You are on page 1of 11

Available online at

ScienceDirect
www.sciencedirect.com

Diabetes & Metabolism 40 (2014) 400–410

Review

Molecular mechanisms of GLUT4 regulation in adipocytes


Mécanismes moléculaires de la régulation de GLUT4 dans l’adipocyte
R. Govers ∗
UMR Inserm U1062, INRA 1260, Aix Marseille University, Campus Timone, Faculty of Medicine, 27, boulevard Jean-Moulin, 13385 Marseille Cedex 5, France
Received 9 December 2013; received in revised form 24 January 2014; accepted 26 January 2014
Available online 20 March 2014

Abstract
Insulin resistance is strongly linked to type 2 diabetes and associated with a reduced uptake of glucose by muscle and adipose tissue. The
transporter that is responsible for this uptake and whose function is disturbed in insulin resistance and type 2 diabetes is GLUT4. In the non-
stimulated state, GLUT4 is efficiently sequestered intracellularly. This retention prevents GLUT4 from reaching the cell surface and transporting
glucose into muscle and fat cells when blood glucose levels are low. After a meal when blood glucose levels rise, insulin is secreted by the
pancreas, which, upon binding to its receptor, triggers an intracellular signaling cascade, leading to the translocation of GLUT4 from intracellular
compartments to the cell surface, resulting in glucose uptake and normalization of the blood glucose levels. Its regulation is dominated by its
localization, efficient intracellular retention and sensitivity to insulin and contraction, which makes GLUT4 an interesting and unique molecule.
These aspects of the intracellular regulation of GLUT4 are described in this review.
© 2014 Elsevier Masson SAS. All rights reserved.

Keywords: GLUT4; Intracellular traffic; Translocation; Regulated exocytosis; Adipocyte

1. GLUT4 in the context of insulin resistance and type 2 GLUT4 is one of the 14 members of the GLUT/SLC2A fam-
diabetes ily of facilitative transmembrane hexose transporters. In addition
to the well-described expression of GLUT4 in skeletal mus-
In healthy individuals, despite alternating periods of fasting cle, cardiomyocytes, and adipose tissue, this transporter is also
and feeding, plasma glucose levels are mostly remarkably well expressed in certain regions of the brain [1], in kidney [2], and
maintained within a narrow range due to the action of the hor- in several tumors, including multiple myeloma [3] and breast
mones insulin and glucagon. In contrast, type 2 diabetes (T2D) cancer cells [4]. The cellular regulation of GLUT4 has been
patients suffer from hyperglycemia that is caused by insulin best studied in muscle and adipose tissue and has appeared to
resistance in combination with insulin levels that are too low to be largely similar in these two tissues. Nevertheless, several
compensate for this resistance. In insulin resistance, the tissues minor and major differences exist. In addition to the contraction-
that are implicated in glucose homeostasis and that are sensitive induced GLUT4-mediated glucose uptake that is specific for
to insulin (i.e. liver, muscle and adipose tissue), no longer effi- muscle, there also exists a tissue-specific regulation of GLUT4
ciently respond to insulin. Hence, at normal insulin levels, the expression. This is evident for example in T2D, where there is
liver continues to synthesize and release glucose, while mus- a reduction in GLUT4 expression (at both mRNA and protein
cle and adipose tissue take up less glucose from the blood. It levels) in adipocytes but not in muscle cells [5]. This may not
is widely accepted that this uptake of glucose is mediated by seem to matter much as in healthy individuals only 15% of the
the insulin-sensitive transporter GLUcose Transporter member blood glucose is absorbed by adipose tissue and the remaining
4 (GLUT4). In muscle, this transporter also takes up glucose 85% by muscle. However, in adipose tissue, GLUT4 function
upon muscle contraction. appears to be extremely important and to play a pivotal role in
glucose-sensing, as its expression level in this tissue is positively

correlated with insulin-sensitivity in muscle and liver, mainly via
Tel.: +33 4 91 29 40 96; fax: +33 4 91 78 21 01.
E-mail address: Roland.Govers@univ-amu.fr

1262-3636/$ – see front matter © 2014 Elsevier Masson SAS. All rights reserved.
http://dx.doi.org/10.1016/j.diabet.2014.01.005
R. Govers / Diabetes & Metabolism 40 (2014) 400–410 401

Fig. 1. GLUT4 translocation in response to insulin stimulation. 3T3-L1 adipocytes were incubated for 20 minutes in the absence (“basal” state, left panel) or presence
of 100 nM insulin (“insulin”, right panel). Cells were then immunolabeled for GLUT4 and analyzed by immunofluorescence microscopy. Of note is the predominant
intracellular (perinuclear) GLUT4 localization in basal adipocytes and cell surface GLUT4 localization in insulin-stimulated cells. Bar: 10 ␮m.

the GLUT4 activity-dependent secretion of adipokines that act cells can be efficiently differentiated into adipocytes in vitro,
on these two tissues [6,7]. express GLUT4 upon differentiation and respond well to insulin
[17].
2. Intracellular localization of GLUT4
2.1. GLUT4 storage vesicles (GSVs)
The intracellular localization of GLUT4 plays a crucial
role in its regulation [8–10] and is disturbed in T2D [11–13]. GSVs are currently widely accepted to be non-endosomal
In non-stimulated (basal) cells, GLUT4 is efficiently retained insulin-sensitive GLUT4-containing vesicles that are relatively
intracellularly, resulting in low cell surface GLUT4 levels, small in size (50–70 nm) [18], smaller than for example endo-
while insulin reduces this retention, thereby largely increas- somal vesicles [19], and sensitive to insulin stimulation and
ing the amount of GLUT4 at the cell surface (Fig. 1). How contraction. GSVs are considered to contain up to half of the
GLUT4 is retained is still largely unclear. GLUT4 may be total cellular GLUT4 pool as ∼ 50% of GLUT4 is localized in
selectively retained in one or several of its intracellular loca- a non-endosomal cell fraction [15,20]. Proteins that are consid-
tions, packaged into a specific insulin-sensitive compartment ered to be enriched, but not uniquely expressed, in GSVs include
that remains static in the absence of insulin, or trafficking within GLUT4, insulin-regulated aminopeptidase (IRAP) [21], sortilin
a dynamic intracellular transport loop that excludes it from (a transmembrane protein involved in the exit of proteins from
recycling endosomal vesicles. These three different possibili- the TGN) [22], low-density lipoprotein receptor-related protein
ties are not mutually exclusive and various pieces of evidence 1 (LRP1, implicated in many physiological processes) [23], and
exist that support all three scenarios. For decades, researchers VAMP2 [24].
have been investigating the nature of the cellular compartments The biogenesis of GSVs appears to be cell-specific as the
that are implicated in intracellular GLUT4 retention and that expression of GLUT4 in fibroblasts (cells that normally do not
are sensitive to insulin stimulation (in adipocytes and mus- express GLUT4) leads to the localization of GLUT4 uniquely
cle) and contraction (in muscle). These compartments have in endosomal structures and not in GSVs [25]. It has been sug-
been collectively termed GLUT4 storage compartments (GSCs) gested that the formation of GSVs or the traffic of GLUT4
and insulin-responsive vesicles (IRVs). More specifically, the into GSVs is primarily induced by the interaction between
non-endosomal vesicular structures that are particularly sen- transmembrane protein sortilin and GLUT4 and that IRAP
sitive to insulin have been termed GLUT4 storage vesicles may be facilitative in this process. Accordingly, the absence
(GSVs) and specialized GLUT4 vesicles [14–16]. In addition of GSVs in fibroblasts is correlated with the absence of sor-
to its storage compartments, GLUT4 is also present in several tilin. Moreover, the ectopic coexpression of sortilin with GLUT4
other cell compartments. These include the plasma membrane, in these cells leads to a shift in the localization of GLUT4
where GLUT4 is transporting glucose, and organelles impli- from larger structures into small vesicles (presumably GSVs
cated in the targeting of GLUT4 into its storage compartments or GSV-like vesicles), stabilization of GLUT4 (preventing its
(Fig. 2). In the following sections, each of these compartments rapid degradation), and an enhancement of insulin-induced
is described in detail as well as the molecular elements that GLUT4 translocation [22,26]. In contrast, the knockdown of
regulate GLUT4 in these compartments of which some are sen- sortilin in adipocytes decreases the amount of GLUT4 in small
sitive to insulin stimulation. Note that most studies described in vesicles (likely to be GSVs), reduces GLUT4 stability, and
this review are based on the use of the 3T3-L1 cell line. These reduces insulin-induced GLUT4 translocation [22]. Intriguingly,
402 R. Govers / Diabetes & Metabolism 40 (2014) 400–410

Fig. 2. Model for intracellular GLUT4 traffic. In non-stimulated adipocytes (left panel), GLUT4 (depicted as triangles) is largely excluded from the plasma membrane
and is predominantly present and retained within GSVs and (subdomains of) recycling endosomes. The TGN generates the GSVs and together with the GSVs and
recycling endosomes, forms an intracellular trafficking loop that contributes to GLUT4 retention. Upon insulin stimulation (right panel), GSVs fuse with the plasma
membrane while also GLUT4 retention in recycling endosomes is reduced, leading to the redistribution of GLUT4 to the plasma membrane. As long as the insulin
stimulus is present, the recruited GLUT4 molecules continue to recycle between the plasma membrane and endosomes, without being sorted into new GSVs. The
thickness of each arrow reflects the relative contribution of the pathway to the intracellular GLUT4 traffic. For reasons of simplicity, the GLUT4 molecules present in
the biosynthetic pathway (up to Golgi stacks) have been omitted. PM: plasma membrane; EE: early (sorting) endosome; RE: recycling endosome; TGN: trans-Golgi
network; GSVs: GLUT4 storage vesicles.

in insulin resistance and diabetes, sortilin levels are reduced ment and their budding into GSVs. Sortilin could be implicated
[27,28]. This may imply a role for sortilin in the associated in this process which is in line with its role in the segrega-
impairment in GLUT4 translocation. How sortilin is involved tion of regulated from constitutive secretory proteins at the
in the formation of GSVs remains uncertain, but it is possible TGN [32].
that an interaction between the lumenal domains of sortilin and In the basal state, GSVs are largely refrained from fusion with
GLUT4 plays a role [29]. IRAP has also been suggested to the plasma membrane. What is preventing this fusion is unclear
be implicated in the sorting of GLUT4 into GSVs or in the at present though various scenarios have been proposed, which
biogenesis of GSVs (thereby contributing to GLUT4 reten- are not necessarily mutually exclusive. Possibly, in the absence
tion) as IRAP knockdown in vitro induces a redistribution of insulin stimulation, the GSVs, which may be mobile and to
of GLUT4 towards endosomes (that serve as an alternative some extent traffic close to the plasma membrane, do not have
trafficking route when the traffic of GLUT4 to GSVs is dis- the proper signals that allow them to fuse with the plasma mem-
rupted [30]), concomitant with a modest increase in cell surface brane. Also, in the basal state, the plasma membrane itself is not
GLUT4 levels [21]. IRAP ablation in vivo has been described “activated” and hence incapable of receiving incoming GSVs
to decrease GLUT4 expression in muscle and adipose tissue [33]. Alternatively, a large part of the GSVs may be attached
[31]. This supports the idea that IRAP facilitates the sorting of to an immobile intracellular matrix that renders them relatively
GLUT4 into GSVs as the absence of GLUT4 from GSVs is static in the absence of insulin. Various proteins have been sug-
known to reduce the stability of the GLUT4 molecule and to gested to be implicated in this form of retention. One is called
decrease GLUT4 expression levels [22]. Possibly, IRAP facil- TUG (a putative Tether, containing a UBX domain, for GLUT4),
itates the targeting of GLUT4 into GSVs by means of the identified by a functional genetic screen [34]. TUG (reviewed
interaction between their lumenal domains [26]. Apparently, the in detail in [35]) was found to interact directly with GLUT4
various GSV proteins (including LRP-1 [23]) may be present uniquely in the absence of insulin and has been proposed to
in (large) multimeric protein complexes. Knockdown of sev- tether GLUT4 vesicles intracellularly. Insulin would release this
eral of these proteins individually leads to a more pronounced tether, possibly via an intramolecular cleavage within TUG [36]
degradation of the other GSV components. Possibly, the GSV (see also Section 2.5). Given the presence of TUG in an early
components become complexed in the compartment from which (pre-TGN) Golgi compartment where it is involved in Golgi
the GSVs are formed (likely the TGN; Fig. 2), subsequently organization [37] and its interaction with Golgin-160, known
triggering the formation of subdomains within this compart- to play a role in intracellular GLUT4 traffic [36,38], TUG may
R. Govers / Diabetes & Metabolism 40 (2014) 400–410 403

sequester GSVs by linking them to the Golgi matrix. Alterna- findings is that a single insulin stimulation of cardiac myocytes
tively, the data on TUG are consistent with a role for TUG in reduces GLUT4 content uniquely in non-endosomal structures,
the targeting of GLUT4 to GSVs [39]. In line with both pos- while repeated insulin stimulations reduce GLUT4 content
sible roles of TUG are the findings that disruption of cellular in both non-endosomal as well as endosomal compartments,
TUG function in vitro increases the amount of GLUT4 in a con- again arguing in favor of the recruitment of endosomal GLUT4
stitutive endosome-plasma membrane recycling pathway at the [49].
expense of GLUT4 in GSVs (as in normal insulin-stimulated Taken together, these findings indicate that a GLUT4 reten-
cells) [39] and that disruption of TUG function in vivo reduces tion mechanism exists in endosomal compartments but that
fasting plasma glucose and insulin levels in mice [40]. a highly efficient intracellular GLUT4 retention requires the
No matter what the exact mechanism is by which the GSVs presence of GLUT4 in non-endosomal structures as well. The
are sequestrated, it needs to be sensitive to insulin regulation. In mechanism implicated in the retention of GLUT4 in endosomes
support of exocytosis of the entire GSV upon insulin stimulation is unknown. As far as the insulin-induced recruitment of GLUT4
are the findings that insulin reduces the amount of these vesi- is concerned, endosomes may not be the principle GLUT4 donor
cles within the cytoplasm without altering the concentration of site, but may provide the cell system with additional GLUT4
GLUT4 in the GSVs that remain [41] and that the GSVs them- under conditions of high demand, for example under prolonged
selves physically fuse with the plasma membrane [42]. GSV insulin stimulations [19] or under repeated stimulations [49].
exocytosis occurs in a highly regulated fashion in that the amount Upon prolonged insulin stimulation, relatively more intracel-
of GSVs that are recruited correlates with the strength of the lular GLUT4 is in endosomes and less in GSVs [20,45] (Fig. 2).
insulin stimulus [43,44]. Presumably, GLUT4 that is initially recruited from GSVs is
internalized and immediately recycled back to the cell surface
2.2. Presence of GLUT4 in endosomes in vesicles that are likely to be distinct from GSVs and presum-
ably derived from recycling and/or early endosomes. This is
Several pieces of evidence exist that indicate that a part of supported by data that suggest that the GLUT4-containing vesi-
the cellular GLUT4 pool is located in endosomal (non-GSV) cles that fuse with the plasma membrane upon prolonged insulin
compartments and that also this GLUT4 pool is retained intra- stimulation are of endosomal origin [19]. It implies that the same
cellularly and sensitive to insulin stimulation. First, in cultured initially recruited GLUT4 molecules remain recycling, uniquely
adipocytes, about half of the total cellular GLUT4 pool is between the plasma membrane and endosomes, and fits with the
present in endosomal compartments [15,20]. Given the fact that finding that the amount of VAMP2-decorated GSVs decrease
GLUT4 is hardly recycling towards the plasma membrane in with insulin, while the amount of GLUT4 in the remaining
non-stimulated adipocytes [43], a retention mechanism has to be (non-recruited) GSVs remains unchanged [41]. Moreover, it
present in endosomes. Moreover, upon insulin stimulation, much explains why the GLUT4 molecules that recycle between the
more than 50% of the total cellular GLUT4 pool participates in cell surface and intracellular compartments in insulin-stimulated
cell surface recycling [43], indicating that both endosomal and adipocytes do not readily mix with the GLUT4 molecules that
non-endosomal GLUT4 pools contribute. This is further sup- remain retained intracellularly, in particular under conditions of
ported by morphological and cell fractionation studies that have low to moderate insulin stimulation [43,50]. Taken together, this
demonstrated that GLUT4 translocates from both GSVs and has several important consequences. It means that insulin only
endosomes [41,45]. Second, acutely inhibiting protein traffic transiently activates the GSV release mechanism. It also implies
from endosomes in living cells reduces insulin-induced GLUT4 that in insulin-stimulated cells, the intracellular traffic in general
translocation by half [20,46]. Third, in adipocytes, a mutated or specifically that of GLUT4 between endosomes and GSVs,
GLUT4 molecule that is exclusively present in endosomal com- between endosomes and the TGN (that generates the GSVs), or
partments, is still efficiently retained, albeit to a somewhat lesser between the TGN and GSVs, is disrupted.
extent when compared with wild-type GLUT4 [43]. Moreover,
this molecule still translocates upon insulin stimulation [43]. 2.3. Involvement of the TGN (sub)compartment in GLUT4
Fourth, in fibroblast-like preadipocytes and CHO cells, GLUT4 traffic
is exclusively present in endosomes [20]. However, in these
cells GLUT4 is retained intracellularly and also more sensi- The presence of GLUT4 in the TGN has been suggested on
tive to insulin stimulation when compared with the transferrin several occasions and is likely to play a role in its intracellu-
receptor (TfR), a bona fide endosomal marker that recycles con- lar regulation. At the morphological level, a significant amount
stitutively [47,48]. Nevertheless, also here, its retention and of GLUT4 in adipocytes is localized in vesicles that lie in the
insulin-sensitivity are reduced when compared with GLUT4 vicinity of the TGN [51,52], where it colocalizes to a large extent
in adipocytes. Finally, prolonged (but not acute) insulin stim- with the cation-dependent mannose 6-phosphate receptor (CD-
ulation induces the fusion of GLUT4-containing vesicles with MPR), a protein that recycles between the TGN and endosomes
the plasma membrane that are relatively large in size and that [52] and with adaptor protein-1 (AP-1), known to be involved in
resemble endosomal vesicles rather than GSVs, suggesting not the exit of proteins from the TGN [53], but not with TGN proteins
only that insulin recruits GLUT4 from both GSVs and endo- TGN38 and furin. Given that the TGN is subcompartmentalized
somes but also that there is a temporal shift in the recruitment [54–56], GLUT4 is likely to be present in a specific subdomain
from both storage compartments [19]. Possibly related to these of the TGN [14]. This GLUT4 pool is not depleted upon insulin
404 R. Govers / Diabetes & Metabolism 40 (2014) 400–410

stimulation [52], suggesting an insulin-independent equilibrium plasma membrane component [33]. Interestingly, these effects
between in- and out-going GLUT4 and a possible involvement of insulin on GLUT4 at the plasma membrane are impaired
of this GLUT4 pool in an intracellular cycle between TGN and in adipocytes rendered insulin-resistant in vitro as well as in
endosomes (Fig. 2). adipocytes obtained from insulin-resistant human subjects
Next, it has been shown that a large amount of GLUT4 is [30,71]. Functionally, these two insulin actions (GSV brake
recycling via the TGN in cardiomyocytes [57], while IRAP, release and stimulation of GSV tethering, docking and fusion)
GLUT4 fellow traveller, has been shown to traffic through the lead to an insulin dose-dependent increase in the size of the cell
TGN after retrieval from the plasma membrane in adipocytes surface recycling GLUT4 pool [43,44,72] and in an increased
[58]. GLUT4 exocytosis rate in the recycling pool [44].
Finally, various Golgi/TGN proteins have been shown to be Another action of insulin that may contribute to increased cell
implicated in intracellular GLUT4 traffic. Sortilin [22] and GGA surface GLUT4 levels in adipocytes is its effect on GLUT4 endo-
proteins [59,60], involved in the exit of certain proteins from the cytosis, as insulin may modestly decrease GLUT4 endocytosis
TGN, have a clear role in the retention of GLUT4, likely by reg- rates. Nevertheless, this insulin action remains controversial
ulating the traffic of GLUT4 from the TGN into the GSVs. Also [73–76]. The insulin-induced reduction in GLUT4 endocytosis
the TGN t-SNARE proteins syntaxin 6 and syntaxin 16 have may be linked to the existence of multiple GLUT4 internaliza-
been indicated to be involved in GLUT4 traffic (see Section 2.8 tion pathways. In adipocytes, GLUT4 has been demonstrated
“SNARE proteins”) and may be colocalizing with GLUT4 in the to be internalized by both AP-2/clathrin- and cholesterol-
TGN subdomain that is distinct from the one containing TGN38. dependent mechanisms that act independently [76,77], whereas
Other Golgi proteins that have been found to be involved in the constitutively recycling TfR is internalized uniquely via the
GLUT4 traffic include Golgin-97 [61], Golgin-160 [38] and clathrin-dependent pathway. Insulin has been shown to induce
the IRAP-interacting proteins p115 [62] and tankyrase [63]. a shift in GLUT4 internalization from one mechanism to the
The Golgi matrix protein Golgin-160 may play a role in GSV other [76]. If the kinetics of both pathways is distinct, this could
sequestration by linking GSVs to the Golgi matrix as this protein explain the effect of insulin on the GLUT4 internalization rate.
has been shown to interact with the GLUT4/GSV-sequestering Alternatively (or possibly linked), insulin may affect GLUT4
protein TUG [36] (see Section 2.1). internalization kinetics via its effect on the clustering of GLUT4
In conclusion, GLUT4 is present in the TGN. Its presence molecules at the plasma membrane [78,79].
is most likely related to the role of the TGN in the biogenesis
of GSVs and in the targeting of GLUT4 and other proteins into 2.5. Insulin receptor signaling pathways implicated in
these vesicles. The GSV components are delivered to the TGN GLUT4 translocation
via recycling endosomes, a transport step involving syntaxins 6
and 16, CHC22 (at least in vertebrate cells) [64,65] and possibly The binding of insulin to the insulin receptor (IR) leads to
also Rab11 (see Section 2.7 “Rab proteins”). structural changes within the receptor that induce the trans-
phosphorylation of various tyrosine residues. This results in
2.4. Major insulin actions on GLUT4 traffic the recruitment of IR substrates to the IR, amongst which IR
substrate 1 (IRS1), and their subsequent tyrosine phosphory-
Insulin has been reported to display several effects on lation. Phosphorylated IRS1 activates PI 3-kinase in the inner
GLUT4. First of all, insulin releases GLUT4 from its static com- leaflet of the plasma membrane [80] leading to phosphoryla-
partment, where it is relatively immobile. This likely represents tion of the inositol ring of phosphatidylinositol and the local
the release of GSVs and results in a two- to three-fold increase formation of the lipid second messenger PIP3. This on its turn
in the amount of GLUT4 vesicles that are mobile [66,67] and serves as a recruitment platform for phosphoinositide-dependent
in the redistribution of GLUT4 from a perinuclear location kinase-1 (PDK1) and serine/threonine protein kinase B (PKB,
towards the periphery of the cell, where it accumulates near also known as Akt2 [81]). PKB then becomes phosphorylated by
the plasma membrane [19,30]. Probably related to this release PDK1 as well as by mammalian target of rapamycin complex-
is the insulin-induced increase in mean GLUT4 vesicle speed 2 (mTORC2), which results in its activation. Activated kinase
[67]. In insulin-resistant adipocytes, this insulin effect is partially PKB also binds to intracellular vesicles that contain GLUT4
impaired [30]. [82] and phosphorylates a multitude of substrates. While PKB is
Insulin also displays several major effects at the cell surface. considered to be a key player in insulin-induced GLUT4 translo-
Insulin enhances the process of the docking and fusion of cation [83], PKB-independent pathways exist that contribute to
GLUT4 vesicles with the plasma membrane. More precisely, the full effect of insulin on the redistribution of GLUT4 to the
insulin stimulation results in an increase in the halting of plasma membrane [84]. For instance, PDK1 not only phosphory-
GLUT4 vesicles at the plasma membrane, more efficient tether- lates and activates PKB but also the atypical PKC isoforms ␭ and
ing and docking (reducing un-tethering and un-docking), and a ␨ [85–88]. While both are implicated in GLUT4 translocation,
reduced docking dwell time, concomitant with an enhancement their downstream effectors are currently unknown [88]. Other
of their fusion with the plasma membrane [16,68,69]. The PKB-independent pathways involved in GLUT4 translocation
fusion of GLUT4 vesicles with the plasma membrane appears include phospholipase C (PLC) that interacts with and is acti-
to be the major insulin-sensitive step in the process of tethering, vated by the IR [89], and the IRS1/PI 3-kinase/PKB-independent
docking and fusion [70] and likely implicates the activation of a TC10 pathway [90]. TC10 is a Rho family member GTPase and
R. Govers / Diabetes & Metabolism 40 (2014) 400–410 405

regulates the activity of the exocyst, implicated in the targeting cellular retention of GLUT4 and in the insulin-induced release
of secretory vesicles, via its interaction with exocyst subunit of this retention, leading to the accumulation of GLUT4 near
Exo70 [91]. In particular the TC10␣ isoform appears to be the plasma membrane [30,84]. In addition, AS160 has been
implicated in GLUT4 translocation [92]. Together with a part found to play a role in the docking of GLUT4 vesicles with
of the cellular IR pool, TC10␣ resides to a large extent in lipid the plasma membrane [69,70]. This is likely related to the
rafts [93]. It has been suggested to be regulated by a CrkII-C3G presence of AS160 at the plasma membrane where it binds to
complex that binds indirectly to the IR via CAP, APS, and Cbl negatively charged phospholipids [113]. The fact that AS160
[94,95]. However, subsequent siRNA experiments have chal- knockdown only partially releases GLUT4 retention while
lenged this mode of regulation [96]. More recent data indicate insulin-sensitivity is maintained [112,114] indicates that insulin
that TC10␣ activity is likely to be regulated via its phosphoryla- induces GLUT4 translocation by both AS160-dependent and
tion and that insulin induces TC10␣ phosphorylation by CDK5 -independent mechanisms. Another indication that supports a
[93]. The role of TC10␣ in GLUT4 translocation likely implies role for an AS160/PKB-independent signaling step in GLUT4
the exocyst [91,97], but also the cleavage of TUG, required for translocation comes from studies that demonstrate that the final
the insulin-induced reduction in GSV sequestration [36], as well insulin-enhanced GSV-plasma membrane fusion event is PI 3-
as the insulin-induced rearrangement of cortical actin [93], con- kinase-dependent but PKB/AS160-independent [69,84]. Given
sistent with the role of the cytoskeleton and associated proteins that the major insulin action involves this fusion step [70], it is
in GLUT4 translocation (elegantly reviewed in [98]). evident that AS160 is not the (only) master director of GLUT4
In insulin resistance, impaired GLUT4 translocation has translocation. At present, the molecular mechanism by which
been attributed to defects in signaling by IRS1 [99,100], PKB insulin enhances fusion rates is unknown and remains to be
[101,102], effectors downstream of PKB [103], and TC10 elucidated.
[104,105]. Additional studies are needed to fully elucidate the Several studies have demonstrated that AS160 is not the
mechanisms by which insulin resistance impinges on these (and only effector molecule by which PKB regulates GLUT4 traf-
other) signaling pathways involved in GLUT4 translocation. fic [84,112]. Accordingly, other PKB substrates have been
identified that are implicated in insulin-induced GLUT4 translo-
2.6. PKB substrates cation. These include motor protein myosin Va, that associates
with the actin cytoskeleton upon its PKB-mediated phosphor-
Of the PKB substrates, Akt substrate of 160 kDa (AS160; also ylation [115], Grp1, a guanine nucleotide exchange factor
known as Tbc1d4) is best known and has been shown to be a key (GEF) for ADP-ribosylation factor 6 (ARF6) [116], also
modulator of GLUT4 translocation [106,107]. AS160 has been involved in GLUT4 translocation [117], GSV- and plasma
the first molecule that directly links insulin signaling to intra- membrane-associated lipid-binding protein CDP138 [118], syn-
cellular GLUT4 traffic and hence has gotten a lot of attention. taxin 4-interacting protein Synip, negatively regulating GLUT4
AS160 is a Rab-GTPase-activating protein (GAP [108]) and translocation at the GSV-plasma membrane docking step
active in its dephosphorylated form. Insulin stimulation induces [119–121], and lipid kinase PIKfyve [122].
the phosphorylation of AS160 on at least 5 residues, which
renders its GAP domain inactive [109]. This PKB-mediated 2.7. Rab proteins
phosphorylation event is facilitated by scaffold protein ClipR-59
[110] and leads to an increase in the GTP-bound (active) form of Recently, several studies have focused on the identification
the AS160 Rab substrate(s) and in the redistribution of GLUT4 and functional characterization of the Rab proteins that are
vesicles towards the cell surface. AS160 can interact with GSV potential substrates of AS160. In adipocytes, the Rab protein
compounds IRAP [24] and LRP1 [23] and is present on GLUT4- that is currently considered to be the main substrate of AS160
containing vesicles but dissociates from these vesicles upon is Rab10. In basal (non-stimulated) adipocytes, a constitutively
insulin stimulation [24]. This suggests that its inhibitory action active Rab10 mutant increases GLUT4 cell surface levels, while
on GLUT4 translocation in the absence of insulin requires its knockdown of Rab10, but not that of other in vitro AS160 Rab
presence on GSVs, maintaining a GSV-associated Rab in its substrates, inhibits GLUT4 translocation [123,124]. Moreover,
GDP-bound inactive state. A mutated phosphorylation-deficient the increase in cell surface GLUT4 levels in basal adipocytes due
AS160 molecule is constitutively active and induces a reduction to AS160 knockdown is reduced by a simultaneous knockdown
in both Rab activity and insulin-induced GLUT4 transloca- of Rab10 [114,123] and further enhanced by Rab10 overexpress-
tion [109]. This effect on GLUT4 translocation is dependent ion [114]. This indicates that in basal cells, Rab10 is the Rab
on a functional Rab-GAP domain within AS160, demonstrat- protein maintained in the GDP-bound (inactive) form by AS160
ing that its activity towards Rab proteins is required for its to impose GLUT4 retention. Rab10 probably acts at the level
role in GLUT4 traffic. AS160 knockdown induces a moder- of the GSVs because neither knockdown nor overexpression of
ate increase in basal cell surface GLUT4 levels [24,111,112], Rab10 has any effect on cell surface TfR levels [114,123], while
that cannot be rescued by the expression of an AS160 mutant GLUT4-positive TfR-negative vesicles that fuse with the plasma
lacking a functional GAP domain, confirming once more that membrane in response to insulin (i.e. GSVs) are predominantly
the GAP activity is required for AS160 function [111]. Taken loaded with Rab10 [42,125]. Also in support is the recent identi-
together, these data indicate that AS160 (and hence also its fication of Dennd4C as the guanine nucleotide exchange factor
upstream regulator PKB) plays a crucial role both in the intra- (GEF) for Rab10 (generating the GTP-bound form of Rab10)
406 R. Govers / Diabetes & Metabolism 40 (2014) 400–410

and its implication in GLUT4 translocation [126]. Dennd4C SNAREs) syntaxin 4 and SNAP23 are located on the plasma
is constitutively active and not sensitive to insulin stimula- membrane. It is generally accepted that VAMP2, also known
tion, implying that insulin regulates Rab10 exclusively via as synaptobrevin, is the vesicle SNARE (v-SNARE) present on
AS160 [114]. The increase in basal cell surface GLUT4 levels GSVs, though v-SNAREs VAMP3 and VAMP8 have also been
induced by Rab10 overexpression, AS160 knockdown, or by the suggested to be present on GLUT4 vesicles and to be possibly
combination of both can be further enhanced by insulin [114], implicated in GLUT4 translocation [134–136]. VAMP2 specif-
suggesting the existence of a Rab10/AS160-independent insulin ically displays affinity for the syntaxin 4/SNAP23 complex and
signaling pathway that contributes to GLUT4 translocation. This hence this affinity aids in the regulation of the specificity (i.e.
is further supported by studies based on total internal reflection selectivity) of the fusion process. Together, syntaxin 4, SNAP23
fluorescence (TIRF), a technique that enables the visualization and VAMP2 form a tight four-helix bundle, enabling the docking
of labeled proteins at the cell surface or just beneath (at a distance and fusion of the GLUT4 vesicles with the plasma membrane.
of maximum 200 nanometers from the plasma membrane), that Additional SNARE proteins are involved as well in intracel-
have suggested that Rab10 regulates the recruitment of GLUT4 lular GLUT4 traffic as they are implicated in the other GLUT4
vesicles to the plasma membrane but not their fusion [114]. trafficking steps. One example concerns a TGN t-SNARE-
While Rab10 is the only Rab protein identified on fusion- complex consisting of syntaxins 6 and 16 [137]. These SNAREs
competent TfR-negative GLUT4 vesicles (GSVs), other Rab are present in the TGN as well as on GLUT4-containing vesi-
proteins also seem to be involved in GLUT4 translocation. cles and substantially redistribute to the plasma membrane upon
Rab14 has been indicated to be present on endosome-derived insulin stimulation [58]. Functionally, syntaxins 6 and 16 have
GLUT4 vesicles and to be involved in their traffic. The nature of been suggested to be implicated in the sequestration of GLUT4
the acceptor compartment for the Rab14-decorated GLUT4 vesi- from endosomes into GSVs [137,138]. Given their usual role
cles is unclear at present as these vesicles have been suggested in protein traffic between endosomes and TGN [139], these t-
to be destined for delivery to the plasma membrane [42], GSVs SNAREs are likely to mediate the traffic of GLUT4 to the TGN,
[114] and TGN [127]. Overexpression of a constitutively active from where it can be further sorted into GSVs. Also the syntaxin
Rab14 mutant inhibits GLUT4 translocation [127]. Remark- 16 regulator mVps45, member of the SM (Sec1/Munc18) family
ably, Rab14 knockdown also decreases GLUT4 translocation, of proteins, has been demonstrated to play a role in this pathway
but unlike Rab10, does not reduce the increased basal cell surface [140].
GLUT4 levels in AS160-depleted cells, implying that Rab14 is At present, about ten proteins have been identified that are
not likely to be regulated by AS160 [114]. In support of a role implicated in the regulation of the SNAREs that mediate the
for Rab10 and Rab14 in distinct pathways is the largely addi- docking and fusion of GLUT4 vesicles with the plasma mem-
tive effect of the knockdown of Rab10 and Rab14 on GLUT4 brane. One of them is SM family member munc18c. This protein
translocation [42]. Both Rab10 and Rab14 have been found to is an inhibitory regulator that binds syntaxin 4, thereby reduc-
interact with myosin Va, a molecular motor implicated in the ing the interaction of syntaxin 4 with SNAP23 and VAMP2
final phases of GLUT4 translocation [42]. Other Rab proteins [141,142]. Upon insulin stimulation, the activated insulin recep-
that are implicated in intracellular GLUT4 traffic but that act tor phosphorylates munc18c [143] which reduces its affinity for
most likely at other places than the plasma membrane include syntaxin 4, leading to a decrease in the interaction between
Rab31, found to play a negative role in GLUT4 translocation, munc18c and syntaxin 4 [144], enabling the formation of the
possibly via regulating the fusion of GSVs with endosomes at trimeric SNARE-complex, required for the fusion of GLUT4
the expense of the fusion of GSVs with the plasma membrane vesicles with the plasma membrane. In accordance, Munc18c
[128], Rab5, acting on GLUT4 internalization or on endosomal ablation results in an increased GLUT4 translocation [145],
GLUT4 traffic [129], Rab11, proposed to play a role in the sor- while munc18c overexpression causes insulin resistance [146].
ting of GLUT4 into GSVs [20], possibly by regulating the traffic Another protein that binds and regulates syntaxin 4 is Doc2b
of GLUT4 from recycling endosomes to the TGN, from where [147]. Insulin increases the interaction between Doc2b and syn-
it can be processed into GSVs, Rab4 [130] and Rab8A [42]. taxin 4, thereby enhancing the docking and/or fusion of GSVs
with the plasma membrane [147]. Accordingly, GLUT4 translo-
2.8. SNARE proteins cation is enhanced by Doc2b overexpression and inhibited by
Doc2b knockdown [147]. Recently, Doc2b has been suggested
The docking and fusion of GLUT4-containing vesicles with to enhance the SNARE-mediated fusion process by inducing a
the plasma membrane are regulated by insulin. Especially the local curvature of the target membrane [136].
fusion event is highly insulin-sensitive [70]. The entire process
of tethering, docking and fusion of vesicles with an acceptor 3. Concluding remarks
membrane is a highly coordinated system. The key players
that form the basis of this process are the so-called SNAREs T2D is taking epidemiological proportions and is associated
(soluble N-ethylmaleimide-sensitive-factor attachment protein with insulin resistance, in which muscle and adipose tissue take
receptors) together with the proteins that regulate SNARE up less glucose. Since GLUT4 is the main transporter that is
function [131]. The SNARE proteins involved in the fusion responsible for this uptake, GLUT4 is considered to play an
of GLUT4 vesicles with the plasma membrane are syntaxin essential role in insulin resistance and T2D. While the intracellu-
4, SNAP23, and VAMP2 [132,133]. The target SNAREs (t- lar GLUT4 traffic and its sensitivity to insulin have proven to be
R. Govers / Diabetes & Metabolism 40 (2014) 400–410 407

regulated in a complex manner, our understanding of its itinerary [11] Garvey WT, Maianu L, Zhu JH, Hancock JA, Golichowski AM. Multiple
and of the molecules implicated in the various trafficking steps defects in the adipocyte glucose transport system cause cellular insulin
resistance in gestational diabetes. Heterogeneity in the number and a novel
has been largely improved over the years. However, how GLUT4
abnormality in subcellular localization of GLUT4 glucose transporters.
is efficiently retained intracellularly and how insulin exactly Diabetes 1993;42:1773–85.
impinges on this retention is still largely unknown and remains to [12] Garvey WT, Maianu L, Zhu JH, Brechtel-Hook G, Wallace P, Baron AD.
be clarified. Unveiling these mechanisms remains a future goal Evidence for defects in the trafficking and translocation of GLUT4 glu-
and may aid in the development of novel therapeutic avenues cose transporters in skeletal muscle as a cause of human insulin resistance.
J Clin Invest 1998;101:2377–86.
for the treatment of T2D.
[13] Maier VH, Gould GW. Long-term insulin treatment of 3T3-L1 adipocytes
results in mis-targeting of GLUT4: implications for insulin-stimulated
Disclosure of interest glucose transport. Diabetologia 2000;43:1273–81.
[14] Karylowski O, Zeigerer A, Cohen A, McGraw TE. GLUT4 is retained
The author declares that he has no conflicts of interest con- by an intracellular cycle of vesicle formation and fusion with endosomes.
Mol Biol Cell 2004;15:870–82.
cerning this article.
[15] Livingstone C, James DE, Rice JE, Hanpeter D, Gould GW. Compartment
ablation analysis of the insulin-responsive glucose transporter (GLUT4)
Acknowledgements in 3T3-L1 adipocytes. Biochem J 1996;315:487–95.
[16] Lizunov VA, Matsumoto H, Zimmerberg J, Cushman SW, Frolov VA.
I apologize to the research groups in this field whose excel- Insulin stimulates the halting, tethering, and fusion of mobile GLUT4
vesicles in rat adipose cells. J Cell Biol 2005;169:481–9.
lent work was not cited due to restricted space limitations. I [17] Green H, Kehinde O. An established preadipose cell line and its dif-
am grateful to Teresa Gonzalez for assistance in preparing the ferentiation in culture. II. Factors affecting the adipose conversion. Cell
French abstract. This work is supported by an Inserm Avenir 1975;5:19–27.
grant and by an Alfediam/SFD-Roche Diagnostics award. [18] Stockli J, Fazakerley DJ, James DE. GLUT4 exocytosis. J Cell Sci
2011;124:4147–59.
[19] Xu Y, Rubin BR, Orme CM, Karpikov A, Yu C, Bogan JS, et al. Dual-
Appendix A. Supplementary data mode of insulin action controls GLUT4 vesicle exocytosis. J Cell Biol
2011;193:643–53.
Supplementary data (French abstract) associated with this [20] Zeigerer A, Lampson MA, Karylowski O, Sabatini DD, Adesnik M, Ren
article can be found, in the online version, at http://dx.doi. M, et al. GLUT4 retention in adipocytes requires two intracellular insulin-
regulated transport steps. Mol Biol Cell 2002;13:2421–35.
org/10.1016/j.diabet.2014.01.005.
[21] Jordens I, Molle D, Xiong W, Keller SR, McGraw TE. Insulin-regulated
aminopeptidase is a key regulator of GLUT4 trafficking by controlling
References the sorting of GLUT4 from endosomes to specialized insulin-regulated
vesicles. Mol Biol Cell 2010;21:2034–44.
[1] Apelt J, Mehlhorn G, Schliebs R. Insulin-sensitive GLUT4 glucose trans- [22] Shi J, Kandror KV. Sortilin is essential and sufficient for the forma-
porters are colocalized with GLUT3-expressing cells and demonstrate a tion of Glut4 storage vesicles in 3T3-L1 adipocytes. Dev Cell 2005;9:
chemically distinct neuron-specific localization in rat brain. J Neurosci 99–108.
Res 1999;57:693–705. [23] Jedrychowski MP, Gartner CA, Gygi SP, Zhou L, Herz J, Kandror KV,
[2] Brosius 3rd FC, Briggs JP, Marcus RG, Barac-Nieto M, Charron MJ. et al. Proteomic analysis of GLUT4 storage vesicles reveals LRP1 to be
Insulin-responsive glucose transporter expression in renal microvessels an important vesicle component and target of insulin signaling. J Biol
and glomeruli. Kidney Int 1992;42:1086–92. Chem 2010;285:104–14.
[3] McBrayer SK, Cheng JC, Singhal S, Krett NL, Rosen ST, Shanmugam M. [24] Larance M, Ramm G, Stockli J, van Dam EM, Winata S, Wasinger
Multiple myeloma exhibits novel dependence on GLUT4, GLUT8, and V, et al. Characterization of the role of the Rab GTPase-activating
GLUT11: implications for glucose transporter-directed therapy. Blood protein AS160 in insulin-regulated GLUT4 trafficking. J Biol Chem
2012;119:4686–97. 2005;280:37803–13.
[4] Medina RA, Meneses AM, Vera JC, Guzman C, Nualart F, Astuya A, [25] Lampson MA, Schmoranzer J, Zeigerer A, Simon SM, McGraw
et al. Estrogen and progesterone up-regulate glucose transporter expres- TE. Insulin-regulated release from the endosomal recycling compart-
sion in ZR-75-1 human breast cancer cells. Endocrinology 2003;144: ment is regulated by budding of specialized vesicles. Mol Biol Cell
4527–35. 2001;12:3489–501.
[5] Maianu L, Keller SR, Garvey WT. Adipocytes exhibit abnormal sub- [26] Shi J, Huang G, Kandror KV. Self-assembly of Glut4 storage
cellular distribution and translocation of vesicles containing glucose vesicles during differentiation of 3T3-L1 adipocytes. J Biol Chem
transporter 4 and insulin-regulated aminopeptidase in type 2 diabetes 2008;283:30311–21.
mellitus: implications regarding defects in vesicle trafficking. J Clin [27] Kaddai V, Jager J, Gonzalez T, Najem-Lendom R, Bonnafous S, Tran
Endocrinol Metab 2001;86:5450–6. A, et al. Involvement of TNF-alpha in abnormal adipocyte and muscle
[6] Abel ED, Peroni O, Kim JK, Kim YB, Boss O, Hadro E, et al. Adipose- sortilin expression in obese mice and humans. Diabetologia 2009;52:
selective targeting of the GLUT4 gene impairs insulin action in muscle 932–40.
and liver. Nature 2001;409:729–33. [28] Tsuchiya Y, Hatakeyama H, Emoto N, Wagatsuma F, Matsushita
[7] Yang Q, Graham TE, Mody N, Preitner F, Peroni OD, Zabolotny JM, S, Kanzaki M. Palmitate-induced down-regulation of sortilin and
et al. Serum retinol binding protein 4 contributes to insulin resistance in impaired GLUT4 trafficking in C2C12 myotubes. J Biol Chem
obesity and type 2 diabetes. Nature 2005;436:356–62. 2010;285:34371–81.
[8] Bryant NJ, Govers R, James DE. Regulated transport of the glucose [29] Shi J, Kandror KV. The luminal Vps10p domain of sortilin plays the
transporter GLUT4. Nat Rev Mol Cell Biol 2002;3:267–77. predominant role in targeting to insulin-responsive Glut4-containing vesi-
[9] Rubin BR, Bogan JS. Intracellular retention and insulin-stimulated mobi- cles. J Biol Chem 2007;282:9008–16.
lization of GLUT4 glucose transporters. Vitam Horm 2009;80:155–92. [30] Xiong W, Jordens I, Gonzalez E, McGraw TE. GLUT4 is sorted to vesicles
[10] Huang S, Czech MP. The GLUT4 glucose transporter. Cell Metab whose accumulation beneath and insertion into the plasma membrane
2007;5:237–52.
408 R. Govers / Diabetes & Metabolism 40 (2014) 400–410

are differentially regulated by insulin and selectively affected by insulin the plasma membrane of 3T3-L1 adipocytes. J Cell Biol 1988;106:
resistance. Mol Biol Cell 2010;21:1375–86. 69–76.
[31] Keller SR, Davis AC, Clairmont KB. Mice deficient in the insulin- [52] Martin S, Millar CA, Lyttle CT, Meerloo T, Marsh BJ, Gould GW,
regulated membrane aminopeptidase show substantial decreases in et al. Effects of insulin on intracellular GLUT4 vesicles in adipocytes:
glucose transporter GLUT4 levels but maintain normal glucose homeo- evidence for a secretory mode of regulation. J Cell Sci 2000;113:
stasis. J Biol Chem 2002;277:17677–86. 3427–38.
[32] Briguglio JS, Kumar S, Turkewitz AP. Lysosomal sorting receptors are [53] Martin S, Ramm G, Lyttle CT, Meerloo T, Stoorvogel W, James DE. Bio-
essential for secretory granule biogenesis in Tetrahymena. J Cell Biol genesis of insulin-responsive GLUT4 vesicles is independent of brefeldin
2013;203:537–50. A-sensitive trafficking. Traffic 2000;1:652–60.
[33] Koumanov F, Jin B, Yang J, Holman GD. Insulin signaling meets vesicle [54] Luke MR, Kjer-Nielsen L, Brown DL, Stow JL, Gleeson PA. GRIP
traffic of GLUT4 at a plasma membrane-activated fusion step. Cell Metab domain-mediated targeting of two new coiled-coil proteins. GCC88 and
2005;2:179–89. GCC185, to subcompartments of the trans-Golgi network J Biol Chem
[34] Bogan JS, Hendon N, McKee AE, Tsao TS, Lodish HF. Functional cloning 2003;278:4216–26.
of TUG as a regulator of GLUT4 glucose transporter trafficking. Nature [55] Lock JG, Hammond LA, Houghton F, Gleeson PA, Stow JL. E-cadherin
2003;425:727–33. transport from the trans-Golgi network in tubulovesicular carriers is selec-
[35] Bogan JS. Regulation of glucose transporter translocation in health and tively regulated by golgin-97. Traffic 2005;6:1142–56.
diabetes. Annu Rev Biochem 2012;81:507–32. [56] Park JJ, Gondre-Lewis MC, Eiden LE, Loh YP. A distinct trans-Golgi
[36] Bogan JS, Rubin BR, Yu C, Loffler MG, Orme CM, Belman JP, et al. Endo- network subcompartment for sorting of synaptic and granule proteins in
proteolytic cleavage of TUG protein regulates GLUT4 glucose transporter neurons and neuroendocrine cells. J Cell Sci 2011;124:735–44.
translocation. J Biol Chem 2012;287:23932–47. [57] Slot JW, Garruti G, Martin S, Oorschot V, Posthuma G, Kraegen EW,
[37] Orme CM, Bogan JS. The ubiquitin regulatory X (UBX) domain- et al. Glucose transporter (GLUT-4) is targeted to secretory granules in
containing protein TUG regulates the p97 ATPase and resides at the rat atrial cardiomyocytes. J Cell Biol 1997;137:1243–54.
endoplasmic reticulum-golgi intermediate compartment. J Biol Chem [58] Shewan AM, Van Dam EM, Martin S, Luen TB, Hong W, Bryant NJ, et al.
2012;287:6679–92. GLUT4 recycles via a trans-Golgi network (TGN) subdomain enriched
[38] Williams D, Hicks SW, Machamer CE, Pessin JE. Golgin-160 is required in syntaxins 6 and 16 but not TGN38: involvement of an acidic targeting
for the Golgi membrane sorting of the insulin-responsive glucose trans- motif. Mol Biol Cell 2003;14:973–86.
porter GLUT4 in adipocytes. Mol Biol Cell 2006;17:5346–55. [59] Watson RT, Khan AH, Furukawa M, Hou JC, Li L, Kanzaki M, et al.
[39] Yu C, Cresswell J, Loffler MG, Bogan JS. The glucose transporter 4- Entry of newly synthesized GLUT4 into the insulin-responsive storage
regulating protein TUG is essential for highly insulin-responsive glucose compartment is GGA dependent. Embo J 2004;23:2059–70.
uptake in 3T3-L1 adipocytes. J Biol Chem 2007;282:7710–22. [60] Li LV, Kandror KV. Golgi-localized, gamma-ear-containing, Arf-binding
[40] Loffler MG, Birkenfeld AL, Philbrick KM, Belman JP, Habtemichael protein adaptors mediate insulin-responsive trafficking of glucose trans-
EN, Booth CJ, et al. Enhanced fasting glucose turnover in mice with porter 4 in 3T3-L1 adipocytes. Mol Endocrinol 2005;19:2145–53.
disrupted action of TUG protein in skeletal muscle. J Biol Chem [61] Hatakeyama H, Kanzaki M. Molecular basis of insulin-responsive
2013;288:20135–50. GLUT4 trafficking systems revealed by single molecule imaging. Traffic
[41] Ramm G, Slot JW, James DE, Stoorvogel W. Insulin recruits GLUT4 2011;12:1805–20.
from specialized VAMP2-carrying vesicles as well as from the dynamic [62] Hosaka T, Brooks CC, Presman E, Kim SK, Zhang Z, Breen M, et al.
Endosomal/Trans-golgi network in rat adipocytes. Mol Biol Cell p115 Interacts with the GLUT4 vesicle protein, IRAP, and plays a
2000;11:4079–91. critical role in insulin-stimulated GLUT4 translocation. Mol Biol Cell
[42] Chen Y, Wang Y, Zhang J, Deng Y, Jiang L, Song E, et al. Rab10 and 2005;16:2882–90.
myosin-Va mediate insulin-stimulated GLUT4 storage vesicle transloca- [63] Yeh TY, Sbodio JI, Tsun ZY, Luo B, Chi NW. Insulin-stimulated exocy-
tion in adipocytes. J Cell Biol 2012;198:545–60. tosis of GLUT4 is enhanced by IRAP and its partner tankyrase. Biochem
[43] Govers R, Coster AC, James DE. Insulin increases cell surface GLUT4 J 2007;402:279–90.
levels by dose dependently discharging GLUT4 into a cell surface [64] Vassilopoulos S, Esk C, Hoshino S, Funke BH, Chen CY, Plocik AM,
recycling pathway. Mol Cell Biol 2004;24:6456–66. et al. A role for the CHC22 clathrin heavy-chain isoform in human glucose
[44] Muretta JM, Romenskaia I, Mastick CC. Insulin releases glut4 from metabolism. Science 2009;324:1192–6.
static storage compartments into cycling endosomes and increases the [65] Esk C, Chen CY, Johannes L, Brodsky FM. The clathrin heavy-chain
rate constant for glut4 exocytosis. J Biol Chem 2008;283:311–23. isoform CHC22 functions in a novel endosomal sorting step. J Cell Biol
[45] Blot V, McGraw TE. Molecular mechanisms controlling GLUT4 intra- 2010;188:131–44.
cellular retention. Mol Biol Cell 2008;19:3477–87. [66] Semiz S, Park JG, Nicoloro SM, Furcinitti P, Zhang C, Chawla A, et al.
[46] Millar CA, Shewan A, Hickson GR, James DE, Gould GW. Conventional kinesin KIF5B mediates insulin-stimulated GLUT4 move-
Differential regulation of secretory compartments containing the insulin- ments on microtubules. Embo J 2003;22:2387–99.
responsive glucose transporter 4 in 3T3-L1 adipocytes. Mol Biol Cell [67] Fujita H, Hatakeyama H, Watanabe TM, Sato M, Higuchi H, Kanzaki M.
1999;10:3675–88. Identification of three distinct functional sites of insulin-mediated GLUT4
[47] Lampson MA, Racz A, Cushman SW, McGraw TE. Demonstration of trafficking in adipocytes using quantitative single molecule imaging. Mol
insulin-responsive trafficking of GLUT4 and vpTR in fibroblasts. J Cell Biol Cell 2010;21:2721–31.
Sci 2000;113:4065–76. [68] Huang S, Lifshitz LM, Jones C, Bellve KD, Standley C, Fonseca S, et al.
[48] Zaarour N, Berenguer M, Le Marchand-Brustel Y, Govers R. Deciphering Insulin stimulates membrane fusion and GLUT4 accumulation in clathrin
the role of GLUT4 N-glycosylation in adipocyte and muscle cell models. coats on adipocyte plasma membranes. Mol Cell Biol 2007;27:3456–69.
Biochem J 2012;445:265–73. [69] Jiang L, Fan J, Bai L, Wang Y, Chen Y, Yang L, et al. Direct quantification
[49] Becker C, Sevilla L, Tomas E, Palacin M, Zorzano A, Fischer Y. The endo- of fusion rate reveals a distal role for AS160 in insulin-stimulated fusion
somal compartment is an insulin-sensitive recruitment site for GLUT4 of GLUT4 storage vesicles. J Biol Chem 2008;283:8508–16.
and GLUT1 glucose transporters in cardiac myocytes. Endocrinology [70] Bai L, Wang Y, Fan J, Chen Y, Ji W, Qu A, et al. Dissecting multiple
2001;142:5267–76. steps of GLUT4 trafficking and identifying the sites of insulin action.
[50] Berenguer M, Le Marchand-Brustel Y, Govers R. GLUT4 molecules Cell Metab 2007;5:47–57.
are recruited at random for insertion within the plasma membrane upon [71] Lizunov VA, Lee JP, Skarulis MC, Zimmerberg J, Cushman SW, Stenkula
insulin stimulation. FEBS Lett 2010;584:537–42. KG. Impaired tethering and fusion of GLUT4 vesicles in insulin-resistant
[51] Blok J, Gibbs EM, Lienhard GE, Slot JW, Geuze HJ. Insulin-induced human adipose cells. Diabetes 2013;62:3114–9.
translocation of glucose transporters from post-Golgi compartments to
R. Govers / Diabetes & Metabolism 40 (2014) 400–410 409

[72] Coster AC, Govers R, James DE. Insulin stimulates the entry of GLUT4 [90] Chiang SH, Baumann CA, Kanzaki M, Thurmond DC, Watson RT, Neu-
into the endosomal recycling pathway by a quantal mechanism. Traffic dauer CL, et al. Insulin-stimulated GLUT4 translocation requires the
2004;5:763–71. CAP-dependent activation of TC10. Nature 2001;410:944–8.
[73] Satoh S, Nishimura H, Clark AE, Kozka IJ, Vannucci SJ, Simpson IA, [91] Inoue M, Chang L, Hwang J, Chiang SH, Saltiel AR. The exocyst complex
et al. Use of bismannose photolabel to elucidate insulin-regulated GLUT4 is required for targeting of Glut4 to the plasma membrane by insulin.
subcellular trafficking kinetics in rat adipose cells. Evidence that exocy- Nature 2003;422:629–33.
tosis is a critical site of hormone action. J Biol Chem 1993;268:17820–9. [92] Chang L, Chiang SH, Saltiel AR. TC10alpha is required for insulin-
[74] Yang J, Holman GD. Comparison of GLUT4 and GLUT1 subcellular stimulated glucose uptake in adipocytes. Endocrinology 2007;148:
trafficking in basal and insulin-stimulated 3T3-L1 cells. J Biol Chem 27–33.
1993;268:4600–3. [93] Okada S, Yamada E, Saito T, Ohshima K, Hashimoto K, Yamada M,
[75] Lee W, Ryu J, Spangler RA, Jung CY. Modulation of GLUT4 and et al. CDK5-dependent phosphorylation of the Rho family GTPase
GLUT1 recycling by insulin in rat adipocytes: kinetic analysis based TC10(alpha) regulates insulin-stimulated GLUT4 translocation. J Biol
on the involvement of multiple intracellular compartments. Biochemistry Chem 2008;283:35455–63.
2000;39:9358–66. [94] Liu J, Kimura A, Baumann CA, Saltiel AR. APS facilitates c-Cbl tyrosine
[76] Blot V, McGraw TE. GLUT4 is internalized by a cholesterol- phosphorylation and GLUT4 translocation in response to insulin in 3T3-
dependent nystatin-sensitive mechanism inhibited by insulin. Embo J L1 adipocytes. Mol Cell Biol 2002;22:3599–609.
2006;25:5648–58. [95] Ribon V, Printen JA, Hoffman NG, Kay BK, Saltiel AR. A novel, mul-
[77] Ros-Baro A, Lopez-Iglesias C, Peiro S, Bellido D, Palacin M, Zorzano tifuntional c-Cbl binding protein in insulin receptor signaling in 3T3-L1
A, et al. Lipid rafts are required for GLUT4 internalization in adipose adipocytes. Mol Cell Biol 1998;18:872–9.
cells. Proc Natl Acad Sci U S A 2001;98:12050–5. [96] Mitra P, Zheng X, Czech MP. RNAi-based analysis of CAP, Cbl, and
[78] Stenkula KG, Lizunov VA, Cushman SW, Zimmerberg J. Insulin controls CrkII function in the regulation of GLUT4 by insulin. J Biol Chem
the spatial distribution of GLUT4 on the cell surface through regulation 2004;279:37431–5.
of its postfusion dispersal. Cell Metab 2010;12:250–9. [97] Lizunov VA, Lisinski I, Stenkula K, Zimmerberg J, Cushman SW. Insulin
[79] Lizunov VA, Stenkula K, Troy A, Cushman SW, Zimmerberg J. Insulin regulates fusion of GLUT4 vesicles independent of Exo70-mediated
regulates Glut4 confinement in plasma membrane clusters in adipose tethering. J Biol Chem 2009;284:7914–9.
cells. PLoS One 2013;8:e57559. [98] Hoffman NJ, Elmendorf JS. Signaling, cytoskeletal and membrane
[80] Kotani K, Carozzi AJ, Sakaue H, Hara K, Robinson LJ, Clark SF, et al. mechanisms regulating GLUT4 exocytosis. Trends Endocrinol Metab
Requirement for phosphoinositide 3-kinase in insulin-stimulated GLUT4 2011;22:110–6.
translocation in 3T3-L1 adipocytes. Biochem Biophys Res Commun [99] Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegel-
1995;209:343–8. man BM. IRS-1-mediated inhibition of insulin receptor tyrosine kinase
[81] Hill MM, Clark SF, Tucker DF, Birnbaum MJ, James DE, Macaulay activity in TNF-alpha- and obesity-induced insulin resistance. Science
SL. A role for protein kinase Bbeta/Akt2 in insulin-stimulated GLUT4 1996;271:665–8.
translocation in adipocytes. Mol Cell Biol 1999;19:7771–81. [100] Morino K, Neschen S, Bilz S, Sono S, Tsirigotis D, Reznick RM, et al.
[82] Kupriyanova TA, Kandror KV. Akt-2 binds to Glut4-containing vesicles Muscle-specific IRS-1 Ser->Ala transgenic mice are protected from fat-
and phosphorylates their component proteins in response to insulin. J Biol induced insulin resistance in skeletal muscle. Diabetes 2008;57:2644–51.
Chem 1999;274:1458–64. [101] Teruel T, Hernandez R, Lorenzo M. Ceramide mediates insulin resistance
[83] Ng Y, Ramm G, Lopez JA, James DE. Rapid activation of Akt2 is suffi- by tumor necrosis factor-alpha in brown adipocytes by maintaining Akt
cient to stimulate GLUT4 translocation in 3T3-L1 adipocytes. Cell Metab in an inactive dephosphorylated state. Diabetes 2001;50:2563–71.
2008;7:348–56. [102] Tonks KT, Ng Y, Miller S, Coster AC, Samocha-Bonet D, Iseli TJ,
[84] Gonzalez E, McGraw TE. Insulin signaling diverges into Akt-dependent et al. Impaired Akt phosphorylation in insulin-resistant human muscle is
and -independent signals to regulate the recruitment/docking and the accompanied by selective and heterogeneous downstream defects. Dia-
fusion of GLUT4 vesicles to the plasma membrane. Mol Biol Cell betologia 2013;56:875–85.
2006;17:4484–93. [103] Ng Y, Ramm G, James DE. Dissecting the mechanism of insulin resistance
[85] Kotani K, Ogawa W, Matsumoto M, Kitamura T, Sakaue H, Hino Y, et al. using a novel heterodimerization strategy to activate Akt. J Biol Chem
Requirement of atypical protein kinase C-lambda for insulin stimulation 2010;285:5232–9.
of glucose uptake but not for Akt activation in 3T3-L1 adipocytes. Mol [104] Gupte A, Mora S. Activation of the Cbl insulin signaling pathway in
Cell Biol 1998;18:6971–82. cardiac muscle; dysregulation in obesity and diabetes. Biochem Biophys
[86] Standaert ML, Bandyopadhyay G, Perez L, Price D, Galloway L, Pokle- Res Commun 2006;342:751–7.
povic A, et al. Insulin activates protein kinases C-zeta and C-lambda by an [105] Jun HS, Hwang K, Kim Y, Park T. High-fat diet alters PP2A, TC10, and
autophosphorylation-dependent mechanism and stimulates their translo- CIP4 expression in visceral adipose tissue of rats. Obesity (Silver Spring)
cation to GLUT4 vesicles and other membrane fractions in rat adipocytes. 2008;16:1226–31.
J Biol Chem 1999;274:25308–16. [106] Kane S, Sano H, Liu SC, Asara JM, Lane WS, Garner CC, et al. A method
[87] Braiman L, Alt A, Kuroki T, Ohba M, Bak A, Tennenbaum T, et al. Activa- to identify serine kinase substrates. Akt phosphorylates a novel adipocyte
tion of protein kinase c-zeta induces serine phosphorylation of vamp2 in protein with a Rab GTPase-activating protein (GAP) domain. J Biol Chem
the glut4 compartment and increases glucose transport in skeletal muscle. 2002;277:22115–8.
Mol Cell Biol 2001;21:7852–61. [107] Lansey MN, Walker NN, Hargett SR, Stevens JR, Keller SR. Deletion of
[88] Bandyopadhyay G, Standaert ML, Kikkawa U, Ono Y, Moscat J, Farese Rab-GAP AS160 modifies glucose uptake and GLUT4 translocation in
RV. Effects of transiently expressed atypical (zeta, lambda), conven- primary skeletal muscles and adipocytes and impairs glucose homeosta-
tional (alpha, beta) and novel (delta, epsilon) protein kinase C isoforms sis. Am J Physiol Endocrinol Metab 2012;303:E1273–86.
on insulin-stimulated translocation of epitope-tagged GLUT4 glucose [108] Miinea CP, Sano H, Kane S, Sano E, Fukuda M, Peranen J, et al. AS160,
transporters in rat adipocytes: specific interchangeable effects of protein the Akt substrate regulating GLUT4 translocation, has a functional Rab
kinases C-zeta and C-lambda. Biochem J 1999;337(Pt 3):461–70. GTPase-activating protein domain. Biochem J 2005;391:87–93.
[89] Kayali AG, Eichhorn J, Haruta T, Morris AJ, Nelson JG, Vollenweider [109] Sano H, Kane S, Sano E, Miinea CP, Asara JM, Lane WS, et al. Insulin-
P, et al. Association of the insulin receptor with phospholipase C-gamma stimulated phosphorylation of a Rab GTPase-activating protein regulates
(PLCgamma) in 3T3-L1 adipocytes suggests a role for PLCgamma in GLUT4 translocation. J Biol Chem 2003;278:14599–602.
metabolic signaling by insulin. J Biol Chem 1998;273:13808–18. [110] Ren W, Cheema S, Du K. The association of ClipR-59 protein with AS160
modulates AS160 protein phosphorylation and adipocyte Glut4 protein
membrane translocation. J Biol Chem 2012;287:26890–900.
410 R. Govers / Diabetes & Metabolism 40 (2014) 400–410

[111] Eguez L, Lee A, Chavez JA, Miinea CP, Kane S, Lienhard GE, et al. Full [131] Jewell JL, Oh E, Thurmond DC. Exocytosis mechanisms underlying
intracellular retention of GLUT4 requires AS160 Rab GTPase-activating insulin release and glucose uptake: conserved roles for Munc18c and
protein. Cell Metab 2005;2:263–72. syntaxin 4. Am J Physiol Regul Integr Comp Physiol 2010;298:R517–31.
[112] Brewer PD, Romenskaia I, Kanow MA, Mastick CC. Loss of AS160 Akt [132] Cheatham B, Volchuk A, Kahn CR, Wang L, Rhodes CJ, Klip A.
substrate causes Glut4 protein to accumulate in compartments that are Insulin-stimulated translocation of GLUT4 glucose transporters requires
primed for fusion in basal adipocytes. J Biol Chem 2011;286:26287–97. SNARE-complex proteins. Proc Natl Acad Sci U S A 1996;93:15169–73.
[113] Tan SX, Ng Y, Burchfield JG, Ramm G, Lambright DG, Stockli J, et al. [133] Macaulay SL, Hewish DR, Gough KH, Stoichevska V, MacPherson SF,
The Rab GTPase-activating protein TBC1D4/AS160 contains an atypical Jagadish M, et al. Functional studies in 3T3-L1 cells support a role for
phosphotyrosine-binding domain that interacts with plasma membrane SNARE proteins in insulin stimulation of GLUT4 translocation. Biochem
phospholipids to facilitate GLUT4 trafficking in adipocytes. Mol Cell J 1997;324:217–24.
Biol 2012;32:4946–59. [134] Olson AL, Knight JB, Pessin JE. Syntaxin 4, VAMP2, and/or
[114] Sadacca LA, Bruno J, Wen J, Xiong W, McGraw TE. Specialized sor- VAMP3/cellubrevin are functional target membrane and vesicle SNAP
ting of GLUT4 and its recruitment to the cell surface are independently receptors for insulin-stimulated GLUT4 translocation in adipocytes. Mol
regulated by distinct Rabs. Mol Biol Cell 2013;24:2544–57. Cell Biol 1997;17:2425–35.
[115] Yoshizaki T, Imamura T, Babendure JL, Lu JC, Sonoda N, Olefsky [135] Zhao P, Yang L, Lopez JA, Fan J, Burchfield JG, Bai L, et al. Variations
JM. Myosin 5a is an insulin-stimulated Akt2 (protein kinase Bbeta) in the requirement for v-SNAREs in GLUT4 trafficking in adipocytes. J
substrate modulating GLUT4 vesicle translocation. Mol Cell Biol Cell Sci 2009;122:3472–80.
2007;27:5172–83. [136] Yu H, Rathore SS, Davis EM, Ouyang Y, Shen J. Doc2b promotes
[116] Li J, Malaby AW, Famulok M, Sabe H, Lambright DG, Hsu VW. Grp1 GLUT4 exocytosis by activating the SNARE-mediated fusion reaction
plays a key role in linking insulin signaling to glut4 recycling. Dev Cell in a calcium- and membrane bending-dependent manner. Mol Biol Cell
2012;22:1286–98. 2013;24:1176–84.
[117] Millar CA, Powell KA, Hickson GR, Bader MF, Gould GW. Evidence [137] Perera HK, Clarke M, Morris NJ, Hong W, Chamberlain LH, Gould GW.
for a role for ADP-ribosylation factor 6 in insulin-stimulated glucose Syntaxin 6 regulates Glut4 trafficking in 3T3-L1 adipocytes. Mol Biol
transporter-4 (GLUT4) trafficking in 3T3-L1 adipocytes. J Biol Chem Cell 2003;14:2946–58.
1999;274:17619–25. [138] Proctor KM, Miller SC, Bryant NJ, Gould GW. Syntaxin 16 controls
[118] Xie X, Gong Z, Mansuy-Aubert V, Zhou QL, Tatulian SA, Sehrt D, the intracellular sequestration of GLUT4 in 3T3-L1 adipocytes. Biochem
et al. C2 domain-containing phosphoprotein CDP138 regulates GLUT4 Biophys Res Commun 2006;347:433–8.
insertion into the plasma membrane. Cell Metab 2011;14:378–89. [139] Mallard F, Tang BL, Galli T, Tenza D, Saint-Pol A, Yue X, et al.
[119] Min J, Okada S, Kanzaki M, Elmendorf JS, Coker KJ, Ceresa BP, et al. Early/recycling endosomes-to-TGN transport involves two SNARE-
Synip: a novel insulin-regulated syntaxin-4 binding protein mediating complexes and a Rab6 isoform. J Cell Biol 2002;156:653–64.
GLUT4 translocation in adipocytes. Mol Cell 1999;3:751–60. [140] Roccisana J, Sadler JB, Bryant NJ, Gould GW. Sorting of GLUT4 into its
[120] Okada S, Ohshima K, Uehara Y, Shimizu H, Hashimoto K, Yamada insulin-sensitive store requires the Sec1/Munc18 protein mVps45. Mol
M, et al. Synip phosphorylation is required for insulin-stimulated Glut4 Biol Cell 2013;24:2389–97.
translocation. Biochem Biophys Res Commun 2007;356:102–6. [141] Tamori Y, Kawanishi M, Niki T, Shinoda H, Araki S, Okazawa H,
[121] Yamada E, Okada S, Saito T, Ohshima K, Sato M, Tsuchiya T, et al. et al. Inhibition of insulin-induced GLUT4 translocation by Munc18c
Akt2 phosphorylates Synip to regulate docking and fusion of GLUT4- through interaction with syntaxin4 in 3T3-L1 adipocytes. J Biol Chem
containing vesicles. J Cell Biol 2005;168:921–8. 1998;273:19740–6.
[122] Berwick DC, Dell GC, Welsh GI, Heesom KJ, Hers I, Fletcher LM, et al. [142] Thurmond DC, Ceresa BP, Okada S, Elmendorf JS, Coker K, Pessin JE.
Protein kinase B phosphorylation of PIKfyve regulates the trafficking of Regulation of insulin-stimulated GLUT4 translocation by Munc18c in
GLUT4 vesicles. J Cell Sci 2004;117:5985–93. 3T3-L1 adipocytes. J Biol Chem 1998;273:33876–83.
[123] Sano H, Eguez L, Teruel MN, Fukuda M, Chuang TD, Chavez JA, et al. [143] Schmelzle K, Kane S, Gridley S, Lienhard GE, White FM. Tempo-
Rab10, a target of the AS160 Rab-GAP, is required for insulin-stimulated ral dynamics of tyrosine phosphorylation in insulin signaling. Diabetes
translocation of GLUT4 to the adipocyte plasma membrane. Cell Metab 2006;55:2171–9.
2007;5:293–303. [144] Jewell JL, Oh E, Ramalingam L, Kalwat MA, Tagliabracci VS, Tackett L,
[124] Sano H, Roach WG, Peck GR, Fukuda M, Lienhard GE. Rab10 in insulin- et al. Munc18c phosphorylation by the insulin receptor links cell signaling
stimulated GLUT4 translocation. Biochem J 2008;411:89–95. directly to SNARE exocytosis. J Cell Biol 2011;193:185–99.
[125] Chen Y, Lippincott-Schwartz J. Rab10 delivers GLUT4 storage vesicles [145] Kanda H, Tamori Y, Shinoda H, Yoshikawa M, Sakaue M, Uda-
to the plasma membrane. Commun Integr Biol 2013;6:e23779. gawa J, et al. Adipocytes from Munc18c-null mice show increased
[126] Sano H, Peck GR, Kettenbach AN, Gerber SA, Lienhard GE. Insulin- sensitivity to insulin-stimulated GLUT4 externalization. J Clin Invest
stimulated GLUT4 protein translocation in adipocytes requires the 2005;115:291–301.
Rab10 guanine nucleotide exchange factor Dennd4C. J Biol Chem [146] Spurlin BA, Thomas RM, Nevins AK, Kim HJ, Kim YJ, Noh HL, et al.
2011;286:16541–5. Insulin resistance in tetracycline-repressible Munc18c transgenic mice.
[127] Reed SE, Hodgson LR, Song S, May MT, Kelly EE, McCaffrey MW, Diabetes 2003;52:1910–7.
et al. A role for Rab14 in the endocytic trafficking of GLUT4 in 3T3-L1 [147] Fukuda N, Emoto M, Nakamori Y, Taguchi A, Miyamoto S, Uraki S, et al.
adipocytes. J Cell Sci 2013;126:1931–41. DOC2B: a novel syntaxin-4 binding protein mediating insulin-regulated
[128] Lodhi IJ, Chiang SH, Chang L, Vollenweider D, Watson RT, Inoue M, GLUT4 vesicle fusion in adipocytes. Diabetes 2009;58:377–84.
et al. Gapex-5, a Rab31 guanine nucleotide exchange factor that regulates
Glut4 trafficking in adipocytes. Cell Metab 2007;5:59–72.
[129] Huang J, Imamura T, Olefsky JM. Insulin can regulate GLUT4 internal-
ization by signaling to Rab5 and the motor protein dynein. Proc Natl Acad
Sci U S A 2001;98:13084–9.
[130] Cormont M, Bortoluzzi MN, Gautier N, Mari M, van Obberghen E, Le
Marchand-Brustel Y. Potential role of Rab4 in the regulation of sub-
cellular localization of Glut4 in adipocytes. Mol Cell Biol 1996;16:
6879–86.

You might also like