ArticlePDF Available

Microtubules that form the stationary lattice of muscle fibers are dynamic and nucleated at Golgi elements

Rockefeller University Press
Journal of Cell Biology (JCB)
Authors:
  • Laboratoire Dielen

Abstract and Figures

Skeletal muscle microtubules (MTs) form a nonclassic grid-like network, which has so far been documented in static images only. We have now observed and analyzed dynamics of GFP constructs of MT and Golgi markers in single live fibers and in the whole mouse muscle in vivo. Using confocal, intravital, and superresolution microscopy, we find that muscle MTs are dynamic, growing at the typical speed of ∼9 µm/min, and forming small bundles that build a durable network. We also show that static Golgi elements, associated with the MT-organizing center proteins γ-tubulin and pericentrin, are major sites of muscle MT nucleation, in addition to the previously identified sites (i.e., nuclear membranes). These data give us a framework for understanding how muscle MTs organize and how they contribute to the pathology of muscle diseases such as Duchenne muscular dystrophy.
EB3-GFP shows steady MT dynamics ex vivo and in vivo, whereas GFP-tubulin highlights a durable MT frame. A single image, focused near the surface of a plated fiber expressing EB3-GFP (A1), shows typical puncta. The dynamics can be appreciated in the corresponding time-lapse series (Video 1) and in its projection (A2). Color coding of the projection helps to visualize movement: the first image of the series is colored blue and the last one magenta, as in the bar. EB3-GFP puncta mostly move longitudinally and transversely, as if along parallel and antiparallel tracks (A2, arrows). Some MT intersections (arrowheads) seem to behave as MT nucleation sites. An image from another fiber (B1) and the kymograph of the line between the arrowheads (B2; see Materials and methods) indicate that puncta move at the same speed in either direction on the longitudinal axis (oblique lines; red arrows). EB3-GFP dynamics in vivo (C1 and C2; Videos 2 and 5 and Fig. S2) validate plated fibers in all respects (arrowheads point to nucleation sites). In contrast, GFP-tubulin in plated fibers (D–G) appears static: the color-coded projections (D2, F1, and F2), with the same number of images and frame rate as A2, are practically white. An aster (arrowhead) indicates an MT nucleation site. The kymograph (E2) of the line between the arrowheads (E1) shows stationary MTs, with occasional local movement (arrows). Muscle MTs show dynamic instability (G); the asterisk shows the plus end of a MT growing and shrinking over 80 s. See Tables 1 and S1 for data quantitation and technical parameters. Bars: (A–E) 10 µm; (insets) 2 µm; (kymograph vertical time axes) 60 s.
… 
Content may be subject to copyright.
The Rockefeller University Press $30.00
J. Cell Biol. Vol. 203 No. 2 205–213
www.jcb.org/cgi/doi/10.1083/jcb.201304063 JCB 205
JCB: Report
Correspondence to Evelyn Ralston: evelyn.ralston@nih.gov
Abbreviations used in this paper: AU, Airy units; BfA, brefeldin A; DMD,
Duchenne muscular dystrophy; ERES, ER exit sites; GalT, galactosyl-transferase;
GM, growth media; FDB, flexor digitorum brevis; MAP, MT-associated protein;
MT, microtubule; MTOC, MT-organizing center; NZ, nocodazole; SR, sarco-
plasmic reticulum.
Introduction
Microtubules (MTs) have recently been implicated in the
pathology of Duchenne muscular dystrophy (DMD; Khairallah
et al., 2012), the most common genetic disease of skeletal
muscle. This puts a spotlight on muscle MTs, whose organization
is poorly understood. During myogenesis, coordinated waves of
subcellular remodeling affect MTs as well as MT-organizing
centers (MTOCs), ER exit sites (ERES), and the Golgi complex
(Tassin et al., 1985a,b; Lu et al., 2001; Musa et al., 2003; Bugnard
et al., 2005; Srsen et al., 2009; Zaal et al., 2011). The resulting
organization has little resemblance to that of proliferating
cells. Muscle cultures can be used to study the rst phase of MT
reorganization that takes place during differentiation of myo-
blasts into multinucleated myotubes. But cultured myotubes do
not mature into the bers of which muscle is made. Therefore
most of our knowledge of MTs in adult muscle comes from
immunouorescence images of single bers, hand-teased from
rodent muscles (Ralston, 1993; Ralston et al., 1999, 2001).
Muscle bers are shaped like attened cylinders; their cytoplasm
is mostly lled with actomyosin laments. Between laments and
the plasmalemma there is a thin cytoplasmic layer that contains
nuclei, other organelles, and what we refer to as surface MTs.
Tubulin immunouorescence shows that these MTs form a
grid-like network with very few clear starting or ending points
(Fig. S1 A). There are no clues as to their organization. Also
lacking from these MTs are the asters that typically represent
MT nucleation sites in images of proliferating cells (we call
aster a ower-shaped gure formed by several MTs, which each
have one end anchored to a central point and the other end free
[Fig. S1 A, insets]). In addition, Golgi elements (the small but
numerous Golgi complexes of muscle bers) are positioned at
the vertices of the MT lattice in a unique and unexplained
organization. It became clear that we could only understand
this organization by looking at live cells.
To do so, we have introduced GFP- and mCherry-tagged
MT and Golgi markers into the mouse exor digitorum brevis
(FDB) muscle (Schertzer et al., 2006; Schertzer and Lynch, 2008;
DiFranco et al., 2009; Fig. S1 G). We have characterized their dy-
namics ex vivo, in single bers obtained by enzymatic digestion
of the muscle (Bekoff and Betz, 1977; Rosenblatt et al., 1995),
and in vivo, in muscles of live animals. We show that muscle MTs
are highly dynamic and grow from static Golgi elements. Thus,
Skeletal muscle microtubules (MTs) form a non-
classic grid-like network, which has so far been
documented in static images only. We have now ob-
served and analyzed dynamics of GFP constructs of MT
and Golgi markers in single live fibers and in the whole
mouse muscle in vivo. Using confocal, intravital, and su-
perresolution microscopy, we find that muscle MTs are dy-
namic, growing at the typical speed of 9 µm/min, and
forming small bundles that build a durable network. We
also show that static Golgi elements, associated with the
MT-organizing center proteins -tubulin and pericentrin,
are major sites of muscle MT nucleation, in addition to the
previously identified sites (i.e., nuclear membranes). These
data give us a framework for understanding how muscle
MTs organize and how they contribute to the pathology of
muscle diseases such as Duchenne muscular dystrophy.
Microtubules that form the stationary lattice
of muscle fibers are dynamic and nucleated at
Golgi elements
Sarah Oddoux, Kristien J. Zaal, Victoria Tate, Aster Kenea, Shuktika A. Nandkeolyar, Ericka Reid, Wenhua Liu,
and Evelyn Ralston
Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda,
MD 20892
This article is distributed under the terms of an Attribution–Noncommercial–Share Alike–No
Mirror Sites license for the first six months after the publication date (see http://www.rupress
.org/terms). After six months it is available under a Creative Commons License (Attribution–
Noncommercial–Share Alike 3.0 Unported license, as described at http://creativecommons
.org/licenses/by-nc-sa/3.0/).
THE JOURNAL OF CELL BIOLOGY
JCB • VOLUME 203 • NUMBER 2 • 2013 206
EB3-GFP labeling the plus tips of MTs growing on parallel and
antiparallel tracks. Puncta can be followed on average for 4.3 ± 0.1 s,
i.e., 0.6 µm. Occasional spots (Fig. 1, A, C, and D, arrowheads)
release EB3-GFP again and again, suggesting cytoplasmic MT
nucleation sites. Projections show these spots to be at MT inter-
sections. The mean growth rate of EB3-GFP in muscle bers is
8.6 ± 0.1 µm/min (Table 1), similar to that in Drosophila mela-
nogaster neuronal dendrites (Ori-McKenney et al., 2012).
To validate plated bers as a model for muscle in vivo, we
built a setup for intravital microscopy of the FDB and surround-
ing muscles (Fig. 1 C; Videos 2 and 5; and Fig. S2, A–C). EB3-
GFP dynamics, the presence of antiparallel tracks, and the
existence of cytoplasmic nucleation centers validate all ex vivo
observations. The mean MT growth rate indicated by EB3-GFP
in vivo is 5.0 ± 0.1 µm/min.
GFP-tubulin highlights a stationary,
durable frame
To visualize the entire MTs we then examined GFP-tubulin.
Compared with EB3-GFP, GFP-tubulin in plated bers rst appears
like some other cells (Emov et al., 2007; Rivero et al., 2009;
Ori-McKenney et al., 2012), muscle bers use the Golgi complex
as a MTOC, while forming a MT network unlike any other.
Results and discussion
EB3-GFP reveals a dynamic network of
MTs in live muscle, both ex vivo and in vivo
The MT plus tip end-binding protein EB3 is arguably the best
marker of growing MTs (Stepanova et al., 2003) but its distribu-
tion in muscle bers is unusual because it is found not only at
the tips but anywhere along MTs (Fig. S1 B). Our rst goal was
therefore to clarify EB3 distribution by observing EB3-GFP in
live bers (see Materials and methods and Fig. S1 D2).
In plated bers (Fig. 1, A1 and B1), each of the EB3-GFP
puncta moves (Video 1) mostly longitudinally (parallel to the ber
axis) or transversely (perpendicular). To analyze EB3-GFP dynam-
ics we generated color-coded time-lapse projections (Fig. 1 A2)
and kymograph plots (Fig. 1 B). Puncta move indifferently left or
right (56 vs. 44%; Fig. 1 A2) and up or down, and at similar speeds
in all directions (Fig. 1 B, arrows). These results are consistent with
Figure 1. EB3-GFP shows steady MT dynamics ex vivo and in vivo, whereas GFP-tubulin highlights a durable MT frame. A single image, focused near the
surface of a plated fiber expressing EB3-GFP (A1), shows typical puncta. The dynamics can be appreciated in the corresponding time-lapse series (Video 1)
and in its projection (A2). Color coding of the projection helps to visualize movement: the first image of the series is colored blue and the last one magenta,
as in the bar. EB3-GFP puncta mostly move longitudinally and transversely, as if along parallel and antiparallel tracks (A2, arrows). Some MT intersections
(arrowheads) seem to behave as MT nucleation sites. An image from another fiber (B1) and the kymograph of the line between the arrowheads (B2; see
Materials and methods) indicate that puncta move at the same speed in either direction on the longitudinal axis (oblique lines; red arrows). EB3-GFP dynam-
ics in vivo (C1 and C2; Videos 2 and 5 and Fig. S2) validate plated fibers in all respects (arrowheads point to nucleation sites). In contrast, GFP-tubulin in
plated fibers (D–G) appears static: the color-coded projections (D2, F1, and F2), with the same number of images and frame rate as A2, are practically
white. An aster (arrowhead) indicates an MT nucleation site. The kymograph (E2) of the line between the arrowheads (E1) shows stationary MTs, with
occasional local movement (arrows). Muscle MTs show dynamic instability (G); the asterisk shows the plus end of a MT growing and shrinking over 80 s.
See Tables 1 and S1 for data quantitation and technical parameters. Bars: (A–E) 10 µm; (insets) 2 µm; (kymograph vertical time axes) 60 s.
207
Skeletal muscle microtubule dynamics • Oddoux et al.
These observations imply that muscle MTs are bundled.
However, diffraction-limited microscopy, with a resolution of
250 nm at best, cannot resolve closely spaced MTs, which have
outer diameters of 25 nm. Using G-STED superresolution micro-
scopy (Vicidomini et al., 2013), we reached an improved resolu-
tion of 65 nm and resolved MT tracks into two to four strands,
often torsading around each other (Fig. 3, B and C, red arrow-
heads). With a few exceptions (Fig. 3 C, black arrowhead) the
strands are of even intensity, suggesting that they represent single
MTs. Muscle MTs thus form small bundles with space between
strands for MT-associated proteins (MAPs; Chen et al., 1992).
MT tracks may contain dynamic MTs growing alongside
stable MTs, as suggested by the stationary character of the GFP-
tubulin frame (Fig. 1 D). However, few muscle MTs have the
curly shape and posttranslational tubulin modications typical
of stable MTs (Schulze et al., 1987). Muscle MTs could be stabi-
lized by the muscle-specic isoform of MAP4 (Nguyen et al.,
1997), which lines each muscle MT (Fig. S1 C) and whose
function is not clear (Mangan and Olmsted, 1996; Casey et al.,
2003). However, it is most likely that the stationary MT frame
is entirely composed of dynamic MTs fasciculating along each
other. Although MTs can bear compressive forces and support
a cell structurally (Brangwynne et al., 2006), a dynamic frame
of short MTs may be better suited to contracting bers; indeed,
excessive MT stabilization has been implicated in cardiac
hypertrophy (Sato et al., 1997; Takahashi et al., 2003) and in
DMD (Khairallah et al., 2012).
Muscle MTs nucleate from static
Golgi elements
During muscle differentiation the MTOC redistributes from
centrosomes to nuclear membranes (Tassin et al., 1985a; Lu
et al., 2001; Bugnard et al., 2005; Zaal et al., 2011). EB3-GFP and
immobile, highlighting a frame that remains unchanged for min-
utes (Fig. 1 D1 and Video 3); the color-coded projections are
mostly white (Fig. 1 D2). However, there are pockets of grow-
ing and shrinking MTs (Fig. 1, D2 [inset] and E–G), reecting
dynamic instability (Mitchison and Kirschner, 1984). The mean
growth rate is 5.6 ± 0.2 µm/min. GFP-tubulin, like EB3-GFP,
grows indifferently left or right (45 ± 4 vs. 55 ± 4%), up or down.
When two MTs are on the same stretch of the frame they grow as
frequently in parallel as in antiparallel directions (48 vs. 52%).
In vivo recordings of GFP-tubulin also validated the
ex vivo data (Fig. S2, D and E; and Video 6). GFP-tubulin–
labeled MTs in vivo grow at 3.6 ± 0.2 µm/min. We ascribe the
apparently slower growth rate of GFP-tubulin compared with
that of EB3-GFP to the different methods of analysis (manual
vs. PlusTipTracker; see Materials and methods) and to intrinsic
differences between the two markers. Tubulin is the building
block of MTs, whereas EB3 is in dynamic association with MTs
from which it dissociates when MTs pause or shrink (explaining
why EB3 puncta can only be followed on average for 0.6 µm).
To learn more about the MT frame, we followed FRAP of
regions of interest containing one to three MT tracks (Fig. 2).
Because tubulin subunits are not in dynamic exchange with
MTs, we did not expect true recovery, i.e., a uniform progres-
sive return of the bleached track. Recovery was possible if new
GFP-tubulin–labeled MTs grew along the bleached track, as
occurred, but in only 7 out of 45 MT tracks. However, in half the
regions of interest a new MT entered the box near the bleached
track (Fig. 2 A, box 3 and arrow). Displacement of the bleached
MT portion, which would indicate motor-induced MT trans-
port, was not observed. The FRAP results are consistent with a
frame of stable, immobile MTs and/or of anchored, growing
MTs. Many of the observed MTs show dynamic instability,
which results in repeated loss and gain of the EB3–tip complex.
This can explain the apparent discrepancy between the low
number of GFP-tubulin recoveries and the larger number of
EB3-GFP puncta that would move in comparable ber areas.
The durable MT frame serves as a track
for growing MTs
To clarify the apparent differences between EB3 and tubulin
dynamics, we coexpressed EB3-GFP and mCherry-tubulin
(Fig. 3 A). Each of the markers behaves as if alone, indicating that
one does not interfere with the other and that neither affects
the network as a whole (also see immunoblotting controls in
Fig. S1, E and F). EB3-GFP moves along mCherry-tubulin–
labeled tracks (Fig. 3 A1 [arrows] and Video 4). In addition, multiple
EB3 dots move in the same or in opposite directions on a single
track. The kymograph (Fig. 3 A2) emphasizes both the contrast in
motility and the association of EB3-GFP with mCherry-tubulin.
Figure 2. FRAP showing only growth (or transport) of MTs restores fluor-
escence to the bleached area. (A) GFP-tubulin in a plated FDB fiber before
(prebleach), just after (bleach), and 28 s after photobleaching of two re-
gions of interest surrounded by orange boxes. For quantitation of recovery
(B), the lower box was divided into parts 2 and 3. In seven independent
photobleachings, more than half of the bleached boxes recover some fluor-
escence, as is the case in box 3, by growth or transport of a MT distinct
from the original one (A, arrows). Bar, 2 µm.
Table 1. Measurements of MT growth rate
MT marker Growth rate ± SEM
Plated fibers Intravital
µm/min µm/min
EB3-GFP 8.6 ± 0.1 (n = 463) 5.0 ± 0.1 (n = 220)
GFP-tubulin 5.6 ± 0.2 (n = 49) 3.6 ± 0.2 (n = 31)
JCB • VOLUME 203 • NUMBER 2 • 2013 208
could play a role in GLUT4 translocation (Semiz et al., 2003)
and lysosomal positioning (Fukuda et al., 2006; Korolchuk
et al., 2011).
-Tubulin and pericentrin are associated
with Golgi elements
The MTOC protein -tubulin is required for both centro-
somal and noncentrosomal MT nucleation (Erhardt et al., 2002;
Bugnard et al., 2005; Emov et al., 2007; Rivero et al., 2009;
Ori-McKenney et al., 2012; Zhu and Kaverina, 2013), but it is
anchored by different proteins. In nonmuscle cells, Golgi -
tubulin is linked either to the trans-Golgi by GCC185 and
CLASP2 (Emov et al., 2007) or to the cis-Golgi by GM130
and AKAP450 (Rivero et al., 2009).
By immunouorescence we detected -tubulin on GM130-
labeled Golgi elements of muscle bers (Fig. 4 C and Table 2).
CLASP2 could not be detected at all, and AKAP450 was only
seen in the perinuclear area (Fig. 4 E). However, AKAP450
resembles another MTOC protein, pericentrin. AKAP450 and
pericentrin are large coiled-coil proteins that share a centrosome-
binding sequence (Gillingham and Munro, 2000). We found in
silico that they also share sequences in the GM130-binding re-
gion of AKAP450 (Hurtado et al., 2011). Moreover, AKAP450
and kendrin, a pericentrin isoform, collaborate to bind -tubulin to
centrosomes (Takahashi et al., 2002). Pericentrin could there-
fore replace or supplement AKAP450 in muscle bers. Peri-
centrin was detected in bers and found colocalized with Golgi
elements (Fig. 4 D and Table 3). Although these results are
consistent with cis-Golgi MT nucleation, we have not been
able to conrm the involvement of GM130 because knocking
it down by shRNA was inconclusive. It is possible that both
cis- and trans-Golgi nucleations coexist or that other cis-Golgi
proteins are involved.
Finally, we assessed the motility of Golgi elements. We
coexpressed galactosyl-transferase-mCherry (GalT-mCherry),
GFP-tubulin, however, grow from additional nucleation centers
(Fig. 1 and Videos 1–3), which are at MT intersections, where
we also nd Golgi elements (Fig. 4 A1). To assess whether Golgi
elements serve as nucleation centers, we treated muscle bers
with nocodazole (NZ) to depolymerize MTs, washed NZ away,
and analyzed MT recovery in relation to Golgi elements. After
NZ treatment (Fig. 4 A2), only curly, NZ-resistant MTs are left
(arrowheads), many of which surround nuclei. These MTs are
detyrosylated, i.e., stable (Fig. 4 F). During the rst minutes of
recovery they elongate (Fig. 4 A3, arrowheads), whereas MT
seeds appear around Golgi elements (arrows) and nuclei. These
seeds rapidly grow into well-formed asters, strikingly centered
on Golgi elements (Fig. 4 A4, arrows). MTs from different
nucleation centers become interconnected (Fig. 4 A5) and an
integrated network reforms (Fig. 4 A6). The percentage of Golgi
elements associated with asters progressively decreases (Fig. 4 B),
indicating that the asters become part of the reformed MT net-
work. We conclude that Golgi elements (in addition to the nu-
clear membranes already described in myotubes) nucleate MTs
in muscle bers.
In proliferating cells, the involvement of the Golgi com-
plex in MT nucleation has been conrmed by treatment with
brefeldin A (BfA), which redistributes Golgi components in-
cluding Golgi-nucleated MTs to ERES (Rivero et al., 2009).
In muscle bers we found no convincing effect of BfA, likely
because the Golgi elements are dispersed and colocalized
with ERES even in the absence of BfA (Ralston et al., 1999;
2001; Lu et al., 2001).
Muscle bers thus have at least two categories of MT
nucleation sites, nuclei and Golgi elements. We do not know
whether there are any biochemical or functional differences
between MTs nucleated from these distinct sites, but one can
hypothesize that perinuclear MTs are involved in the position-
ing of nuclei (Elhanany-Tamir et al., 2012; Metzger et al., 2012;
Wilson and Holzbaur, 2012), whereas Golgi-nucleated MTs
Figure 3. Dynamic MTs grow along MTs and
are bundled. EB3-GFP and mCherry-tubulin
were coexpressed and simultaneously im-
aged in plated fibers (A1; Video 4). EB3-GFP
(green) moves along static mCherry-tubulin
tracks (red; A2 kymograph). The asterisk indi-
cates a nucleation spot and the arrows point to
EB3-GFP–labeled MTs growing toward each
other. G-STED superresolution microscopy of
FDB fibers stained for -tubulin resolves MTs
into two or more components (B and C; panels
with blue arrowheads show confocal images;
panels with red arrowheads show the cor-
responding G-STED image at a resolution of
65 nm). A black arrowhead points to a proba-
bly unresolved MT. Bars: (A) 10 µm; (B and C)
2.5 µm; (A2, vertical time axis) 60 s.
209Skeletal muscle microtubule dynamics • Oddoux et al.
and its absence in the mdx mouse prevents MTs from form-
ing an orthogonal grid (Percival et al., 2007; Prins et al., 2009).
However, dystrophin does not line MTs as MAP4 does; instead,
dystrophin appears to dene domains along which MTs grow
preferentially (Prins et al., 2009). Dystrophin may capture MTs;
those that start at an oblique angle (Fig. 1 D2, colored MTs)
often abruptly change their orientation (Fig. 5 B) when they
encounter the transverse bands that contain dystrophin. Studies
of MT dynamics and directionality in mdx muscles will help us
to further develop this model.
Skeletal muscle MTs, like the proverbial canary in the
mine, are affected by practically all physiological and patho-
logical changes in muscle, most likely because they are sen-
sitive to patterned contractions (Ralston et al., 2001). They
reach and dynamically connect all domains of muscle bers.
a marker of the Golgi complex, with GFP-tubulin or EB3-GFP
(Fig. S3, Video 7, and Video 8). In both plated bers (Fig. S3 A)
and in vivo (Fig. S3 B) Golgi elements are static. Thus, the clas-
sic model that MTs position the Golgi complex is turned upside
down in muscle, where, instead, static Golgi elements position
MT nucleation. The lack of motility of muscle Golgi elements
is consistent with their steady positioning along Z bands (Kaisto
and Metsikkö, 2003).
Building a model of muscle MT organization
At this point, we can start building a model (Fig. 5 A). At
steady-state, MTs nucleated from Golgi elements grow along
other dynamic and/or stable MTs to form bundles. To explain
the orthogonal grid of MTs, we must involve dystrophin, the
protein missing in DMD. Dystrophin is a MAP (Prins et al., 2009)
Figure 4. MTs are nucleated on Golgi ele-
ments that concentrate -tubulin and pericen-
trin. To investigate MT nucleation, plated FDB
fibers were treated with NZ to depolymerize
MTs and fixed after different periods of recov-
ery. They were then stained with anti–-tubulin
(-tub) and anti-GM130 (gm) to label MTs
and Golgi elements. In a control fiber (A1),
Golgi elements are along MTs, especially at
crossings, and around nuclei. After NZ, before
recovery (A2), only a few curly MTs remain (ar-
rowheads). These contain both detyrosylated
and tyrosylated tubulins (detyr-tub and Y-tub; F),
indicating the presence of both stable and
dynamic MTs. In the first minute of recovery,
NZ-resistant MTs elongate (A3, arrowheads)
and MT seeds appear around Golgi elements
(A3, arrows). A few minutes later, full asters
centered on Golgi elements become prominent
(A4, arrows). MTs then progressively reform a
network (A5 and A6). During early recovery,
90% of Golgi elements are at the center
of MT asters; at steady-state, only 10% are
(B; data are from a single representative ex-
periment out of three; n = 100, from five fibers,
for each time point). Fibers at early stages of
recovery (as shown in A3) were stained with
anti-GM130 and with antibodies against -
tubulin (-tub), pericentrin, and AKAP450.
-Tubulin and pericentrin are concentrated
on Golgi elements of MT seeds (C1–C4 and
D1–D4); AKAP450 is only detected around
nuclei (E1–E3). Most MT seeds are associ-
ated with Golgi elements ± -tubulin and some
with -tubulin alone (C5; Table 2). Similar
results are obtained for quantitation with peri-
centrin (D5; Table 3; two independent ex-
periments, nine fibers, 400 MT seeds for C5,
and 322 MT seeds for D5). Bars: (A–F) 10 µm;
(C4 and D4) 2 µm.
Table 2. Association of MT seeds with GM130 and -tubulin
Associated with Fraction of MT seeds ± SEM
%
GM130 + -tubulin 20.1 ± 2.8
GM130 only 25.4 ± 2.9
-Tubulin only 14.1 ± 1.7
None 40.4 ± 4.0
Based on two experiments and 322 MT seeds.
Table 3. Association of MT seeds with GM130 and pericentrin
Associated with Fraction of MT seeds ± SEM
%
GM130 + pericentrin 45.5 ± 4.9
GM130 only 6.1 ± 0.8
Pericentrin only 13.6 ± 1.5
None 34.8 ± 4.5
Based on two experiments and 400 MT seeds.
JCB • VOLUME 203 • NUMBER 2 • 2013 210
AKAP350 from J. Goldenring (Medical College of Georgia, Augusta, GA;
Shanks et al., 2002). Other antibodies were purchased commercially:
rat anti-tyrosylated tubulin from Accurate; rabbit anti–-tubulin and mouse
anti-GAPDH from Abcam; mouse anti–-tubulin, rabbit anti-acetylated tubu-
lin, and rabbit anti–-tubulin from Sigma-Aldrich; rabbit anti-detyrosylated
tubulin from EMD Millipore; mouse anti-GM130 and mouse anti-pericentrin
from BD; rabbit anti-GFP from Cell Signaling Technology; and rabbit anti-
pericentrin from Covance. Goat anti–mouse and anti–rat conjugated with
DyLight 488, 549, and 647 were obtained from Jackson ImmunoResearch
Laboratories, Inc. and goat anti–rabbit conjugated with Alexa 488, 546,
568, and 647 were obtained from Molecular Probes. For Western blot anal-
yses we used horseradish peroxidase–conjugated goat anti–rabbit and goat
anti–mouse purchased from Bio-Rad Laboratories and Alexa 680–conjugated
goat anti–rat and anti–mouse antibodies purchased from Invitrogen.
Plasmids
All plasmids expressed in this study are based on pEGFP-N1 or pEGFP-
C1 vectors (Takara Bio Inc.). p-EB3-GFP-N1 cDNA was a gift from A.
Akhmanova (Stepanova et al., 2003), p-EGFP-tubulin-C1 was obtained
from Takara Bio Inc., and p-mCherry-tubulin-C1 was constructed from p-eGFP-
tubulin-C1 and p-mCherry-C1. p-GalT-mCherry-N1 was constructed from
p-GalT-eGFP-N1 (Zaal et al., 1999) and p-mCherry-N1 (a gift from G. Patterson
[National Institute of Biomedical Imaging and Bioengineering, Bethesda, MD]).
p-EGFP-EMTB-N1 was subcloned from p-EMTB-N1 (a gift from C. Bulinski
[Columbia University, New York, NY]).
cDNA injection and electroporation into mouse muscles
All animal protocols were reviewed and approved by the National Institute
of Arthritis and Musculoskeletal and Skin Diseases Animal Care and Use
Committee. Mice were C57BL/6 (The Jackson Laboratory), 6 to 8 wk old
unless otherwise mentioned. To obtain cDNA expression we followed the
protocol of DiFranco et al. (2009) with a few modifications. Mice were
anesthetized with 4% isoflurane throughout the procedure and received a
subcutaneous injection of 0.05 mg/kg buprenorphine-HCl to avoid pain.
To loosen the extracellular matrix and allow the plasmid to reach the
FDB muscle, which extends along the sole of the foot, 10 µl of 0.5 U/µl
(0.36 mg/ml) hyaluronidase was injected through the skin at the heel.
After 1 h, 20–50 µg of endotoxin-free plasmid was injected (Genewiz) at
5 mg/ml in sterile DPBS. After 15 min, acupuncture needles (0.20 ×
25 mm, Tai Chi; Lhasa OMS) were placed under the skin at the heel and at
the base of the toes and connected to an ECM 830 BTX electroporator
(BTX Harvard Apparatus). Six pulses of 20 ms each at 1 Hz were applied
to yield an electric field of 75 V/cm. 5–7 days later the animal was killed
to collect muscles or prepared for intravital imaging. Expression of the
injected cDNA is easily verified under fluorescence illumination on the dis-
section microscope (Fig. S1 G). The efficiency of expression ranges from
20 to 80% of the fibers depending on the plasmid.
Selection of EB3-GFP and GFP-tubulin constructs to track MTs
in muscle fibers
In search of suitable MT markers we expressed GFP-tubulin (Fig. S1 D1),
EB3-GFP (Fig. S1 D2), and the MT-binding domain of the MAP ensconsin
(EMTB-GFP; Fig. S1 D3). Our criteria for accepting a construct were as fol-
lows: pattern and location indistinguishable from native MTs (Fig. S1 A),
even at moderate expression levels, normal appearance of the whole MT
network, and useful signal-to-noise ratio. EB3-GFP and GFP-tubulin satisfied
all criteria. Regrettably, EMTB (Faire et al., 1999) caused abnormalities of
MTs in all but the lowest expressing fibers. We verified by immunoblotting
(Fig. S1 F) that GFP-tubulin undergoes detyrosylation and acetylation, the
normal posttranslational modifications of muscle tubulin (Gundersen et al.,
1989), and that EB3-GFP overexpression does not affect the posttransla-
tional modifications of tubulin (Fig. S1 E). We also checked by immunofluor-
escence that the expression of the GFP constructs does not alter the
respective patterns of detyrosylated and tyrosylated tubulin. Finally we veri-
fied that mCherry constructs gave results similar to those with the corre-
sponding GFP construct.
Intravital imaging
Mice injected with cDNA and electroporated as described in a previous
paragraph were anesthetized by intraperitoneal injection of 75 mg/kg
of sodium pentobarbital. A flap of skin was removed from the plant of
the foot to expose the FDB. The mouse was placed in a tub-shaped stage
insert (custom designed at the National Institute of Arthritis and Musculo-
skeletal and Skin Diseases for the TCS SP5 confocal microscope [Leica]),
the bottom of which was made of a no. 1.5 coverglass. The exposed FDB,
MTs are moored by Golgi elements through ERES to the ER/
sarcoplasmic reticulum, which extends longitudinally in the
myobrillar core (Kaisto and Metsikkö, 2003). They interact with
the triad junctions of T-tubules and sarcoplasmic reticulum
(Fourest-Lieuvin et al., 2012), with dystrophin, and also with
muscle-specic protein networks involved in the maintenance
of sarcomeric organization (Ayalon et al., 2008, 2011; Randazzo
et al., 2013). For a long time, the relevance of muscle MTs
and the consequences of their perturbations were not clear. Now
muscle MTs are nally starting to receive long overdue attention.
Materials and methods
Antibodies and other reagents
Several antibodies were gifts: rabbit anti–mouse/human detyrosylated
tubulin from G. Cooper IV and T. Galien (Veterans Affairs Medical Center,
Charleston, SC; Sato et al., 1997); rabbit anti–mouse/human EB3 from
A. Akhmanova (Utrecht University, Utrecht, Netherlands; Stepanova et al.,
2003); rabbit anti–mouse mMAP4 from J. Olmsted (University of Rochester,
Rochester, NY; Casey et al., 2003); and rabbit anti–human AKAP450/
Figure 5. Model of MT organization in skeletal muscle fibers at steady-
state. (A) MTs nucleating from Golgi elements grow parallel or antiparal-
lel to existing MTs, thereby forming small bundles, which are guided or
restricted by the dystrophin bands positioned along Z lines, M bands, and
longitudinal stripes. MTs starting at an oblique angle reorient upon con-
tact with other MTs or dystrophin bands, as observed (B) in GFP-tubulin–
expressing fibers. The MT lattice that results is both durable and dynamic.
Bars, 2 µm.
211Skeletal muscle microtubule dynamics • Oddoux et al.
detection on the gated HyD detectors of a TCS SP8 X G-STED system
(Leica) with pulsed white light laser excitation. The conditions for confocal
imaging were identical to those for time-lapse series, i.e., pinhole opened
to 1.5 AU to increase brightness and depth of field, whereas G-STED
parameters were optimized for best resolution, i.e., pinhole between 0.5
and 1.0 AU.
Image rendering and analysis
Two techniques were used to make dynamics perceptible in single static
images: color coding and kymograph plotting. Color coding of confocal
time series was done with Photoshop CS5. Each image of the 38-s time se-
ries had its red, green, and blue levels set to obtain a progressive change
of colors, the first frame being blue (red = 0, green = 0, and blue = 255)
and the last frame magenta (red = 255, green = 0, and blue = 255). A
moving object appears rainbow colored in projection whereas a station-
ary object is white. Kymographs were done on image stacks in ImageJ
with the Reslice tool. The position of the line that is extracted from each
image and repeated in the kymograph is indicated by two arrowheads on
one of the images. An object that moves along the selected line during the
recording of the series appears as an oblique dash in the kymograph, a
stationary object on the line appears as a vertical line, and an object that
crosses the line appears as a single dot.
EB3-GFP speed was analyzed with PlusTipTracker, a MatLab-based
open source software (Applegate et al., 2011). The tracks were detected
with PlusTip GetTracks, with the following parameters, adapted depending
on the quality of the movie: gap length = 3, angle = 10–15, radius range =
1–8, fluctuation radius = 2, and maximum shrinkage factor = 1.5. Each
track was checked by hand with PlusTip SeeTracks to avoid false positives.
GFP-tubulin MT growth rate was analyzed with ImageJ. The path covered
was tracked, measured manually, and converted to a growth rate in mi-
crometers per minute.
To quantitate the association of nascent MTs with Golgi elements,
-tubulin, and/or pericentrin (Fig. 4, C5 and D5; and Tables 2 and 3),
we examined the tubulin staining of triple-stained immunofluorescence im-
ages while hiding the other channels, marked the position of presumed MT
seeds (at least three MT fragments), and then examined whether Golgi and
-tubulin or pericentrin staining were present. Because MT seeds at early
recovery times are less organized than the later asters, it is possible to count
as seeds the simple crossing of two MTs. It is likely that this explains the rel-
atively high percentage of MT seeds not linked to any of the components.
Immunoblots
Fibers were prepared as described in a previous paragraph but instead
of being plated they were rinsed three times in DPBS, lysed in 40 µl of
loading buffer (National Diagnostics), and boiled. Protein concentration
was assayed with the 2D Quant Kit (GE Healthcare). Proteins were sepa-
rated on 10% acrylamide gels and immunoblotted according to standard
procedures. Protein bands were detected either with the Odyssey infrared
imaging system (Li-Cor) or with photographic film.
Online supplemental material
Fig. S1 presents background and quality control data: immunofluorescence
of muscle fibers stained for tubulin, EB3, and MAP4, and characterization
of muscle fibers expressing GFP constructs by fluorescence and immuno-
blotting. Fig. S2 illustrates the setup for and images from intravital imaging of
EB3-GFP and GFP-tubulin. Fig. S3 illustrates simultaneous recordings of the
Golgi marker GalT-mCherry and of a MT marker, ex vivo and in vivo. Videos
provide examples of time-lapse recordings and are essential to perceive MT
dynamics. Videos 1 and 2 present EB3-GFP dynamics ex vivo and in vivo,
respectively. Video 3 shows GFP-tubulin ex vivo. Video 4 shows simultaneous
EB3-GFP and mCherry-tubulin recordings ex vivo. Video 5 shows a more ex-
tensive muscle area expressing EB3-GFP in vivo. Video 6 shows GFP-tubulin
in vivo. Video 7 shows simultaneous GFP-tubulin and GalT-mCherry record-
ings ex vivo, and Video 8 shows EB3-GFP and GalT-mCherry in vivo. On-
line supplemental material is available at http://www.jcb.org/cgi/content/
full/jcb.201304063/DC1. Additional data are available in the JCB Data-
Viewer at http://dx.doi.org/10.1083/jcb.201304063.dv.
We wish to acknowledge the late George Cooper IV for generous advice
over the years. We thank all colleagues who shared reagents; Jonathan Boyd
and Geoff Daniels for essential help with G-STED imaging; Gary Melvin for
design and fabrication of hardware for intravital imaging; and Nina Raben,
Rachel Myerowitz, Dan Sackett, and Andrew Milgroom for critical reading of
the manuscript.
facing the objective lens, was held on the coverglass by a small lever arm
(Fig. S3, A and B). The mouse temperature was maintained at 37°C with a
heating lamp, and breathing of the animal was monitored visually through-
out the experiment.
FDB fiber preparation
Mice were killed by CO2 followed by cervical dislocation. FDB muscles
were dissected in sterile DPBS under an MZ FLIII dissecting microscope
(Leica), rinsed in sterile DMEM, and incubated with rotation for 3 h at 37°C
in DMEM containing 1.5 mg/ml type I collagenase from Clostridium histo-
lyticum (Sigma-Aldrich) and 1 mg/ml BSA. Fibers were then freed from the
muscle by trituration and plated on Mattek dishes or Lab-Tek chambered
coverglass that had been coated for 1 h with a 1:10 dilution of Matrigel
(BD). Fibers were plated in 0.1 ml of growth media (GM), consisting of
DMEM supplemented with 20% FBS and 0.2% chicken embryo extract.
After 2 h they were fed with GM supplemented with penicillin-streptomycin.
The fibers were used within 24 h, ahead of the reorganization of MT and
other cytoskeletal components, which occurs after denervation or when
patterned activity is modified (Ralston et al., 2001, 2006). The only dam-
age directly linked to the collagenase treatment is a degradation of the
fine structure of the neuromuscular junction. We have not searched for
neuromuscular junction–specific MTs (Schmidt et al., 2012). FDB fibers
are predominantly type IIA (intermediate type). Their MTs are organized
similarly to those in fast-twitch fibers (Ralston et al., 1999).
Drug treatments
To prevent MT polymerization and induce loss of MTs, plated fibers were
incubated for 4 h at 37°C in 4 µg/ml NZ (Sigma-Aldrich) in GM. After
washing out the drug with GM at 37°C, MTs were left to recover at 37°C
in GM for 2 min to 24 h. Fibers were then fixed for staining, either with
methanol at 20°C or with 4% PFA (Electron Microscopy Sciences). The
Golgi complex was disrupted with 5 µg/ml BfA (Sigma-Aldrich) at 37°C
for 1 h. For unrecovered NZ or BfA controls, the drug was added to PFA;
alternately, methanol was used for fixation. When combined with NZ treat-
ment, BfA was added for the last hour of the 4-h incubation with NZ and
throughout the washout and fixation process.
Immunofluorescence
Fixed fibers were blocked for 2 h at RT in PBS containing either 5% BSA,
1% normal goat serum, and 0.04% saponin or the blocking reagent from
the Mouse On Mouse basic kit (Vector Laboratories). Fibers were then incu-
bated with primary antibodies for 2 h at RT (or overnight at 4°C) and with
secondary antibodies for 2 h at RT, counterstained with Hoechst 33342,
and mounted in Vectashield (Vector Laboratories). Rinses between and
after antibody incubations were three times 5 min with PBS containing
0.04% saponin.
Microscopy
Confocal images were collected using a 63× 1.4 NA or a 40× 1.25 NA
oil immersion objective lenses on a TCS SP5 (Leica) driven by the LAS AF
2.6.1 software or on an LSM 780 confocal microscope (Carl Zeiss) driven
by Zen 2011. In both cases, images were collected in sequential scanning
to avoid cross talk. Live fibers were imaged in phenol red–free GM with
25 mM Hepes at 37°C, using a Tokai Hit heated stage insert on the SP5
and a Pecon Lab-Tek S1 stage insert on the LSM 780. We used a confocal
pinhole between 1 and 3 Airy units (AU) to increase depth of field and sig-
nal intensity while limiting potential laser damage to live fibers.
For FRAP sequences on the LSM 780, we wanted to achieve close
to complete photobleaching of target regions without damaging them. We
tested different protocols and settled on 10 bleaching iterations with a
40-mW Argon laser at 80%. We were satisfied that no damage was done
because MTs that had been photobleached continued to grow outside of
the bleached area.
Unless otherwise mentioned, the images shown are single frames.
Camera icons refer to the corresponding video, available online. Projec-
tions of images were done as “maximum” projections. Images were ex-
ported in 8-bit TIF format and linearly adjusted using Photoshop CS5, and
then cropped and resized if needed for composing montages. Some mov-
ies were processed in ImageJ (National Institutes of Health) with an image
stabilizer and Kalman filter. For black and white images, the grayscale
was inverted to facilitate viewing. Settings used for still images and videos
can be found in Table S1.
Superresolution microscopy
Fibers stained with anti-tubulin followed by Alexa 488 or 647 goat anti–
mouse IgG were mounted in Prolong Gold. They were imaged using time-gated
JCB • VOLUME 203 • NUMBER 2 • 2013 212
directional cell migration and ciliogenesis. J. Cell Biol. 193:917–933.
http://dx.doi.org/10.1083/jcb.201011014
Kaisto, T., and K. Metsikkö. 2003. Distribution of the endoplasmic reticulum
and its relationship with the sarcoplasmic reticulum in skeletal myobers.
Exp. Cell Res. 289:47–57. http://dx.doi.org/10.1016/S0014-4827(03)
00231-3
Khairallah, R.J., G. Shi, F. Sbrana, B.L. Prosser, C. Borroto, M.J. Mazaitis, E.P.
Hoffman, A. Mahurkar, F. Sachs, Y. Sun, et al. 2012. Microtubules under-
lie dysfunction in duchenne muscular dystrophy. Sci. Signal. 5:ra56.
http://dx.doi.org/10.1126/scisignal.2002829
Korolchuk, V.I., S. Saiki, M. Lichtenberg, F.H. Siddiqi, E.A. Roberts, S. Imarisio,
L. Jahreiss, S. Sarkar, M. Futter, F.M. Menzies, et al. 2011. Lysosomal
positioning coordinates cellular nutrient responses. Nat. Cell Biol. 13:
453–460. http://dx.doi.org/10.1038/ncb2204
Lu, Z., D. Joseph, E. Bugnard, K.J. Zaal, and E. Ralston. 2001. Golgi com-
plex reorganization during muscle differentiation: visualization in living
cells and mechanism. Mol. Biol. Cell. 12:795–808. http://dx.doi.org/10
.1091/mbc.12.4.795
Mangan, M.E., and J.B. Olmsted. 1996. A muscle-specic variant of microtubule-
associated protein 4 (MAP4) is required in myogenesis. Development.
122:771–781.
Metzger, T., V. Gache, M. Xu, B. Cadot, E.S. Folker, B.E. Richardson, E.R.
Gomes, and M.K. Baylies. 2012. MAP and kinesin-dependent nuclear
positioning is required for skeletal muscle function. Nature. 484:120–
124. http://dx.doi.org/10.1038/nature10914
Mitchison, T., and M. Kirschner. 1984. Dynamic instability of microtubule
growth. Nature. 312:237–242. http://dx.doi.org/10.1038/312237a0
Musa, H., C. Orton, E.E. Morrison, and M. Peckham. 2003. Microtubule assem-
bly in cultured myoblasts and myotubes following nocodazole induced
microtubule depolymerisation. J. Muscle Res. Cell Motil. 24:301–308.
http://dx.doi.org/10.1023/A:1025477807393
Nguyen, H.L., S. Chari, D. Gruber, C.M. Lue, S.J. Chapin, and J.C. Bulinski.
1997. Overexpression of full- or partial-length MAP4 stabilizes micro-
tubules and alters cell growth. J. Cell Sci. 110:281–294.
Ori-McKenney, K.M., L.Y. Jan, and Y.N. Jan. 2012. Golgi outposts shape
dendrite morphology by functioning as sites of acentrosomal micro-
tubule nucleation in neurons. Neuron. 76:921–930. http://dx.doi.org/10
.1016/j.neuron.2012.10.008
Percival, J.M., P. Gregorevic, G.L. Odom, G.B. Banks, J.S. Chamberlain, and
S.C. Froehner. 2007. rAAV6-microdystrophin rescues aberrant Golgi
complex organization in mdx skeletal muscles. Trafc. 8:1424–1439.
http://dx.doi.org/10.1111/j.1600-0854.2007.00622.x
Prins, K.W., J.L. Humston, A. Mehta, V. Tate, E. Ralston, and J.M. Ervasti. 2009.
Dystrophin is a microtubule-associated protein. J. Cell Biol. 186:363–
369. http://dx.doi.org/10.1083/jcb.200905048
Ralston, E. 1993. Changes in architecture of the Golgi complex and other subcel-
lular organelles during myogenesis. J. Cell Biol. 120:399–409. http://dx
.doi.org/10.1083/jcb.120.2.399
Ralston, E., Z. Lu, and T. Ploug. 1999. The organization of the Golgi complex
and microtubules in skeletal muscle is ber type-dependent. J. Neurosci.
19:10694–10705.
Ralston, E., T. Ploug, J. Kalhovde, and T. Lømo. 2001. Golgi complex, endoplas-
mic reticulum exit sites, and microtubules in skeletal muscle bers are or-
ganized by patterned activity. J. Neurosci. 21:875–883.
Ralston, E., Z. Lu, N. Biscocho, E. Soumaka, M. Mavroidis, C. Prats, T. Lømo,
Y. Capetanaki, and T. Ploug. 2006. Blood vessels and desmin control the
positioning of nuclei in skeletal muscle bers. J. Cell. Physiol. 209:874–
882. http://dx.doi.org/10.1002/jcp.20780
Randazzo, D., E. Giacomello, S. Lorenzini, D. Rossi, E. Pierantozzi, B. Blaauw,
C. Reggiani, S. Lange, A.K. Peter, J. Chen, and V. Sorrentino. 2013.
Obscurin is required for ankyrinB-dependent dystrophin localization and
sarcolemma integrity. J. Cell Biol. 200:523–536. http://dx.doi.org/10
.1083/jcb.201205118
Rivero, S., J. Cardenas, M. Bornens, and R.M. Rios. 2009. Microtubule
nucleation at the cis-side of the Golgi apparatus requires AKAP450
and GM130. EMBO J. 28:1016–1028. http://dx.doi.org/10.1038/
emboj.2009.47
Rosenblatt, J.D., A.I. Lunt, D.J. Parry, and T.A. Partridge. 1995. Culturing satel-
lite cells from living single muscle ber explants. In Vitro Cell. Dev. Biol.
Anim. 31:773–779. http://dx.doi.org/10.1007/BF02634119
Sato, H., T. Nagai, D. Kuppuswamy, T. Narishige, M. Koide, D.R. Menick,
and G. Cooper IV. 1997. Microtubule stabilization in pressure over-
load cardiac hypertrophy. J. Cell Biol. 139:963–973. http://dx.doi.org/
10.1083/jcb.139.4.963
Schertzer, J.D., and G.S. Lynch. 2008. Plasmid-based gene transfer in mouse
skeletal muscle by electroporation. Methods Mol. Biol. 433:115–125.
http://dx.doi.org/10.1007/978-1-59745-237-3_7
This work was supported by the Intramural Research Program of the
National Institute of Arthritis and Musculoskeletal and Skin Diseases.
Submitted: 9 April 2013
Accepted: 18 September 2013
References
Applegate, K.T., S. Besson, A. Matov, M.H. Bagonis, K. Jaqaman, and G.
Danuser. 2011. plusTipTracker: Quantitative image analysis software for
the measurement of microtubule dynamics. J. Struct. Biol. 176:168–184.
http://dx.doi.org/10.1016/j.jsb.2011.07.009
Ayalon, G., J.Q. Davis, P.B. Scotland, and V. Bennett. 2008. An ankyrin-based
mechanism for functional organization of dystrophin and dystroglycan.
Cell. 135:1189–1200. http://dx.doi.org/10.1016/j.cell.2008.10.018
Ayalon, G., J.D. Hostettler, J. Hoffman, K. Kizhatil, J.Q. Davis, and V. Bennett.
2011. Ankyrin-B interactions with spectrin and dynactin-4 are required
for dystrophin-based protection of skeletal muscle from exercise in-
jury. J. Biol. Chem. 286:7370–7378. http://dx.doi.org/10.1074/jbc
.M110.187831
Bekoff, A., and W.J. Betz. 1977. Physiological properties of dissociated muscle
bres obtained from innervated and denervated adult rat muscle. J. Physiol.
271:25–40.
Brangwynne, C.P., F.C. MacKintosh, S. Kumar, N.A. Geisse, J. Talbot,
L. Mahadevan, K.K. Parker, D.E. Ingber, and D.A. Weitz. 2006.
Microtubules can bear enhanced compressive loads in living cells because
of lateral reinforcement. J. Cell Biol. 173:733–741. http://dx.doi.org/
10.1083/jcb.200601060
Bugnard, E., K.J. Zaal, and E. Ralston. 2005. Reorganization of microtubule
nucleation during muscle differentiation. Cell Motil. Cytoskeleton. 60:1–
13. http://dx.doi.org/10.1002/cm.20042
Casey, L.M., H.D. Lyon, and J.B. Olmsted. 2003. Muscle-specic microtubule-
associated protein 4 is expressed early in myogenesis and is not sufcient
to induce microtubule reorganization. Cell Motil. Cytoskeleton. 54:317–
336. http://dx.doi.org/10.1002/cm.10105
Chen, J., Y. Kanai, N.J. Cowan, and N. Hirokawa. 1992. Projection domains of
MAP2 and tau determine spacings between microtubules in dendrites and
axons. Nature. 360:674–677. http://dx.doi.org/10.1038/360674a0
DiFranco, M., M. Quinonez, J. Capote, and J. Vergara. 2009. DNA transfection
of mammalian skeletal muscles using in vivo electroporation. J. Vis. Exp.
19:32.
Emov, A., A. Kharitonov, N. Emova, J. Loncarek, P.M. Miller, N. Andreyeva,
P. Gleeson, N. Galjart, A.R. Maia, I.X. McLeod, et al. 2007. Asymmetric
CLASP-dependent nucleation of noncentrosomal microtubules at the
trans-Golgi network. Dev. Cell. 12:917–930. http://dx.doi.org/10.1016/j
.devcel.2007.04.002
Elhanany-Tamir, H., Y.V. Yu, M. Shnayder, A. Jain, M. Welte, and T. Volk.
2012. Organelle positioning in muscles requires cooperation between two
KASH proteins and microtubules. J. Cell Biol. 198:833–846. http://dx
.doi.org/10.1083/jcb.201204102
Erhardt, M., V. Stoppin-Mellet, S. Campagne, J. Canaday, J. Mutterer, T. Fabian,
M. Sauter, T. Muller, C. Peter, A.M. Lambert, and A.C. Schmit. 2002. The
plant Spc98p homologue colocalizes with gamma-tubulin at microtubule
nucleation sites and is required for microtubule nucleation. J. Cell Sci.
115:2423–2431.
Faire, K., C.M. Waterman-Storer, D. Gruber, D. Masson, E.D. Salmon, and J.C.
Bulinski. 1999. E-MAP-115 (ensconsin) associates dynamically with micro-
tubules in vivo and is not a physiological modulator of microtubule dy-
namics. J. Cell Sci. 112:4243–4255.
Fourest-Lieuvin, A., J. Rendu, A. Osseni, K. Pernet-Gallay, D. Rossi, S. Oddoux,
J. Brocard, V. Sorrentino, I. Marty, and J. Fauré. 2012. Role of triadin in
the organization of reticulum membrane at the muscle triad. J. Cell Sci.
125:3443–3453. http://dx.doi.org/10.1242/jcs.100958
Fukuda, T., L. Ewan, M. Bauer, R.J. Mattaliano, K. Zaal, E. Ralston, P.H. Plotz,
and N. Raben. 2006. Dysfunction of endocytic and autophagic pathways
in a lysosomal storage disease. Ann. Neurol. 59:700–708. http://dx.doi
.org/10.1002/ana.20807
Gillingham, A.K., and S. Munro. 2000. The PACT domain, a conserved centro-
somal targeting motif in the coiled-coil proteins AKAP450 and pericen-
trin. EMBO Rep. 1:524–529.
Gundersen, G.G., S. Khawaja, and J.C. Bulinski. 1989. Generation of a stable,
posttranslationally modied microtubule array is an early event in myo-
genic differentiation. J. Cell Biol. 109:2275–2288. http://dx.doi.org/10
.1083/jcb.109.5.2275
Hurtado, L., C. Caballero, M.P. Gavilan, J. Cardenas, M. Bornens, and R.M.
Rios. 2011. Disconnecting the Golgi ribbon from the centrosome prevents
213Skeletal muscle microtubule dynamics • Oddoux et al.
Schertzer, J.D., D.R. Plant, and G.S. Lynch. 2006. Optimizing plasmid-based
gene transfer for investigating skeletal muscle structure and function.
Mol. Ther. 13:795–803. http://dx.doi.org/10.1016/j.ymthe.2005.09.019
Schmidt, N., S. Basu, S. Sladecek, S. Gatti, J. van Haren, S. Treves, J. Pielage, N.
Galjart, and H.R. Brenner. 2012. Agrin regulates CLASP2-mediated cap-
ture of microtubules at the neuromuscular junction synaptic membrane.
J. Cell Biol. 198:421–437. http://dx.doi.org/10.1083/jcb.201111130
Schulze, E., D.J. Asai, J.C. Bulinski, and M. Kirschner. 1987. Posttranslational
modication and microtubule stability. J. Cell Biol. 105:2167–2177.
http://dx.doi.org/10.1083/jcb.105.5.2167
Semiz, S., J.G. Park, S.M. Nicoloro, P. Furcinitti, C. Zhang, A. Chawla, J.
Leszyk, and M.P. Czech. 2003. Conventional kinesin KIF5B mediates in-
sulin-stimulated GLUT4 movements on microtubules. EMBO J. 22:2387–
2399. http://dx.doi.org/10.1093/emboj/cdg237
Shanks, R.A., B.T. Steadman, P.H. Schmidt, and J.R. Goldenring. 2002.
AKAP350 at the Golgi apparatus. I. Identication of a distinct Golgi ap-
paratus targeting motif in AKAP350. J. Biol. Chem. 277:40967–40972.
http://dx.doi.org/10.1074/jbc.M203307200
Srsen, V., X. Fant, R. Heald, C. Rabouille, and A. Merdes. 2009.
Centrosome proteins form an insoluble perinuclear matrix during
muscle cell differentiation. BMC Cell Biol. 10:28. http://dx.doi.org/
10.1186/1471-2121-10-28
Stepanova, T., J. Slemmer, C.C. Hoogenraad, G. Lansbergen, B. Dortland, C.I.
De Zeeuw, F. Grosveld, G. van Cappellen, A. Akhmanova, and N. Galjart.
2003. Visualization of microtubule growth in cultured neurons via the use
of EB3-GFP (end-binding protein 3-green uorescent protein). J. Neurosci.
23:2655–2664.
Takahashi, M., A. Yamagiwa, T. Nishimura, H. Mukai, and Y. Ono. 2002.
Centrosomal proteins CG-NAP and kendrin provide microtubule nucle-
ation sites by anchoring gamma-tubulin ring complex. Mol. Biol. Cell.
13:3235–3245. http://dx.doi.org/10.1091/mbc.E02-02-0112
Takahashi, M., H. Shiraishi, Y. Ishibashi, K.L. Blade, P.J. McDermott, D.R.
Menick, D. Kuppuswamy, and G. Cooper IV. 2003. Phenotypic conse-
quences of beta1-tubulin expression and MAP4 decoration of micro-
tubules in adult cardiocytes. Am. J. Physiol. Heart Circ. Physiol. 285:
H2072–H2083.
Tassin, A.M., B. Maro, and M. Bornens. 1985a. Fate of microtubule-organizing
centers during myogenesis in vitro. J. Cell Biol. 100:35–46. http://dx.doi
.org/10.1083/jcb.100.1.35
Tassin, A.M., M. Paintrand, E.G. Berger, and M. Bornens. 1985b. The Golgi
apparatus remains associated with microtubule organizing centers dur-
ing myogenesis. J. Cell Biol. 101:630–638. http://dx.doi.org/10.1083/
jcb.101.2.630
Vicidomini, G., A. Schönle, H. Ta, K.Y. Han, G. Moneron, C. Eggeling, and
S.W. Hell. 2013. STED nanoscopy with time-gated detection: theoretical
and experimental aspects. PLoS ONE. 8:e54421. http://dx.doi.org/
10.1371/journal.pone.0054421
Wilson, M.H., and E.L. Holzbaur. 2012. Opposing microtubule motors drive ro-
bust nuclear dynamics in developing muscle cells. J. Cell Sci. 125:4158–
4169. http://dx.doi.org/10.1242/jcs.108688
Zaal, K.J., C.L. Smith, R.S. Polishchuk, N. Altan, N.B. Cole, J. Ellenberg, K.
Hirschberg, J.F. Presley, T.H. Roberts, E. Siggia, et al. 1999. Golgi mem-
branes are absorbed into and reemerge from the ER during mitosis. Cell.
99:589–601. http://dx.doi.org/10.1016/S0092-8674(00)81548-2
Zaal, K.J., E. Reid, K. Mousavi, T. Zhang, A. Mehta, E. Bugnard, V. Sartorelli,
and E. Ralston. 2011. Who needs microtubules? Myogenic reorganization
of MTOC, Golgi complex and ER exit sites persists despite lack of nor-
mal microtubule tracks. PLoS ONE. 6:e29057. http://dx.doi.org/10.1371/
journal.pone.0029057
Zhu, X., and I. Kaverina. 2013. Golgi as an MTOC: making microtubules for its
own good. Histochem. Cell Biol. 140:361–367. http://dx.doi.org/10
.1007/s00418-013-1119-4
... In the myofiber, microtubules form a grid that contains longitudinal components that are parallel along the axis of the fiber, and perpendicular transverse components across the fiber. This organization of microtubules into lattice-like arrays is depicted in Figure 1a,b [1,2]. Microtubule assembly nucleates at microtubule-organizing centers (MTOCs) at the microtubule minus-end. ...
... The presence of transverse microtubules, intersecting with longitudinal microtubules, suggests an additional nucleation center beyond the well-defined MTOC that surrounds the nuclear envelope. Golgi bodies within myofibers are located along the intersections of transverse and longitudinal microtubules [1], as well as along stabilized microtubules [42]. Additionally, Golgi bodies wrap around the nucleus and associate with gamma-tubulin and pericentrin, both of which are components of the MTOC. ...
... Additionally, Golgi bodies wrap around the nucleus and associate with gamma-tubulin and pericentrin, both of which are components of the MTOC. Thus, it is not surprising that Golgi bodies have been shown to serve as additional microtubule nucleation sites in myofibers [1]. ...
Article
Full-text available
The contractile cells of skeletal muscles, called myofibers, are elongated multinucleated syncytia formed and maintained by the fusion of proliferative myoblasts. Human myofibers can be hundreds of microns in diameter and millimeters in length. Myofibers are non-mitotic, obviating the need for microtubules in cell division. However, microtubules have been adapted to the unique needs of these cells and are critical for myofiber development and function. Microtubules in mature myofibers are highly dynamic, and studies in several experimental systems have demonstrated the requirements for microtubules in the unique features of muscle biology including myoblast fusion, peripheral localization of nuclei, assembly of the sarcomere, transport and signaling. Microtubule-binding proteins have also been adapted to the needs of the skeletal muscle including the expression of skeletal muscle-specific protein isoforms generated by alternative splicing. Here, we will outline the different roles microtubules play in skeletal muscle cells, describe how microtubule abnormalities can lead to muscle disease and discuss the broader implications for microtubule function.
... Unique to striated muscle and neurons, centrosomal proteins are recruited to the NE by association with AKAP6, which together with AKAP9, are anchored by nesprin-1α2 to form an NE-MTOC [52,53]. Centrosomal proteins and MT nucleation are additionally observed at the Golgi apparatus which is uniquely distributed around the nucleus of striated muscle cells, yet the integration and contribution NE-MTOC activity at the Golgi is poorly understood [54][55][56]. ...
Article
Full-text available
Nesprins (nuclear envelope spectrin repeat proteins) are multi-isomeric scaffolding proteins. Giant nesprin-1 and -2 localise to the outer nuclear membrane, interact with SUN (Sad1p/UNC-84) domain-containing proteins at the inner nuclear membrane to form the LInker of Nucleoskeleton and Cytoskeleton (LINC) complex, which, in association with lamin A/C and emerin, mechanically couples the nucleus to the cytoskeleton. Despite ubiquitous expression of nesprin giant isoforms, pathogenic mutations in nesprin-1 and -2 are associated with tissue-specific disorders, particularly related to striated muscle such as dilated cardiomyopathy and Emery-Dreifuss muscular dystrophy. Recent evidence suggests this muscle-specificity might be attributable in part, to the small muscle specific isoform, nesprin-1α2, which has a novel role in striated muscle function. Our current understanding of muscle-specific functions of nesprin-1 and its isoforms will be summarised in this review to provide insight into potential pathological mechanisms of nesprin-related muscle disease and may inform potential targets of therapeutic modulation.
... 79,80 Apart from the main Golgi apparatus surrounding the nucleus, additional small Golgi elements are scattered around the cell, which also act as MT-organizing centres. 81 Hence, the MT-organizing centres at the Golgi apparatus and Golgi elements allow distribution of proteins across the cell via MTs. The MT minus-end is relatively static, and MT dynamic behaviour is therefore largely restricted to the other end of the MT: the MT plus-end. ...
Article
Full-text available
The cardiac sodium channel NaV1.5 is an essential modulator of cardiac excitability, with decreased NaV1.5 levels at the plasma membrane and consequent reduction in sodium current (INa) leading to potentially lethal cardiac arrhythmias. NaV1.5 is distributed in a specific pattern at the plasma membrane of cardiomyocytes, with localization at the crests, grooves, and T-tubules of the lateral membrane, and particularly high levels at the intercalated disc region. NaV1.5 forms a large macromolecular complex with and is regulated by interacting proteins, some of which are specifically localised at either the lateral membrane or intercalated disc. One of the NaV1.5 trafficking routes is via microtubules (MTs), which are regulated by MT plus-end tracking proteins (+TIPs). In our search for mechanisms involved in targeted delivery of NaV1.5, we here provide an overview of previously demonstrated interactions between NaV1.5 interacting proteins and +TIPs, which potentially (in)directly impact on NaV1.5 trafficking. Strikingly, +TIPs interact extensively with several intercalated disc- and lateral membrane-specific NaV1.5 interacting proteins. Recent work indicates that this interplay of +TIPs and NaV1.5 interacting proteins mediates the targeted delivery of NaV1.5 at specific cardiomyocyte subcellular domains, while also being potentially relevant for the trafficking of other ion channels. These observations are especially relevant for diseases associated with loss of NaV1.5 specifically at the lateral membrane (such as Duchenne muscular dystrophy), or at the intercalated disc (for example, arrhythmogenic cardiomyopathy), and open up potential avenues for development of new anti-arrhythmic therapies.
... Unique to striated muscle and neurons, centrosomal proteins are recruited to the NE by association with AKAP6, which together with AKAP9, are anchored by nesprin-1α2 to form an NE-MTOC [52,53]. Centrosomal proteins and MT nucleation are additionally observed at the Golgi apparatus which is uniquely distributed around the nucleus of striated muscle cells, yet the integration and contribution NE-MTOC activity at the Golgi is poorly understood [54][55][56]. ...
... However, TubA treatment did not affect the overall α-tubulin abundance (P > 0.05 compared with mdx-veh mice; Fig. 1d). In healthy muscle, the microtubule network forms a grid lattice with longitudinal, transverse, and perinuclear microtubules [78][79][80] . Here, in WT-CTL mice, we observed that transverse and longitudinal microtubules are regularly spaced by ∼2 µm and ∼5 µm, respectively (see arrowheads Fig. 3a, and Supplementary Fig. 3c-e). ...
Article
Full-text available
The absence of dystrophin in Duchenne muscular dystrophy disrupts the dystrophin-associated glycoprotein complex resulting in skeletal muscle fiber fragility and atrophy, associated with fibrosis as well as microtubule and neuromuscular junction disorganization. The specific, non-conventional cytoplasmic histone deacetylase 6 (HDAC6) was recently shown to regulate acetylcholine receptor distribution and muscle atrophy. Here, we report that administration of the HDAC6 selective inhibitor tubastatin A to the Duchenne muscular dystrophy, mdx mouse model increases muscle strength, improves microtubule, neuromuscular junction, and dystrophin-associated glycoprotein complex organization, and reduces muscle atrophy and fibrosis. Interestingly, we found that the beneficial effects of HDAC6 inhibition involve the downregulation of transforming growth factor beta signaling. By increasing Smad3 acetylation in the cytoplasm, HDAC6 inhibition reduces Smad2/3 phosphorylation, nuclear translocation, and transcriptional activity. These findings provide in vivo evidence that Smad3 is a new target of HDAC6 and implicate HDAC6 as a potential therapeutic target in Duchenne muscular dystrophy. Here, authors show that Smad3 acetylation via HDAC6 inhibition reverses Duchenne muscular dystrophy-like symptoms in the mdx mouse model, suggesting a potential therapeutic target for the disorder.
... Desmin depletion, on the other hand, virtually eliminated the typical Z-disk bias for pauses, rescues, or fewer catastrophes. Initiations had a strong Z-disk bias regardless of intervention, which likely reflects nucleating events from microtubule organizing centers at Golgi outposts proximal to the Z-disk that are not affected by these manipulations [26]. ...
Article
Full-text available
In heart failure, an increased abundance of post-translationally detyrosinated microtubules stiffens the cardiomyocyte and impedes its contractile function. Detyrosination promotes interactions between microtubules, desmin intermediate filaments, and the sarcomere to increase cytoskeletal stiffness, yet the mechanism by which this occurs is unknown. We hypothesized that detyrosination may regulate the growth and shrinkage of dynamic microtubules to facilitate interactions with desmin and the sarcomere. Through a combination of biochemical assays and direct observation of growing microtubule plus-ends in adult cardiomyocytes, we find that desmin is required to stabilize growing microtubules at the level of the sarcomere Z-disk, where desmin also rescues shrinking microtubules from continued depolymerization. Further, reducing detyrosination (i.e. tyrosination) below basal levels promotes frequent depolymerization and less efficient growth of microtubules. This is concomitant with tyrosination promoting the interaction of microtubules with the depolymerizing protein complex of end-binding protein 1 (EB1) and CAP-Gly domain-containing linker protein 1 (CLIP1/CLIP170). The dynamic growth and shrinkage of tyrosinated microtubules reduce their opportunity for stabilizing interactions at the Z-disk region, coincident with tyrosination globally reducing microtubule stability. These data provide a model for how intermediate filaments and tubulin detyrosination establish long-lived and physically reinforced microtubules that stiffen the cardiomyocyte and inform both the mechanism of action and therapeutic index for strategies aimed at restoring tyrosination for the treatment of cardiac disease.
Article
Full-text available
This article critically evaluates the multifunctional role of the Golgi apparatus within neurological paradigms. We succinctly highlight its influence on neuronal plasticity, development, and the vital trafficking and sorting mechanisms for proteins and lipids. The discourse further navigates to its regulatory prominence in neurogenesis and its implications in Alzheimer's Disease pathogenesis. The emerging nexus between the Golgi apparatus and SARS-CoV-2 underscores its potential in viral replication processes. This consolidation accentuates the Golgi apparatus's centrality in neurobiology and its intersections with both neurodegenerative and viral pathologies. In essence, understanding the Golgi's multifaceted functions harbors profound implications for future therapeutic innovations in neurological and viral afflictions.
Article
Full-text available
Microtubules serve as tracks for long-range intracellular trafficking of glucose transporter 4 (GLUT4), but the role of this process in skeletal muscle and insulin resistance is unclear. Here, we used fixed and live-cell imaging to study microtubule-based GLUT4 trafficking in human and mouse muscle fibers and L6 rat muscle cells. We found GLUT4 localized on the microtubules in mouse and human muscle fibers. Pharmacological microtubule disruption using Nocodazole (Noco) prevented long-range GLUT4 trafficking and depleted GLUT4-enriched structures at microtubule nucleation sites in a fully reversible manner. Using a perifused muscle-on-a-chip system to enable real-time glucose uptake measurements in isolated mouse skeletal muscle fibers, we observed that Noco maximally disrupted the microtubule network after 5 min without affecting insulin-stimulated glucose uptake. In contrast, a 2h Noco treatment markedly decreased insulin responsiveness of glucose uptake. Insulin resistance in mouse muscle fibers induced either in vitro by C2 ceramides or in vivo by diet-induced obesity, impaired microtubule-based GLUT4 trafficking. Transient knockdown of the microtubule motor protein kinesin-1 protein KIF5B in L6 muscle cells reduced insulin-stimulated GLUT4 translocation while pharmacological kinesin-1 inhibition in incubated mouse muscles strongly impaired insulin-stimulated glucose uptake. Thus, in adult skeletal muscle fibers, the microtubule network is essential for intramyocellular GLUT4 movement, likely functioning to maintain an insulin-responsive cell-surface recruitable GLUT4 pool via kinesin-1 mediated trafficking.
Article
The synchronized distribution of centrosomal and genetic materials to the dividing daughter cells is critically important and depends on precisely orchestrated processes on structural and molecular levels. Structural and functional relationships between the nucleus and centrosomes facilitate cellular communication and coordination of cell cycle control and progression which becomes especially important during the transition from interphase to mitosis when synchrony between centrosomes and nuclear events is critical.
Chapter
Microtubules are stiff yet dynamic intracellular polymers. Differentiated cells leverage these properties, using microtubules to facilitate transport, polarization, and force generation. Accordingly, different cell types establish unique and distinct microtubule organization using a variety of microtubule organizing centers (MTOCs) to enable diverse cell functions. This article will review general properties of microtubules and the subcellular structures they build and highlight the diversity and function of cell-scale microtubule networks across differentiated cells.
Article
Full-text available
Several microtubule binding proteins, including CLIP-170 (cytoplasmic linker protein-170), CLIP-115, and EB1 (end-binding protein 1), have been shown to associate specifically with the ends of growing microtubules in non-neuronal cells, thereby regulating microtubule dynamics and the binding of microtubules to protein complexes, organelles, and membranes. When fused to GFP (green fluorescent protein), these proteins, which collectively are called +TIPs (plus end tracking proteins), also serve as powerful markers for visualizing microtubule growth events. Here we demonstrate that endogenous +TIPs are present at distal ends of microtubules in fixed neurons. Using EB3-GFP as a marker of microtubule growth in live cells, we subsequently analyze microtubule dynamics in neurons. Our results indicate that microtubules grow slower in neurons than in glia and COS-1 cells. The average speed and length of EB3-GFP movements are comparable in cell bodies, dendrites, axons, and growth cones. In the proximal region of differentiated dendrites ∼65% of EB3-GFP movements are directed toward the distal end, whereas 35% are directed toward the cell body. In more distal dendritic regions and in axons most EB3-GFP dots move toward the growth cone. This difference in directionality of EB3-GFP movements in dendrites and axons reflects the highly specific microtubule organization in neurons. Together, these results suggest that local microtubule polymerization contributes to the formation of the microtubule network in all neuronal compartments. We propose that similar mechanisms underlie the specific association of CLIPs and EB1-related proteins with the ends of growing microtubules in non-neuronal and neuronal cells.
Article
Full-text available
We have probed the relationship between tubulin posttranslational modification and microtubule stability, using a variation of the antibody-blocking technique. In human retinoblastoma cells we find that acetylated and detyrosinated microtubules represent congruent subsets of the cells' total microtubules. We also find that stable microtubules defined as those that had not undergone polymerization within 1 h after injection of biotin-tubulin were all posttranslationally modified; furthermore dynamic microtubules were all unmodified. We therefore conclude that in these cells the stable, acetylated, and detyrosinated microtubules represent the same subset of the cells' total network. Posttranslational modification, however, is not a prerequisite for microtubule stability and vice versa. Potorous tridactylis kidney cells have no detectable acetylated microtubules but do have a sizable subset of stable ones, and chick embryo fibroblast cells are extensively modified but have few stable microtubules. We conclude that different cell types can create specific microtubule subsets by modulating the relative rates of posttranslational modification and microtubule turnover.
Article
Full-text available
Obscurin is a large myofibrillar protein that contains several interacting modules, one of which mediates binding to muscle-specific ankyrins. Interaction between obscurin and the muscle-specific ankyrin sAnk1.5 regulates the organization of the sarcoplasmic reticulum in striated muscles. Additional muscle-specific ankyrin isoforms, ankB and ankG, are localized at the subsarcolemma level, at which they contribute to the organization of dystrophin and β-dystroglycan at costameres. In this paper, we report that in mice deficient for obscurin, ankB was displaced from its localization at the M band, whereas localization of ankG at the Z disk was not affected. In obscurin KO mice, localization at costameres of dystrophin, but not of β-dystroglycan, was altered, and the subsarcolemma microtubule cytoskeleton was disrupted. In addition, these mutant mice displayed marked sarcolemmal fragility and reduced muscle exercise tolerance. Altogether, the results support a model in which obscurin, by targeting ankB at the M band, contributes to the organization of subsarcolemma microtubules, localization of dystrophin at costameres, and maintenance of sarcolemmal integrity.
Article
Full-text available
In a stimulated emission depletion (STED) microscope the region in which fluorescence markers can emit spontaneously shrinks with continued STED beam action after a singular excitation event. This fact has been recently used to substantially improve the effective spatial resolution in STED nanoscopy using time-gated detection, pulsed excitation and continuous wave (CW) STED beams. We present a theoretical framework and experimental data that characterize the time evolution of the effective point-spread-function of a STED microscope and illustrate the physical basis, the benefits, and the limitations of time-gated detection both for CW and pulsed STED lasers. While gating hardly improves the effective resolution in the all-pulsed modality, in the CW-STED modality gating strongly suppresses low spatial frequencies in the image. Gated CW-STED nanoscopy is in essence limited (only) by the reduction of the signal that is associated with gating. Time-gated detection also reduces/suppresses the influence of local variations of the fluorescence lifetime on STED microscopy resolution.
Article
Full-text available
Striated muscle fibers are characterized by their tightly organized cytoplasm. Here, we show that the Drosophila melanogaster KASH proteins Klarsicht (Klar) and MSP-300 cooperate in promoting even myonuclear spacing by mediating a tight link between a newly discovered MSP-300 nuclear ring and a polarized network of astral microtubules (aMTs). In either klar or msp-300(ΔKASH), or in klar and msp-300 double heterozygous mutants, the MSP-300 nuclear ring and the aMTs retracted from the nuclear envelope, abrogating this even nuclear spacing. Anchoring of the myonuclei to the core acto-myosin fibrillar compartment was mediated exclusively by MSP-300. This protein was also essential for promoting even distribution of the mitochondria and ER within the muscle fiber. Larval locomotion is impaired in both msp-300 and klar mutants, and the klar mutants were rescued by muscle-specific expression of Klar. Thus, our results describe a novel mechanism of nuclear spacing in striated muscles controlled by the cooperative activity of MSP-300, Klar, and astral MTs, and demonstrate its physiological significance.
Article
Microtubules (MTs) have been implicated to function in the change of cell shape and intracellular organization that occurs during myogenesis. However, the mechanism by which MTs are involved in these morphogenetic events is unclear. As a first step in elucidating the role of MTs in myogenesis, we have examined the accumulation and subcellular distribution of posttranslationally modified forms of tubulin in differentiating rat L6 muscle cells, using antibodies specific for tyrosinated (Tyr), detyrosinated (Glu), and acetylated (Ac) tubulin. Both Glu and Ac tubulin are components of stable MTs, whereas Tyr tubulin is the predominant constituent of dynamic MTs. In proliferating L6 myoblasts, as in other types of proliferating cells, the level of Glu tubulin was very low when compared with the level of Tyr tubulin. However, when we shifted proliferating L6 cells to differentiation media, we observed a rapid accumulation of Glu tubulin in cellular MTs. By immunofluorescence, the increase in Glu tubulin was first detected in MTs of prefusion myoblasts and was specifically localized to MTs that were associated with elongating portions of the cell. MTs in the multinucleated myotubes observed at later stages of differentiation maintained the elevated level of Glu tubulin that was observed in the prefusion myoblasts. When cells at early stages of differentiation (less than 1 d after switching the culture medium) were immunostained for Glu tubulin and the muscle-specific marker, muscle myosin, we found that the increase in Glu tubulin preceded the accumulation of muscle myosin. Thus, the elaboration of Glu MTs is one of the very early events in myogenesis. Ac tubulin also increased during L6 myogenesis; however, the increase in acetylation occurred later in myogenesis, after fusion had already occurred. Because detyrosination was temporally correlated with early events of myogenesis, we examined the mechanism responsible for the accumulation of Glu tubulin in the MTs of prefusion myoblasts. We found that an increase in the stability of L6 cell MTs occurred at the onset of differentiation, suggesting that the early increase in detyrosination that we observed is a manifestation of a decrease in MT dynamics in elongating myoblasts. We conclude that the establishment of an oriented array of microtubules heightened in its stability and its level of posttranslationally modified subunits may be involved in the subcellular remodeling that occurs during myogenesis.
Article
In vitro myogenesis involves a dramatic reorganization of the microtubular network, characterized principally by the relocalization of microtubule nucleating sites at the surface of the nuclei in myotubes, in marked contrast with the classical pericentriolar localization observed in myoblasts (Tassin, A. M., B. Maro, and M. Bornens, 1985, J. Cell Biol., 100:35-46). Since a spatial relationship between the Golgi apparatus and the centrosome is observed in most animal cells, we have decided to follow the fate of the Golgi apparatus during myogenesis by an immunocytochemical approach, using wheat germ agglutinin and an affinity-purified anti-galactosyltransferase. We show that Golgi apparatus in myotubes displays a perinuclear distribution which is strikingly different from the polarized juxtanuclear organization observed in myoblasts. As a result, the Golgi apparatus in myotubes is situated close to the microtubule organizing center (MTOC), the cis-side being situated at a fixed distance from the nuclear envelope, a situation which suggests the existence of a structural association between the Golgi apparatus and the nuclear periphery. This is supported by experiments of microtubule depolymerization by nocodazole, in which a minimal effect was observed on Golgi apparatus localization in myotubes in contrast with the dramatic scattering observed in myoblasts. In both cell types, electron microscopy reveals that microtubule disruption generates individual dictyosomes; this suggests that the connecting structures between dictyosomes are principally affected. This structural dependency of the Golgi apparatus upon microtubules is not apparently accompanied by a reverse dependency of MTOC structure or function upon Golgi apparatus activity. Golgi apparatus modification by monensin, as effective in myotubes as in myoblasts, is without apparent effect on MTOC localization or activity and on microtubule stability. The main result of our study is to show that in a cell type where the MTOC is dissociated from centrioles and where antero-posterior polarity has disappeared, the association between the Golgi apparatus and the MTOC is maintained. The significance of such a tight association is discussed.
Article
We have probed the relationship between tubulin posttranslational modification and microtubule stability, using a variation of the antibody-blocking technique. In human retinoblastoma cells we find that acetylated and detyrosinated microtubules represent congruent subsets of the cells' total microtubules. We also find that stable microtubules defined as those that had not undergone polymerization within 1 h after injection of biotin-tubulin were all posttranslationally modified; furthermore dynamic microtubules were all unmodified. We therefore conclude that in these cells the stable, acetylated, and detyrosinated microtubules represent the same subset of the cells' total network. Posttranslational modification, however, is not a prerequisite for microtubule stability and vice versa. Potorous tridactylis kidney cells have no detectable acetylated microtubules but do have a sizable subset of stable ones, and chick embryo fibroblast cells are extensively modified but have few stable microtubules. We conclude that different cell types can create specific microtubule subsets by modulating the relative rates of posttranslational modification and microtubule turnover.
Article
In cells, microtubules (MTs) are nucleated at MT-organizing centers (MTOCs). The centrosome-based MTOCs organize radial MT arrays, which are often not optimal for polarized trafficking. A recently discovered subset of non-centrosomal MTs nucleated at the Golgi has proven to be indispensable for the Golgi organization, post-Golgi trafficking and cell polarity. Here, we summarize the history of this discovery, known molecular prerequisites of MT nucleation at the Golgi and unique functions of Golgi-derived MTs.
Article
Microtubule nucleation is essential for proper establishment and maintenance of axons and dendrites. Centrosomes, the primary site of nucleation in most cells, lose their function as microtubule organizing centers during neuronal development. How neurons generate acentrosomal microtubules remains unclear. Drosophila dendritic arborization (da) neurons lack centrosomes and therefore provide a model system to study acentrosomal microtubule nucleation. Here, we investigate the origin of microtubules within the elaborate dendritic arbor of class IV da neurons. Using a combination of in vivo and in vitro techniques, we find that Golgi outposts can directly nucleate microtubules throughout the arbor. This acentrosomal nucleation requires gamma-tubulin and CP309, the Drosophila homolog of AKAP450, and contributes to the complex microtubule organization within the arbor and dendrite branch growth and stability. Together, these results identify a direct mechanism for acentrosomal microtubule nucleation within neurons and reveal a function for Golgi outposts in this process.