ArticlePDF Available

LC3-dependent Intracellular Membrane Tubules Induced by γ-Protocadherins A3 and B2

Authors:

Abstract and Figures

Clustered protocadherins (Pcdhs) are a family of cadherin-like molecules arranged in gene clusters (α, β, and γ). γ-Protocadherins (Pcdh-γs) are involved in cell-cell interactions, but their prominent intracellular distribution in vivo and different knock-out phenotypes suggest that these molecules participate in still unidentified processes. We found using correlative light and electron microscopy that Pcdh-γA3 and -γB2, but not -γC4, -α1, or N-cadherin, generate intracellular juxtanuclear membrane tubules when expressed in cells. These tubules recruit the autophagy marker MAP1A/1B LC3 (LC3) but are not associated with autophagic vesicles. Lipidation of LC3 is required for its coclustering with Pcdh-γ tubules, suggesting the involvement of an autophagic-like molecular cascade. Expression of wild-type LC3 with Pcdh-γA3 increased tubule length whereas expression of lipidation-defective LC3 decreased tubule length relative to Pcdh-γA3 expressed alone. The tubules were found to emanate from lysosomes. Deletion of the luminal/extracellular domain of Pcdh-γA3 preserved lysosomal targeting but eliminated tubule formation whereas cytoplasmic deletion eliminated both lysosomal targeting and tubule formation. Deletion of the membrane-proximal three cadherin repeats resulted in tubes that were narrower than those produced by full-length molecules. These results suggest that Pcdh-γA and -γB families can influence the shape of intracellular membranes by mediating intraluminal interactions within organelles.
Content may be subject to copyright.
Rachel B. Hazan and Greg R. Phillips
Yildirim, Rebecca Lee, Kimita Suyama,
Fernández-Monreal, Twethida Oung, Murat
Hugo H. Hanson, Semie Kang, Mónica
INTERACTIONS
and B2: A ROLE FOR INTRALUMINAL
-Protocadherins A3γTubules Induced by
LC3-dependent Intracellular Membrane
Cell Biology:
doi: 10.1074/jbc.M109.092031 originally published online May 3, 2010
2010, 285:20982-20992.J. Biol. Chem.
10.1074/jbc.M109.092031Access the most updated version of this article at doi:
.JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
Supplemental material:
http://www.jbc.org/jbc/suppl/2010/05/03/M109.092031.DC1.html
http://www.jbc.org/content/285/27/20982.full.html#ref-list-1
This article cites 48 references, 21 of which can be accessed free at
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
LC3-dependent Intracellular Membrane Tubules Induced by
-Protocadherins A3 and B2
A ROLE FOR INTRALUMINAL INTERACTIONS
*
S
Received for publication, December 4, 2009, and in revised form, April 6, 2010 Published, JBC Papers in Press, May 3, 2010, DOI 10.1074/jbc.M109.092031
Hugo H. Hanson
‡1
, Semie Kang
‡1
,Mo´ nica Ferna´ ndez-Monreal
, Twethida Oung
, Murat Yildirim
, Rebecca Lee
,
Kimita Suyama
§
, Rachel B. Hazan
§
, and Greg R. Phillips
‡2
From the
Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029 and the
§
Department of
Pathology, Albert Einstein College of Medicine, Bronx, New York 10461
Clustered protocadherins (Pcdhs) are a family of cadherin-
like molecules arranged in gene clusters (
,
, and
).
-Pro-
tocadherins (Pcdh-
s) are involved in cell-cell interactions, but
their prominent intracellular distribution in vivo and different
knock-out phenotypes suggest that these molecules participate
in still unidentified processes. We found using correlative light
and electron microscopy that Pcdh-
A3 and -
B2, but not -
C4,
-
1, or N-cadherin, generate intracellular juxtanuclear mem-
brane tubules when expressed in cells. These tubules recruit the
autophagy marker MAP1A/1B LC3 (LC3) but are not associated
with autophagic vesicles. Lipidation of LC3 is required for its
coclustering with Pcdh-
tubules, suggesting the involvement of
an autophagic-like molecular cascade. Expression of wild-type
LC3 with Pcdh-
A3 increased tubule length whereas expression
of lipidation-defective LC3 decreased tubule length relative to
Pcdh-
A3 expressed alone. The tubules were found to emanate
from lysosomes. Deletion of the luminal/extracellular domain
of Pcdh-
A3 preserved lysosomal targeting but eliminated
tubule formation whereas cytoplasmic deletion eliminated both
lysosomal targeting and tubule formation. Deletion of the mem-
brane-proximal three cadherin repeats resulted in tubes that
were narrower than those produced by full-length molecules.
These results suggest that Pcdh-
A and -
B families can influ-
ence the shape of intracellular membranes by mediating intralu-
minal interactions within organelles.
Clustered protocadherins (Pcdhs)
3
comprise 50 different
cadherin-like transmembrane proteins arranged into three
genomic clusters, termed
,
, and
(1, 2). The Pcdh genes
within the
and
clusters, numbering 14 and 22, respectively,
share constant cytoplasmic domains within their respective
clusters by alternative splicing (3). The Pcdh-
s participate in
cell-cell interactions (4, 5) with homophilic properties (6) and
have been localized at synapses (68), but also have a promi-
nent intracellular distribution in cultured cells and in vivo (6, 8).
For Pcdh-
s, conflicting evidence has been presented as to
whether they mediate cell-cell interactions (9–11), but a syn-
aptic localization has been reported (1).
Complete genetic deletion of Pcdh-
s reduced the number of
synaptic specializations in spinal cord but also caused substan-
tial apoptosis of spinal cord interneurons (7, 12). Conditional
knock out in retina also increased apoptosis of some retinal cell
types but, unlike in spinal cord, did not affect synaptic connec-
tivity (13). Conditional knock out in astrocytes did not cause
apoptosis but delayed synaptic development (14). The multiple
effects of Pcdh-
deletion may indicate still unidentified cellu-
lar roles for these molecules.
We show here that Pcdh-
A3 and -
B2 generate intracellu-
lar membrane tubules that recruit the autophagic vesicle pro-
tein LC3, the mammalian homologue of yeast Atg8. LC3 is lipi-
dated via a reaction similar to the ubiquitin ligase cascade and
targets to nascent autophagosomes where it is thought to sta-
bilize the phagophore (15, 16) or promote its closure (17). How
LC3 carries out this function(s) is completely unknown. LC3 is
a member of a family of proteins including GABARAP, GEC-1,
and GATE-16 (18), all of which are implicated in protein or
vesicle trafficking. We found that LC3 recruitment to Pcdh-
-
induced tubules was dependent on LC3 lipidation. Coexpres-
sion of Pcdh-
A3 with lipidation-defective LC3 reduced tubule
length whereas expression of wild-type LC3 increased tubule
length compared with Pcdh-
A3 expressed alone. The tubules
emanated from lysosomes. The luminal/extracellular domain
of Pcdh-
A3 was found to be required for tubule formation,
and shortening its length resulted in narrower tubules. Thus,
Pcdh-
s, in concert with LC3 family members, could partici-
pate in the biogenesis of their own trafficking organelles via
intraluminal homophilic interactions. The effect of LC3 pertur-
bation on tubule length is consistent with a role for LC3 in
promoting or stabilizing membrane expansion.
EXPERIMENTAL PROCEDURES
Plasmid Constructs—Pcdh-
A3-GFP has been described (8).
Pcdh-
B2-YFP and Pcdh-
C4-YFP were provided by Dr.
Joshua Weiner (University of Iowa). Luminal/extracellular
deleted Pcdh-
A3-GFP was provided by Drs. Marcus Frank and
Ingrid Haas (Max Planck Institute). Pcdh-
1-GFP was pro-
vided by Dr. Qiang Wu (Shanghai Jiao Tong University).
*This work was supported by National Institutes of Health Grant NS051238
and an Irma T. Hirschl award (to G. R. P.).
S
The on-line version of this article (available at http://www.jbc.org) contains
supplemental Figs. 1– 4.
1
Both authors contributed equally to this work.
2
To whom correspondence should be addressed: Dept. of Neuroscience,
Mount Sinai School of Medicine, Box 1065, One Gustave L. Levy Place, New
York, NY 10029. Fax: 212-659-8574; E-mail: greg.phillips@mssm.edu.
3
The abbreviations used are: Pcdh, protocadherin; GFP, green fluorescent
protein; YFP, yellow fluorescent protein; RFP, red fluorescent protein;
N-cad, N-cadherin; LC3, MAP 1A/1B LC3; CLEM, correlative light and elec-
tron microscopy; wt, wild type; ER, endoplasmic reticulum; CCD, cytoplas-
mic domain; ECD, extracellular domain.
THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 285, NO. 27, pp. 20982–20992, July 2, 2010
© 2010 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.
20982 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285NUMBER 27• JULY 2, 2010
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
N-cadherin (N-cad)-Venus was provided by Dr. Hidekazu
Tanaka (Osaka University School of Medicine). RFP-LC3 was
provided by Dr. Zhenyue Yue (Mount Sinai School of Medi-
cine). hcRed-LC3wt and hcRed-LC3G were provided by Dr.
Isei Tanida (National Institute of Infectious Diseases, Tokyo,
Japan). Cytoplasmic domain-deleted Pcdh-
A3-GFP (CCD-
GFP) has been described (6). Pcdh-
A3-GFP with the three
membrane-proximal cadherin repeats deleted was generated
by PCR amplification of the distal three cadherin repeats and
ligating to the cytoplasmic, transmembrane, and extracellular
membrane-proximal segment just after the end of the sixth
cadherin repeat (corresponding to amino acid 667) using an
introduced Sal1 site to fuse the segments.
Cell Cultures, Transfection, and Immunostaining—Human
embryonic kidney (HEK293) cells were grown in Dulbecco’s
modified Eagle’s medium with 10% fetal bovine serum. Cells
were plated on 35-mm live imaging dishes with gridded glass
bottoms (Mattek) for analysis by correlative light and electron
microscopy (CLEM). Cells were plated on 25-mm coverslips in
6-well plates for light microscopy alone. Cells were transfected
with 4
g of each plasmid using Lipofectamine 2000 (Invitro-
gen) according to the manufacturer’s instructions. For lysoso-
mal inhibition, after transfection cells were treated overnight
with 20 mMNH
4
Cl. Primary neurons were grown and trans-
fected as described (6). Cells were fixed and immunostained
exactly as described (6) except that the concentration of Triton
X-100 was 0.2%.
Antibodies—Antibodies to the constant cytoplasmic domain
of mouse Pcdh-
s and to the cytoplasmic domain of N-cad have
been described (8, 19, 20). Antibodies to the constant cytoplas-
mic domain of mouse Pcdh-
s were generated using GST fused
to the entire Pcdh-
constant cytoplasmic domain. Polyclonal
anti-GFP was from Clontech. Chicken antibodies to MAP2
were from Covance. Monoclonal anti-KDEL was from Stress-
gen. Monoclonal anti-LAMP-2 (H4B4, developed by J. T.
August and J. E. K. Hildreth) was from the Developmental Stud-
ies Hybridoma Bank developed under the auspices of the
NICHD and maintained by the University of Iowa, Department
of Biology.
Microscopy—Detailed methods for CLEM after confocal
microscopy have been described (21–23). For Pcdhs or N-cad
transfected alone, cells were fixed 24 h after transfection with
4% glutaraldehyde in 0.1 Msodium cacodylate buffer with 1 mM
CaCl
2
. For cotransfection with hcRedLC3wt or G constructs,
cells were imaged live prior to fixation with glutaraldehyde
because we found hcRed fluorescence to be incompatible with
glutaraldehyde fixation. Confocal stacks of transfected cells
were acquired on a Zeiss LSM 510 META microscope. Differ-
ential interference contrast and fluorescent images were
acquired. The location of the cell was documented with respect
to the coverslip grid using brightfield illumination. The cells
were then fixed if imaged live and processed for transmission
electron microscopy. The material was washed in sodium caco-
dylate buffer and treated with 1% osmium tetroxide, 1.5%
potassium ferracyanide in 0.1 Mcacodylate buffer for1hat4°C.
The cells were then dehydrated in solutions of ethanol at
increasing concentrations of 50%, 60%, and 70%, kept in 2 ml of
3% uranyl acetate in 70% ethanol for 12 h at 4 °C, washed in 70%
ethanol, and further dehydrated with increasing concentra-
tions of 80%, 90%, and 100% ethanol. After dehydration, cells
were infiltrated with a 1:1 solution of resin (Embed 812 kit;
Electron Microscopy Sciences) and 100% ethanol for 24 h at
room temperature. After infiltration, the resin/ethanol was
replaced with a 1-ml layer of pure resin, and an open ended
embedding capsule was placed on the dish surrounding the cell
of interest. The resin was then hardened in a vacuum oven at
65 °C for 8 –12 h. After the first layer was solidified, the capsule
was topped off with more resin and put back in the oven for
another 8 –12 h. To separate the block from the dish, a hot plate
was heated to 60 °C, and the dish was placed on a preheated hot
plate for exactly 3 min. The dish was removed from the hot
plate and the capsule carefully peeled free from the dish.
The imaged cell was relocated in the block face and sectioned
through. Sections were contrasted with lead citrate and uranyl
acetate, and serial sections of the cell of interest were docu-
mented at magnifications of 10,000, 15,000, and 30,000.
Confocal, differential interference contrast, and transmission
electron microscopy images were realigned and oriented using
nuclear and other morphological landmarks. Immunoelectron
microscopy was performed as described (8).
FIGURE 1. Pcdh-
s generate electron-dense membrane profiles when
expressed in cells. A, three cells are labeled in the images. Cells 1 and 2 have
been transfected with Pcdh-
B2-YFP, whereas cell 3 remains untransfected.
Pcdh-
B2-YFP accumulates in a line at the junction between cells 1 and 2
(arrowheads) but does not accumulate at the junction between cells 1 and 3
(arrows), consistent with its participation in a transcellular interaction
between cells 1 and 2. Band C, interfaces between cells 1 and 2 and cells 1 and
3 were examined by CLEM. The membranes at the junction between cells 1
and 2 were significantly more electron-dense (B) relative to the membranes at
the junction between cells 1 and 3 (C).
Pcdh-
Intracellular Tubules
JULY 2, 2010VOLUME 285• NUMBER 27 JOURNAL OF BIOLOGICAL CHEMISTRY 20983
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
Pcdh-
Intracellular Tubules
20984 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285NUMBER 27• JULY 2, 2010
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
Quantification—Induction of LC3 clustering by Pcdh-
s,
-
1, or N-cad was quantified as follows. Confocal sections of
random cells doubly transfected with RFP-LC3 and Pcdh or
N-cad GFP/YFP/Venus constructs were acquired on a Zeiss
LSM 410 microscope. It was then determined whether RFP-
LC3 in each cell was punctate or diffuse. At least 75 cells were
scored for each condition over three independent experiments,
and the number of punctate LC3 cells was averaged. All condi-
tions were counted blindly. ImageJ was used on imported trans-
mission electron microscopy images to determine tubule
length and diameter and averaged from 20–30 tubules from
two cells/condition. A ttest was used to determine significance.
For immunoelectron microscopic quantification of anti-Pcdh-
-labeled organelles, the overall number of gold particles/or-
ganelle was determined, and for each organelle, the orientation
of particles was designated as “paired,” representing an adhe-
sive-like conformation, or “unpaired.” The ratio of unpaired to
paired labeled organelles was determined from 51 labeled
organelles.
RESULTS
Pcdh-
s Generate Electron-dense Membrane Appositions
Pcdh-
-GFP fusions are functional in vivo (7, 12–14, 24). When
expressed in cultured hippocampal
neurons and cell lines, Pcdh-
-
GFPs exhibited an intracellular dis-
tribution that shifted to the cell sur-
face at cell-cell contacts upon deletion
of the cytoplasmic region (6), indicat-
ing specific retention signals within
the cytoplasmic domain. To deter-
mine the identity of the compart-
ments that harbor full-length intra-
cellular Pcdh-
s, we performed
CLEM (21, 23) of transiently trans-
fected cells. This allowed the precise
relocalization of GFP-tagged Pcdh-
s under the electron microscope.
Although largely intracellular, in
some cases
B2-YFP (Fig. 1A,
arrowheads) was found at the inter-
face between two transfected cells
(Fig. 1A, cells 1 and 2). Pcdh-
A3-
GFP was previously observed to tar-
get to cell-cell interfaces (5, 6).
B2-
YFP never accumulated at the
interface between a transfected cell
and a nontransfected cell (Fig. 1A,
cells 1 and 3). Both Pcdh-
-positive
and -negative cell-cell interfaces
were relocated under the electron microscope by CLEM. The
B2-YFP positive interface between cell 1 and cell 2 (Fig. 1B)
was more electron-dense with rigid alignment of plasma mem-
branes compared with the
B2-YFP-negative interface between
cell 1 and the nonexpressing cell 3 (Fig. 1C).
Pcdh-
A3 and -
B2 Induce Intracellular Tubules—We took
advantage of the property that Pcdh-
s render membranes
electron-dense to identify the intracellular organelles that har-
bor the A, B, and C subclasses of Pcdh-
s and compare these
with a Pcdh-
family member (Pcdh-
1) and the classical cad-
herin, N-cad. Three representatives of the Pcdh-
cluster,
A3-
GFP,
B2-YFP, and
C4-YFP, were compared with each other
and with a member of the
cluster, Pcdh-
1-GFP (
1-GFP), as
well as the classical N-cad-Venus. Intracellular juxtanuclear
accumulations of
A3-GFP were present in most transfected
cells (Fig. 2A,box). The area corresponding to
A3-GFP fluo-
rescence (Fig. 2A,boxes) was analyzed by CLEM in individual
cells (Fig. 2, A,right panel, and B). In all cells, precisely at
the area of
A3-GFP fluorescence, electron-dense membrane
tubules were observed that were completely absent in untrans-
fected cells. High magnification of the
A3-GFP induced
tubules (Fig. 2B) revealed a diameter of 60–70 nm. These
FIGURE 2. Pcdh-
A3 and -
B2 but not -
C4 generate intracellular tubules. Aand B,
A3-GFP accumulates mostly intracellularly in HEK293 cells (6). A, cell
expressing
A3-GFP was identified by confocal microscopy (left and center) and processed for CLEM. Boxed region in confocal micrographs was visualized by
electron microscopy (right). This region contained part of the nucleus, several mitochondria, and other tubulovesicular organelles resembling ER, which were
also found in nontransfected cells. Darker, electron-dense tubule-shaped organelles, that were never found in untransfected cells, were found in the region of
A3-GFP expression. B, high magnification images are shown of electron-dense organelles generated by
A3-GFP. The tubules measured 60 –70 nm across
and were in some cases observed to emanate from other round organelles of 100 –250 nm in diameter, which contained smaller structures inside (arrow-
head). C,
B2-YFP also generated tubules at sites of intracellular accumulation and was also sometimes associated with other organelles (arrowhead). D,in
contrast,
C4-YFP did not generate tubules at sites of intracellular accumulation but rather produced membrane sheets adjacent to the nucleus. N, nucleus; M,
mitochondria.
FIGURE 3. Pcdh-
1-GFP and N-cad-Venus do not produce intracellular tubules. A,
1-GFP, a representative
of the
cluster, exhibited almost exclusive intracellular accumulation (left). Low magnification electron
micrograph of boxed region (middle). When visualized by electron microscopy,
1-GFP produced elon-
gated rough ER-like organelles that were found associated with ribosomes (right,arrowheads). B, although
found more frequently at cell-cell interfaces, N-cad-Venus could be found intracellularly in some cells
(box,left). Low magnification electron micrograph of boxed region (middle). Examination of intracellular
N-cad-Venus regions revealed membrane whorls and numerous smaller vesicles that lacked ribosomes
(right). N, nucleus; M, mitochondria.
Pcdh-
Intracellular Tubules
JULY 2, 2010VOLUME 285• NUMBER 27 JOURNAL OF BIOLOGICAL CHEMISTRY 20985
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
tubules were distinct from rough and smooth ER-like profiles
that were oftentimes found in the vicinity of the tubules but
were also observed in untransfected cells. In some cases the
tubules were observed to emanate from 100–250 nm vesicular
organelles that also contained smaller dot-like profiles (Fig. 2B,
right panel,arrowheads).
Intracellular accumulations were also observed for
B2-YFP
and
C4-YFP (Fig. 2, Cand D, respectively) and were examined
by CLEM. Tubules similar to those found in
A3-GFP-trans-
fected cells were found in
B2-YFP-transfected cells (Fig. 2C).
In contrast,
C4-YFP did not induce
the formation of tubules (Fig. 2D)
but instead accumulated in layered
membrane sheets similar to the type
of accumulations observed for
some ER proteins (25, 26). Unlike
A3-GFP-induced tubules, the
C4-YFP-induced sheets were con-
tinuous through several serial sec-
tions, demonstrating that
C4-YFP
induces accumulation of sheets
rather than tubules (supple-
mental Fig. 1). In addition,
C4-YFP
sheets were found to be continuous
with the nuclear envelope, suggest-
ing an ER origin (supplemental
Fig. 1).
By comparison,
1-GFP was al-
most exclusively intracellular and
absent at cell-cell contacts (Fig. 3A)
in agreement with its apparent
lack of adhesive activity (9, 10). By
CLEM,
1-GFP showed ER-like
accumulations that, at higher mag-
nification, appeared to be associated
with ribosomes (Fig. 3A,right panel),
suggesting an expansion of rough
ER. N-cad-Venus was more promi-
nent at cell junctions (not shown)
but also could be found intracellu-
larly (Fig. 3B). CLEM of N-cad-
Venus intracellular accumulations
showed a different ER-like profile
with wrapped sheets and vacuoles
(Fig. 3B,right panel) that, unlike
1-GFP, did not associate with ribo-
somes. These whorled membranes
have also been shown to be associ-
ated with accumulations due to ER
expansion (25, 26). Thus,
A3-GFP
and
B2-YFP produce a specific
type of intracellular tubule versus
the ER and sheet-like accumula-
tions observed for the other
constructs.
A3-GFP and
B2-YFP Cocluster
with LC3—To determine the origin
of the tubules within the secretory
pathway, we searched for organelle markers that might colocal-
ize with
A3-GFP- and
B2-YFP-induced tubules (see
supplemental Fig. 2). The autophagy protein LC3, when fused
to either GFP or RFP, has been shown to be a reliable marker for
autophagosomes in cultured cells and in vivo (18, 27). We found
that both
A3-GFP and
B2-YFP induced the clustering of, and
colocalized with, RFP-LC3 (Fig. 4A). In contrast, neither
C4-
YFP,
1-GFP, nor N-cad-Venus affected the clustering of RFP-
LC3, which remained diffuse in transfected cells (Fig. 4A).
Quantification showed that 70% of cells transfected with
FIGURE 4. Pcdh-
A3 and -
B2 induce clustering of, and colocalize with, LC3. A, the indicated constructs
(top panels) were transfected together with RFP-LC3 (middle panels). All Pcdh-
constructs generated clear
intracellular accumulations (arrows and arrowheads). Only
A3-GFP and
B2-YFP induced clustering of, and
colocalized with, RFP-LC3 (arrowheads).
C4-YFP did not induce clustering of RFP-LC3 despite its intracellular
location (arrows).
1-GFP and N-cad-Venus also did not affect the distribution of RFP-LC3. B, the percentage of
cells with punctate RFP-LC3 was determined from three independent experiments. Error bars, S.E. C, colocal-
ization of Pcdh-
A3-GFP and RFP-LC3 in neurons transfected with the two constructs at 12 days in vitro is
shown. Blue channel is MAP2 immunostaining. Arrowheads indicate colocalized profiles. Inset shows codistri-
bution of the two constructs in an organelle-like profile.
Pcdh-
Intracellular Tubules
20986 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285NUMBER 27• JULY 2, 2010
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
A3-GFP or
B2-YFP and RFP-LC3 exhibited clustered LC3
(Fig. 4B). All constructs evaluated were expressed at compara-
ble levels in cells (see supplemental Fig. 3). Furthermore, no
accumulation of autophagic vesicles or autolysosomes, which
have been visualized using the same electron microscopic prep-
aration method (22, 23), could account for the high levels of
clustered LC3 in
A3-GFP- or
B2-YFP-transfected cells (see
Fig. 2).
A3-GFP, when cotransfected with RFP-LC3 into cul-
tured hippocampal neurons at 12 days in vitro, also produced
intracellular clusters, some of which were found to cocluster
with RFP-LC3 (Fig. 4C,arrowheads and inset).
It is known that LC3 clustering artifacts due to protein aggre-
gation can lead to misidentification of LC3 puncta (28, 29). LC3
is lipidated at the glycine residue near its carboxyl terminus via
a reaction similar to the ubiquitin ligase pathway, and this lipi-
dation is required for LC3 targeting in autophagy (29). Nonspe-
cific clustering of LC3 at protein aggregates lacks the require-
ment for lipidation of LC3 (29). We sought to determine
whether LC3 lipidation is required for clustering with
A3-
GFP. Wild-type hcRed-LC3 or lipidation-defective hcRed-
LC3-G (29) was transfected with
A3-GFP, and the extent of
LC3 clustering was evaluated quantitatively (Fig. 5). Wild-type
hcRed-LC3 was found to cocluster
with
A3-GFP (Fig. 5A,left panels)
similar to RFP-LC3 (see Fig. 4). In
contrast, hcRed-LC3-G did not
cocluster with
A3-GFP (Fig. 5A,
right panels). Quantitative analysis
demonstrated that
A3-GFP in-
duced the formation of wild-type
hcRed-LC3 clusters but not that of
hcRed-LC3-G. Thus, lipidation of
LC3 is required for the colocaliza-
tion of LC3 with Pcdh-
-induced
intracellular tubules.
LC3 Mediates Pcdh-
Intracellu-
lar Tubule Elongation from Ly-
sosomes—We suspected that wild-
type and lipidation-defective LC3
might differentially affect tubule
formation or elongation when coex-
pressed with
A3-GFP. To test this,
we compared the tubules produced
by
A3-GFP alone or in the pres-
ence of wild-type hcRed-LC3 or
hcRed-LC3-G. In contrast to the
relatively long electron-dense tu-
bules formed by
A3-GFP in the
presence of wild-type hcRed-LC3
(Fig. 6A), the electron-dense tubules
were significantly shorter in the
presence of hcRed-LC3-G (Fig.
6B). Instead of tubules, most of the
electron-dense organelles in hcRed-
LC3-G/
A3-GFP-cotransfected
cells consisted of vesicles similar to
the type that were often connected
to the tubules in RFP-LC3 and
A3-
FIGURE 5. Lipidation-defective LC3 does not colocalize with
A3-GFP.
A,
A3-GFP was cotransfected with wild-type hcRed tagged LC3 (hcRed-
LC3wt) or similarly tagged LC3 truncated at the glycine residue near the car-
boxyl terminus (hcRed-LC3G), the site for lipid conjugation. hcRed-LC3wt
was clustered and colocalized with
A3-GFP, whereas hcRed-LC3G was not.
B, results were quantified as the percentage of cells with punctate LC3 from
three independent experiments. Error bars, S.E.
FIGURE 6. hcRed-LC3G shortens
A3-GFP-induced tubule length. A,
A3-GFP was cotransfected with
hcRed-LC3wt and processed for CLEM. B, cotransfection of hcRed-LC3G with
A3-GFP and CLEM. Intracellular
accumulation of
A3-GFP (green channel) did not recruit hcRed-LC3G(red channel), and CLEM of this site
revealed fewer tubules that were shorter in length than those observed when hcRred-LC3wt was cotrans-
fected. C, quantification of tubule length for
A3-GFP cotransfected with hcRed-LC3wt or Gversus
A3-GFP
alone. Results were averaged from 20 –30 tubules from two different cells/condition. A ttest was used to
determine significance. **, p0.001; *, p0.05. Error bars, S.D. N, nucleus.
Pcdh-
Intracellular Tubules
JULY 2, 2010VOLUME 285• NUMBER 27 JOURNAL OF BIOLOGICAL CHEMISTRY 20987
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
GFP-cotransfected cells (see Fig. 2B,right panel). Quantifica-
tion revealed an almost 50% reduction in tubule length in cells
transfected with
A3-GFP plus hcRed-LC3-G relative to
gA3-GFP plus wild-type hcRed-LC3. Cells transfected with
A3-GFP alone had a tubule length intermediate to the two
(Fig. 6C). Thus, LC3 promotes
A3-GFP tubule elongation in a
lipidation-dependent fashion.
In cells cotransfected with hcRed-LC3-G and
A3-GFP,
the predominant electron-dense organelles were 100–250-nm
vesicles (Fig. 7A,right panel) rather than tubules (Fig. 7A,left
panel), suggesting that the initial substrates for tubule forma-
tion may be a subclass of vesicular organelles that sprout
tubules when Pcdh-
s accumulate in them. We sought to deter-
mine the types of organelles that harbor Pcdh-
s by immuno-
staining of
A3-GFP-transfected cells with subcellular markers
including those for ER, Golgi, early endosomes, and late endo-
somes/lysosomes (supplemental Fig. 2). We observed a signifi-
cant colocalization of Pcdh-
s with anti-LAMP-2 immuno-
staining (Fig. 7Band supplemental Fig. 2). Both
A3-GFP and
B2-YFP, constructs that induced tubules, exhibited significant
colocalization with LAMP-2 (supplemental Fig. 4). Thus, in
these cells, the combined data sug-
gest that Pcdh-
A and B, but not C,
families, together with LC3, induce
the sprouting of tubules as they
accumulate in late endosomes/lyso-
somes in a manner dependent on
LC3 lipidation.
To verify the lysosomal origin
of
A3-GFP-induced tubules, we
treated
A3-GFP-transfected cells
with NH
4
Cl, an inhibitor of lysoso-
mal acidification. In this case, large
vacuoles (Fig. 7C) were induced
with a reduction in tubules, indi-
cating that tubulogenesis is tied to
lysosomal function. A few tubules
(Fig. 7C,arrowhead) were observed,
however, to emanate from the
enlarged vacuoles (Fig. 7C,arrow).
Differential Effect of Luminal/
Extracellular and Cytoplasmic Do-
main Deletion on Lysosomal Tar-
geting and Intracellular Tubule
Biogenesis—The luminal/extracellu-
lar domains of Pcdh-
A3 and Pcdh-
B2, but not Pcdh-
C4, were shown
to participate in cell-cell interactions
with homophilic properties, whereas
the cytoplasmic domains promoted
retention (6). We sought to deter-
mine whether the luminal/extracel-
lular and/or cytoplasmic domains
mediate tubule formation. Cyto-
plasmic-deleted
A3-GFP (CCD-
GFP) was mostly found at the cell
surface (6), but intracellular accu-
mulations could be found in a few
transfected cells (Fig. 8A,arrows). These never induced cluster-
ing of RFP-LC3, nor did they colocalize with LAMP-2 (Fig. 8A,
arrows). In contrast, when the luminal/extracellular domain
was deleted (ECD-GFP; Fig. 8A,arrowheads), both RFP-LC3
clustering and LAMP-2 colocalization were observed. CLEM
showed that ECD-GFP accumulated in distorted electron-
dense organelles that contained smaller profiles similar to those
found in lysosomes (Fig. 8B,bottom,arrows). In this case, no
tubules were observed. In contrast, CCD-GFP generated
membrane whorls (Fig. 8B,top) similar to the type (25, 26)
produced upon expression of some ER-targeted proteins.
CCD-GFP-induced intracellular membrane whorls were pos-
itively immunolabeled with anti-KDEL antibodies (Fig. 8C,
arrowheads), confirming their ER origin.
Shortening the Luminal/Extracellular Domain Decreases
Tubule Width—The above results suggested that the cytoplas-
mic domain promotes lysosomal targeting whereas the lumi-
nal/extracellular domain is required for tubulogenesis, poten-
tially by mediating intraluminal interactions. It is known that
cadherins mediate their adhesive interactions via the most dis-
tal extracellular moiety, the first cadherin repeat (30–32). We
FIGURE 7. Lysosomes are the substrates for tubulogenesis. A, electron-dense organelles in
A3-GFP and
hcRed-LC3G-cotransfected cells resemble lysosomes (right panel,arrow)versus organelles from
A3-GFP-
tranfected cells in which the tubules (left panel,arrow) retained electron-dense properties whereas lysosomal-
like organelles were less electron-dense. B, colocalization of
A3-GFP with lysosomal marker LAMP-2 indicates
Pcdh-
s target to lysosomes, implicating them as the substrate for tubule growth. C,NH
4
Cl inhibition of
lysosomal acidification reduces tubulogenesis. Enlarged vacuoles (arrow) only occasionally sprouted tubules
(arrowhead). N, nucleus; M, mitochondria.
Pcdh-
Intracellular Tubules
20988 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285NUMBER 27• JULY 2, 2010
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
deleted the three membrane-proximal cadherin repeats (cad-
herin repeats 46, EC46-GFP; see supplemental Fig. 3)to
determine whether the distal part of the extracellular domain
might be involved in tubulogenesis. We found that this con-
struct induced clustering of RFP-LC3 and colocalized with
LAMP-2 (Fig. 9A). CLEM of intracellular EC46-GFP re-
vealed that juxtanuclear tubules
were generated, but with an appar-
ent narrower width. We quantified
the width of tubules generated by
A3-GFP,
B2-YFP, and EC4–6-
GFP (Fig. 9B) and found that
although both
A3-GFP and
B2-
YFP generated tubules of identical
width (65 nm), EC46-GFP-
generated tubules were significantly
narrower (40 nm) (Fig. 9C). Thus,
the molecular length of the luminal/
extracellular domain is correlated
with the diameter of the intracellu-
lar tubules, suggesting an intralumi-
nal interaction between Pcdh-
molecules on apposing membranes
stabilizing tubule structure.
Pcdh-
s Delinate Subdomains of
Tubulovesciular Organelles in Vivo
By immunoelectron microscopy,
Pcdh-
s were shown to have a pro-
minent intracellular component in
vivo (6, 8). In many cases, Pcdh-
labeled constrictions of, or emana-
tions from, organelle profiles (Fig.
10A,arrowheads), in a paired arrange-
ment on opposing membranes (Fig.
10B,left panel). When quantified,
Pcdh-
-labeled organelles had this
arrangement 55% of the time versus
45% of organelles with unpaired
gold particles (Fig. 10B,right panel).
These results are consistent with a
role for Pcdh-
-mediated intralu-
minal interactions in organelle
dynamics in vivo (Fig. 10C).
DISCUSSION
The cellular function of all of the
Pcdhs has been poorly defined (33),
unlike the well characterized classi-
cal cadherins. The Pcdh-
s can
mediate transcellular interactions
with homophilic properties (5, 6)
and, for the Pcdh-
s, less is known
about their functions at the cellular
level, but these most likely do not
mediate transcellular interactions
(9, 10, 34). Knock-out phenotypes
for Pcdh-
s are consistent with
transcellular interactions as one fea-
ture but also point to other modes of action (7, 12–14, 24, 35).
Pcdh-
s exhibit a prominent intracellular distribution in cul-
tured neurons and in neural tissue (6, 8). Here, we show that
Pcdh-
A3 and -
B2 generate, from substrate organelles, intra-
cellular tubules that require the autophagic molecule LC3 for
their elongation. Based on the accumulated data, we speculate
FIGURE 8. Deletion of the luminal/extracellular and cytoplasmic domains of Pcdh-
A3 differentially
affect lysosomal localization, LC3 clustering, and tubule formation. A, intracellular accumulations of
CCD-GFP, when found, did not induce clustering of RFP-LC3 and did not colocalize with LAMP-2. In contrast,
the luminal/extracellular deletion (ECD-GFP) did induce clustering of RFP-LC3 and colocalized with LAMP-2.
B, electron micrographs of ECD-GFP and CCD-GFP intracellular accumulations are shown. ECD-GFP was
found in clustered juxtanuclear organelles that contained smaller particles in the lumen (arrows), suggestive of
distorted lysosomes whereas CCD-GFP generated larger membrane whorls that resembled ER distortion as
has been observed previously (25, 26). C, intracellular CCD-GFP induced membrane whorls labeled positive
with anti-KDEL antibodies (arrowhead). N, nucleus.
Pcdh-
Intracellular Tubules
JULY 2, 2010VOLUME 285• NUMBER 27 JOURNAL OF BIOLOGICAL CHEMISTRY 20989
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
that Pcdh-
s have the ability to generate their own trafficking
organelles at intracellular sites where they accumulate by alter-
ing membrane shape or curvature and promoting intraluminal
interactions.
Nonconventional Roles for Pcdh-
s—The predominant phe-
notype when the Pcdh-
cluster is inactivated in neurons is cell
death (7, 12, 13, 24), and this could be either cell-autonomous
or non-cell-autonomous (13, 24). Prevention of neuron death
in Pcdh-
-deleted animals by dou-
ble knock out with Bax revealed syn-
aptic defects in spinal cord (12) pre-
sumably due to disrupted Pcdh-
transcellular interactions. However,
completely normal retinal connec-
tivity was restored by rescue from
apoptosis in Pcdh-
retinal knock
outs (13), indicating that retinal
connectivity has no requirement for
Pcdh-
s despite the high expression
of the molecules there. The exis-
tence of two, apparently separable,
phenotypes, neuronal death and
synapse loss, indicates the potential
for multiple functions for Pcdh-
s
in various neuronal systems. Either
or both synapse development and
cell survival could be dependent on
intracellular trafficking of Pcdh-
s.
There is a growing list of mole-
cules that influence intracellular
membrane shape, and multiple ER-
resident transmembrane proteins
that drive the structural dynamics of
ER are now beginning to be elabo-
rated (36), including the reticulon
family which can transform the ER
from sheets into tubules (37). It is
possible that Pcdh-
s could define
a subdomain of membranes in
organelles in which they accumu-
late and modify these membranes in
a manner similar to the reticulons
(37, 38). Pcdh-
s lack a cleavable
prodomain (39), that for classical
cadherins blocks adhesion during
intracellular transport (40), and
therefore its membrane-membrane
adhesive activity is likely to be active
inside the lumen of intracellular
compartments where it could par-
ticipate in the stabilization of
organelle shape (Fig. 10C). An adhe-
sive-like mechanism for mainte-
nance of ER shape has previously
been proposed (36) and could also
participate in the stabilization of the
tubule morphology. Such Pcdh-
-
generated tubules might emanate
from any organelle that harbors them in neurons.
Nonautophagic Role for LC3—LC3 is the mammalian homo-
logue of yeast Atg8 which was identified as a gene required for
proper formation of autophagic vesicles or vacuoles in yeast.
LC3 was first identified as an 18-kDa protein that copurified
with microtubules and with MAP1A/1B (41). LC3 was found to
translocate to autophagosomes and can be present on the cyto-
solic and luminal sides (27). In mammals, LC3 has three other
FIGURE 9. Shortening of the extracellular domain decreases tubule width. Cadherin repeats 4 –6 were
removed, leaving only cadherin repeats 1–3 in the extracellular domain (EC4 – 6-GFP; see supple-
mental Fig. 3). A,EC4 –6-GFP induces clustering of RFP-LC3 and colocalizes with LAMP-2. B, low (left) and high
(center) magnification electron micrographs of intracellular tubules induced by EC4 – 6-GFP are shown. Right,
widths of tubules generated by
A3-GFP,
B2-GFP and EC4 –6-GFP constructs were quantified. Decrease in
width of EC4 –6-GFP tubules was statistically significant (ttest, p0.0001). Widths of tubules induced by
A3-GFP and
B2-GFP were not different (p0.2). Error bars, S.D. C, high magnification of images illustrates
widths of
A3-GFP (left),
B2-GFP (center), and EC4 –6-GFP induced tubules (right). N, nucleus.
Pcdh-
Intracellular Tubules
20990 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285NUMBER 27• JULY 2, 2010
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
homologues, GABARAP, GEC-1, and GATE-16, which also
translocate to autophagosomes upon lipidation (18). The exact
role of LC3/Atg8 and related proteins in autophagy is still not
clear. One study showed that the size of autophagic vacuoles
depends on Atg8 levels in yeast (15). Knock outs of different
genes of the autophagic cascade, which eliminate LC3 family
lipidation, were shown to alter the shape or size of autophagic
vesicles, or prevent their closure, but not eliminate them (17).
We found that Pcdh-
-induced intracellular tubules are
shorter in the presence of the lipidation-defective LC3. Our
results support the idea, previously suggested, that LC3 con-
trols the rate of intracellular membrane expansion (15), which
may indicate a generalized role for LC3 in nonautophagic pro-
cesses, some of which have recently begun to be elaborated (42)
and are consistent with a suggested role for LC3 family mem-
bers in membrane and receptor trafficking (43–48). It is possi-
ble that intracellular Pcdh-
s may
be involved in regulating LC3 fami-
ly-mediated organelle trafficking in
addition to their roles in cell-cell
interactions.
Whether or not the Pcdh-
-in-
duced tubules represent a precur-
sor to autophagic vesicles or
another type of organelle that uti-
lizes LC3 for its biogenesis remains
to be conclusively determined, but
autophagy is unlikely given that
autophagic vesicles were never
found in the vicinity of intracellular
Pcdh-
s and that the tubules origi-
nate from lysosomes. The tubules
observed in the present study do
bear a resemblance, tubules con-
nected to vesicular profiles, to those
found using biochemical purifica-
tion of LC3-containing membranes
(49). Because no increases in auto-
phagic vesicles were observed in
Pcdh-
-transfected cells it is likely
that the tubules observed are one of
the few examples of nonautophagic
organelles that utilize LC3 for
its biogenesis. Alternatively, the
tubules could be an arrested stage
of autophagic organelle biogenesis
prior to the generation of the phago-
phore. Regardless of whether the
tubules are autophagy-related or
-unrelated, the observation that cer-
tain Pcdh-
s can induce organelle
formation in an LC3-dependent
fashion points to a potential novel
intracellular role for the molecules
in trafficking in addition to cell-cell
interactions.
Mechanism for Trafficking of Rec-
ognition Units—The diversity of
Pcdh-
s in the nervous system generated by alternative splicing
(3) strongly suggests a role in cell surface recognition. It is
possible that through intracellular homophilic interactions,
Pcdh-
trafficking might be directed into “packets” (Fig. 10C)
that when inserted into the plasma membrane contain a homo-
geneous population of a single Pcdh-
isoform. The addition of
such recognition packets to the surface may have more imme-
diate consequences on synaptic development versus slow accu-
mulation of individual Pcdh-
molecules at cell-cell interac-
tions sites in neurons via conventional secretory mechanisms.
Acknowledgments—We thank Drs. Qiang Wu, Isei Tanida, Joshua
Weiner, Hidekazu Tanaka, Marcus Frank, Ingrid Haas, and Zhenyu
Yue for reagents; Drs. Ana Maria Cuervo and Zhenyu Yue for helpful
discussions; and William G. Janssen for technical advice.
FIGURE 10. Arrangement of Pcdh-
s within organelle profiles. A, immunoelectron microscopy of adult
hippocampal CA1 region with antibodies to Pcdh-
constant intracellular domain. Pcdh-
s have a prominent
intracellular distribution (8) and label tubulovesicular organelles in axons and dendrites. Pairing of gold parti-
cles (arrowheads) at apparent constriction sites suggests the possibility of an adhesive conformation within
organelles. Such paired labeling of gold particles has also been observed at sites of transcellular interactions
(8). Pcdh-
s were also observed in lysosomal-like organelles (bottom left). B, example of organelle labeled in
paired (left,arrowheads) or unpaired (right) configuration. C, model for intraluminal interactions mediated by
Pcdh-
s. Interactions could be stabilized by LC3 (squares). Homophilic intraluminal interactions may facilitate
the budding of new organelles containing high concentrations of a single Pcdh-
isoform.
Pcdh-
Intracellular Tubules
JULY 2, 2010VOLUME 285• NUMBER 27 JOURNAL OF BIOLOGICAL CHEMISTRY 20991
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
REFERENCES
1. Kohmura, N., Senzaki, K., Hamada, S., Kai, N., Yasuda, R., Watanabe, M.,
Ishii, H., Yasuda, M., Mishina, M., and Yagi, T. (1998) Neuron 20,
1137–1151
2. Wu, Q., and Maniatis, T. (1999) Cell 97, 779 –790
3. Tasic, B., Nabholz, C. E., Baldwin, K. K., Kim, Y., Rueckert, E. H., Ribich,
S. A., Cramer, P., Wu, Q., Axel, R., and Maniatis, T. (2002) Mol. Cell 10,
21–33
4. Obata, S., Sago, H., Mori, N., Rochelle, J. M., Seldin, M. F., Davidson, M., St
John, T., Taketani, S., and Suzuki, S. T. (1995) J. Cell Sci. 108, 3765–3773
5. Frank, M., Ebert, M., Shan, W., Phillips, G. R., Arndt, K., Colman, D. R.,
and Kemler, R. (2005) Mol. Cell. Neurosci. 29, 603–616
6. Ferna´ndez-Monreal, M., Kang, S., and Phillips, G. R. (2009) Mol. Cell.
Neurosci. 40, 344–353
7. Wang, X., Weiner, J. A., Levi, S., Craig, A. M., Bradley, A., and Sanes, J. R.
(2002) Neuron 36, 843–854
8. Phillips, G. R., Tanaka, H., Frank, M., Elste, A., Fidler, L., Benson, D. L., and
Colman, D. R. (2003) J. Neurosci. 3, 5096–5104
9. Mutoh, T., Hamada, S., Senzaki, K., Murata, Y., and Yagi, T. (2004) Exp.
Cell Res. 94, 494–508
10. Morishita, H., Umitsu, M., Murata, Y., Shibata, N., Udaka, K., Higuchi, Y.,
Akutsu, H., Yamaguchi, T., Yagi, T., and Ikegami, T. (2006) J.Biol.Chem.
281, 33650–33663
11. Triana-Baltzer, G. B., and Blank, M. (2006) J. Neurobiol. 66, 393– 407
12. Weiner, J. A., Wang, X., Tapia, J. C., and Sanes, J. R. (2005) Proc. Natl.
Acad. Sci. U.S.A. 102, 8–14
13. Lefebvre, J. L., Zhang, Y., Meister, M., Wang, X., and Sanes, J. R. (2008)
Development 135, 4141–4151
14. Garrett, A. M., and Weiner, J. A. (2009) J. Neurosci. 29, 11723–11731
15. Xie, Z., Nair, U., and Klionsky, D. J. (2008) Mol. Biol. Cell 19, 3290 –3298
16. Xie, Z., Nair, U., Geng, J., Szefler, M. B., Rothman, E. D., and Klionsky, D. J.
(2009) Autophagy, 217–220
17. Fujita, N., Hayashi-Nishino, M., Fukumoto, H., Omori, H., Yamamoto, A.,
Noda, T., and Yoshimori, T. (2008) Mol.Biol. Cell 19, 4651–4659
18. Tanida, I., Ueno, T., and Kominami, E. (2004) Int. J. Biochem. Cell Biol. 36,
2503–2518
19. Tanaka, H., Shan, W., Phillips, G. R., Arndt, K., Bozdagi, O., Shapiro, L.,
Huntley, G. W., Benson, D. L., and Colman, D. R. (2000) Neuron 25,
93–107
20. Phillips, G. R., Huang, J. K., Wang, Y., Tanaka, H., Shapiro, L., Zhang, W.,
Shan, W. S., Arndt, K., Frank, M., Gordon, R. E., Gawinowicz, M. A., Zhao,
Y., and Colman, D. R. (2001) Neuron 32, 63–77
21. Polishchuk, R. S., Polishchuk, E. V., Marra, P., Alberti, S., Buccione, R.,
Luini, A., and Mironov, A. A. (2000) J. Cell Biol. 148, 45–58
22. Razi, M., Chan, E. Y., and Tooze, S. A. (2009) J. Cell Biol. 185, 305–321
23. Razi, M., and Tooze, S. A. (2009) Methods Enzymol. 452, 261–275
24. Prasad, T., Wang, X., Gray, P. A., and Weiner, J. A. (2008) Development
135, 4153–4164
25. Snapp, E. L., Hegde, R. S., Francolini, M., Lombardo, F., Colombo, S.,
Pedrazzini, E., Borgese, N., and Lippincott-Schwartz, J. (2003) J.Cell Biol.
63, 257–269
26. Lingwood, D., Schuck, S., Ferguson, C., Gerl, M. J., and Simons, K. (2009)
J. Biol. Chem. 284, 12041–12048
27. Kabeya, Y., Mizushima, N., Ueno, T., Yamamoto, A., Kirisako, T., Noda,
T., Kominami, E., Ohsumi, Y., and Yoshimori, T. (2000) EMBO J. 19,
5720–5728
28. Kuma, A., Matsui, M., and Mizushima, N. (2007) Autophagy 3, 323–328
29. Tanida, I., Yamaji, T., Ueno, T., Ishiura, S., Kominami, E., and Hanada, K.
(2008) Autophagy 4, 131–134
30. Shapiro, L., Fannon, A. M., Kwong, P. D., Thompson, A., Lehmann, M. S.,
Gru¨bel, G., Legrand, J. F., Als-Nielsen, J., Colman, D. R., and Hendrickson,
W. A. (1995) Nature 374, 327–337
31. Pertz, O., Bozic, D., Koch, A. W., Fauser, C., Brancaccio, A., and Engel, J.
(1999) EMBO J. 18, 1738–1747
32. Patel, S. D., Ciatto, C., Chen, C. P., Bahna, F., Rajebhosale, M., Arkus, N.,
Schieren, I., Jessell, T. M., Honig, B., Price, S. R., and Shapiro, L. (2006) Cell
124, 1255–1268
33. Jontes, J. D., and Phillips, G. R. (2006) Trends Neurosci. 29, 186 –191
34. Emond, M. R., and Jontes, J. D. (2008) Dev. Biol. 321, 175–187
35. Su, H., Marcheva, B., Meng, S., Liang, F. A., Kohsaka, A., Kobayashi, Y., Xu,
A. W., Bass, J., and Wang, X. (2010) Dev. Biol. 339, 38–50
36. Shibata, Y., Voeltz, G. K., and Rapoport, T. A. (2006) Cell 126, 435– 439
37. Voeltz, G. K., Prinz, W. A., Shibata, Y., Rist, J. M., and Rapoport, T. A.
(2006) Cell 124, 573–586
38. Hu, J., Shibata, Y., Voss, C., Shemesh, T., Li, Z., Coughlin, M., Kozlov,
M. M., Rapoport, T. A., and Prinz, W. A. (2008) Science 319, 1247–1250
39. Koch, A. W., Farooq, A., Shan, W., Zeng, L., Colman, D. R., and Zhou,
M. M. (2004) Structure 12, 793–805
40. Ozawa, M., and Kemler, R. (1990) J. Cell Biol. 111, 1645–1650
41. Kuznetsov, S. A., and Gelfand, V. I. (1987) FEBS Lett. 212, 145–148
42. Baisamy, L., Cavin, S., Jurisch, N., and Diviani, D. (2009) J. Biol. Chem. 284,
28232–28242
43. Elazar, Z., Scherz-Shouval, R., and Shorer, H. (2003) Biochim. Biophys.
Acta 1641, 145–156
44. Scherz-Shouval, R., Sagiv, Y., Shorer, H., and Elazar, Z. (2003) J. Biol.
Chem. 278, 14053–14058
45. Chen, Z. W., Chang, C. S., Leil, T. A., and Olsen, R. W. (2007) J. Neurosci.
27, 6655–6663
46. Leil, T. A., Chen, Z. W., Chang, C. S., and Olsen, R. W. (2004) J. Neurosci.
24, 11429–11438
47. Wang, Y., Dun, S. L., Huang, P., Chen, C., Chen, Y., Unterwald, E. M., Dun,
N. J., Van Bockstaele, E. J., and Liu-Chen, L. Y. (2006) Neuroscience 140,
1265–1276
48. Marsden, K. C., Beattie, J. B., Friedenthal, J., and Carroll, R. C. (2007)
J. Neurosci. 27, 14326–14337
49. Gao, W., Kang, J. H., Liao, Y., Ding, W. X., Gambotto, A. A., Watkins, S. C.,
Liu, Y. J., Stolz, D. B., and Yin, X. M. (2010) J. Biol. Chem. 285, 1371–1383
Pcdh-
Intracellular Tubules
20992 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285NUMBER 27• JULY 2, 2010
at Adelphi Univ on July 18, 2014http://www.jbc.org/Downloaded from
... Previous results showed that Pcdh-γA3 traffics to intracellular compartments that resemble the late endosome at the electron microscopy level in HEK 293 cells (Fernández-Monreal et al., 2009;Hanson et al., 2010;O'Leary et al., 2011). To confirm the trafficking pathway for Pcdh-γA3, cells were cotransfected with a plasmid coding for the full length molecule or the cytoplasmic deletion, fused to RFP (γA3 FL and γA3 Δ190, respectively), together with plasmids coding for markers of early, late, recycling endosomes (Rab5, Rab7, or Rab11, respectively), fused to GFP ( Figures 1A, B). ...
... Previous studies have shown that intact Pcdh-γs are inefficiently expressed on the surfaces of many transfected cell types as well as endogenous Pcdh-γs in vivo, when compared to classical cadherins (Phillips et al., 2003;Fernández-Monreal et al., 2009;Hanson et al., 2010;O'Leary et al., 2011;Shonubi et al., 2015). Rather it has been found that these proteins are trafficked in the endosomal system, at least in cell lines, but also most likely in neurons both in culture and in vivo. ...
... Full length mouse Pcdh-γA3-GFP, -RFP, cytoplasmic deletions and γA3 stub-GFP constructs have been described previously Hanson et al., 2010;O'Leary et al., 2011;Shonubi et al., 2015). γA3 stub-RFP and new γA3 stub deletions from the C-terminus in the γA3 stub backbone were generated by PCR and subcloning. ...
Article
Full-text available
The family of ∼60 clustered protocadherins (Pcdhs) are cell adhesion molecules encoded by a genomic locus that regulates expression of distinct combinations of isoforms in individual neurons resulting in what is thought to be a neural surface “barcode” which mediates same-cell interactions of dendrites, as well as interactions with other cells in the environment. Pcdh mediated same-cell dendrite interactions were shown to result in avoidance while interactions between different cells through Pcdhs, such as between neurons and astrocytes, appear to be stable. The cell biological mechanism of the consequences of Pcdh based adhesion is not well understood although various signaling pathways have been recently uncovered. A still unidentified cytoplasmic regulatory mechanism might contribute to a “switch” between avoidance and adhesion. We have proposed that endocytosis and intracellular trafficking could be part of such a switch. Here we use “stub” constructs consisting of the proximal cytoplasmic domain (lacking the constant carboxy-terminal domain spliced to all Pcdh-γs) of one Pcdh, Pcdh-γA3, to study trafficking. We found that the stub construct traffics primarily to Rab7 positive endosomes very similarly to the full length molecule and deletion of a substantial portion of the carboxy-terminus of the stub eliminates this trafficking. The intact stub was found to be ubiquitinated while the deletion was not and this ubiquitination was found to be at non-lysine sites. Further deletion mapping of the residues required for ubiquitination identified potential serine phosphorylation sites, conserved among Pcdh-γAs, that can reduce ubiquitination when pseudophosphorylated and increase surface expression. These results suggest Pcdh-γA ubiquitination can influence surface expression which may modulate adhesive activity during neural development.
... After release by cleavage, the ICD can translocate to the nucleus to regulate gene expression, be intracellularly trafficked within the endosomal system, or be degraded by the proteasome (Phillips et al., 2003;Haas et al., 2005;Fernández-Monreal et al., 2009;Hanson et al., 2010;Bouillot et al., 2011;O'Leary et al., 2011;Shonubi et al., 2015) ( Figure 4A). While nuclear localization was substantiated for γand α-Pcdhs (Haas et al., 2005;Bonn et al., 2007;Emond and Jontes, 2008), the target genes or nuclear interactions of Pcdh ICDs are so far poorly characterized. ...
... Moreover, the ICD of γ-Pcdhs was demonstrated to be required for intracellular trafficking and cell surface delivery of these Pcdhs, and a conserved 26-residue ICD segment, known as the variable cytoplasmic domain (VCD) motif, was proven to be crucial for endolysosomal targeting (Fernández-Monreal et al., 2009O'Leary et al., 2011;Shonubi et al., 2015). Furthermore, it was shown that colocalization of γ-PcdhA and γ-PcdhB isoforms with the endolysosomal markers autophagy protein LC3 and lysosome associated membrane protein 2 (LAMP-2) are ICDdependent Hanson et al., 2010). Initially, Pcdhs that do not engage in trans binding at the synaptic membrane were postulated to be endocytosed and stored in intracellular organelles to be eventually recycled (Phillips et al., 2003;Jontes and Phillips, 2006;Fernández-Monreal et al., 2010). ...
Article
Full-text available
Protocadherins (Pcdhs) are cell adhesion molecules that belong to the cadherin superfamily, and are subdivided into clustered (cPcdhs) and non-clustered Pcdhs (ncPcdhs) in vertebrates. In this review, we summarize their discovery, expression mechanisms, and roles in neuronal development and cancer, thereby highlighting the context-dependent nature of their actions. We furthermore provide an extensive overview of current structural knowledge, and its implications concerning extracellular interactions between cPcdhs, ncPcdhs, and classical cadherins. Next, we survey the known molecular action mechanisms of Pcdhs, emphasizing the regulatory functions of proteolytic processing and domain shedding. In addition, we outline the importance of Pcdh intracellular domains in the regulation of downstream signaling cascades, and we describe putative Pcdh interactions with intracellular molecules including components of the WAVE complex, the Wnt pathway, and apoptotic cascades. Our overview combines molecular interaction data from different contexts, such as neural development and cancer. This comprehensive approach reveals potential common Pcdh signaling hubs, and points out future directions for research. Functional studies of such key factors within the context of neural development might yield innovative insights into the molecular etiology of Pcdh-related neurodevelopmental disorders.
... While Keeler et al. (2015) found that the C-terminal motif interacts with phosphatidyl inositol phospholipids (but not lysophospholipids or sphingolipids) and that PKC phosphorylation of S119 abrogates this binding, there are no data yet on the functional role this plays. One possible role could be in regulating g-Pcdh protein trafficking, given that removal of the cytoplasmic domain has been observed to increase cell surface expression, likely due to removal of ER-retention signals (Fernandez-Monreal et al., 2009;Hanson et al., 2010;O'Leary et al., 2011;Schreiner & Weiner, 2010;Thu et al., 2014). Several other studies have shown that a portion of g-Pcdh protein is associated with membrane vesicles of the endolysosome system, specifically late endosomes Phillips et al., 2003). ...
Preprint
Full-text available
The Pcdhg gene cluster encodes 22 γ-Protocadherin (γ-Pcdh) cell adhesion molecules that critically regulate multiple aspects of neural development, including neuronal survival, dendritic and axonal arborization, and synapse formation and maturation. Each γ-Pcdh isoform has unique protein domains--a homophilically interacting extracellular domain and a juxtamembrane cytoplasmic domain--as well as a C-terminal cytoplasmic domain shared by all isoforms. The extent to which isoform-specific vs. shared domains regulate distinct γ-Pcdh functions remains incompletely understood. Our previous in vitro studies identified PKC phosphorylation of a serine residue within a shared C-terminal motif as a mechanism through which γ-Pcdh promotion of dendrite arborization via MARCKS is abrogated. Here, we used CRISPR/Cas9 genome editing to generate two new mouse lines expressing only non-phosphorylatable γ-Pcdhs, due either to a serine-to-alanine mutation (PcdhgS/A) or to a 15-amino acid C-terminal deletion resulting from insertion of an early stop codon (PcdhgCTD). Both lines are viable and fertile, and the density and maturation of dendritic spines remains unchanged in both PcdhgS/A and PcdhgCTD cortex. Dendrite arborization of cortical pyramidal neurons, however, is significantly increased in both lines, as are levels of active MARCKS. Intriguingly, despite having significantly reduced levels of γ-Pcdh proteins, the PcdhgCTD mutation yields the strongest phenotype, with even heterozygous mutants exhibiting increased arborization. The present study confirms that phosphorylation of a shared C-terminal motif is a key γ-Pcdh negative regulation point, and contributes to a converging understanding of γ-Pcdh family function in which distinct roles are played by both individual isoforms and discrete protein domains.
... In our results, FRET signals were not observed in intracellular spaces. Previous research using electron microscopies has shown that Pcdhγ proteins generate tubule structures in intracellular spaces and suggested that the structure consists of homophilically interacting Pcdhγ proteins (27,33). There are two possible reasons for these contradictory observations. ...
Article
Clustered protocadherin (Pcdh) functions as a cell recognition molecule through the homophilic interaction in the central nervous system. However, its interactions have not yet been visualized in neurons. We previously reported PcdhγB2-Förster resonance energy transfer (FRET) probes to be applicable only to cell lines. Herein, we designed γB2-FRET probes by fusing FRET donor and acceptor fluorescent proteins to a single γB2 molecule and succeeded in visualizing γB2 homophilic interaction in cultured hippocampal neurons. The γB2-FRET probe localized in the soma and neurites, and FRET signals, which were observed at contact sites between neurites, eliminated by ethylene glycol tetraacetic acid (EGTA) addition. Live imaging revealed that the FRET-negative γB2 signals rapidly moved along neurites and soma, whereas the FRET-positive signals remained in place. We observed that the γB2 proteins at synapses rarely interact homophilically. The γB2-FRET probe might allow us to elucidate the function of the homophilic interaction and the cell recognition mechanism.
... The fluorescence was linearly unmixed with autofluorescence, FRET−, and FRET+ ( Figure S3B) (See STAR Methods). Pcdhγ proteins show a dot-like localization (2,(23)(24)(25)27). We transfected the plasmid encoding the combined γB2FL-FRET probe into cultured hippocampal neurons along with mCherry or iRFP670 to visualize cell morphology ( Figure 3A). ...
Preprint
Clustered protocadherin (Pcdh) functions as a cell recognition molecule through the homophilic interaction in CNS. However, its interactions have yet not been visualized in neurons. We previously reported PcdhγB2-FRET probes to be applicable only for cell lines. Herein, we newly designed PcdhγB2-FRET probes by fusing FRET donor and acceptor fluorescent proteins to a single PcdhγB2 molecule and succeeded in visualizing PcdhγB2 homophilic interaction in cultured hippocampal neurons. The γB2-FRET probe localized in the soma and neurites, and FRET signals were observed at contact sites between neurites and eliminated by EGTA addition. Live imaging revealed that the FRET-negative γB2 signals were rapidly moving along neurites and soma, whereas the FRET-positive signals remained in place. We observed that the γB2 proteins at synapses rarely interact homophilically. The γB2-FRET probe would allow us to elucidate the function of the homophilic interaction and the cell recognition mechanism. Significance Statement We visualize the Pcdh homophilic interaction using a novel FRET-based probe, and reveal that the homophilically interacting Pcdh proteins are found at contact sites between the neurites and roots of neurites from the soma, and are stable at a location. Additionally, in neurons, Pcdh proteins are located at synapses but rarely interact homophilically.
... Both mechanisms are interconnected by several molecular nodes and a close cross-talk exists [54]. In direct proximity of the VTRNA-1 locus is the proto-cadherin cluster that encodes for the protocadherin family, which is involved in autophagy [126]. Therefore, it seems indicative that also vault RNAs might be involved in autophagy [52] (Fig. 1). ...
Article
Full-text available
Non-coding RNAs are important regulators of differentiation during embryogenesis as well as key players in the fine-tuning of transcription and furthermore, they control the post-transcriptional regulation of mRNAs under physiological conditions. Deregulated expression of non-coding RNAs is often identified as one major contribution in a number of pathological conditions. Non-coding RNAs are a heterogenous group of RNAs and they represent the majority of nuclear transcripts in eukaryotes. An evolutionary highly conserved sub-group of non-coding RNAs is represented by vault RNAs, named since firstly discovered as component of the largest known ribonucleoprotein complexes called “vault”. Although they have been initially described 30 years ago, vault RNAs are largely unknown and their molecular role is still under investigation. In this review we will summarize the known functions of vault RNAs and their involvement in cellular mechanisms.
... So far, no functional relationship between vault RNAs and the syntenically conserved genes has been uncovered. However, with the newly described link between vault RNAs and autophagy (see below), it is noteworthy that members of the protocadherin family have also been described to associate with autophagy-related proteins and to influence lysosome targeting [22]. Additional clues that could explain the syntenic conservation might be uncovered in the future. ...
Article
Full-text available
RNA-binding proteins typically change the fate of RNA, such as stability, translation or processing. Conversely, we recently uncovered that the small non-coding vault RNA 1-1 (vtRNA1-1) directly binds to the autophagic receptor p62/SQSTM1 and changes the protein’s function. We refer to this process as ‘riboregulation’. Here, we discuss this newly uncovered vault RNA function against the background of three decades of vault RNA research. We highlight the vtRNA1-1-p62 interaction as an example of riboregulation of a key cellular process.
Article
In the last decades, the advancements of microscopes technology, together with the development of new imaging approaches, are trying to address some biological questions that have been unresolved in the past: the need to combine in the same analysis temporal, functional and morphological information on the biological sample has become pressing. For this reason, the use of correlative microscopy, in which two or more imaging techniques are combined in the same analysis, is getting increasingly widespread. In fact, correlative microscopy can overcome limitations of a single imaging method, giving access to a larger amount of information from the same specimen. However, correlative microscopy can be challenging, and appropriate protocols for sample preparation and imaging methods must be selected. Here we review the state of the art of correlating electron microscopy with different imaging methods, focusing on sample preparation, tools, and labeling methods, with the aim to provide a comprehensive guide for those scientists who are approaching the field of correlative methods.
Article
Full-text available
The clustered protocadherins (cPcdhs) are a subfamily of type I single-pass transmembrane cell adhesion molecules predominantly expressed in the brain. Their stochastic and combinatorial expression patterns encode highly diverse neural identity codes which are central for neuronal self-avoidance and non-self discrimination in brain circuit formation. In this review, we first briefly outline mechanisms for generating a tremendous diversity of cPcdh cell-surface assemblies. We then summarize the biological functions of cPcdhs in a wide variety of neurodevelopmental processes, such as neuronal migration and survival, dendritic arborization and self-avoidance, axonal tiling and even spacing, and synaptogenesis. We focus on genetic, epigenetic, and 3D genomic dysregulations of cPcdhs that are associated with various neuropsychiatric and neurodevelopmental diseases. A deeper understanding of regulatory mechanisms and physiological functions of cPcdhs should provide significant insights into the pathogenesis of mental disorders and facilitate development of novel diagnostic and therapeutic strategies.
Article
Full-text available
La liste complète des auteurs est disponible sur la notice WOS : http://gateway.isiknowledge.com/gateway/Gateway.cgi?&GWVersion=2&SrcAuth=INRA&SrcApp=INRA&DestLinkType=FullRecord&DestApp=WOS&KeyUT=ISI:000253008800003
Article
Full-text available
Gamma-protocadherins (Pcdh-γs) are good candidates to mediate specificity in synaptogenesis but their role in cell-cell interactions is a matter of debate. We proposed that Pcdh-γs modify preformed synapses via trafficking of Pcdh-γs-containing organelles, insertion into synaptic membranes and homophilic transcellular interaction. Here we provide evidence in support of this model. We show for the first time that Pcdh-γs have homophilic properties and that they accumulate at dendro-dendritic and axo-dendritic interfaces during neuronal development. Pcdh-γs are maintained in a substantial mobile intracellular pool in dendrites and cytoplasmic deletion shifts the molecule to the surface and reduces the number and velocity of the mobile packets. We monitored Pcdh-γ temporal and spatial dynamics in transport organelles. Pcdh-γ organelles bud and fuse with stationary clusters near synapses. These results suggest that Pcdh-γ-mediated cell-cell interactions in synapse development or maintenance are tightly regulated by control of intracellular trafficking via the cytoplasmic domain.
Article
Full-text available
The hypothalamic neuronal circuits that modulate energy homeostasis become mature and functional during early postnatal life. However, the molecular mechanism underlying this developmental process remains largely unknown. Here we use a mouse genetic approach to investigate the role of gamma-protocadherins (Pcdh-gammas) in hypothalamic neuronal circuits. First, we show that rat insulin promoter (RIP)-Cre conditional knockout mice lacking Pcdh-gammas in a broad subset of hypothalamic neurons are obese and hyperphagic. Second, specific deletion of Pcdh-gammas in anorexigenic proopiomelanocortin (POMC) expressing neurons also leads to obesity. Using cell lineage tracing, we show that POMC and RIP-Cre expressing neurons do not overlap but interact with each other in the hypothalamus. Moreover, excitatory synaptic inputs are reduced in Pcdh-gamma deficient POMC neurons. Genetic evidence from both knockout models shows that Pcdh-gammas can regulate POMC neuronal function autonomously and non-autonomously through cell-cell interaction. Taken together, our data demonstrate that Pcdh-gammas regulate the formation and functional integrity of hypothalamic feeding circuitry in mice.
Article
Full-text available
Autophagosomes and their precursors are best defined by electron microscopy but may also be traced in living cells based on the distribution of specific autophagy molecules. LC3, the most commonly examined autophagy marker in mammalian cells, labels structures that are frequently manifested as dots or rings using light microscopy; however, the nature of these structures is not entirely clear. We reported here a novel approach to examine the LC3-positive compartment in cell-free lysates, which revealed that they were actually tubulovesicular structures with considerable heterogeneity. Using affinity purification, we isolated these membranes for electron microscopy, which indicated that they possessed ultrastructural features consistent with autophagosomal membranes at various maturation stages. Further biochemical and proteomics analyses demonstrated the presence of multiple autophagy-related and other functional molecules. The different distribution patterns of Atg5, Atg16, Atg9, and p62/SQSTM1 on the LC3-positive compartment provided new clues on how these molecules might be involved in the dynamics of the autophagosomal membranes. Finally, several morphologically unique groups of LC3-positive membranes were categorized. Their topological configurations suggested that double-membrane vesicles could be derived from single membrane compartments via different means, including tubule-to-vesicle conversion, whose presence was supported by live cell imaging. These findings thus provide new information on the dynamics of the autophagosomal compartment.
Article
Full-text available
Recent studies indicate that astrocytes, whose processes enwrap synaptic terminals, promote synapse formation both by releasing soluble factors and through contact-dependent mechanisms. Although astrocyte-secreted synaptogenic factors have been identified, the molecules underlying perisynaptic astroctye-neuron contacts are unknown. Here we show that the gamma-protocadherins (gamma-Pcdhs), a family of 22 neuronal adhesion molecules encoded by a single gene cluster, are also expressed by astrocytes and localize to their perisynaptic processes. Using cocultures in which either astrocytes or neurons are Pcdh-gamma-null, we find that astrocyte-neuron gamma-Pcdh contacts are critical for synaptogenesis in developing cultures. Synaptogenesis can eventually proceed among neurons cocultured with Pcdh-gamma-null astrocytes, but only if these neurons themselves express the gamma-Pcdhs. Consistent with this, restricted mutation of the Pcdh-gamma cluster in astrocytes in vivo significantly delays both excitatory and inhibitory synapse formation. Together, these results identify the first known contact-dependent mechanism by which perisynaptic astrocyte processes promote synaptogenesis.
Article
Full-text available
AKAP-Lbc is a member of the A-kinase anchoring protein (AKAP) family that has been recently associated with the development of pathologies, such as cardiac hypertrophy and cancer. We have previously demonstrated that, at the molecular level, AKAP-Lbc functions as a guanine nucleotide exchange factor (GEF) that promotes the specific activation of RhoA. In the present study, we identified the ubiquitin-like protein LC3 as a novel regulatory protein interacting with AKAP-Lbc. Mutagenesis studies revealed that LC3, through its NH(2)-terminal alpha-helical domain, interacts with two binding sites located within the NH(2)-terminal regulatory region of AKAP-Lbc. Interestingly, LC3 overexpression strongly reduced the ability of AKAP-Lbc to interact with RhoA, profoundly impairing the Rho-GEF activity of the anchoring protein and, as a consequence, its ability to promote cytoskeletal rearrangements associated with the formation of actin stress fibers. Moreover, AKAP-Lbc mutants that fail to interact with LC3 show a higher basal Rho-GEF activity as compared with the wild type protein and become refractory to the inhibitory effect of LC3. This suggests that LC3 binding maintains AKAP-Lbc in an inactive state that displays a reduced ability to promote downstream signaling. Collectively, these findings provide evidence for a previously uncharacterized role of LC3 in the regulation of Rho signaling and in the reorganization of the actin cytoskeleton.
Article
Full-text available
Autophagy, an intracellular degradative pathway, maintains cell homeostasis under normal and stress conditions. Nascent double-membrane autophagosomes sequester and enclose cytosolic components and organelles, and subsequently fuse with the endosomal pathway allowing content degradation. Autophagy requires fusion of autophagosomes with late endosomes, but it is not known if fusion with early endosomes is essential. We show that fusion of AVs with functional early endosomes is required for autophagy. Inhibition of early endosome function by loss of COPI subunits (beta', beta, or alpha) results in accumulation of autophagosomes, but not an increased autophagic flux. COPI is required for ER-Golgi transport and early endosome maturation. Although loss of COPI results in the fragmentation of the Golgi, this does not induce the formation of autophagosomes. Loss of COPI causes defects in early endosome function, as both transferrin recycling and EGF internalization and degradation are impaired, and this loss of function causes an inhibition of autophagy, an accumulation of p62/SQSTM-1, and ubiquitinated proteins in autophagosomes.
Article
Full-text available
Cell membranes predominantly consist of lamellar lipid bilayers. When studied in vitro, however, many membrane lipids can exhibit non-lamellar morphologies, often with cubic symmetries. An open issue is how lipid polymorphisms influence organelle and cell shape. Here, we used controlled dimerization of artificial membrane proteins in mammalian tissue culture cells to induce an expansion of the endoplasmic reticulum (ER) with cubic symmetry. Although this observation emphasizes ER architectural plasticity, we found that the changed ER membrane became sequestered into large autophagic vacuoles, positive for the autophagy protein LC3. Autophagy may be targeting irregular membrane shapes and/or aggregated protein. We suggest that membrane morphology can be controlled in cells.
Article
A family of mammalian protocadherin (Pcdh) proteins is encoded by three closely linked gene clusters (α, β, and γ). Multiple α and γ Pcdh mRNAs are expressed in distinct patterns in the nervous system and are generated by alternative pre-mRNA splicing between different “variable” exons and three “constant” exons within each cluster. We show that each Pcdh variable exon is preceded by a promoter and that promoter choice determines which variable exon is included in a Pcdh mRNA. In addition, we provide evidence that alternative splicing of variable exons within a gene cluster occurs via a cis-splicing mechanism. However, virtually every variable exon can engage in trans-splicing with constant exons from another cluster, albeit at a far lower level.
Article
How is the characteristic shape of a membrane bound organelle achieved? We have used an in vitro system to address the mechanism by which the tubular network of the endoplasmic reticulum (ER) is generated and maintained. Based on the inhibitory effect of sulfhydryl reagents and antibodies, network formation in vitro requires the integral membrane protein Rtn4a/NogoA, a member of the ubiquitous reticulon family. Both in yeast and mammalian cells, the reticulons are largely restricted to the tubular ER and are excluded from the continuous sheets of the nuclear envelope and peripheral ER. Upon overexpression, the reticulons form tubular membrane structures. The reticulons interact with DP1/Yop1p, a conserved integral membrane protein that also localizes to the tubular ER. These proteins share an unusual hairpin topology in the membrane. The simultaneous absence of the reticulons and Yop1p in S. cerevisiae results in disrupted tubular ER. We propose that these "morphogenic" proteins partition into and stabilize highly curved ER membrane tubules.
Article
Autophagy is an intracellular degradative pathway that is essential for cellular homeostasis. Efficient autophagy ultimately relies on the ability of the cell to form autophagosomes, and the efficiency of lysosomal enzymes and lipid hydrolases contained within the autolysosome to degrade sequestered cytosolic material and organelles and recycle these nutrients back to the cytosol. Several assays and techniques to monitor autophagy are available, and these can be quantitative or qualitative, biochemical or morphological. Here we describe a method for monitoring the autophagic process that is based on morphology and the application of both light and electron microscopy, called correlative light and electron microscopy, or CLEM. CLEM provides an advance over either technique (light or electron microscopy) alone and can be performed on any cell or tissue sample, which can be grown or mounted on a gridded coverslip or support compatible with light microscopy. CLEM gives a broad low magnification overview of the cell, allowing an assessment of both spatial and temporal events, as well as providing high-resolution information about individual autophagosomes or single compartments.