Você está na página 1de 16

Biol. Cell (2010) 102, 581–591 (Printed in Great Britain) doi:10.

1042/BC20100046
Research article

Spatial organization of the


transforming MHC class II
compartment
Hezder E. van Nispen tot Pannerden*†, Willie J. Geerts*‡, Monique J. Kleijmeer*† and Harry F.G. Heijnen*§1
*Institute of Biomembranes, Padualaan 8, 3584 CH Utrecht, The Netherlands, †Cell Microscopy Center, Department of Cell Biology,
University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands, ‡Department of Molecular Cell Biology, Utrecht
University, Utrecht, The Netherlands, and §Laboratory of Clinical Chemistry and Haematology, University Medical Center Utrecht,
Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
www.biolcell.org

Background information. DC (dendritic cells) continuously capture pathogens and process them into small pep-
tides within the endolysosomal compartment, the MIIC (MHC class II-containing compartment). In MIICs peptides
are loaded on to MHC class II and rapidly redistributed to the cell surface. This redistribution is accompanied
by profound changes of the MIICs into tubular structures. An emerging concept is that MIIC tubulation provides
a means to transport MHC class II–peptide complexes to the cell surface, either directly or through vesicular
intermediates. To obtain spatial information on the reorganization of the MIICs during DC maturation, we per-
formed electron tomography on cryo-immobilized and freeze-substituted mouse DCs after stimulation with LPS
(lipopolysaccharide).
Results. In non-stimulated DCs, MIICs are mostly spherical. After 3 h of LPS stimulation, individual MIICs transform
into tubular structures. Three-dimensional reconstruction showed that the MIICs frequently display fusion profiles
and after 6 h of LPS stimulation, MIICs become more interconnected, thereby creating large MIIC reticula. Micro-
tubules and microfilaments align these MIICs and reveal physical connections. In our tomograms we also identified a
separate population of MIIC-like intermediates, particularly at extended ends of MIIC tubules and in close proximity
to the trans-Golgi network. No fusion events were captured between reticular MIICs and the plasma membrane.
Conclusions. Our results indicate that MIICs have the capacity to fuse together, whereby the cytoskeleton possibly
provides a scaffold for the MIIC shape change and directionality. MIIC-like intermediates may represent MHC class
Biology of the Cell

II carriers.

Introduction oid organs and interact with antigen-specific CD4+


Immature DCs (dendritic cells) principally reside in T-cells to induce an immune response. It has been well
peripheral tissues and continuously survey for patho- established that the major sites of antigen processing
gens (e.g. Sallusto et al., 1995). On activation by and peptide loading in APCs (antigen-presenting
pathogen-derived antigens, DCs migrate to lymph- cells) are the late-endosomal and lysosomal com-
partments, collectively termed MIICs (MHC class
II-containing compartments) (Neefjes et al., 1990;
1 To whom correspondence should be addressed (email Watts, 1997; Mellman et al., 1998; Trombetta and
h.f.g.heijnen@umcutrecht.nl).
Key words: dendritic cell, electron microscopy, MHC class II-containing Mellman, 2005). MIICs have been extensively charac-
compartment (MIIC), spatial organization, three-dimensional analysis, terized in several APCs, including B-cells (Kleijmeer
tomography.
Abbreviations used: APC, antigen-presenting cell; DC, dendritic cell; ET, et al., 1997) and DCs (Kleijmeer et al., 2001).
electron tomography; HPF-FS, high-pressure freezing and freeze substitution; They appear as spherical multivesicular bodies, but
IEM, immunoelectron microscopy; IF, immunofluorescence; LPS,
lipopolysaccharide; MIIC, MHC class II-containing compartment; MT,
may also have a multilaminar appearance. Previous
microtubule; TGN, trans -Golgi network. three-dimensional analyses showed that the luminal

www.biolcell.org | Volume 102 (11) | Pages 581–591 581


H.E. van Nispen tot Pannerden and others

membrane sheets and vesicles are not connected to the indicating that MIICs fuse together. Increased ex-
limiting membrane of the MIIC (Murk et al., 2003b; posure to LPS causes the MIIC profiles to become
Murk et al., 2004). Most of the intracellular pool of more interconnected, thereby creating large MIIC
MHC class II molecules in immature DCs is localized networks. The reticular MIICs are surrounded by a
to the MIIC internal vesicles (Kleijmeer et al., 2001). complex network of MTs and microfilaments with
On DC activation, peptide-loaded MHC class II mo- which physical connections exist, thereby providing a
lecules are redistributed to the cell surface, together scaffold for their structural alteration. We also found
with a set of co-stimulatory molecules, thereby en- isolated MIIC intermediates, distinct from the re-
abling their interaction with CD4+ T-cells (Hart, ticulum, which may represent MHC class II carrier
1997; Kleijmeer et al., 2001). vesicles en route to the cell surface.
The exact pathway by which MHC class II–peptide
complexes are transported to the cell surface is
still not completely understood. Several independent Results
studies have shown that during activation and on in- To obtain a detailed three-dimensional view on the
teraction of DCs with CD4+ T-cells, MIICs undergo membrane reorganization of transforming MIICs,
major structural changes and transform into tubular cultured D1 cells were left untreated or stimu-
compartments (Kleijmeer et al., 2001; Boes et al., lated with LPS for 3 and 6 h respectively and
2002; Chow et al., 2002). These tubules are believed subjected to HPF-FS (high-pressure freezing and
to be formed in an MT (microtubule)-dependent way freeze substitution), followed by plastic embed-
(Boes et al., 2002) and contain high amounts of MHC ding and ET (electron tomography) analysis. Stim-
class II on their limiting membranes. Back-fusion ulated cells were carefully selected on 200–300 nm
of the MIIC internal membranes with the limit- thick serial sections on the basis of their character-
ing membrane was suggested to facilitate peptide istic morphology (i.e. dendritic profile). The stim-
loading and transport of the MHC class II–peptide ulation response of the D1 cells was verified by
complex towards the plasma membrane (Kleijmeer determining the cell surface expression of MHC
et al., 2001). Back-fusion may promote the associ- class II molecules using confocal microscopy and IEM
ation of MHC class II with the chaperone protein (immunoelectron microscopy) analyses (see Supple-
HLA-DM, which resides predominantly on the lim- mentary Figure S1 and S2 at http://www.biolcell.
iting membrane of MIICs (Kleijmeer et al., 2001). org/boc/102/boc1020581add.htm). ET does not al-
This would facilitate the release of the CLIP (MHC low identification of MIICs by immunolabelling.
class II-associated invariant-chain peptide) from the Our knowledge of the recognition of MIICs in APCs
peptide-binding cleft of MHC class II (Kropshofer comes from a previous detailed IEM analysis, carried
et al., 1999; Alfonso and Karlsson, 2000), thus en- out in our laboratory. From these studies we know
abling peptide loading. Back-fusion also generates that the MHC class II compartments belong to the
the membrane supply necessary for the MIIC tubula- regular endocytic pathway and include late endo-
tion and outgrowth towards the plasma membrane. somes and lysosomes (Geuze, 1998). It is based on
The final step in MHC class II transport would then this knowledge that we have selected the structures
be fusion of the MIIC tubules with the plasma mem- as MIICs in our three-dimensional reconstructions.
brane and/or the budding and subsequent fusion of As a rule we simultaneously carried out IEM
transport vesicles with the plasma membrane. Part experiments and compared immunolabelled MIICs
of the MIIC luminal membranes are also secreted as in thin frozen sections with those in our tomo-
exosomes when MIICs fuse with the plasma mem- grams. Two distinct markers of early stages (invari-
brane, a phenomenon that has been documented as a ant chain) and intermediate and late MIICs (HLA-
constitutive process in B-cells (Raposo et al., 1996) DM) were used (Geuze, 1998). DCs were stimu-
and DCs (Buschow et al., 2005). lated for 6 h with LPS, the same conditions as those
We present here the three-dimensional reconstruc- used for the three-dimensional analysis. Labelling
tion of transformed MIICs in maturating DCs. On of HLA-DM was absent from early compartments
stimulation with LPS (lipopolysaccharide), MIICs be- (low vesicle number) and abundant in multivesicu-
come tubular and frequently reveal fusion profiles, lar (intermediate and late) MIICs (see Supplementary

582 
C The Authors Journal compilation 
C 2010 Portland Press Limited
Three-dimensional organization of the transforming MIIC Research article
Figures S3A and S3B at http://www.biolcell.org/ the networks analysed exhibit internal membranes
boc/102/boc1020581add.htm). Vice versa, invariant that have the morphological characteristics of late
chain was abundant in the early compartments and MIICs. Within these networks we detected in-
absent from multivesicular MIICs (Supplementary tact, but also partially degraded vesicles, tubular
Figures S3C and S3D). Based on the ultrastructural membrane structures and isolated membrane sheets
criteria and the immunogold labelling, we conclude (Figures 2C and 2D; see Supplementary Movie 2 at
that the multivesicular and multilaminar structures http://www.biolcell.org/boc/102/boc1020581add.
in our tomograms represent intermediate and late htm). The last two features are reminiscent of the
stages of MIICs. To maximally capture membrane morphology of internal membranes in multilam-
continuities, three-dimensional reconstructions were inar lysosomes. This suggests that MIIC networks
carried out in large-volume tomograms, covering fuse with multilaminar lysosomes. Representative ex-
areas where MIICs are positioned both at TGN (trans- amples of fusion profiles and neck regions are shown
Golgi network) exit sites and in close proximity to in Figure 3. In Figures 3(A)–3(C), two interconnect-
the plasma membrane. Figures 1(A) and 1(B) repres- ing MIICs are shown after 3 h of LPS stimulation (see
ent an example of such a tomogram after 6 h of LPS also Supplementary Movie 3 at http://www.biolcell.
stimulation, covering a total volume of 23.6 μm3 . org/boc/102/boc1020581add.htm). The compart-
The MIICs in our tomograms appear as spher- ment comprises two distinct MIIC profiles, con-
ical and elongated tubular organelles (Figure 1B, nected through a ∼ 25 nm wide fusion pore,
coloured asterisks, highlighted in Figures 1C–1E). and is completely captured within the tomo-
Tubular MIICs were more regularly shaped in the gram. Interconnecting MIICs are generally larger
HPF-FS samples (Figures 1C and 1F) as com- after 6 h LPS (Figures 3D–3H; see Supplement-
pared with those observed in ultrathin cryosec- ary Movie 4 at http://www.biolcell.org/boc/102/
tions from chemically fixed cells (Figure 4D in boc1020581add.htm), and reveal a multi-domain
Kleijmeer et al., 2001). Fusion/fission profiles of structure and multiple neck regions (inset to
the internal membranes with the limiting mem- Figure 3H; see Supplementary Movie 5 at http://
brane of the tubular MIICs were occasionally ob- www.biolcell.org/boc/102/boc1020581add.htm).
served (results not shown). Tubular MIICs are dis- Semi-quantitative analysis of the MIIC profiles
tinct from the tubular elements emerging from within our tomograms revealed that after 3 and
early endosomes, which are also encountered in LPS- 6 h of LPS stimulation respectively, 12% (n = 23)
stimulated cells, albeit at low frequency (see Sup- and 22% (n = 23) of the MIIC profiles are con-
plementary Figure S4 at http://www.biolcell.org/ nected to a network (Figure 3I). MIICs in non-
boc/102/boc1020581add.htm). These early endo- stimulated cells are often completely captured within
somes exhibit a characteristic spherical morphology our tomograms, and no fusion profiles or inter-
with low number of luminal vesicles and abundant connections between them were observed (see Sup-
invariant chain (see also Supplementary Figure S3). plementary Figure S5 at http://www.biolcell.org/
The tubular extensions are significantly smaller than boc/102/boc1020581add.htm). Their average vol-
the MIIC tubules detected after LPS stimulation (av- ume was 1.6×107 nm3 (n = 18). In contrast, the
erage diameter 50+ −11 nm, n = 11 nm versus 153 + − MIIC networks in LPS-stimulated cells were much
57 nm, n = 6, P = 0.015). The tubular MIICs cap- larger (>1.4×108 nm3 , n = 26) and were never cap-
tured in our tomograms could reach lengths of at tured completely in our tomograms. We therefore
least 3 μm. conclude that MIICs must fuse together on LPS stim-
ulation.
MIICs become organized into a reticular network
When membranes of MIICs were traced manually, it
became apparent that the individual MIIC profiles are
often interconnected and form reticular membrane Fusion of MIICs is crucial for MHC class II
networks (Figures 1D, 1E, 1G and 1H; Figures 2A transport to the cell surface
and 2B; see Supplementary Movie 1 at http://www. Bafilomycin is known to inhibit lysosomal acidifica-
biolcell.org/boc/102/boc1020581add.htm). Most of tion and thereby the fusion of late endosomes with

www.biolcell.org | Volume 102 (11) | Pages 581–591 583


H.E. van Nispen tot Pannerden and others

Figure 1 Tubular and reticular MIICs in a stimulated D1 cell


(A) Overview of an HPF-FS, 6 h LPS-stimulated D1 cell. Boxes indicate the area where tomograms have been taken. PM, plasma
membrane; G, Golgi area. (B) Slice of the montaged tomogram. The asterisks indicate MIIC profiles, all containing internal
membranes. The MIICs marked with bright green asterisks represent tubular profiles; all other coloured asterisks belong to MIIC
networks. Bright green, pink and yellow indicate the MIICs that are shown in larger magnification in (C–E). (C, F) Tomographic
slice and reconstructed model of two tubular-shaped MIICs. (D, E) Interconnected MIIC profiles, corresponding to the models
shown in (G, H). (G, H) Reconstructed models of the two reticular MIICs shown in (D, E). Scale bars: (A, B) 1 μm and (C–E)
500 nm.

lysosomes in other cell types (Yamamoto et al., 1998). of bafilomycin A1 . As expected, LPS alone induces
To check the importance of MIIC fusion for trans- transport of MHC class II from intracellular stores
port of MHC class II to the cell surface, we incub- to the cell surface (Supplementary Figure S1 and
ated D1 cells with LPS in the presence or absence S2). On treatment with bafilomycin A1 , most MHC

584 
C The Authors Journal compilation 
C 2010 Portland Press Limited
Three-dimensional organization of the transforming MIIC Research article
Figure 2 Examples of MIICs containing various shapes of internal membranes
Cells were stimulated for 6 h with LPS. (A) A reticular MIIC with traced membranes. Next to the network an intermediate MIIC
can be seen (iMIIC). (B) Reconstructed model of the reticular MIIC in (A). (C) Intact luminal membranes and membrane sheets
(arrowheads) are detected. The later ones are suggestive of membrane degradation activity. (D) Tomographic slices through an
MIIC displaying tubular-shaped luminal membranes. The white arrowheads point to a tubular membrane in cross section. Scale
bars: (A, B) 250 nm and (C, D) 100 nm.

class II remained intracellular (see Supplementary (blue arrowheads in Figure 4A) and ∼ 7 nm wide
Figures S6A and S6B at http://www.biolcell. microfilamentous structures (pink double arrow-
org/boc/102/boc1020581add.htm), associated with heads in Figures 4A and 4B) appear in close
accumulated luminal membranes in MIICs (Supple- proximity to the MIIC networks, thereby form-
mentary Figure S6C). These results suggest that the ing a scaffold of cytoskeleton elements (see the
luminal pH of MIICs and fusion of MIICs are im- model in Figure 4C and Supplementary Movie 6
portant determinants for the translocation of MHC at http://www.biolcell.org/boc/102/boc1020581add.
class II molecules to the cell surface. htm). Short and long MTs appeared arranged par-
allel to the tubular MIIC profiles. Also in IF
Spatial organization of MIIC with cytoskeleton (results not shown) and in immunogold-labelled
elements cryosections of LPS-stimulated DCs, we found close
Recent studies in DCs using IF (immunofluores- association of α-tubulin with the MIICs (Figure 4D).
cence) have established that the profound changes The MTs showed closed capped ends (Figure 4E)
from spherical MIIC into long tubular structures as well as frayed open ends (Figure 4F) and some
highly depend on intact MTs. A close association revealed a kinked appearance (Figure 3G, arrows).
of MIICs with the cytoskeleton was confirmed Physical linkages were detected between the MIIC
after three-dimensional reconstruction analysis. MTs limiting membrane and microfilamentous structures

www.biolcell.org | Volume 102 (11) | Pages 581–591 585


H.E. van Nispen tot Pannerden and others

Figure 3 Fusion and neck profiles within the MIIC network


Cells were stimulated for 3 h (A–C) and 6 h (D–H) with LPS. (A, D, G) Tomographic slices through fusing MIICs. Arrowheads mark
small membrane continuities between adjacent MIICs. The arrows in (G) point out to a kinked MT. (B, E) Series of tomographic
slices showing the small connecting pore depicted in (A, D). (C, F) Reconstructed models and the view through the fusion pore
(arrow, view inset from the right to left). (H) Detail of a neck region in the MIIC network (double arrowhead), and complete model
of the MIIC network (inset). Note the multidomain structure of the MIIC with multiple interconnections (see also Supplementary
Movie 5 at http://www.biolcell.org/boc/102/boc1020581add.htm). (I) Relative number of MIIC profiles that represent individual
MIICs or that are connected to a network, control, n = 18; 3 h LPS, n = 23; 6 h LPS, n = 73. Scale bars, 100 nm.

(Figure 4B, black arrowheads, average distance of org/boc/102/boc1020581add.htm). These MIIC pro-
filament to MIIC was 16+
−5 nm, n = 20), as well as files had diameters ranging from 50 to 200 nm (av-
the MTs (Figures 4G and 4H, black arrowheads). erage volume 2.1×106 nm3 , n = 37) and were fre-
quently found positioned in the close vicinity of the
Small MIIC intermediates extended poles of the reticular MIICs (Figure 5E) and
In our tomograms, we also encountered a large appeared also in the Golgi area. They often contained
number of isolated MIIC profiles, which appeared internal membranes and partially degraded material
not connected to the tubular and reticular MIIC such as short isolated membrane sheets (Figures 5A–
networks (‘iMIIC’ in Figure 2A; Figure 5A–5D; 5D). The small MIIC structures were also detected in
see Supplementary Movie 7 at http://www.biolcell. thin frozen sections of chemically fixed D1 cells, as

586 
C The Authors Journal compilation 
C 2010 Portland Press Limited
Three-dimensional organization of the transforming MIIC Research article
Figure 4 Physical linkages connect transforming MIICs with cytoskeleton elements
Cells were stimulated for 6 h with LPS. (A) A tubular part of the MIIC (yellow asterisks) is flanked by an MT (blue arrowheads)
and microfilaments (pink double arrowheads). (B) Highlighted subdomain of the same MIIC showing distinct connections (black
arrowheads) between microfilaments and the MIICs limiting membrane. (C) Reconstructed side view of the MIIC reticulum
(yellow asterisks in A) with associated MTs and microfilaments. (D) Chemically fixed cryosection of a stimulated D1 cell (6 h
LPS). Immunogold labelling of α-tubulin associated with an MIIC. (E) MT with a capped end (arrow). (F) MT with a frayed end
(arrows). (G, H) Tomographic slices through longitudinal (G) and cross-sectioned (H) orientated MT displaying physical linkages
to the limiting membranes of MIICs (black arrowheads). Scale bars: (A) 250 nm, (B, E–H) 100 nm and (D) 200 nm.

verified by immunogold labelling using anti-MHC study the three-dimensional architecture of the endo-
class II antibodies (Figure 5F). However, due to the lysosomal system in DCs. In immature D1 cells, early
limited Z resolution of thin frozen sections, we can- endosomes and MIICs are globular shaped, some-
not exclude that at least a part of them represent times with small tubular extensions (Murk et al.,
cross-sectioned MIIC tubules. The positioning of a 2004). Based on the morphological criteria (i.e. mul-
population of small MIIC profiles within the Golgi tivesicular and multilaminar morphology), and the
area could point towards early stages in the forma- characteristic immunolabelling patterns of invariant
tion of MIICs. Although MIIC intermediates were chain, HLA-DM and MHC class II, we here show
sometimes closely positioned to trans-Golgi stacks, that the multivesicular and multilaminar structures
we have not found direct membrane continuities with presented in our tomograms are intermediate/late
the TGN. MIICs. Our three-dimensional analysis revealed that
after LPS stimulation, MIICs form large intercon-
necting membrane networks with alternating spher-
Discussion ical and tubular domains. This typical spatial mor-
In the present study we have generated large-scale phology of the MIIC, the large increase in their size
three-dimensional cell volumes from HPF-FS LPS- and the presence of small membrane openings con-
stimulated D1 cells to define the spatial membrane necting them indicate that MIICs have the capacity
architecture of transformed MIICs. The HPF-FS tech- to fuse with each other during DC maturation. This
nology preserves the cellular fine structure in a close- fusion of individual MIICs contributes to the forma-
to-live condition, and has previously been used to tion of large extended and reticular MIICs. Whether

www.biolcell.org | Volume 102 (11) | Pages 581–591 587


H.E. van Nispen tot Pannerden and others

Figure 5 Small MIIC profiles not connected to a network cells (Wubbolts et al., 1996), B-cells and mouse DCs
Cells were stimulated for 6 h with LPS. (A–D) Gallery of small (Boes et al., 2002; Chow et al., 2002). It was pro-
isolated MIICs. The diameter of these MIICs varies from 50 posed that tubular MIICs extending towards the cell
to 200 nm (average volume 2.1×106 nm3 , n = 37). Note that periphery could facilitate the transfer of MHC class II
some of the internal membrane structures are incompletely to the plasma membrane, either directly or through
closed vesicles. (E) Series of tomographic slices showing an vesicular intermediates budding from the tubules.
isolated MIIC profile in close apposition to an MIIC reticulum. An important reason to use HPF-FS and ET analyses
(F) MHC class II immunogold-labelled cryosection of a small is the ability to capture the rapid membrane dynam-
MIIC intermediate. G, Golgi. Scale bars (A–D) 100 nm and ics that occur during MIIC transformation. When
(E–F) 200 nm. analysing large-scale cell volumes in the cell peri-
phery, we found no membrane connections of reticu-
lar MIICs with the cell surface. In a previous study,
we reported on MHC II-containing structures prox-
imate to the tips of MIIC tubules. These structures
exhibited a diameter similar to that of the tubules,
suggesting that these could derive from the tubules
(Figure 3D in Kleijmeer et al., 2001). We here con-
firm the existence of such isolated vesicular MIIC
profiles in maturating D1 cells, and their close posi-
tioning to tubular/reticular MIICs. The fact that they
contain signs of internal membrane degradation may
indeed be an indication of their origin from the MIIC
tubules/reticula and represent a population of MHC
class II carrier vesicles en route to the cell surface. Be-
sides at the polar ends of tubular profiles of the MIIC
reticulum, isolated MIIC vesicles were also detected
in the vicinity of TGN exit sites, where they often
exhibited close membrane apposition to trans-Golgi
stacks. The isolated MIIC vesicles may thus also rep-
resent early stages in the formation of the MIICs.
the reticular MIICs are further interconnected and However, when analysing many TGN exit sites, we
form a large reticulum throughout the cell requires were not able to detect any membrane continuities
whole cell reconstruction analysis, which was not es- between MIIC intermediates and trans-Golgi stacks.
tablished in the present study. It is of course possible that despite our large-scale
Previous studies have suggested that back-fusion volume analysis, such continuities have still escaped
of the luminal membranes with the limiting mem- our detection.
brane of the MIICs is responsible for MIIC tubu- The question arises what is the advantage of MIIC
lation and timed translocation of MHC class II to fusion and the formation of an elaborate MIIC net-
the cell surface (Kleijmeer et al., 2001). Continuit- work, for the transport of MHC class II to the cell sur-
ies between luminal vesicles and the limiting mem- face. MIIC-derived tubules are long and directional
brane of the MIIC tubular reticulum were occasion- when DCs are co-cultured with T-cells (Boes et al.,
ally observed in tomograms of LPS-stimulated D1 2002; Vyas et al., 2007). Shorter and more randomly
cells. However, we could not establish if these profiles oriented MIIC tubules are observed on Toll-like re-
represent inward budding of newly formed vesicles ceptor ligation (Vyas et al., 2007). This may suggest
or back-fusion of pre-existing vesicles as suggested that formation of a reticulum such as observed here in
previously (Kleijmeer et al., 2001). cultured D1 cells represents a transitional stage and
Using GFP (green fluorescent protein)-tagged requires other ‘extracellular’ stimuli (i.e. from inter-
MHC class II, fusion of class II-containing vesicles acting T-cells) to ultimately form the long tubules
with the cell surface has been reported in cultured observed in co-cultures (Boes et al., 2002; Bertho

588 
C The Authors Journal compilation 
C 2010 Portland Press Limited
Three-dimensional organization of the transforming MIIC Research article
et al., 2003; Vyas et al., 2007). Fusion between MIICs For HPF-FS experiments the Petri dishes contained golden grids
may also be important for ‘temporal’ regulation of the layered with a Formvar film and a thin layer of gelatin (Murk
et al., 2003b). To stimulate the D1 cells, LPS (Sigma–Aldrich,
internal pH required for optimal processing of anti- St. Louis, MO, U.S.A.) was added to the medium (final concen-
gens. Both in macrophages and DCs, fusion of pha- tration 10 μg/ml) for 3 or 6 h as described previously (Kleijmeer
gosomes with lysosome-related organelles has been et al., 2001). The maturation state of the cells was verified mor-
described as a mechanism to prevent full degradation phologically and after determination of the cell surface expression
of antigens (Savina et al., 2006; Jancic et al., 2007; of MHC class II by IF and IEM analyses. In some experiments, the
cells were exposed to 100 nM bafilomycin A1 (Sigma–Aldrich)
Mantegazza et al., 2008). Absence of fusion could lead in DMSO (final concentration 0.1%) half an hour before and
to accelerated acidification and degradation of anti- during LPS stimulation.
genic peptides. It is well established that treatment of
APCs with alkalinization drugs results in impaired HPF-FS
peptide loading and T-cell response (Savina et al., Cryo-immobilization and freeze substitution were performed es-
2006; Jancic et al., 2007; Mantegazza et al., 2008). sentially as described before (Murk et al., 2003b). In short,
Stimulation of D1 cells with LPS in the presence of cells adhering to the grids were sandwiched between two alu-
minium cups (flat side towards the grid) and immediately cryo-
bafilomycin A1 completely blocked the delivery of immobilized using a high-pressure freezer (Leica HPF; Leica
MHC class II to the cell surface and resulted in the Microsystems, Vienna, Austria; now M. Wohlwend, Sannwald,
intracellular accumulation of MIICs. These observa- Switzerland). The samples were transferred to the freeze sub-
tions indicate that MIIC fusion and MHC class II stitution apparatus (Leica AFS; Leica Microsystems), containing
precooled anhydrous acetone as the substitution fluid, and fixed
transport to the cell surface are related to the luminal with 0.5% osmium tetroxide and 0.25% glutaraldehyde in an-
pH of the MIICs. Formation of MIIC tubules has hydrous acetone at a temperature of − 90◦ C for 48 h. The tem-
been demonstrated to depend on intact MTs (Vyas perature was increased by 1◦ C/h to − 30◦ C and this temperature
et al., 2007). In our electron tomograms we found was further maintained for 8 h. The substitution medium was
washed away by anhydrous acetone and the cells and grids were
MTs and intermediate filaments flanking the reticu- embedded in Epon.
lar MIIC. These MTs were often kinked and fragmen-
ted, and exhibited flaired and capped ends, suggest-
Tomography and data analysis
ing that they may have been severed during MIIC Sections of 200–400 nm thickness were collected on 1 hole
transformation. Our three-dimensional analysis also Formvar-coated copper grids, and post-stained with uranyl acet-
revealed that physical linkages exist between the lim- ate and lead citrate. Fiducial markers (10 or 15 nm gold particles)
iting membrane of the reticular MIICs and MTs and were applied to both sides of the section and one extra layer of
Formvar was applied on top of the section for stability. Cells
intermediate filaments, suggesting that fusion and displaying the typical activated dendritic phenotype were pre-
tubulation of MIICs depend on these cytoskeleton selected using a Jeol 1010 electron microscope (Jeol). Dual-
elements. axis tilt series of selected MIICs were then recorded using
In summary, our ET analysis provides insight into a Tecnai 20 transmission electron microscope (FEI) equipped
the mechanism of MIIC reorganization during DC with a slow scan CCD camera (charge-coupled-device camera;
Temcam F214; TVIPS, Gauting, Germany), as previously de-
maturation. We have shown that intermediate and scribed (Murk et al., 2003a, 2004). Tilt series were acquired
late MIICs are capable of fusing together to form re- using a single-axis high-tilt tomography holder (model 670;
ticular membrane networks. We also show that phys- Gatan, München, Germany) or a Fischione tomography holder
ical linkages exist between the transforming MIICs (model 2020; Fischione Instruments, K-Vision BV, Huizen,
The Netherlands). After the first tilt series the specimens were
and MTs and intermediate filaments. Furthermore, manually rotated over an angle of 90◦ , and the second tilt
we have confirmed the existence of vesicular MIIC series were acquired. Then, 1000×1000 images were recor-
profiles, possibly representing MHC class II carriers ded automatically with Xplore 3D software (FEI) with an
en route to the plasma membrane or early stages in angular range from − 60◦ to +60◦ with 1◦ increment. Tilt
series were aligned with ETomo, a program from the IMOD
MIIC formation. program package (Kremer et al., 1996; Mastronarde, 1997;
http://bio3d.colorado.edu/imod/), using the fiducial markers,
and combined into one three-dimensional volume. First, aligned
Materials and methods single-axis tomograms were computed by resolution-weighted
Cell culture and cell treatments back-projection. These two volumes were combined into one
The mouse DC cell line D1 was cultured as described previously final volume. In some cases (e.g. Figure 1), tomograms of serial
(Winzler et al., 1997). The cells were grown in Petri dishes. sections were stitched together using the ETomo join applic-
For IF experiments the Petri dishes contained glass coverslips. ation. After reconstruction each tomogram was unpacked into

www.biolcell.org | Volume 102 (11) | Pages 581–591 589


H.E. van Nispen tot Pannerden and others

tiff format and manually stitched together with adjacent tomo- scope. The acquisition settings were kept the same for the dif-
grams using Adobe Photoshop CS3. The tiff files were stacked ferent conditions within each experiment. The subcellular dis-
back into a three-dimensional volume (per section) and the three tribution pattern of MHC class II was quantified in control and
serial sections were joined together using ETomo join. For ex- LPS-stimulated cells (+
−bafilomycin A1 ) by counting five to eight
ample, the tomogram presented in Figure 1 is composed of random fields reaching a total number of ∼ 100 cells per con-
one single-axis and 12 dual-axis tilt series spread over three dition with respectively intracellular, mixed and predominantly
serial sections, resulting in a reconstructed volume of 23.6 μm3 . surface-expressed MHC class II (n = 100 cells).
Within the IMOD program, MIIC membranes were manually
traced to form a three-dimensional representation of the mem- Author contribution
branes. The MIICs were identified by morphological criteria, i.e.
membrane-bound organelles containing luminal membranes, as The concept of the research was created by Monique
also recognized by MHC class II immunolabelling in thin frozen Kleijmeer and Harry Heijnen, and the design and
sections. supervision of the research was by Harry Heijnen.
Volumes of the modelled organelles were measured using the Experimental work was carried out by Hezder van
IMOD info option in IMOD. Quantification of MIIC profiles
was done on randomly selected tomographic slices from sev- Nispen tot Pannerden, and Willie Geerts participated
eral tomograms and different maturation stages (3 and 6 h LPS in the acquisition of the three-dimensional data. Data
stimulation) of the D1 cells. For each MIIC profile we analysed analysis, interpretation and writing of the manuscript
whether the structure was connected to an MIIC network or were carried out by Hezder van Nispen tot Pannerden
represented an isolated spherical/tubular MIIC.
The statistical significance for the quantification of tubule and Harry Heijnen.
diameter was assessed by a two-tailed unpaired Student’s t test.
The difference in tubule diameter was considered statistically Acknowledgements
significant when P < 0.05. We are grateful to V. Kondylis for his help with con-
focal microscopy, G. Posthuma for useful discussions
IEM
For detection of MHC class II, monoclonal rat anti-mouse MHC
on quantification and S. van Dijk for her assistance
class II antibody was used [M5/114 (Bhattacharya et al., 1981); with ultrathin sectioning and immunolabelling. We
BD PharMingen, San Diego, CA, U.S.A.]. Mouse anti-α-tubulin also thank M. van Peski and R. Scriwanek for their
was obtained from Sigma–Aldrich. Rat anti-mouse H2DM help with preparation of the Movies and Figures and
(2E5A) directed to the HLA-DM β-chain and rat anti-mouse
invariant chain (In-1) have been described previously (Kleijmeer
J.L. Murk for his help with sample preparation. We
et al., 2001). For immunogold labelling, cells were fixed in a mix- thank Dr P. Ricciardi-Castagnoli (Department of Bio-
ture of 2% (w/v) paraformaldehyde and 0.2% glutaraldehyde in technology and Bioscience, University of Milano-
0.1 M phosphate buffer (0.1 M NaH2 PO4 and 0.1 M Na2 HPO4 Bicocca, Milan, Italy) for providing us with D1 cell
in aqua dest). The cells were washed with PBS/lysine to quench line.
free aldehydes, then embedded in gelatin (Liou et al., 1997) and
infiltrated in 2.3 M sucrose, followed by rapid freezing in liquid
N2 . Then, 50 nm thick cryosections were cut at − 120◦ C us- Funding
ing an Ultracut-S ultra microtome (Leica Microsystems, Vienna, This work was supported by The Netherlands Organ-
Austria). The sections were collected on Formvar-coated grids
using a mixture of 1.8% methylcellulose and 2.3 M sucrose (Liou isation for Scientific Research [grant number ALW
et al., 1996), and incubated with primary antibodies and Protein 813.08.001 (to H.E.v.N.t.P.)].
A–gold (Slot et al., 1991). As bridging antibodies, rabbit anti-
mouse IgG and rabbit anti-rat IgG were used. Immunogold
labelling was performed using 10 nm gold particles (Slot and
References
Alfonso, C. and Karlsson, L. (2000) Nonclassical MHC class II
Geuze, 1985). After labelling, the sections were fixed with 1%
molecules. Ann. Rev. Immunol. 18, 113–142
glutaraldehyde, counterstained with uranyl acetate, and embed- Bertho, N., Cerny, J., Kim, Y.M., Fiebiger, E., Ploegh, H. and Boes, M.
ded in methylcellulose-uranyl acetate. The sections were viewed (2003) Requirements for T cell-polarized tubulation of class II+
in a Jeol 1200CX electron microscope (Jeol). compartments in dendritic cells. J. Immunol. 171, 5689–5696
Bhattacharya, A., Dorf, M.E. and Springer, T.A. (1981) A shared
Immunofluorescence alloantigenic determinant on Ia antigens encoded by the I-A and
Control and LPS-stimulated D1 cells were fixed with 4% para- I-E subregions: evidence for I region gene duplication. J. Immunol.
formaldehyde in PBS. The cells were washed with PBS and 127, 2488–2495
Boes, M., Cerny, J., Massol, R., Op den Brouw, M., Kirchhausen, T.,
quenched with NaBH4 (final concentration 1 μg/ml in PBS) fol-
Chen, J. and Ploegh, H.L. (2002) T-cell engagement of dendritic
lowed by permeabilization in 0.5% Triton X-100 and blocked cells rapidly rearranges MHC class II transport. Nature 418,
with fish skin gelatin (0.2%). The samples were incubated with 983–988
rat anti-mouse MHC class II antibody, followed by secondary Buschow, S.I., Liefhebber, J.M., Wubbolts, R. and Stoorvogel, W.
anti-rat IgG coupled with Alexa Fluor® 568 (Molecular Probes). (2005) Exosomes contain ubiquitinated proteins. Blood Cells Mol.
Images were acquired using a Zeiss LSM-510 confocal micro- Dis. 35, 398–403

590 
C The Authors Journal compilation 
C 2010 Portland Press Limited
Three-dimensional organization of the transforming MIIC Research article
Chow, A., Toomre, D., Garrett, W. and Mellman, I. (2002) Dendritic Murk, J.L., Posthuma, G., Koster, A.J., Geuze, H.J., Verkleij, A.J.,
cell maturation triggers retrograde MHC class II transport from Kleijmeer, M.J. and Humbel, B.M. (2003b) Influence of aldehyde
lysosomes to the plasma membrane. Nature 418, 988–994 fixation on the morphology of endosomes and lysosomes:
Geuze, H.J. (1998) The role of endosomes and lysosomes in MHC quantitative analysis and electron tomography. J. Micr. 212,
class II functioning. Immun. Today 19, 282–287 81–90
Hart, D.N. (1997) Dendritic cells: unique leukocyte populations which Neefjes, J.J., Stollorz, V., Peters, P.J., Geuze, H.J. and Ploegh, H.L.
control the primary immune response. Blood 90, 3245–3287 (1990) The biosynthetic pathway of MHC class II but
Jancic, C., Savina, A., Wasmeier, C., Tolmachova, T., El-Benna, J., not class I molecules intersects the endocytic route. Cell 61,
Dang, P.M., Pascolo, S., Gougerot-Pocidalo, M.A., Raposo, G., 171–183
Seabra, M.C. and Amigorena, S. (2007) Rab27a regulates Raposo, G., Nijman, H.W., Stoorvogel, W., Liejendekker, R., Harding,
phagosomal pH and NADPH oxidase recruitment to dendritic cell C.V., Melief, C.J. and Geuze, H.J. (1996) B lymphocytes secrete
phagosomes. Nature Cell Biol. 9, 367–378 antigen-presenting vesicles. J. Exp. Med. 183, 1161–1172
Kleijmeer, M., Ramm, G., Schuurhuis, D., Griffith, J., Rescigno, M., Sallusto, F., Cella, M., Danieli, C. and Lanzavecchia, A. (1995)
Ricciardi-Castagnoli, P., Rudensky, A.Y., Ossendorp, F., Melief, Dendritic cells use macropinocytosis and the mannose
C.J., Stoorvogel, W. and Geuze, H.J. (2001) Reorganization of receptor to concentrate macromolecules in the major
multivesicular bodies regulates MHC class II antigen presentation histocompatibility complex class II compartment: downregulation
by dendritic cells. J. Cell Biol. 155, 53–63 by cytokines and bacterial products. J. Exp. Med. 182,
Kleijmeer, M.J., Morkowski, S., Griffith, J.M., Rudensky, A.Y. and 389–400
Geuze, H.J. (1997) Major histocompatibility complex class II Savina, A., Jancic, C., Hugues, S., Guermonprez, P., Vargas, P.,
compartments in human and mouse B lymphoblasts represent Moura, I.C., Lennon-Dumenil, A.M., Seabra, M.C., Raposo, G. and
conventional endocytic compartments. J. Cell Biol. 139, 639–649 Amigorena, S. (2006) NOX2 controls phagosomal pH to regulate
Kremer, J.R., Mastronarde, D.N. and McIntosh, J.R. (1996) Computer antigen processing during crosspresentation by dendritic cells.
visualization of three-dimensional image data using IMOD. J. Cell 126, 205–218
Struct. Biol. 116, 71–76 Slot, J.W. and Geuze, H.J. (1985) A new method of preparing gold
Kropshofer, H., Hammerling, G.J. and Vogt, A.B. (1999) The impact of probes for multiple-labeling cytochemistry. Eur. J. Cell Biol. 38,
the non-classical MHC proteins HLA-DM and HLA-DO on loading 87–93
of MHC class II molecules. Immunol. Rev. 172, 267–278 Slot, J.W., Geuze, H.J., Gigengack, S., Lienhard, G.E. and James,
Liou, W., Geuze, H.J., Geelen, M.J. and Slot, J.W. (1997) The D.E. (1991) Immuno-localization of the insulin regulatable glucose
autophagic and endocytic pathways converge at the nascent transporter in brown adipose tissue of the rat. J. Cell Biol. 113,
autophagic vacuoles. J. Cell Biol. 136, 61–70 123–135
Liou, W., Geuze, H.J. and Slot, J.W. (1996) Improving structural Trombetta, E.S. and Mellman, I. (2005) Cell biology of antigen
integrity of cryosections for immunogold labeling. Histochem. Cell processing in vitro and in vivo. Ann. Rev. Immunol. 23, 975–1028
Biol. 106, 41–58 Vyas, J.M., Kim, Y.M., Artavanis-Tsakonas, K., Love, J.C., Van Der
Mantegazza, A.R., Savina, A., Vermeulen, M., Perez, L., Geffner, J., Veen, A.G. and Ploegh, H.L. (2007) Tubulation of class II MHC
Hermine, O., Rosenzweig, S.D., Faure, F. and Amigorena, S. (2008) compartments is microtubule dependent and involves multiple
NADPH oxidase controls phagosomal pH and antigen cross- endolysosomal membrane proteins in primary dendritic cells.
presentation in human dendritic cells. Blood 112, 4712–4722 J. Immunol. 178, 7199–7210
Mastronarde, D.N. (1997) Dual-axis tomography: an approach with Watts, C. (1997) Capture and processing of exogenous antigens for
alignment methods that preserve resolution. J. Struct. Biol. 120, presentation on MHC molecules. Ann. Rev. Immunol. 15, 821–850
343–352 Winzler, C., Rovere, P., Rescigno, M., Granucci, F., Penna, G., Adorini,
Mellman, I., Turley, S.J. and Steinman, R.M. (1998) Antigen L., Zimmermann, V.S., Davoust, J. and Ricciardi-Castagnoli, P.
processing for amateurs and professionals. Trends Cell Biol. 8, (1997) Maturation stages of mouse dendritic cells in growth
231–237 factor-dependent long-term cultures. J. Exp. Med. 185, 317–328
Murk, J.L., Humbel, B.M., Ziese, U., Griffith, J.M., Posthuma, G., Wubbolts, R., Fernandez-Borja, M., Oomen, L., Verwoerd, D.,
Slot, J.W., Koster, A.J., Verkleij, A.J., Geuze, H.J. and Kleijmeer, Janssen, H., Calafat, J., Tulp, A., Dusseljee, S. and Neefjes, J.
M.J. (2003a) Endosomal compartmentalization in three (1996) Direct vesicular transport of MHC class II molecules from
dimensions: implications for membrane fusion. Proc. Natl. Acad. lysosomal structures to the cell surface. J. Cell Biol. 135,
Sci. U.S.A. 100, 13332–13337 611–622
Murk, J.L., Lebbink, M.N., Humbel, B.M., Geerts, W.J., Griffith, J.M., Yamamoto, A., Tagawa, Y., Yoshimori, T., Moriyama, Y., Masaki, R.
Langenberg, D.M., Verreck, F.A., Verkleij, A.J., Koster, A.J., Geuze, and Tashiro, Y. (1998) Bafilomycin A1 prevents maturation of
H.J. and Kleijmeer, M.J. (2004) 3-D structure of multilaminar autophagic vacuoles by inhibiting fusion between
lysosomes in antigen presenting cells reveals trapping of MHC II autophagosomes and lysosomes in rat hepatoma cell line, H-4-II-E
on the internal membranes. Traffic 5, 936–945 cells. Cell Struct. Funct. 23, 33–42

Received 13 April 2010/13 August 2010; accepted 16 August 2010


Published as Immediate Publication 16 August 2010, doi:10.1042/BC20100046

www.biolcell.org | Volume 102 (11) | Pages 581–591 591


Biol. Cell (2010) 102, 581–591 (Printed in Great Britain) doi:10.1042/BC20100046

Supplementary online data


Spatial organization of the transforming MHC class II
compartment
Hezder E. van Nispen tot Pannerden*†, Willie J. Geerts*‡, Monique J. Kleijmeer*† and Harry F.G. Heijnen*§1
*Institute of Biomembranes, Padualaan 8, 3584 CH Utrecht, The Netherlands, †Cell Microscopy Center, Department of Cell Biology,
University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands, ‡Department of Molecular Cell Biology, Utrecht
University, Utrecht, The Netherlands, and §Laboratory of Clinical Chemistry and Haematology, University Medical Center Utrecht,
Heidelberglaan 100, 3584 CX Utrecht, The Netherlands

Figure S1 MHCII transport to the cell surface after LPS


stimulation
Representative confocal images showing immunostaining of
MHC class II in untreated D1 cells (A) and cells stimulated for
6 h with LPS (B). After 6 h of LPS stimulation, much of the
MHC class II has reached the plasma membrane. Scale bar,
10 μm.

1 To whom correspondence should be addressed (email


h.f.g.heijnen@umcutrecht.nl).

www.biolcell.org | Volume 102 (11) | Pages 581–591


H.E. van Nispen tot Pannerden and others

Figure S2 Identification of MIICs by immunogold labelling


(A) Non-stimulated control D1 cell. MHC class II localizes to the luminal and limiting membranes of the spherical MIIC, and is
virtually absent from the plasma membrane (PM). (B) After 6 h of LPS treatment, MHC class II is both found on the plasma
membrane and associated with irregularly shaped MIICs. Scale bars, 200 nm.


C The Authors Journal compilation 
C 2010 Portland Press Limited
Three-dimensional organization of the transforming MIIC

Figure S3 Identification of early and late MIICs


D1 cells were stimulated for 6 h with LPS. (A, B) Two subsequent cryosections showing abundant labelling of HLA-DM on the
limiting membrane of multivesicular phenotypes representing intermediate and late MIICs. Note the absence of label in the early
MIICs with low vesicle number. Arrows indicate interconnecting MIICs. (C, D) Vice versa, early compartments with sparse luminal
vesicles (EE) contain abundant invariant chain (Ii), whereas multivesicular MIIC phenotypes, representing the late stages, are
devoid of invariant chain. Note also the abundant labelling of invariant chain in the ER (arrowheads). Two subsequent cryosections
are shown. PM, plasma membrane; MIIC, MHC class II compartment; ER, endoplasmic reticulum; EE, early endosomes. Scale
bars: (A, B) 500 nm and (C, D) 200 nm.

www.biolcell.org | Volume 102 (11) | Pages 581–591


H.E. van Nispen tot Pannerden and others

Figure S4 Early endosomes


(A) Tubular structures emerging from early endosomes (EE) in a 3 h LPS-stimulated D1 cell. (B) Two models of early endosomes
[the pink model represents the early endosomes in (A)]. Gold colour indicates internal vesicles. Arrowheads indicate tubules
emerging from the early endosomes. Scale bars, 100 nm.

Figure S5 Series of tomographic slices through two MIICs in non-stimulated D1 cells


The closely positioned MIICs are not interconnected and are both completely captured within the tomogram. Inset: 3D model.
Scale bar, 100 nm.


C The Authors Journal compilation 
C 2010 Portland Press Limited
Three-dimensional organization of the transforming MIIC

Figure S6 Effect of bafilomycin A1 on MHCII transport


(A) Representative confocal image showing immunostaining of MHC class II in D1 cells stimulated for 6 h with LPS in the
presence of bafilomycin A1 . MHC class II remains mostly intracellular (compare with non-stimulated cells in Supplementary
Figure S2A). (B) Semi-quantitative evaluation of the MHC class II distribution in control and LPS-stimulated D1 cells (in the
presence or absence of bafilomycin A1 ). The percentages of D1 cells exhibiting the indicated staining patterns of MHC class II
are shown: mostly intracellular, partially transported to the cell surface, or mostly expressed on the cell surface (n = 100 cells
per condition). (C) Example of MIIC profile after 6 h of LPS stimulation in the presence of bafilomycin A1 . Accumulated luminal
vesicles contain large numbers of MHC class II (10 nm gold). Scale bars: (A) 10 μm and (C) 200 nm.

Received 13 April 2010/13 August 2010; accepted 16 August 2010


Published as Immediate Publication 16 August 2010, doi:10.1042/BC20100046

www.biolcell.org | Volume 102 (11) | Pages 581–591

Você também pode gostar