasm. Our with the other members: ABCD1, ABCD2, and ABCD3
current studies extend these findings by reporting the recom- (Wanders et al., 2007). Interaction analyses performed on
binant production of LMBD1 and ABCD4; even with our other members of this subfamily support our conclusions that
optimized protocol, the overall yields of these membrane ABCD4 forms homodimers. Using yeast two-hybrid
proteins were limited (100 mg/l of culture) but both were approaches, Liu and co-workers (Liu et al., 1999) showed
highly pure (495%) and stable in solution – essential criteria that the cytoplasmic C-terminal NBD of ABCD1, ABCD2,
for structural and biophysical analyses. and ABCD3 mediates homo- and heterodimerization in all
Because of limited structure-function studies that have been possible combinations. In another study, ABCD1 and
published to date, our understanding of LMBD1 and ABCD4 ABCD3, isolated from mouse liver, formed homodimers
has been primarily derived from homologous proteins. (Guimaraes et al., 2004). Live cell FRET microscopy
LMBD1 shares homology with the lipocalin-1 interacting identified homodimerization of ABCD1 and ABCD3 in
membrane receptor (LIMR). LIMR is widely expressed across addition to the formation of an ABCD1-ABCD3 heterodi-
tissues and localizes to the plasma membrane (Wojnar et al., meric complex (Hillebrand et al., 2007).
DOI: 10.3109/09687688.2014.990998 Interactions: LMBD1, ABCD4, MMACHC 259
Figure 6. Representative SPR for LMBD1
and ABCD4 binding to amine-coupled
MMACHC and MMADHC. Kinetics of
buffer (black), BSA (red), ABCD4 (green), or
LMBD1 (blue), each at 100 nM, binding to
amine-coupled MMACHC (A, 700 RU) or
MMADHC (B, 200 RU) at 25 ml/min (1 min
association + 5 min dissociation). Multi-cycle
titrations of ABCD4 (C) and LMBD1 (E)
binding (0–100 nM, all 2-fold serial) to
immobilized MMACHC (700 RU) at 25 ml/
min (10 min association + 20 min dissoci-
ation). Corresponding binding isotherms in
which steady-state binding responses were
plotted versus analyte concentrations (circles;
D, ABCD4; F, LMBD1) and fit to the
‘‘steady-state affinity’’ model in
BIAevaluation (lines).
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15
For personal use only.

Our SPR analyses provided direct evidence for binding MMACHC-MMADHC interaction (Deme et al., 2012; Plesa
between LMBD1 and ABCD4. Given that previous SPR et al., 2011). To date, a BtuF-like lysosomal Cbl binding
literature indicated similar outcomes using direct or indirect protein has not yet been identified in mammals; significantly,
immobilization of membrane proteins (Vigonsky et al., 2013), cultured cblF or cblJ fibroblasts accumulate non-protein
we chose direct amine-coupling (over indirect Ni-NTA bound Cbl, which argues against the presence of a lysosomal
capture) to minimize sample consumption. The high-affinity Cbl-binding protein. Instead, we propose that Cbl transport
interaction that we detected between ABCD4 and LMBD1 across the lysosome is facilitated by a high affinity complex
(i.e. KD 10 nM by SPR) was characterized by a slow formed between LMBD1 and ABCD4.
dissociation rate. Accordingly, stable complex formation Our phage display predictions indicate that cytoplasmic
between ABCD4 and LMBD1 may be an important step for regions of LMBD1 and ABCD4 can recruit MMACHC. We
mediating intracellular vitamin B12 transport. In the context identified three cytoplasmic loop regions between TM helices
of Cbl transport, the Escherichia coli ABC transporter 3–4 and 5–6 on LMBD1. There are no known point mutations
BtuC2D2 mediates Cbl import into the cytoplasm of E. coli within these regions in patients with the cblF defect. Rather,
by forming a complex with the periplasmic Cbl-binding all known mutations in LMBRD1 result in protein truncation
protein BtuF. On its own, BtuC2D2 has poor affinity for Cbl (Armour et al., 2013; Miousse et al., 2011; Rutsch et al.,
which necessitates the delivery of Cbl by BtuF. Similar to our 2009). For ABCD4, we identified four regions within the
ABCD4-LMBD1 outcomes, BtuF binds BtuC2D2 with high cytoplasmic C-terminal NBD. These regions did not over-
affinity and their interaction is characterized by a slow lap with the putative motifs required for ATP hydrolysis,
dissociation rate constant using SPR (Lewinson et al., 2010). but two overlapped with known mutations in ABCD4
Slow dissociation may therefore be an inherent property of (p.G443_S485del and p.E583LfsX9) that would effectively
interactions involved in the early transport of Cbl, given that abrogate the proposed binding residues. The predictive power
we also observed slow dissociation when characterizing the of phage display is illustrated in our previous work where we
260 J. C. Deme et al. Mol Membr Biol, 2014; 31(7–8): 250–261

proposed binding interfaces between MMACHC and provide the framework for functional assays that will provide
MMADHC and then performed SPR (in vitro) and bacterial insight into the activity of LMBD1 and ABCD4, thus
two hybrid (in vivo) analyses to provide direct evidence for advancing our basic understanding of early intracellular
their interaction (Deme et al., 2012; Plesa et al., 2011). Five vitamin B12 metabolism.
proposed MMACHC-binding regions were mapped to residues
C-terminal to Glu142 on MMADHC (Plesa et al., 2011). An Acknowledgements
independent group (Gherasim et al., 2013a) recently corrobo-
The authors wish to thank the following colleagues at the
rated our findings by demonstrating that MMACHC can bind
Structural Genomics Consortium, Oxford: Leela Shrestha for
to an MMADHC variant lacking 115 N-terminal residues.
baculovirus construction and initial detergent screening of
We now communicate phage display results that predict
LMBD1, Rod Chalk and Georgina Berridge for mass
MMACHC-binding sites on ABCD4 and LMBD1. Our current
spectrometry services, and Shubhashish Mukhopadhyay for
SPR analyses provide unequivocal, direct evidence that both of
cell culture expression and training.
these membrane proteins can interact with MMACHC in a
specific, dose-dependent manner. While parallel analyses do
not support MMADHC as a partner protein for ABCD4 or Funding
LMBD1, we performed control titrations to cross-validate our Research was supported by operating grants to J.W.C. from the
previously published SPR data for MMACHC-MMACHC and Natural Sciences and Engineering Research Council [NSERC;
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15

MMACHC-MMADHC (Deme et al., 2012; Plesa et al., 2011). RGPIN 7289-06-07] and to D.S.R. from the Canadian
Given that the functional integrity of our MMACHC and Institutes of Health Research [CIHR; MOP-15078]. The
MMADHC preparations was intact, it is interesting to note that McGill SPR Facility thanks the Canada Foundation for
the affinity of MMACHC for ABCD4 or LMBD1 (KD 65 nM Innovation [CFI grant to Montreal Integrated Genomics
and KD 14 nM, respectively) is stronger compared to Group for Research on Infectious Pathogens] and Canadian
MMACHC for MMADHC (KD 360 nM). Thus, the inter- Institutes of Health Research [CIHR Research Resource Grant
action of MMACHC with these lysosomal exporters would be 200610PRG-165657-PRG-CFAA-11449] for infrastructure
favoured over cytoplasmic MMADHC should they be mutually support. The Structural Genomics Consortium (SGC) is a
exclusive. registered charity [no. 1097737] that receives funds from
Our evidence for the recruitment of MMACHC to the AbbVie, Canadian Institutes for Health Research, Genome
For personal use only.

cytoplasmic side of lysosomal exporters agrees with recent Canada, GlaxoSmithKline, Eli Lilly Canada, the Novartis
proposals (Froese & Gravel, 2010; Rutsch et al., 2011). Research Foundation, Pfizer, Takeda, Canada Foundation for
Such proposals argue that MMACHC recruitment allows the Innovation, the Ontario Ministry of Economic Development
vectorial delivery of Cbl following transport across the and Innovation, and the Wellcome Trust [092809/Z/10/Z].
lysosomal membrane, preventing dilution and exposure of
the reactive cofactor to the cellular milieu. In the cytoplasm, Declaration of interest
MMACHC catalyzes the conversion of ‘‘base-on’’ Cbl to the
‘‘base-off’’ state; however, the reaction rate is slow (20 min) The authors report no conflicts of interest. The authors alone
(Koutmos et al., 2011). It has been proposed that the acidic are responsible for the content and writing of the paper.
environment of the lysosomal lumen promotes the ‘‘base-off’’
state and that this Cbl conformation is maintained following
lysosomal export by binding directly to MMACHC, effectively References
circumventing the slow MMACHC-catalyzed reaction Armour CM, Brebner A, Watkins D, Geraghty MT, Chan A, Rosenblatt
(Banerjee, 2006; Gherasim et al., 2013a). While LMBD1 is DS. 2013. A patient with an inborn error of vitamin B12 metabolism
speculated to form the transport channel for this process (cblF) detected by newborn screening. Pediatrics 132:e257–e261.
Banerjee R. 2006. B12 trafficking in mammals: A for coenzyme escort
(Coelho et al., 2012), we cannot discount that MMACHC service. ACS Chem Biol 1:149–159.
interacts with both putative exporters given the similar binding Boulter JM, Wang DN. 2001. Purification and characterization of human
affinities detected. Cbl binding to MMACHC may trigger erythrocyte glucose transporter in decylmaltoside detergent solution.
dissociation from LMBD1/ABCD4 and subsequent recruit- Protein Expr Purif 22:337–348.
Chapel A, Kieffer-Jaquinod S, Sagne C, Verdon Q, Ivaldi C, Mellal M,
ment to MMADHC; the Cbl-dependence of the MMACHC- et al. 2013. An extended proteome map of the lysosomal membrane
MMADHC interaction has recently been reported (Gherasim reveals novel potential transporters. Mol Cell Proteomics 12:
et al., 2013a). 1572–1588.
Coelho D, Kim JC, Miousse IR, Fung S, Du Moulin M, Buers I, et al.
2012. Mutations in ABCD4 cause a new inborn error of vitamin B12
Conclusions metabolism. Nat Genet 44:1152–1155.
Deme JC, Miousse IR, Plesa M, Kim JC, Hancock MA, Mah W, et al.
Results from this study are consistent with the channeling and 2012. Structural features of recombinant MMADHC isoforms and
chaperoning of Cbl that is mediated through protein–protein their interactions with MMACHC, proteins of mammalian vitamin
B12 metabolism. Mol Genet Metab 107:352–362.
interactions (Banerjee, 2006; Gherasim et al., 2013b). To Duong F. 2003. Binding, activation and dissociation of the dimeric SecA
date, research on LMBD1 and ABCD4 has been mostly ATPase at the dimeric SecYEG translocase. EMBO J 22:4375–4384.
limited to studies in cultured cells. For the first time, our Froese DS, Gravel RA. 2010. Genetic disorders of vitamin B12
study describes the recombinant production of LMBD1 and metabolism: Eight complementation groups – eight genes. Expert
Rev Mol Med 12:e37.
ABCD4 and protein complexes formed between these lyso- Gherasim C, Hannibal L, Rajagopalan D, Jacobsen DW, Banerjee R.
somal membrane proteins and with MMACHC. Our results 2013a. The C-terminal domain of CblD interacts with CblC and
DOI: 10.3109/09687688.2014.990998 Interactions: LMBD1, ABCD4, MMACHC 261
influences intracellular cobalamin partitioning. Biochimie 95: human proteins participating in intracellular vitamin B12 metabolism.
1023–1032. Mol Genet Metab 102:139–148.
Gherasim C, Lofgren M, Banerjee R. 2013b. Navigating the B12 road: Rath A, Glibowicka M, Nadeau VG, Chen G, Deber CM. 2009.
Assimilation, delivery, and disorders of cobalamin. J Biol Chem 288: Detergent binding explains anomalous SDS-PAGE migration of
13186–13193. membrane proteins. Proc Natl Acad Sci USA 106:1760–1765.
Guimaraes CP, Domingues P, Aubourg P, Fouquet F, Pujol A, Jimenez- Rosenblatt DS, Hosack A, Matiaszuk NV, Cooper BA, Laframboise R.
Sanchez G, et al. 2004. Mouse liver PMP70 and ALDP: Homomeric 1985. Defect in vitamin B12 release from lysosomes: Newly described
interactions prevail in vivo. Biochim Biophys Acta 1689:235–243. inborn error of vitamin B12 metabolism. Science 228:1319–1321.
Hesselink RW, Findlay JB. 2013. Expression, characterization and ligand Rutsch F, Gailus S, Miousse IR, Suormala T, Sagne C, Toliat MR, et al.
specificity of lipocalin-1 interacting membrane receptor (LIMR). Mol 2009. Identification of a putative lysosomal cobalamin exporter
Membr Biol 30:327–337. altered in the cblF defect of vitamin B12 metabolism. Nat Genet 41:
Hillebrand M, Verrier SE, Ohlenbusch A, Schafer A, Soling HD, 234–239.
Wouters FS, Gartner J. 2007. Live cell FRET microscopy: Homo- and Rutsch F, Gailus S, Suormala T, Fowler B. 2011. LMBRD1: The gene for
heterodimerization of two human peroxisomal ABC transporters, the the cblF defect of vitamin B12 metabolism. J Inherit Metab Dis 34:
adrenoleukodystrophy protein (ALDP, ABCD1) and PMP70 121–126.
(ABCD3). J Biol Chem 282:26997–27005. Schuck P. 2000. Size-distribution analysis of macromolecules by
Hodgkin DC, Kamper J, Mackay M, Pickworth J, Trueblood KN, White sedimentation velocity ultracentrifugation and Lamm equation
JG. 1956. Structure of vitamin B12. Nature 178:64–66. modeling. Biophys J 78:1606–1619.
Holzinger A, Kammerer S, Roscher AA. 1997. Primary structure of Shani N, Jimenez-Sanchez G, Steel G, Dean M, Valle D. 1997.
human PMP69, a putative peroxisomal ABC-transporter. Biochem Identification of a fourth half ABC transporter in the human
Biophys Res Commun 237:152–157. peroxisomal membrane. Hum Mol Genet 6:1925–1931.
Kashiwayama Y, Seki M, Yasui A, Murasaki Y, Morita M, Yamashita Y,
Mol Membr Biol Downloaded from informahealthcare.com by University of Sydney on 01/02/15

Slotboom DJ, Duurkens RH, Olieman K, Erkens GB. 2008. Static light
et al. 2009. 70-kDa peroxisomal membrane protein related protein scattering to characterize membrane proteins in detergent solution.
(P70R/ABCD4) localizes to endoplasmic reticulum not peroxisomes, Methods 46:73–82.
and NH2-terminal hydrophobic property determines the subcellular Stubbe J. 1994. Binding site revealed of nature’s most beautiful cofactor.
localization of ABC subfamily D proteins. Exp Cell Res 315:190–205. Science 266:1663–1664.
Kim JC, Lee NC, Hwu PW, Chien YH, Fahiminiya S, Majewski J, et al. Tseng LT, Lin CL, Tzen KY, Chang SC, Chang MF. 2013. LMBD1
2012. Late onset of symptoms in an atypical patient with the cblJ
serves as a specific adaptor for insulin receptor internalization. J Biol
inborn error of vitamin B12 metabolism: Diagnosis and novel mutation
Chem 288:32424–32432.
revealed by exome sequencing. Mol Genet Metab 107:664–668.
van der Does C, De Keyzer J, Van Der Laan M, Driessen AJ. 2003.
Koutmos M, Gherasim C, Smith JL, Banerjee R. 2011. Structural basis
Reconstitution of purified bacterial preprotein translocase in lipo-
of multifunctionality in a vitamin B12-processing enzyme. J Biol
Chem 286:29780–19787. somes. Methods Enzymol 372:86–98.
Lewinson O, Lee AT, Locher KP, Rees DC. 2010. A distinct mechanism Vassiliadis A, Rosenblatt DS, Cooper BA, Bergeron JJ. 1991. Lysosomal
cobalamin accumulation in fibroblasts from a patient with an inborn
For personal use only.

for the ABC transporter BtuCD-BtuF revealed by the dynamics of


complex formation. Nat Struct Mol Biol 17:332–338. error of cobalamin metabolism (cblF complementation group):
Liu LX, Janvier K, Berteaux-Lecellier V, Cartier N, Benarous R, Visualization by electron microscope radioautography. Exp Cell Res
Aubourg P. 1999. Homo- and heterodimerization of peroxisomal ATP- 195:295–302.
binding cassette half-transporters. J Biol Chem 274:32738–32743. Vigonsky E, Ovcharenko E, Lewinson O. 2013. Two molybdate/
Maillard AP, Lalani S, Silva F, Belin D, Duong F. 2007. Deregulation of tungstate ABC transporters that interact very differently with their
the SecYEG translocation channel upon removal of the plug domain. substrate binding proteins. Proc Natl Acad Sci USA 110:5440–5445.
J Biol Chem 282:1281–1287. Wanders RJ, Visser WF, Van Roermund CW, Kemp S, Waterham HR.
Mandava S, Makowski L, Devarapalli S, Uzubell J, Rodi DJ. 2004. 2007. The peroxisomal ABC transporter family. Pflugers Arch 453:
RELIC – a bioinformatics server for combinatorial peptide analysis 719–734.
and identification of protein-ligand interaction sites. Proteomics 4: Watkins D, Rosenblatt DS. 1986. Failure of lysosomal release of vitamin
1439–1460. B12: A new complementation group causing methylmalonic aciduria
Miousse IR, Watkins D, Rosenblatt DS. 2011. Novel splice site (cblF). Am J Hum Genet 39:404–408.
mutations and a large deletion in three patients with the cblF inborn Wojnar P, Lechner M, Merschak P, Redl B. 2001. Molecular cloning of a
error of vitamin B12 metabolism. Mol Genet Metab 102:505–507. novel lipocalin-1 interacting human cell membrane receptor using
Myszka DG. 1999. Improving biosensor analysis. J Mol Recognit 12: phage display. J Biol Chem 276:20206–20212.
279–284. Xu J, Liu Y, Yang Y, Bates S, Zhang JT. 2004. Characterization of
Plesa M, Kim J, Paquette SG, Gagnon H, Ng-Thow-Hing C, Gibbs BF, oligomeric human half-ABC transporter ATP-binding cassette G2.
et al. 2011. Interaction between MMACHC and MMADHC, two J Biol Chem 279:19781–19789.

Supplementary material available online


Supplementary Figures 1 and 2
Supplementary Tables 1 and 2.

You might also like