Insulin rapidly stimulates GLUT4 translocation and glucose transport in fat and muscle cells. Signals from the occupied insulin receptor are translated into downstream signalling changes in serine/threonine kinases within timescales of seconds, and this is followed by delivery and accumulation of the glucose transporter GLUT4 at the plasma membrane. Kinetic studies have led to realisation that there are distinct phases of this stimulation by insulin. There is a rapid initial burst of GLUT4 delivered to the cell surface from a subcellular reservoir compartment and this is followed by a steady-state level of continuing stimulation in which GLUT4 recycles through a large itinerary of subcellular locations. Here, we provide an overview of the phases of insulin stimulation of GLUT4 translocation and the molecules that are currently considered to activate these trafficking steps. Furthermore, we suggest how use of new experimental approaches together with phospho-proteomic data may help to further identify mechanisms for activation of these trafficking processes.

In the 100th year of the discovery of insulin we reflect on a famous cartoon of the 1970s in which ‘Chuck’ describes the black box theory of insulin action and the following sequence: insulin binds to its receptor; something happens; cellular effects [1]. Since that time, some 45 years later, the black box ‘something happens’ could be replaced by ‘lots of things happen, but not all of them happen quickly’. So, what can we now learn from the available catalogue of necessary proteins, their subcellular compartments, and the kinetics of the insulin response?

Pioneering cell biology studies [2,3] revealed that insulin stimulates glucose transport into fat and muscle cells through translocation of the glucose transporter from intracellular stores to the plasma membrane (PM). Since the discovery of GLUT4 in the late 1980s [4–6], studies have shed light on the GLUT4 complex intracellular trafficking itinerary that effectively excludes GLUT4 from the PM in the absence of insulin and provides a large reservoir of GLUT4 that can be readily mobilised in response to insulin. Very early in these studies it became evident that insulin stimulation of glucose uptake and GLUT4 translocation is a very fast process, and that maximum glucose uptake is achieved within 10 min of insulin binding to its receptor (Table 1).

Table 1.
Key signalling and trafficking components in GLUT4 translocation. See key at foot of Table p5
processkey effectorsinsulin regulatedPI3K dependentAkt dependenttime scale of activation including phosphorylation (full activation unless otherwise indicated)
Phosphorylation times from ref. [36]
Overall process 
Glucose transport  Yes Yes Yes ½ time = 5–10 min 
Signalling 
Formation of the IR–IRS–PI3K complex* Insulin receptor, IRS, PI3K [36,106,107Yes   <1 min
InsR and IRS1/2 pTyr in 15 s 
IP3 synthesis at the PM* PI3K Class 1a [13,33,108–112Yes Yes  <1 min
½ time — 2.4 s [33
Akt activation* Akt, PDK1, mTORC2 [16,36,107Yes Yes
Yes 
Yes
Yes 
Akt translocation <1 min; Akt phosphorylation <1 min (Akt2 pThr309 in 15 s; pSer474 in 30 s); Akt activation (substrate phosphorylation) <1 min (30 s) [36]
mTORC2 multiple phosphosites, full activation within 5 min 
Actin remodelling EHD1* [113], EHBP1 [113], EHD2 [106,114Yes Yes Yes pTyr453 EHD2 within 5 min [106]
EHD1 — pSer456 in 20 min
EHBP1 — pSer1061 in 30 s
EHD2 — pSer451 in 15 s — only PI3K-dependent 
CAP, TC10* [74,115,116Yes No
No 
No
No 
CAP — multiple Ser/Thr phosphosites some, as Ser287, are activated in 5 min and are PI3K- and Akt-dependent perhaps via a feedback loop.
TC10 — pSer9 in 1 min
No time course of GTP loading reported 
Tropomodulin3 [117Yes Yes
No 
Yes
No 
pSer71 in 2–5 min 
Tropomyosin3.1 [118Yes N.R.
No 
N.R.
No 
pSer51 in 20–60 min 
CLASP2 (microtubules, membrane ruffles) [119,120Yes No No Several Ser phosphosites in 2–60 min
Serine phosphorylation via MAPK [120
Rac1 [121,122Yes Yes No NPD 
Rab13, MICAL-L2, Actinin4* [123Yes Yes
Yes 
No
No 
2–5 min time course of GTP loading
Rab13 pTyr5 in 15 s pSer178 in 10 min
MICAL-L2-pSer143 in 15 s;
Actinin4 — pSer424 in 5 min (transient) 
GLUT4 vesicle trafficking 
Translocation of GSVs to PM and tethering of GSV at PM TBC1D4* [34,35,58,100,124–127Yes Yes
Yes 
Yes
Yes 
TBC1D4 phosphorylation 15–30 s on different Ser/Thr residues
Interaction with 14-3-3 [58,125,127], IRAP [100,128
Rab10 (GTP loading) [67No N.R.
Yes 
N.R.
No 
pTyr6 in 15 s; Slower pThr73 in 10 min 
DENND4c (Rab GEF) [92No (GEF activity) N.R.
Yes 
N.R.
Yes 
pSer1043, pSer1096 and pSer1321 insulin-sensitive phosphorylation.
Multiple phosphosites; pSer 1289 fastest in 2–5 min 
Sec16 [63,129,130Yes N.R.
Yes 
Yes
No 
Sec16a multiple phosphosites. Fastest pSer2311 in 2 min
Sec16b pSer182/pSer185 Moderate activation 30 s 
TBC1D1 [127,131–133Yes

 
Yes Yes 5 phosphosites detected, only pSer231 is insulin sensitive, but not above threshold value of 0.5 
Rab8 and Rab13 [123,134Yes N.R.
No 
N.R.
No 
2–5 min time course of GTP loading
Rab8: Transient phosphorylation pSer181/185 30 s to 1 min
Rab13 see above 
MyoVA [68,135,123,134Yes Yes
No 
Yes
No 
Akt substrate pSer1650 in 5–10 min
pSer600 and pSer1650 in 10 min 
Synip [69Yes Yes
No 
Yes
No 
Akt substrate, phosphorylation in 5–10 min
Only pSer12 at 60 min 
RGC2/AS250 (RalGAP)* [71Yes Yes
Yes 
Yes
Yes 
Akt substrate, phosphorylation within 5 min
Multiple phosphosites. Fastest Thr715 in 15 s + pSer486/696/765 in 30 s 
RalA [71,75Yes Yes Yes <5 min for GTP loading
NISPD 
Exocyst (Exo70, Exo 84 [77,96,136], Sec6 [137], Sec8 [138]) Yes No
No 
N.R.
No 
Exo 84 is dependent on transient phosphorylation (2–5 min) of TBK1.
Exo70 pSer 242/245/250 at 60 min; Sec 8 pSer32 in 5–10 min. Exo84 NISPD 
Myo1C [75,139–141,142Yes Yes
No 
No
No 
Phosphorylation 2–5 min
Small activation at 2 min pSer375 
CAMKII [141Yes N.R.
No 
No
No 
Several phosphosites activated in 60 min 
Rab3(B,D), Noc2 [73Yes No No <5 min for GTP loading
NPD
Rab3GAP1 and Rab3IP are phosphorylated within 30 s 
GSV docking, fusion with PM and dispersal of GLUT4 in the PM Munc18c* [72,86,90,91,106Yes No
No 
No
No 
pTyr 5 min
pSer588 in 15 s to 2 min 
Doc2B [143Yes N.R. N.R. NPD 
Tomosyn1/2* [70,97,144,145Yes Yes
No 
Yes
No 
Akt substrate Ser783[70]. Timing <5 min
pSer724/786/783 all within 15–30 s 
Syntaxin4* [79,146Yes Yes
Yes 
Yes
Yes 
Stx4 pY [36,106] 30 s for full activation
pTyr251 30 s;
Multiples phosphosites: Ser208 in 15 s; pSer248 transient 2–5 min but no PI3K or Akt dependence 
SNAP23* [80,81N.R. N.R.
No 
N.R.
No 
pThr24 transient 30 s to 2 min 
Vamp2 [57,78,147N.R. N.R.
Yes 
N.R.
Yes 
pSer48 in 5–10 min 
Complexin-2 [148Yes N.R.
No 
No
No 
pSer93 in 30 s 
NSF and αSNAP* [93,95,149,150N.R. N.R.
No 
N.R.
No 
NSF: pSer207 in 15 s
αSNAP: Ser24 in 20–60min 
EFR3 and PI 4-kinase IIIα* [66N.R. N.R.
Yes 
N.R.
Yes 
EFR3: very low Ser360/363 in 15 s 
GLUT4 internalisation (endocytosis) Clathrin [98,151Yes N.R.
No 
N.R.
No 
Heavy chain: pTyr434 in 15 s 
Dynamin-2* [152–154Yes N.R.
No 
N.R.
No 
NPD but Dynamin-1 (Dnm) detected with multiple phosphosites — some in 15 s or 30 s 
CIP4/2 [74]
N-WASp, CIP4* [155
Yes
Yes 
N.R.
No 
N.R.
No 
CIP4 Multiple phosphosites pSer296 in 15 s
N-WASP: pSer426 1–2 min 
Cholesterol, nystatin endocytosis [101Yes N.R. N.R.  
EEA1, Rab5, GAPEX5, Rab31 [156–158Yes N.R.
Yes 
N.R.
No 
EEA1: pSer720 in 10 and 60 min;
Rab5a: pSer193 — 60min
GAPEX5: Multiple phosphosites pSer742, pSer1017 in 5–10 min Rab31: NPD 
Ub, USP25* [159,160N.R. N.R. N.R. USP25: Transient pThr61 at 30 s 
Caveolae* [116,161–163No N.R.
Yes 
N.R.
No 
Multiple phosphosites on Caveolin-1 pTyr phosphorylated within 30 s 
GLUT4 return to GSV compartment and sorting away from recycling endosomes LRP1 [164Yes N.R.
Yes 
N.R.
Yes 
pSer4524 and pThr4525 at 60 min 
IRAP [165–169Yes Yes
Yes 
Yes
Yes 
pSer51 in 5 min; pTyr70 high 1–10 min transient 
Sortilin [170–173Yes N.R.
No 
N.R.
No 
pSer824 marginal (∼0.5) 15–20 min 
GGA, AP1* [174–177N.R. N.R.
Yes 
N.R.
No 
GGA: pSer414/418 in 5 min LY only
AP1(subunit mu): pThr152/154 in 30 s and 15 s resp. LY only
Other AP1 subunit NPD 
ESCRT [178N.R. N.R.
No 
N.R.
No 
Chmp3 and Vps4 NPD
Chmp2b phosphorylated in 15 s 
Retromer [166,179,180N.R. N.R.
Yes 
N.R.
Yes 
Vps35: pSer7 at 60 min
Vps26a NPD
Vps26b: multiple phosphosites and pSer302 in 2–5 min
Vps29 NPD
Snx3 and 27: some phosphorylation in 60 min. Not PI3K- or Akt-dependent 
Golgin97 and Vti1a [170N.R. N.R. N.R. NISPD 
Syntaxin 6* [181], Syntaxin 16 [182], mVps45 [183Yes (Stx16) N.R.
No† 
N.R.
No 
Stx16 dephosphorylation within 30min [181]
Stx16 some phosphorylation detected at long timepoints 60 min. Dephosphorylation for Ser35 detected since 15 s 
Rab11, Rip11* [184,185Yes Yes
Yes 
No
Yes 
Rab11b: Multiple phosphosites some activated at 2–10 min
Rip11: Multiple phosphosites, some pSer188 and 480 very fast 15 s 
Rab4, KIF13 (motor protein) [135,186–188Yes Yes
Yes 
N.R.
No 
Slow GTP loading over 45 min
Rab4 NPD
Kif13: Fast 30 s multiple sites pThr1717 
Rab14 [55,189N.R. N.R.
No 
N.R.
Yes 
2 min pSer97 
TUG [190–192N.R. N.R.
Yes 
N.R.
No 
Proteolytic cleavage in 10 min
pSer279 at 5 min 
TRARG1(TUSC5) [193,194N.R. N.R.
Yes 
N.R.
Yes 
Multiple phosphosites Ser56, 70 and 72 in 20–60 min 
TBC1D13, Rab35 [195N.R. N.R.
No 
N.R.
Yes 
TBC1D13: Ser184 in 20–60 min 
CHC22 [196N.R. N.R. N.R. NPD 
Budding of GSV, PLD [61,62Yes N.R. N.R. PLD1 — NISPD 
Delivery of newly synthesised GLUT4 to GSVs and maintaining insulin responsiveness Sortilin [170–172Yes N.R. N.R. See above 
syntaxin 16, mVps45, syntaxin 6 [181–183Yes (Stx16) N.R. N.R. Stx16 dephosphorylation within 30 min [181]. See above 
GGA, AP1 [174–177N.R. N.R. N.R. See above 
ACAP1, Arf6 [197Yes N.R. N.R. NPD 
VAMP7 (TI-VAMP), VAMP4 [198N.R. N.R.
Yes 
N.R.
No 
Vamp4 — Multiple phosphosites pSer17/20 in 1–5 min 
Axin, TNKS [199–201N.R. N.R. N.R. Axin1 — multiple sites but below 0.5 
P115 [202N.R. N.R.
No 
N.R.
No 
pSer717 slow 60 min 
Sec16 [63,129,130N.R. N.R. N.R. See above 
CHC22 [196N.R. N.R. N.R. NPD 
Transfected GLUT4 becoming insulin responsive N.R. N.R. N.R. 6–9 h [174]
3.5 h in primary adipocytes [152
processkey effectorsinsulin regulatedPI3K dependentAkt dependenttime scale of activation including phosphorylation (full activation unless otherwise indicated)
Phosphorylation times from ref. [36]
Overall process 
Glucose transport  Yes Yes Yes ½ time = 5–10 min 
Signalling 
Formation of the IR–IRS–PI3K complex* Insulin receptor, IRS, PI3K [36,106,107Yes   <1 min
InsR and IRS1/2 pTyr in 15 s 
IP3 synthesis at the PM* PI3K Class 1a [13,33,108–112Yes Yes  <1 min
½ time — 2.4 s [33
Akt activation* Akt, PDK1, mTORC2 [16,36,107Yes Yes
Yes 
Yes
Yes 
Akt translocation <1 min; Akt phosphorylation <1 min (Akt2 pThr309 in 15 s; pSer474 in 30 s); Akt activation (substrate phosphorylation) <1 min (30 s) [36]
mTORC2 multiple phosphosites, full activation within 5 min 
Actin remodelling EHD1* [113], EHBP1 [113], EHD2 [106,114Yes Yes Yes pTyr453 EHD2 within 5 min [106]
EHD1 — pSer456 in 20 min
EHBP1 — pSer1061 in 30 s
EHD2 — pSer451 in 15 s — only PI3K-dependent 
CAP, TC10* [74,115,116Yes No
No 
No
No 
CAP — multiple Ser/Thr phosphosites some, as Ser287, are activated in 5 min and are PI3K- and Akt-dependent perhaps via a feedback loop.
TC10 — pSer9 in 1 min
No time course of GTP loading reported 
Tropomodulin3 [117Yes Yes
No 
Yes
No 
pSer71 in 2–5 min 
Tropomyosin3.1 [118Yes N.R.
No 
N.R.
No 
pSer51 in 20–60 min 
CLASP2 (microtubules, membrane ruffles) [119,120Yes No No Several Ser phosphosites in 2–60 min
Serine phosphorylation via MAPK [120
Rac1 [121,122Yes Yes No NPD 
Rab13, MICAL-L2, Actinin4* [123Yes Yes
Yes 
No
No 
2–5 min time course of GTP loading
Rab13 pTyr5 in 15 s pSer178 in 10 min
MICAL-L2-pSer143 in 15 s;
Actinin4 — pSer424 in 5 min (transient) 
GLUT4 vesicle trafficking 
Translocation of GSVs to PM and tethering of GSV at PM TBC1D4* [34,35,58,100,124–127Yes Yes
Yes 
Yes
Yes 
TBC1D4 phosphorylation 15–30 s on different Ser/Thr residues
Interaction with 14-3-3 [58,125,127], IRAP [100,128
Rab10 (GTP loading) [67No N.R.
Yes 
N.R.
No 
pTyr6 in 15 s; Slower pThr73 in 10 min 
DENND4c (Rab GEF) [92No (GEF activity) N.R.
Yes 
N.R.
Yes 
pSer1043, pSer1096 and pSer1321 insulin-sensitive phosphorylation.
Multiple phosphosites; pSer 1289 fastest in 2–5 min 
Sec16 [63,129,130Yes N.R.
Yes 
Yes
No 
Sec16a multiple phosphosites. Fastest pSer2311 in 2 min
Sec16b pSer182/pSer185 Moderate activation 30 s 
TBC1D1 [127,131–133Yes

 
Yes Yes 5 phosphosites detected, only pSer231 is insulin sensitive, but not above threshold value of 0.5 
Rab8 and Rab13 [123,134Yes N.R.
No 
N.R.
No 
2–5 min time course of GTP loading
Rab8: Transient phosphorylation pSer181/185 30 s to 1 min
Rab13 see above 
MyoVA [68,135,123,134Yes Yes
No 
Yes
No 
Akt substrate pSer1650 in 5–10 min
pSer600 and pSer1650 in 10 min 
Synip [69Yes Yes
No 
Yes
No 
Akt substrate, phosphorylation in 5–10 min
Only pSer12 at 60 min 
RGC2/AS250 (RalGAP)* [71Yes Yes
Yes 
Yes
Yes 
Akt substrate, phosphorylation within 5 min
Multiple phosphosites. Fastest Thr715 in 15 s + pSer486/696/765 in 30 s 
RalA [71,75Yes Yes Yes <5 min for GTP loading
NISPD 
Exocyst (Exo70, Exo 84 [77,96,136], Sec6 [137], Sec8 [138]) Yes No
No 
N.R.
No 
Exo 84 is dependent on transient phosphorylation (2–5 min) of TBK1.
Exo70 pSer 242/245/250 at 60 min; Sec 8 pSer32 in 5–10 min. Exo84 NISPD 
Myo1C [75,139–141,142Yes Yes
No 
No
No 
Phosphorylation 2–5 min
Small activation at 2 min pSer375 
CAMKII [141Yes N.R.
No 
No
No 
Several phosphosites activated in 60 min 
Rab3(B,D), Noc2 [73Yes No No <5 min for GTP loading
NPD
Rab3GAP1 and Rab3IP are phosphorylated within 30 s 
GSV docking, fusion with PM and dispersal of GLUT4 in the PM Munc18c* [72,86,90,91,106Yes No
No 
No
No 
pTyr 5 min
pSer588 in 15 s to 2 min 
Doc2B [143Yes N.R. N.R. NPD 
Tomosyn1/2* [70,97,144,145Yes Yes
No 
Yes
No 
Akt substrate Ser783[70]. Timing <5 min
pSer724/786/783 all within 15–30 s 
Syntaxin4* [79,146Yes Yes
Yes 
Yes
Yes 
Stx4 pY [36,106] 30 s for full activation
pTyr251 30 s;
Multiples phosphosites: Ser208 in 15 s; pSer248 transient 2–5 min but no PI3K or Akt dependence 
SNAP23* [80,81N.R. N.R.
No 
N.R.
No 
pThr24 transient 30 s to 2 min 
Vamp2 [57,78,147N.R. N.R.
Yes 
N.R.
Yes 
pSer48 in 5–10 min 
Complexin-2 [148Yes N.R.
No 
No
No 
pSer93 in 30 s 
NSF and αSNAP* [93,95,149,150N.R. N.R.
No 
N.R.
No 
NSF: pSer207 in 15 s
αSNAP: Ser24 in 20–60min 
EFR3 and PI 4-kinase IIIα* [66N.R. N.R.
Yes 
N.R.
Yes 
EFR3: very low Ser360/363 in 15 s 
GLUT4 internalisation (endocytosis) Clathrin [98,151Yes N.R.
No 
N.R.
No 
Heavy chain: pTyr434 in 15 s 
Dynamin-2* [152–154Yes N.R.
No 
N.R.
No 
NPD but Dynamin-1 (Dnm) detected with multiple phosphosites — some in 15 s or 30 s 
CIP4/2 [74]
N-WASp, CIP4* [155
Yes
Yes 
N.R.
No 
N.R.
No 
CIP4 Multiple phosphosites pSer296 in 15 s
N-WASP: pSer426 1–2 min 
Cholesterol, nystatin endocytosis [101Yes N.R. N.R.  
EEA1, Rab5, GAPEX5, Rab31 [156–158Yes N.R.
Yes 
N.R.
No 
EEA1: pSer720 in 10 and 60 min;
Rab5a: pSer193 — 60min
GAPEX5: Multiple phosphosites pSer742, pSer1017 in 5–10 min Rab31: NPD 
Ub, USP25* [159,160N.R. N.R. N.R. USP25: Transient pThr61 at 30 s 
Caveolae* [116,161–163No N.R.
Yes 
N.R.
No 
Multiple phosphosites on Caveolin-1 pTyr phosphorylated within 30 s 
GLUT4 return to GSV compartment and sorting away from recycling endosomes LRP1 [164Yes N.R.
Yes 
N.R.
Yes 
pSer4524 and pThr4525 at 60 min 
IRAP [165–169Yes Yes
Yes 
Yes
Yes 
pSer51 in 5 min; pTyr70 high 1–10 min transient 
Sortilin [170–173Yes N.R.
No 
N.R.
No 
pSer824 marginal (∼0.5) 15–20 min 
GGA, AP1* [174–177N.R. N.R.
Yes 
N.R.
No 
GGA: pSer414/418 in 5 min LY only
AP1(subunit mu): pThr152/154 in 30 s and 15 s resp. LY only
Other AP1 subunit NPD 
ESCRT [178N.R. N.R.
No 
N.R.
No 
Chmp3 and Vps4 NPD
Chmp2b phosphorylated in 15 s 
Retromer [166,179,180N.R. N.R.
Yes 
N.R.
Yes 
Vps35: pSer7 at 60 min
Vps26a NPD
Vps26b: multiple phosphosites and pSer302 in 2–5 min
Vps29 NPD
Snx3 and 27: some phosphorylation in 60 min. Not PI3K- or Akt-dependent 
Golgin97 and Vti1a [170N.R. N.R. N.R. NISPD 
Syntaxin 6* [181], Syntaxin 16 [182], mVps45 [183Yes (Stx16) N.R.
No† 
N.R.
No 
Stx16 dephosphorylation within 30min [181]
Stx16 some phosphorylation detected at long timepoints 60 min. Dephosphorylation for Ser35 detected since 15 s 
Rab11, Rip11* [184,185Yes Yes
Yes 
No
Yes 
Rab11b: Multiple phosphosites some activated at 2–10 min
Rip11: Multiple phosphosites, some pSer188 and 480 very fast 15 s 
Rab4, KIF13 (motor protein) [135,186–188Yes Yes
Yes 
N.R.
No 
Slow GTP loading over 45 min
Rab4 NPD
Kif13: Fast 30 s multiple sites pThr1717 
Rab14 [55,189N.R. N.R.
No 
N.R.
Yes 
2 min pSer97 
TUG [190–192N.R. N.R.
Yes 
N.R.
No 
Proteolytic cleavage in 10 min
pSer279 at 5 min 
TRARG1(TUSC5) [193,194N.R. N.R.
Yes 
N.R.
Yes 
Multiple phosphosites Ser56, 70 and 72 in 20–60 min 
TBC1D13, Rab35 [195N.R. N.R.
No 
N.R.
Yes 
TBC1D13: Ser184 in 20–60 min 
CHC22 [196N.R. N.R. N.R. NPD 
Budding of GSV, PLD [61,62Yes N.R. N.R. PLD1 — NISPD 
Delivery of newly synthesised GLUT4 to GSVs and maintaining insulin responsiveness Sortilin [170–172Yes N.R. N.R. See above 
syntaxin 16, mVps45, syntaxin 6 [181–183Yes (Stx16) N.R. N.R. Stx16 dephosphorylation within 30 min [181]. See above 
GGA, AP1 [174–177N.R. N.R. N.R. See above 
ACAP1, Arf6 [197Yes N.R. N.R. NPD 
VAMP7 (TI-VAMP), VAMP4 [198N.R. N.R.
Yes 
N.R.
No 
Vamp4 — Multiple phosphosites pSer17/20 in 1–5 min 
Axin, TNKS [199–201N.R. N.R. N.R. Axin1 — multiple sites but below 0.5 
P115 [202N.R. N.R.
No 
N.R.
No 
pSer717 slow 60 min 
Sec16 [63,129,130N.R. N.R. N.R. See above 
CHC22 [196N.R. N.R. N.R. NPD 
Transfected GLUT4 becoming insulin responsive N.R. N.R. N.R. 6–9 h [174]
3.5 h in primary adipocytes [152

Key molecules reported to modulate GLUT4 trafficking and their activation time scale and phosphorylation status in response to insulin stimulation and sensitivity to PI3K/Akt inhibitors (where known).

Italics text —proteins reported to have a role in skeletal muscle; N.R. not reported.

Bold text phosphorylation times and PI3K and Akt inhibitors data from Humphrey et al. [36]. Only values above the threshold of 0.5 log2 of the ratio Insulin-stimulated/unstimulated cells were reported.

NISPD no insulin-sensitive phosphosites detected (protein listed in the database as a phosphorylated protein but no kinetic data available).

NPD no phosphorylation detected (protein not listed in the database).

*

Fast phosphorylation, less than 1 min, according to Humphrey et al. [36].

As the inhibition of phosphorylation in [36] was assessed only at a time point of 20 min the effect of the inhibitors on transient phosphorylation at earlier timepoints or phosphorylation events occurring at later timepoints >20 min cannot be assessed.

In this perspective, we discuss how the different ‘phases’ of the GLUT4 trafficking responses to insulin and review kinetic considerations for how insulin signalling may co-ordinate multiple steps in GLUT4 traffic. We discuss methodological bottlenecks to a more complete understanding of the dynamics, molecules and systems involved in GLUT4 traffic, and suggest how we might accelerate identification of which trafficking steps, and regulatory proteins, are involved in signalling to the key phases of insulin action on GLUT4.

The proximal insulin signalling pathway required for GLUT4 translocation has been well characterised and very extensively reviewed elsewhere [7]. We, therefore, highlight only the basics of insulin signalling to GLUT4. Studies using the PI3K inhibitors wortmannin [8] and LY294002 [9] and an inhibitory and inactive PI3K p85 subunit [10,11] were among the first to show that PI3K activity is essential for insulin-stimulated glucose transport and GLUT4 translocation. Since then, studies using isoform-specific PI3K inhibitors have established that the main PI3K isoform involved is 1A [12,13]. The most important signalling event downstream of PI3K for GLUT4 translocation is the activation of the serine–threonine protein kinase Akt or protein kinase B (PKB) [14,15]. Akt is recruited to PIP3 at the PM through its PH domain with a similar time course to PIP3 generation [16], within ∼40 s of insulin binding to its receptor (Table 1), and this leads to Akt activation through the action of two kinases phosphoinositide-dependent kinase 1 (PDK1) [17,18] and mTOR complex 2 (mTORC2) [19]. Phosphorylation of Akt at both PDK1 and mTORC2 activating sites is required for full Akt activation [20,21]. Initial research on the role of Akt in insulin-stimulated glucose uptake and GLUT4 translocation relied extensively on genetic approaches including expression of constitutively active and membrane associated forms of Akt isoforms (Akt1, 2 or 3), siRNA knockdown in cell lines or knockout mouse models [22–26]. These demonstrated that Akt2 isoform expression and activity correlated with insulin-sensitive glucose uptake in insulin-sensitive tissues and is preferentially recruited to the PM in response to insulin stimulation [22,27,28]. More recently, specific inhibitors of Akt were developed (Akti-1/2 [29] and MK-2206 [30–32]), which confirmed that Akt activity is required for the very acute stimulation of glucose transport and GLUT4 translocation in response to insulin. Overall, these studies have provided strong evidence that the PI3K–Akt signalling pathway is very rapidly activated in response to insulin (Table 1), and is necessary and sufficient for insulin-stimulated GLUT4 translocation [24,33].

One of the fascinating features of insulin-regulated GLUT4 trafficking is that it requires the integration of signal transduction and subcellular protein traffic. But how does insulin signalling directly affect specific GLUT4 trafficking processes? Given the acute timeframe of insulin responses, and the key role for Akt in insulin-regulated glucose transport, the most likely mechanism is via protein phosphorylation. The discovery of the Rab-GTPase activating protein (GAP) TBC1D4 (AS160) as an Akt substrate established the first direct link between the insulin signalling and Rab-mediated control of membrane vesicle traffic [34,35]. Crucially, TBC1D4 phosphorylation is sensitive to PI3K and Akt inhibition, and occurs within 15 s of insulin stimulation, is maximal by 1 min (Table 1) [36]. Furthermore, a phosphorylation-dead mutant of TBC1D4 (termed TBC1D4-4P), has revealed that TBC1D4 phosphorylation by Akt is required for insulin-stimulated GLUT4 traffic [35]. These data, along with the kinetics of signalling to TBC1D4, are in keeping with a critical role for Akt-mediated TBC1D4 phosphorylation in acute insulin-stimulated GLUT4 translocation. TBC1D4 is a Rab-GAP, and negatively regulates GLUT4 translocation through inhibiting its substrates Rab10 and Rab14 in adipocytes (Rab8 and Rab13 are similarly implicated in muscle cells, Table 1). TBC1D4 maintains these Rabs in their inactive, GDP-bound, form, and inactivation of TBC1D4 following insulin signalling to TBC1D4 leads to GTP loading and Rab-mediated GLUT4 translocation.

In early versions of the glucose transporter translocation model, it was assumed that the glucose transporters were delivered to the PM and then remained there until insulin dissociated from its receptor [2]. Using the hexose photolabel ATB-BMPA [8,37–40], some of the first studies on the kinetics of GLUT4 translocation revealed that GLUT4 continuously recycled from the PM to an endosome compartment and a sequestered intracellular-storage compartment even in the presence of insulin. These studies suggested at least 3-compartments are required to adequately account for GLUT4 traffic. They revealed that the main effect of insulin was to accelerate the rate of exocytosis from this sequestered compartment [37,40]. There also appeared to be small reduction in the endocytosis rate constant following insulin treatment. However, detailed follow-up studies revealed that, when considering GLUT4 endosomal recycling, there was no insulin-dependent reduction in GLUT4 endocytosis [41].

In addition to insulin primarily targeting GLUT4 exocytosis, studies on photolabel-tagged GLUT4 revealed that there are (at least) two phases to the insulin response. First, there is a fast acute phase of the response to insulin where, within ≈3 min, 50% of GLUT4 is delivered in a burst to the cell surface [37,42,43], likely from its specialised storage compartment termed GLUT4 storage vesicles (GSVs). Second, there is steady-state phase, which can last for hours, where elevated PM GLUT4 abundance is maintained via GLUT4 recycling to the PM with a half time of ≈8 min [37,42,43].

The concept of two phases of the GLUT4 trafficking response to insulin, and of insulin signalling increasing GLUT4 exocytosis rates, are consistent with experimental data from total internal reflectance fluorescence microscopy (TIRF-M). TIRF-M has allowed study of the kinetics of GLUT4 movement close to the PM [44–48], and the application and use of pH-sensitive GLUT4 constructs (e.g. phluorin-GLUT4) [45,49] has enabled study of GSV fusion at this site. Studies in adipocytes have revealed a burst of GLUT4 vesicle fusion events in the first 5–10 min directly after insulin addition. This burst of fusion activity then falls away to a steady-state rate of vesicle fusion that remains higher than observed in unstimulated cells [45,49].

The presence of two phases to the insulin response was also supported by studies led by James and Coster using antibody binding to epitope-tagged GLUT4 reaching the cell surface of 3T3-L1 cells to monitor GLUT4 traffic [50–52]. These studies revealed that GLUT4 appeared to be released to the cell surface in a graded manner to increasing doses of insulin, with a distinct plateauing and saturation of antibody binding in the basal state and at submaximal insulin levels. In the basal state only ≈20% of GLUT4 recycled and bound cell surface antibody while 80% appeared to remain in a non-recycling compartment. Submaximal levels of insulin lead to a quantal increase in the antibody saturation point, but a maximal insulin stimulation was necessary to mobilise all the available GLUT4. The concept of a static-retention GSV compartment is interesting since it suggests that insulin acts to release GLUT4 from a highly sequestered and non-recycling pool, effectively increasing the mobility of an additional proportion of sequestered GLUT4 available to traffic to the PM.

Building on these data on the quasi-static GSV compartment, Mastick and co-workers [41,53], together with Coster and co-workers [54–56], developed a 4-compartment dynamic-retention kinetic model for GLUT4 traffic. The main feature of this model is that of two exocytotic routes to the PM, one from a sequestered GLUT4 compartment and one from an endosome recycling compartment (ERC) (Figure 1). In the absence of insulin, it is proposed that GLUT4 exocytosis mainly occurs through the recycling endosome route (≈20% of GLUT4), with the bulk of the GLUT4 (≈80%) sequestered and partitioned from this route into the more slowly recycling GSV compartment. This 4-compartment model can be successfully used to simulate quantal effects in release of GLUT4 and plateauing of cell surface antibody binding to GLUT4. Insulin signalling is proposed to stimulate both the release of GLUT4 from its sequestered compartment (a TBC1D4/Rab10 regulated step) and fusion of GSVs with the PM [54–56]. These modelling studies also accounted for insulin responsive traffic of transferrin receptors from sorting endosomes and LRP1 traffic through an ERC [41]. Both endosome ligand recycling compartments were kinetically separate from the sequestered GLUT4 storage compartment.

GLUT4 sequestration and exocytosis.

Figure 1.
GLUT4 sequestration and exocytosis.

The schematic diagram represents an extension of the 3-compartment sequestration model [42] and the 4-compartment dynamic-retention model [54,55]). The compartments considered in modelling of kinetics of GLUT4 traffic are the plasma membrane (PM), a sequestered GLUT4 vesicle (GSV) compartment, sorting endosomes (SE) and perinuclear compartments (PNC) and an endosome recycling compartment (ERC). The GSV reservoir is supplied by vesicles from SE and PNC, but the proportion of vesicles supplied by these compartments will vary in different cell types. In basal 3T3-L1 cells ∼80% of the GLUT4 is present in GSV, while 20% of GLUT4 is recycled from SE through ERC to the PM [54]. We suggest here that a retention-catalyst C is responsible for the partitioning and saturation of the sequestered GLUT4 compartment. It is proposed that C catalyses formation of GSV. In addition, C is released from the GSV compartment when insulin signalling leads to vesicle fusion with the PM. The rate constants linking these compartments are: kseq for movements at the saturable step and requiring C; kf for fusion of docked vesicles with the plasma membrane — and associated with desaturation of the GSV compartment and release or recycling of C; krd for reversal of docking without fusion — this step involves vesicles that sample docking sites but do not fuse; krc for recycling of GLUT4 from SE through ERC and to the PM.

Figure 1.
GLUT4 sequestration and exocytosis.

The schematic diagram represents an extension of the 3-compartment sequestration model [42] and the 4-compartment dynamic-retention model [54,55]). The compartments considered in modelling of kinetics of GLUT4 traffic are the plasma membrane (PM), a sequestered GLUT4 vesicle (GSV) compartment, sorting endosomes (SE) and perinuclear compartments (PNC) and an endosome recycling compartment (ERC). The GSV reservoir is supplied by vesicles from SE and PNC, but the proportion of vesicles supplied by these compartments will vary in different cell types. In basal 3T3-L1 cells ∼80% of the GLUT4 is present in GSV, while 20% of GLUT4 is recycled from SE through ERC to the PM [54]. We suggest here that a retention-catalyst C is responsible for the partitioning and saturation of the sequestered GLUT4 compartment. It is proposed that C catalyses formation of GSV. In addition, C is released from the GSV compartment when insulin signalling leads to vesicle fusion with the PM. The rate constants linking these compartments are: kseq for movements at the saturable step and requiring C; kf for fusion of docked vesicles with the plasma membrane — and associated with desaturation of the GSV compartment and release or recycling of C; krd for reversal of docking without fusion — this step involves vesicles that sample docking sites but do not fuse; krc for recycling of GLUT4 from SE through ERC and to the PM.

Close modal

It is noted in Supplementary S1a that the simpler 3-compartment sequestration model can also be used to simulate plateauing of antibody binding responses. This occurs when the model includes a endosome recycling step (as discussed in [42]) and therefore two exocytosis steps (one from sequestered GSV and one from endosomes). However, none of the three models above has given a mechanistic basis for the level of saturation of antibody binding and for partitioning of compartments in the basal state. We suggest that the concentration of sequestered GLUT4 is dependent on the level of a retention-catalyst (such as a vesicle coat protein or a retaining Rab protein), and include this suggestion as an extension to the 3-compartment model [42]. This additional feature is shown schematically in Figure 1, and further developed in Supplementary 1b. In the basal state, the availability of the retention-catalyst limits the concentration of GLUT4 in the non-recycling (or slowly recycling) compartment (at ≈80% in 3T3-L1 cells). In this basal state the level of free catalyst approaches zero, as all the available catalyst is bound to the GSV. It is proposed that insulin signalling leads to a release of GLUT4 vesicles from the retention-catalyst for fusion with the PM in a rapid burst. This leads to increased delivery of GLUT4 at the PM, but also releases the retention-catalyst to regenerate the GSV compartment. This updated 3-compartment model provides an approach to directly link insulin signalling molecules and catalysis to compartmentalisation and movement of GLUT4 vesicles.

In addition to the observations of the phases of the insulin response, TIRF-M studies have provided detailed analysis of GSVs behaviour at the cell surface (e.g. trafficking to the cell periphery, docking, fusion) and in the PM bilayer (e.g. dispersal). These studies have revealed that insulin acts to increase vesicle docking and fusion at the PM. Movement of GLUT4-containing vesicles into the TIRF zone was found to be wortmannin-insensitive. However, wortmannin markedly reduced GLUT4 vesicle docking, demonstrating that the capture of GSVs by the PM is dependent on PIP3 signalling [44]. A cell-free fusion assay, which combines separate PM, intracellular vesicle and cytoplasm fractions from basal or insulin-stimulated cells, also revealed that insulin facilitates fusion of GSVs with the PM. In this assay, wortmannin was inhibitory in the overall cell-free fusion system [57,58]. This assay does not distinguish whether the docking or fusion step is involved in the inhibitory response to PI3K-inhbition, but these data are consistent with the docking/fusion of GLUT4 vesicles at the PM as a key regulated process downstream of PIP3 synthesis.

There is also evidence that TBC1D4 may play a role in the docking/fusion of GSVs at the PM. First, the phospho-tyrosine binding domains of TBC1D4 inhibited GLUT4 vesicle fusion with the PM in a cell-free assay [58]. Second, TIRF-M studies revealed that TBC1D4-4P markedly reduced GLUT4 vesicle docking and consequently fusion [59,60]

Away from actions at the PM several studies have shown insulin effects on vesicle budding from the perinuclear compartment [61,62]. Brumfield et al. [63] provided evidence that insulin/Akt signalling promoted GLUT4 mobilisation from the perinuclear region. Using a photoconvertible GLUT4 reporter, they measured a ∼1.5-fold increase in the presence of insulin in GLUT4 movement from this region over a time course (20 min) consistent with a role in delivering GLUT4 to the PM. Because photoconversion can be confined to specific subcellular locations this technique can, to some extent, be considered as a providing a separation and isolation from other ongoing processes in other locations. In addition, the Akt inhibitor MK2206 inhibited this insulin response. Knockdown of TBC1D4 increased GLUT4 mobilisation from the perinuclear region to the PM [63]. In contrast, knockdown of the TBC1D4 cognate Rab, Rab10, had the opposite effect. This suggests that insulin signalling to TBC1D4/Rab10 promotes GLUT4 delivery to the PM to sustain the insulin response. Together, these data support a role for GLUT4 vesicle delivery to the cell periphery as an insulin-regulated step. However, the extent of insulin activation (fold-change) of this perinuclear step is smaller than those at the PM.

The experimental data outlined above suggest that PI3K–Akt signalling acts at multiple sites in GLUT4 trafficking, including traffic from the perinuclear region and GLUT4 vesicle docking and fusion at the PM. In addition, modelling and direct experimental evidence have suggested a role for TBC1D4 in release of GLUT4 towards the cell periphery [63], the sorting of GLUT4 into GSVs [56], release of GSVs to the PM [59] together with GSV interactions with the PM [44,57–60]. Does insulin signalling act via PI3K–Akt–(TBC1D4) to target several different processes in GLUT4 traffic?

In section ‘Considering kinetics of insulin-stimulated GLUT4 translocation’, we have proposed an updated model of GLUT4 traffic that includes a ‘retention-catalyst’ that is required for GSV formation and sequestration. Importantly, this catalyst is limiting, so that the size of the GSV pool of GLUT4 is finite. This ‘retention-catalyst’ could be the TBC1D4–Rab10 couple. If this is the case, then PI3K–Akt catalysed suppression of TBC1D4 Rab-GAP activity and increased Rab-GTP loading will have two immediate consequences: (1) release of GSVs for fusion with the PM and (2) desaturation of the GSV compartment and refilling of this compartment. As such, this proposed catalytic cycle involving a retention-catalyst (Figure 1) may offer advantages in terms of fully understanding the apparent multiple sites of action of insulin action occurring through TBC1D4.

However, control of GLUT4 by TBC1D4 alone provides an incomplete picture of insulin action on GLUT4 traffic. Some studies indicate that vesicle fusion can be perturbed or activated independently of TBC1D4-driven vesicle docking [28,60,64]. Indeed, many molecules that are involved in SNARE complex formation and actin remodelling could be involved in activation of fusion independent of the involvement of TBC1D4 in vesicle docking. These proteins are targets of insulin-stimulated phospho-serine/threonine and of phospho-tyrosine kinase activities (Table 1 and section ‘Identifying insulin control of GLUT4 traffic via phosphorylation’).

In addition to these findings related to GLUT4 vesicle docking and fusion, detailed analysis of GLUT4 movement within the PM using TIRF-M suggests that insulin regulates GLUT4 dispersal from fusion sites [47,49,65,66]. The significance of this process for GLUT4 catalytic activity needs further resolution and new methods to study these steps independently of other events at the PM are needed. To some extent, an altered dispersal within the PM may be a prelude to GLUT4 interaction with clathrin lattices and internalisation [49].

A major goal of the field remains to understand how insulin signalling controls GLUT4 traffic at the molecular level. TBC1D4 is the most highly studied substrate of insulin signalling in this context, and the data generated on TBC1D4 highlights the power of combining effective experimental tools to manipulate insulin signalling (e.g. TBC1D4-4P mutant) with experimental systems that discern specific GLUT4 trafficking processes such as vesicle docking.

As described above, insulin regulates GSV interactions and fusion with the PM. Consistent with this, some proteins implicated in GSV docking/fusion are direct Akt substrates or regulated downstream of Akt substrates. These include Rab10 [67], myosin Va [68], synip [69], tomosyn [70] and RalA [71]. In contrast, the activities of other mediators of GLUT4 vesicle-PM interactions (such as Munc18c [72], Rab3 [73], TC10 [74] and exocyst components [75–77]) are regulated by insulin, but not affected by PI3K or Akt inhibitors (Table 1). In addition to insulin regulation of serine/threonine kinases, tyrosine kinases, including the insulin receptor are also implicated in the important docking and fusion steps of insulin action. The formation of the ternary SNARE complex at the PM is reported to be targeted by insulin signalling via Munc18c. The GSV docking and the fusion steps involve the formation of the SNARE complex between the t-SNAREs syntaxin4 and SNAP23 [78–81] at the PM and v-SNARE protein VAMP2 on GSVs. The formation of this complex is stimulated by insulin [82], which fits with the stimulatory effect of insulin on GSV docking and fusion reported in the TIRF studies and indicated as being critical sites in kinetic modelling studies. This process is modulated by Munc18c [82–87], which is now known to be a direct target of the tyrosine kinase activity of the insulin receptor [88]. Furthermore, an interesting and powerful new approach to studying and isolating insulin action on SNARE proteins has been developed using proximity ligation assays. This approach has been used to show that insulin-stimulated Munc18c phosphorylation is required for SNARE assembly [89]. Phosphomimetic mutants (Y219E or Y521E), but not phosphodeficient mutants (Y219F or Y521F), rescued defective insulin-regulated GLUT4 traffic in Munc18c knockdown adipocytes, suggesting that phosphorylation of this protein promotes SNARE complex formation [72,90,91]. In addition to Munc18c, Syntaxin 4 is phosphorylated in response to insulin, and both Syntaxin 4 and Munc18c are phosphorylated rapidly, with time courses of seconds and minutes (detailed in Table 1). These data are consistent with a role for signalling to these proteins in the burst of GSV exocytosis upon insulin stimulation.

Phospho-proteomics data provide unbiased insight into new sites of insulin action. Global analysis of the kinetics and PI3K/Akt dependence of the insulin signalling network in 3T3-L1 adipocytes revealed very rapid activation of the PI3K–Akt pathway (within 1 min) and that PI3K activation is required for a large proportion (∼66%) of all insulin-stimulated protein phosphorylation/dephosphorylation events in these cells [36]. Furthermore, this study uncovered the remarkable breadth of the insulin signalling network with approximately 5000 phosphosites on 2000 proteins that are regulated by insulin. This list contains phosphosites on known regulators of GLUT4 traffic that are not functionally characterised (Table 1), including some on Rabs and their regulatory or effector proteins. Rabs are G-proteins and their activity can be controlled by insulin through combinations of phosphorylation of GEF and GAP proteins that modulate GTP loading and by direct phosphorylation of the Rab themselves. Many of these phosphorylation reactions occur rapidly (Table 1). We know that the Rab10 GAP (TBC1D4) and GEF (DENND4C) are targeted by insulin signalling [35,92], but phospho-proteomics has revealed that Rab10 itself also undergoes insulin-regulated phosphorylation [36] (Table 1). It will be of interest to ascertain how Rab10 phosphorylation influences Rab10 GTP loading and activity. Some data suggest that Rab10 on GLUT4 vesicles may not be GTP-loaded in response to insulin [67]. However, it is technically challenging to determine Rab protein GTP loading status and we require more refined techniques for determining both levels of the GTP loading and the activation of subpopulations of Rab proteins within cells. This is also relevant to additional Rabs since many Rabs and Rab effectors are phosphorylated rapidly by insulin (Table 1) including Rab11/Rip11, Rab3/Rab3Gap1/Rab3ip and RAB13/MICAL-L2. Rab5/RABEX5 is also subject to insulin-stimulated phosphorylation, albeit with slower kinetics (Table 1).

Overall, there are many very rapidly phosphorylated insulin-regulated proteins that are known regulators of GLUT4 traffic (Table 1). However, we note that phospho-proteome screening has revealed phosphosites on proteins that are implicated in GLUT4 vesicle traffic but have not yet been extensively studied as targets of insulin action. Many of these under-studied proteins are rapidly phosphorylated by insulin signalling often in <1 min (see asterisk in Table 1). It is, therefore, clear that phospho-proteomic analyses are an invaluable resource, but also present the challenge of firstly filtering out sites of greatest interest, and then understanding the functional significance of these phosphorylation events in GLUT4 translocation.

Method development to allow distinct GLUT4 trafficking processes to be studied in isolation and in sufficient kinetic detail are critical for us to fully map the major sites of insulin action in GLUT4 trafficking. We propose that by focusing on insulin-regulated trafficking steps, and identifying putative regulatory signalling events from new or existing phospho-proteomics data, we will accelerate experimental elucidation of the mechanism of insulin action.

One avenue for method development we are particularly interested in is cell-free experimental systems. Although difficult to set up, they have yielded big advances in the study of isolated SNARE involvement in neuronal fusion systems [93–95]. This has now been further expanded and the SNARES involved in GSV fusion have been successfully used for characterising proteins involved in the docking and fusion events [94,96,97]. With the advent of a range of more sensitive fluorescent molecules and fluorescent nanodots, perhaps new and improved cell-free assays of insulin action on fusion can be developed to provide the mechanistic isolation of individual reactions that are not possible in the whole cell setting. The PM lawn assay and permeabilised cell assays [98] also have potential as cell-free approaches to studying vesicle docking and fusion. Lawns which retain activity after attachment to coverslips may be accessible to live TIRF microscopy. Isolated adipocyte attached to coverslips form regular arrays of isolated PM sheets because of the regular packing of large cells. These cells can readily be lysed to remove the bulk of cell content [43]. GLUT4 vesicle reactions with isolated lawns could be followed under conditions in which the cytosol components of the fusion reaction are modified.

One caveat to use phospho-proteomics data is that there are currently very few studies on separately manipulating specific phosphorylation sites on target proteins or of identifying and separating functional from non-functional phosphorylation [99]. These studies will be vital, especially for proteins with multiple insulin-regulated phosphorylation sites. A nice example of studying isolated processes is a recent series of experiments in which recombinant full length TBC1D4 was incubated with isolated and purified specific kinases [100]. This study showed the preference of specific phosphorylation sites for the upstream kinases Akt and AMPK, but also that a major effect of TBC1D4 phosphorylation induced by these kinases was a reduction in TBC1D4 binding to IRAP. This study supports the model whereby insulin signalling disrupts TBC1D4 binding to GSVs (via IRAP). Overall, phospho-proteomics datasets are a valuable resource to find new regulatory mechanisms underpinning insulin-stimulated GLUT4 traffic, but it will require substantial effort to assess the role of high priority phosphorylation sites in insulin signalling to GLUT4. Related to this, we should not lose sight of the usefulness of small molecule inhibitors as an important part of the data presented in this review came from their use. Although it is important to be mindful of off-target effects [31], small molecules generally act rapidly and can be used on timescales similar to that at which insulin acts to provide clear data on the involvement of PI3K or Akt, for example, in the insulin response.

We suggest that considering retention-catalysts may help begin to unpick how insulin signalling regulates steps of GLUT4 traffic. Here, we propose that saturation of the GSV compartment occurs because of a limit in retention-catalyst availability. Exploration of the consequences of saturation of membrane traffic compartments and steps could be more extensively studied in future and could apply to several (or even all) steps in complex pathways. Their consideration may simplify the considerations of membrane compartment partitioning (and the associated rate constants for traffic through these compartments), as compartment size may be restricted by separate facilitators at each step. For example, clathrin lattices [98] and clathrin vesicle coating could conceivably be limited at the PM and lead to partitioning between a clathrin and non-clathrin routes for endocytosis [101]. This concept also has implications in experimental situations systems in which a recombinant mediator is introduced. The endogenous saturation point for a compartment may be exceeded and there may be overflow into compartments that would not normally be used in an unperturbed system. For example, in studies in which GLUT4 has been overexpressed [102], it exceeds the capacity of the insulin-regulated storage system and more GLUT4 is diverted to a recycling system that is not insulin regulated. The GLUT4 is diverted to the PM and raises basal levels of unregulated traffic.

From kinetic data available to-date, it is clear that GSV docking and fusion are major sites of insulin regulation in adipocytes. Furthermore, evidence from human adipose tissue suggests that the tethering/docking and fusion steps are impaired in insulin resistant adipocytes [103]. It is important to note that these kinetic considerations also apply for insulin action in muscle cells (the major site of insulin-stimulated glucose uptake). GLUT4 vesicles were found to be tethered (or docked) at the sarcolemma and transverse tubule membranes in the absence of insulin, and fused with the limiting membrane from these sites in response to insulin [46,104,105]. These data together suggest that it is critical to focus attention on molecules and docking/fusion processes at the PM in both fat and muscle. This will help to map out how insulin signalling impinges upon delivery of GLUT4 to its functionally and physiologically important location and delivers enhanced glucose transport into insulin target cells.

The authors declare that there are no competing interests associated with the manuscript.

D.J.F. is supported by an MRC Career Development Award (MR/S007091/1) G.D.H. is supported by Leverhulme Trust (EM-2018-039) and F.K. is supported by MRC grant MR/P002927/1 and Diabetes U.K. grant 19/0005983.

Open access for this article was enabled by the participation of University of Bath in an all-inclusive Read & Publish pilot with Portland Press and the Biochemical Society under a transformative agreement with JISC.

ERC

endosome recycling compartment

GSVs

GLUT4 storage vesicles

mTORC2

mTOR complex 2

PDK1

phosphoinositide-dependent kinase 1

PM

plasma membrane

TIRF-M

total internal reflectance fluorescence microscopy

1
Kahn
,
C.R.
(
1979
)
What is the molecular basis for the action of insulin?
Trends Biochem. Sci.
4
,
N263
N266
2
Cushman
,
S.W.
and
Wardzala
,
L.J.
(
1980
)
Potential mechanism of insulin action on glucose transport in the isolated rat adipose cell. Apparent translocation of intracellular transport systems to the plasma membrane
.
J. Biol. Chem.
255
,
4758
4762
3
Suzuki
,
K.
and
Kono
,
T.
(
1980
)
Evidence that insulin causes translocation of glucose transport activity to the plasma membrane from an intracellular storage site
.
Proc. Natl Acad. Sci. U.S.A.
77
,
2542
2545
4
James
,
D.E.
,
Strube
,
M.
and
Mueckler
,
M.
(
1989
)
Molecular cloning and characterization of an insulin-regulatable glucose transporter
.
Nature
338
,
83
87
5
Charron
,
M.J.
,
Brosius
, III,
F.C.
,
Alper
,
S.L.
and
Lodish
,
H.F.
(
1989
)
A glucose transport protein expressed predominately in insulin-responsive tissues
.
Proc. Natl Acad. Sci. U.S.A.
86
,
2535
2539
6
Birnbaum
,
M.J.
(
1989
)
Identification of a novel gene encoding an insulin-responsive glucose transporter protein
.
Cell
57
,
305
315
7
Klip
,
A.
,
McGraw
,
T.E.
and
James
,
D.E.
(
2019
)
Thirty sweet years of GLUT4
.
J. Biol. Chem.
294
,
11369
11381
8
Clarke
,
J.F.
,
Young
,
P.W.
,
Yonezawa
,
K.
,
Kasuga
,
M.
and
Holman
,
G.D.
(
1994
)
Inhibition of the translocation of GLUT1 and GLUT4 in 3T3-L1 cells by the phosphatidylinositol 3-kinase inhibitor, wortmannin
.
Biochem. J.
300
(
Pt 3
),
631
635
9
Cheatham
,
B.
,
Vlahos
,
C.J.
,
Cheatham
,
L.
,
Wang
,
L.
,
Blenis
,
J.
and
Kahn
,
C.R.
(
1994
)
Phosphatidylinositol 3-kinase activation is required for insulin stimulation of pp70 S6 kinase, DNA synthesis, and glucose transporter translocation
.
Mol. Cell. Biol.
14
,
4902
4911
10
Quon
,
M.J.
,
Chen
,
H.
,
Ing
,
B.L.
,
Liu
,
M.L.
,
Zarnowski
,
M.J.
,
Yonezawa
,
K.
et al (
1995
)
Roles of 1-phosphatidylinositol 3-kinase and ras in regulating translocation of GLUT4 in transfected rat adipose cells
.
Mol. Cell. Biol.
15
,
5403
5411
11
Katagiri
,
H.
,
Asano
,
T.
,
Inukai
,
K.
,
Ogihara
,
T.
,
Ishihara
,
H.
,
Shibasaki
,
Y.
et al (
1997
)
Roles of PI 3-kinase and Ras on insulin-stimulated glucose transport in 3T3-L1 adipocytes
.
Am. J. Physiol.
272
,
E326
E331
12
Knight
,
Z.A.
,
Gonzalez
,
B.
,
Feldman
,
M.E.
,
Zunder
,
E.R.
,
Goldenberg
,
D.D.
,
Williams
,
O.
et al (
2006
)
A pharmacological map of the PI3-K family defines a role for p110alpha in insulin signaling
.
Cell
125
,
733
747
13
Chaussade
,
C.
,
Rewcastle
,
G.W.
,
Kendall
,
J.D.
,
Denny
,
W.A.
,
Cho
,
K.
,
Gronning
,
L.M.
et al (
2007
)
Evidence for functional redundancy of class IA PI3K isoforms in insulin signalling
.
Biochem. J.
404
,
449
458
14
Kohn
,
A.D.
,
Summers
,
S.A.
,
Birnbaum
,
M.J.
and
Roth
,
R.A.
(
1996
)
Expression of a constitutively active Akt Ser/Thr kinase in 3T3-L1 adipocytes stimulates glucose uptake and glucose transporter 4 translocation
.
J. Biol. Chem.
271
,
31372
31378
15
Tanti
,
J.F.
,
Grillo
,
S.
,
Gremeaux
,
T.
,
Coffer
,
P.J.
,
Van Obberghen
,
E.
and
Le Marchand-Brustel
,
Y.
(
1997
)
Potential role of protein kinase B in glucose transporter 4 translocation in adipocytes
.
Endocrinology
138
,
2005
2010
16
Norris
,
D.M.
,
Yang
,
P.
,
Krycer
,
J.R.
,
Fazakerley
,
D.J.
,
James
,
D.E.
and
Burchfield
,
J.G.
(
2017
)
An improved Akt reporter reveals intra- and inter-cellular heterogeneity and oscillations in signal transduction
.
J. Cell Sci.
130
,
2757
2766
17
Alessi
,
D.R.
,
James
,
S.R.
,
Downes
,
C.P.
,
Holmes
,
A.B.
,
Gaffney
,
P.R.J.
,
Reese
,
C.B.
et al (
1997
)
Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Ba
.
Curr. Biol.
7
,
261
269
18
Stokoe
,
D.
,
Stephens
,
L.R.
,
Copeland
,
T.
,
Gaffney
,
P.R.
,
Reese
,
C.B.
,
Painter
,
G.F.
et al (
1997
)
Dual role of phosphatidylinositol-3,4,5-trisphosphate in the activation of protein kinase B
.
Science
277
,
567
570
19
Sarbassov
,
D.D.
,
Guertin
,
D.A.
,
Ali
,
S.M.
and
Sabatini
,
D.M.
(
2005
)
Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex
.
Science
307
,
1098
1101
20
Kearney
,
A.L.
,
Cooke
,
K.C.
,
Norris
,
D.M.
,
Zadoorian
,
A.
,
Krycer
,
J.R.
,
Fazakerley
,
D.J.
et al (
2019
)
Serine 474 phosphorylation is essential for maximal Akt2 kinase activity in adipocytes
.
J. Biol. Chem.
294
,
16729
16739
21
Alessi
,
D.R.
,
Andjelkovic
,
M.
,
Caudwell
,
B.
,
Cron
,
P.
,
Morrice
,
N.
,
Cohen
,
P.
et al (
1996
)
Mechanism of activation of protein kinase B by insulin and IGF-1
.
EMBO J.
15
,
6541
6551
22
Cho
,
H.
,
Mu
,
J.
,
Kim
,
J.K.
,
Thorvaldsen
,
J.L.
,
Chu
,
Q.
,
Crenshaw
,
E.B.
et al (
2001
)
Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKB beta)
.
Science
292
,
1728
1731
23
Chen
,
W.S.
,
Xu
,
P.Z.
,
Gottlob
,
K.
,
Chen
,
M.L.
,
Sokol
,
K.
,
Shiyanova
,
T.
et al (
2001
)
Growth retardation and increased apoptosis in mice with homozygous disruption of the Akt1 gene
.
Genes Dev.
15
,
2203
2208
24
Ng
,
Y.
,
Ramm
,
G.
,
Lopez
,
J.A.
and
James
,
D.E.
(
2008
)
Rapid activation of Akt2 is sufficient to stimulate GLUT4 translocation in 3T3-L1 adipocytes
.
Cell Metab.
7
,
348
356
25
Jiang
,
Z.Y.
,
Zhou
,
Q.L.
,
Coleman
,
K.A.
,
Chouinard
,
M.
,
Boese
,
Q.
and
Czech
,
M.P.
(
2003
)
Insulin signaling through Akt/protein kinase B analyzed by small interfering RNA-mediated gene silencing
.
Proc. Natl Acad. Sci. U.S.A.
100
,
7569
7574
26
Lai
,
K.M.
,
Gonzalez
,
M.
,
Poueymirou
,
W.T.
,
Kline
,
W.O.
,
Na
,
E.
,
Zlotchenko
,
E.
et al (
2004
)
Conditional activation of Akt in adult skeletal muscle induces rapid hypertrophy
.
Mol. Cell. Biol.
24
,
9295
9304
27
Bae
,
S.S.
,
Cho
,
H.
,
Mu
,
J.
and
Birnbaum
,
M.J.
(
2003
)
Isoform-specific regulation of insulin-dependent glucose uptake by Akt/protein kinase B
.
J. Biol. Chem.
278
,
49530
49536
28
Gonzalez
,
E.
and
McGraw
,
T.E.
(
2006
)
Insulin signaling diverges into Akt-dependent and -independent signals to regulate the recruitment/docking and the fusion of GLUT4 vesicles to the plasma membrane
.
Mol. Biol. Cell
17
,
4484
4493
29
Green
,
C.J.
,
Goransson
,
O.
,
Kular
,
G.S.
,
Leslie
,
N.R.
,
Gray
,
A.
,
Alessi
,
D.R.
et al (
2008
)
Use of Akt inhibitor and a drug-resistant mutant validates a critical role for protein kinase B/Akt in the insulin-dependent regulation of glucose and system A amino acid uptake
.
J. Biol. Chem.
283
,
27653
27667
30
Yap
,
T.A.
,
Yan
,
L.
,
Patnaik
,
A.
,
Fearen
,
I.
,
Olmos
,
D.
,
Papadopoulos
,
K.
et al (
2011
)
First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors
.
J. Clin. Oncol.
29
,
4688
4695
31
Tan
,
S.
,
Ng
,
Y.
and
James
,
D.E.
(
2011
)
Next-generation Akt inhibitors provide greater specificity: effects on glucose metabolism in adipocytes
.
Biochem. J.
435
,
539
544
32
Lai
,
Y.C.
,
Liu
,
Y.
,
Jacobs
,
R.
and
Rider
,
M.H.
(
2012
)
A novel PKB/Akt inhibitor, MK-2206, effectively inhibits insulin-stimulated glucose metabolism and protein synthesis in isolated rat skeletal muscle
.
Biochem J.
447
,
137
147
33
Xu
,
Y.
,
Nan
,
D.
,
Fan
,
J.
,
Bogan
,
J.S.
and
Toomre
,
D.
(
2016
)
Optogenetic activation reveals distinct roles of PIP3 and Akt in adipocyte insulin action
.
J. Cell Sci.
129
,
2085
2095
34
Miinea
,
C.P.
,
Sano
,
H.
,
Kane
,
S.
,
Sano
,
E.
,
Fukuda
,
M.
,
Peranen
,
J.
et al (
2005
)
AS160, the Akt substrate regulating GLUT4 translocation, has a functional Rab GTPase-activating protein domain
.
Biochem. J.
391
,
87
93
35
Sano
,
H.
,
Kane
,
S.
,
Sano
,
E.
,
Miinea
,
C.P.
,
Asara
,
J.M.
,
Lane
,
W.S.
et al (
2003
)
Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation
.
J. Biol. Chem.
278
,
14599
14602
36
Humphrey
,
S.J.
,
Yang
,
G.
,
Yang
,
P.
,
Fazakerley
,
D.J.
,
Stockli
,
J.
,
Yang
,
J.Y.
et al (
2013
)
Dynamic adipocyte phosphoproteome reveals that Akt directly regulates mTORC2
.
Cell Metab.
17
,
1009
1020
37
Satoh
,
S.
,
Nishimura
,
H.
,
Clark
,
A.E.
,
Kozka
,
I.J.
,
Vannucci
,
S.J.
,
Simpson
,
I.A.
et al (
1993
)
Use of bismannose photolabel to elucidate insulin-regulated GLUT4 subcellular trafficking kinetics in rat adipose cells. Evidence that exocytosis is a critical site of hormone action
.
J. Biol. Chem.
268
,
17820
17829
38
Holman
,
G.D.
,
Kozka
,
I.J.
,
Clark
,
A.E.
,
Flower
,
C.J.
,
Saltis
,
J.
,
Habberfield
,
A.D.
et al (
1990
)
Cell surface labeling of glucose transporter isoform GLUT4 by bis-mannose photolabel. Correlation with stimulation of glucose transport in rat adipose cells by insulin and phorbol ester
.
J. Biol. Chem.
265
,
18172
18179
39
Yang
,
J.
,
Clark
,
A.E.
,
Harrison
,
R.
,
Kozka
,
I.J.
and
Holman
,
G.D.
(
1992
)
Trafficking of glucose transporters in 3T3-L1 cells. Inhibition of trafficking by phenylarsine oxide implicates a slow dissociation of transporters from trafficking proteins
.
Biochem. J.
281
(
Pt 3
),
809
817
40
Yang
,
J.
and
Holman
,
G.D.
(
1993
)
Comparison of GLUT4 and GLUT1 subcellular trafficking in basal and insulin-stimulated 3T3-L1 cells
.
J. Biol. Chem.
268
,
4600
4603
41
Habtemichael
,
E.N.
,
Brewer
,
P.D.
,
Romenskaia
,
I.
and
Mastick
,
C.C.
(
2011
)
Kinetic evidence that Glut4 follows different endocytic pathways than the receptors for transferrin and alpha2-macroglobulin
.
J. Biol. Chem.
286
,
10115
10125
42
Holman
,
G.D.
,
Lo Leggio
,
L.
and
Cushman
,
S.W.
(
1994
)
Insulin-stimulated GLUT4 glucose transporter recycling. A problem in membrane protein subcellular trafficking through multiple pools
.
J. Biol. Chem.
269
,
17516
17524
43
Yang
,
J.
,
Hodel
,
A.
and
Holman
,
G.D.
(
2002
)
Insulin and isoproterenol have opposing roles in the maintenance of cytosol pH and optimal fusion of GLUT4 vesicles with the plasma membrane
.
J. Biol. Chem.
277
,
6559
6566
44
Bai
,
L.
,
Wang
,
Y.
,
Fan
,
J.
,
Chen
,
Y.
,
Ji
,
W.
,
Qu
,
A.
et al (
2007
)
Dissecting multiple steps of GLUT4 trafficking and identifying the sites of insulin action
.
Cell Metab.
5
,
47
57
45
Burchfield
,
J.G.
,
Lu
,
J.
,
Fazakerley
,
D.J.
,
Tan
,
S.X.
,
Ng
,
Y.
,
Mele
,
K.
et al (
2013
)
Novel systems for dynamically assessing insulin action in live cells reveals heterogeneity in the insulin response
.
Traffic
14
,
259
273
46
Lizunov
,
V.A.
,
Stenkula
,
K.G.
,
Lisinski
,
I.
,
Gavrilova
,
O.
,
Yver
,
D.R.
,
Chadt
,
A.
et al (
2012
)
Insulin stimulates fusion, but not tethering, of GLUT4 vesicles in skeletal muscle of HA-GLUT4-GFP transgenic mice
.
Am. J. Physiol. Endocrinol. Metab.
302
,
E950
E960
47
Lizunov
,
V.A.
,
Stenkula
,
K.
,
Troy
,
A.
,
Cushman
,
S.W.
and
Zimmerberg
,
J.
(
2013
)
Insulin regulates Glut4 confinement in plasma membrane clusters in adipose cells
.
PLoS One
8
,
e57559
48
Lizunov
,
V.A.
,
Matsumoto
,
H.
,
Zimmerberg
,
J.
,
Cushman
,
S.W.
and
Frolov
,
V.A.
(
2005
)
Insulin stimulates the halting, tethering, and fusion of mobile GLUT4 vesicles in rat adipose cells
.
J. Cell Biol.
169
,
481
489
49
Stenkula
,
K.G.
,
Lizunov
,
V.A.
,
Cushman
,
S.W.
and
Zimmerberg
,
J.
(
2010
)
Insulin controls the spatial distribution of GLUT4 on the cell surface through regulation of its postfusion dispersal
.
Cell Metab.
12
,
250
259
50
Govers
,
R.
,
Coster
,
A.C.
and
James
,
D.E.
(
2004
)
Insulin increases cell surface GLUT4 levels by dose dependently discharging GLUT4 into a cell surface recycling pathway
.
Mol. Cell. Biol.
24
,
6456
6466
51
Coster
,
A.C.
,
Govers
,
R.
and
James
,
D.E.
(
2004
)
Insulin stimulates the entry of GLUT4 into the endosomal recycling pathway by a quantal mechanism
.
Traffic
5
,
763
771
52
Fazakerley
,
D.J.
,
Holman
,
G.D.
,
Marley
,
A.
,
James
,
D.E.
,
Stockli
,
J.
and
Coster
,
A.C.
(
2010
)
Kinetic evidence for unique regulation of GLUT4 trafficking by insulin and AMP-activated protein kinase activators in L6 myotubes
.
J. Biol. Chem.
285
,
1653
1660
53
Brewer
,
P.D.
,
Romenskaia
,
I.
,
Kanow
,
M.A.
and
Mastick
,
C.C.
(
2011
)
Loss of AS160 Akt substrate causes Glut4 protein to accumulate in compartments that are primed for fusion in basal adipocytes
.
J. Biol. Chem.
286
,
26287
26297
54
Brewer
,
P.D.
,
Habtemichael
,
E.N.
,
Romenskaia
,
I.
,
Mastick
,
C.C.
and
Coster
,
A.C.
(
2014
)
Insulin-regulated Glut4 translocation: membrane protein trafficking with six distinctive steps
.
J. Biol. Chem.
289
,
17280
17298
55
Brewer
,
P.D.
,
Habtemichael
,
E.N.
,
Romenskaia
,
I.
,
Coster
,
A.C.
and
Mastick
,
C.C.
(
2016
)
Rab14 limits the sorting of Glut4 from endosomes into insulin-sensitive regulated secretory compartments in adipocytes
.
Biochem. J.
473
,
1315
1327
56
Brewer
,
P.D.
,
Habtemichael
,
E.N.
,
Romenskaia
,
I.
,
Mastick
,
C.C.
and
Coster
,
A.C.
(
2016
)
Glut4 is sorted from a Rab10 GTPase-independent constitutive recycling pathway into a highly insulin-responsive Rab10 GTPase-dependent sequestration pathway after adipocyte differentiation
.
J. Biol. Chem.
291
,
773
789
57
Koumanov
,
F.
,
Jin
,
B.
,
Yang
,
J.
and
Holman
,
G.D.
(
2005
)
Insulin signaling meets vesicle traffic of GLUT4 at a plasma-membrane-activated fusion step
.
Cell Metab.
2
,
179
189
58
Koumanov
,
F.
,
Richardson
,
J.D.
,
Murrow
,
B.A.
and
Holman
,
G.D.
(
2011
)
AS160 phosphotyrosine-binding domain constructs inhibit insulin-stimulated GLUT4 vesicle fusion with the plasma membrane
.
J. Biol. Chem.
286
,
16574
16582
59
Tan
,
S.X.
,
Ng
,
Y.
,
Burchfield
,
J.G.
,
Ramm
,
G.
,
Lambright
,
D.G.
,
Stockli
,
J.
et al (
2012
)
The Rab GTPase-activating protein TBC1D4/AS160 contains an atypical phosphotyrosine-binding domain that interacts with plasma membrane phospholipids to facilitate GLUT4 trafficking in adipocytes
.
Mol. Cell. Biol.
32
,
4946
4959
60
Jiang
,
L.
,
Fan
,
J.
,
Bai
,
L.
,
Wang
,
Y.
,
Chen
,
Y.
,
Yang
,
L.
et al (
2008
)
Direct quantification of fusion rate reveals a distal role for AS160 in insulin-stimulated fusion of GLUT4 storage vesicles
.
J. Biol. Chem.
283
,
8508
8516
61
Kristiansen
,
S.
and
Richter
,
E.A.
(
2002
)
GLUT4-containing vesicles are released from membranes by phospholipase D cleavage of a GPI anchor
.
Am. J. Physiol. Endocrinol. Metab.
283
,
E374
E382
62
Xu
,
Z.
and
Kandror
,
K.V.
(
2002
)
Translocation of small preformed vesicles is responsible for the insulin activation of glucose transport in adipose cells. Evidence from the in vitro reconstitution assay
.
J. Biol. Chem.
277
,
47972
47975
63
Brumfield
,
A.
,
Chaudhary
,
N.
,
Molle
,
D.
,
Wen
,
J.
,
Graumann
,
J.
and
McGraw
,
T.E.
(
2021
)
Insulin-promoted mobilization of GLUT4 from a perinuclear storage site requires RAB10
.
Mol. Biol. Cell
32
,
57
73
64
Lopez
,
J.A.
,
Burchfield
,
J.G.
,
Blair
,
D.H.
,
Mele
,
K.
,
Ng
,
Y.
,
Vallotton
,
P.
et al (
2009
)
Identification of a distal GLUT4 trafficking event controlled by actin polymerization
.
Mol. Biol. Cell
20
,
3918
3929
65
Gao
,
L.
,
Chen
,
J.
,
Gao
,
J.
,
Wang
,
H.
and
Xiong
,
W.
(
2017
)
Super-resolution microscopy reveals the insulin-resistance-regulated reorganization of GLUT4 on plasma membranes
.
J. Cell Sci.
130
,
396
405
66
Koester
,
A.M.
,
Laidlaw
,
K.M.
,
Morris
,
S.
,
Cutiongco
,
M.F.A.
,
Stirrat
,
L.
,
Gadegaard
,
N.
et al (
2021
)
EFR3 and phosphatidylinositol 4-kinase IIIα regulate insulin-stimulated glucose transport and GLUT4 dispersal in 3T3-L1 adipocytes
.
bioRxiv
2021.2006.2002.446733
67
Sano
,
H.
,
Roach
,
W.G.
,
Peck
,
G.R.
,
Fukuda
,
M.
and
Lienhard
,
G.E.
(
2008
)
Rab10 in insulin-stimulated GLUT4 translocation
.
Biochem. J.
411
,
89
95
68
Yoshizaki
,
T.
,
Imamura
,
T.
,
Babendure
,
J.L.
,
Lu
,
J.C.
,
Sonoda
,
N.
and
Olefsky
,
J.M.
(
2007
)
Myosin 5a is an insulin-stimulated Akt2 (protein kinase Bbeta) substrate modulating GLUT4 vesicle translocation
.
Mol. Cell. Biol.
27
,
5172
5183
69
Yamada
,
E.
,
Okada
,
S.
,
Saito
,
T.
,
Ohshima
,
K.
,
Sato
,
M.
,
Tsuchiya
,
T.
et al (
2005
)
Akt2 phosphorylates Synip to regulate docking and fusion of GLUT4-containing vesicles
.
J. Cell Biol.
168
,
921
928
70
Nagano
,
K.
,
Takeuchi
,
H.
,
Gao
,
J.
,
Mori
,
Y.
,
Otani
,
T.
,
Wang
,
D.
et al (
2015
)
Tomosyn is a novel Akt substrate mediating insulin-dependent GLUT4 exocytosis
.
Int. J. Biochem. Cell Biol.
62
,
62
71
71
Chen
,
X.W.
,
Leto
,
D.
,
Xiong
,
T.
,
Yu
,
G.
,
Cheng
,
A.
,
Decker
,
S.
et al (
2011
)
A Ral GAP complex links PI 3-kinase/Akt signaling to RalA activation in insulin action
.
Mol. Biol. Cell
22
,
141
152
72
Jewell
,
J.L.
,
Oh
,
E.
,
Ramalingam
,
L.
,
Kalwat
,
M.A.
,
Tagliabracci
,
V.S.
,
Tackett
,
L.
et al (
2011
)
Munc18c phosphorylation by the insulin receptor links cell signaling directly to SNARE exocytosis
.
J. Cell Biol.
193
,
185
199
73
Koumanov
,
F.
,
Pereira
,
V.J.
,
Richardson
,
J.D.
,
Sargent
,
S.L.
,
Fazakerley
,
D.J.
and
Holman
,
G.D.
(
2015
)
Insulin regulates Rab3-Noc2 complex dissociation to promote GLUT4 translocation in rat adipocytes
.
Diabetologia
58
,
1877
1886
74
Chang
,
L.
,
Adams
,
R.D.
and
Saltiel
,
A.R.
(
2002
)
The TC10-interacting protein CIP4/2 is required for insulin-stimulated Glut4 translocation in 3T3L1 adipocytes
.
Proc. Natl Acad. Sci. U.S.A.
99
,
12835
12840
75
Chen
,
X.W.
,
Leto
,
D.
,
Chiang
,
S.H.
,
Wang
,
Q.
and
Saltiel
,
A.R.
(
2007
)
Activation of RalA is required for insulin-stimulated Glut4 trafficking to the plasma membrane via the exocyst and the motor protein Myo1c
.
Dev. Cell
13
,
391
404
76
Chen
,
X.W.
,
Leto
,
D.
,
Xiao
,
J.
,
Goss
,
J.
,
Wang
,
Q.
,
Shavit
,
J.A.
et al (
2011
)
Exocyst function is regulated by effector phosphorylation
.
Nat. Cell Biol.
13
,
580
588
77
Uhm
,
M.
,
Bazuine
,
M.
,
Zhao
,
P.
,
Chiang
,
S.H.
,
Xiong
,
T.
,
Karunanithi
,
S.
et al (
2017
)
Phosphorylation of the exocyst protein Exo84 by TBK1 promotes insulin-stimulated GLUT4 trafficking
.
Sci. Signal.
10
,
eaah5085
78
Cheatham
,
B.
,
Volchuk
,
A.
,
Kahn
,
C.R.
,
Wang
,
L.
,
Rhodes
,
C.J.
and
Klip
,
A.
(
1996
)
Insulin-stimulated translocation of GLUT4 glucose transporters requires SNARE-complex proteins
.
Proc. Natl Acad. Sci. U.S.A.
93
,
15169
15173
79
Volchuk
,
A.
,
Wang
,
Q.
,
Ewart
,
H.S.
,
Liu
,
Z.
,
He
,
L.
,
Bennett
,
M.K.
et al (
1996
)
Syntaxin 4 in 3T3-L1 adipocytes: regulation by insulin and participation in insulin-dependent glucose transport
.
Mol. Biol. Cell
7
,
1075
1082
80
Rea
,
S.
,
Martin
,
L.B.
,
McIntosh
,
S.
,
Macaulay
,
S.L.
,
Ramsdale
,
T.
,
Baldini
,
G.
et al (
1998
)
Syndet, an adipocyte target SNARE involved in the insulin-induced translocation of GLUT4 to the cell surface
.
J. Biol. Chem.
273
,
18784
18792
81
Macaulay
,
S.L.
,
Rea
,
S.
,
Gough
,
K.H.
,
Ward
,
C.W.
and
James
,
D.E.
(
1997
)
Botulinum E toxin light chain does not cleave SNAP-23 and only partially impairs insulin stimulation of GLUT4 translocation in 3T3-L1 cells
.
Biochem. Biophys. Res. Commun.
237
,
388
393
82
Kioumourtzoglou
,
D.
,
Gould
,
G.W.
and
Bryant
,
N.J.
(
2014
)
Insulin stimulates syntaxin4 SNARE complex assembly via a novel regulatory mechanism
.
Mol. Cell. Biol.
34
,
1271
1279
83
Araki
,
S.
,
Tamori
,
Y.
,
Kawanishi
,
M.
,
Shinoda
,
H.
,
Masugi
,
J.
,
Mori
,
H.
et al (
1997
)
Inhibition of the binding of SNAP-23 to syntaxin 4 by Munc18c
.
Biochem. Biophys. Res. Commun.
234
,
257
262
84
Khan
,
A.H.
,
Thurmond
,
D.C.
,
Yang
,
C.
,
Ceresa
,
B.P.
,
Sigmund
,
C.D.
and
Pessin
,
J.E.
(
2001
)
Munc18c regulates insulin-stimulated glut4 translocation to the transverse tubules in skeletal muscle
.
J. Biol. Chem.
276
,
4063
4069
85
Tamori
,
Y.
,
Kawanishi
,
M.
,
Niki
,
T.
,
Shinoda
,
H.
,
Araki
,
S.
,
Okazawa
,
H.
et al (
1998
)
Inhibition of insulin-induced GLUT4 translocation by Munc18c through interaction with syntaxin4 in 3T3-L1 adipocytes
.
J. Biol. Chem.
273
,
19740
19746
86
Thurmond
,
D.C.
,
Ceresa
,
B.P.
,
Okada
,
S.
,
Elmendorf
,
J.S.
,
Coker
,
K.
and
Pessin
,
J.E.
(
1998
)
Regulation of insulin-stimulated GLUT4 translocation by Munc18c in 3T3L1 adipocytes
.
J. Biol. Chem.
273
,
33876
33883
87
Kanda
,
H.
,
Tamori
,
Y.
,
Shinoda
,
H.
,
Yoshikawa
,
M.
,
Sakaue
,
M.
,
Udagawa
,
J.
et al (
2005
)
Adipocytes from Munc18c-null mice show increased sensitivity to insulin-stimulated GLUT4 externalization
.
J. Clin. Invest.
115
,
291
301
88
Oh
,
E.
and
Thurmond
,
D.C.
(
2006
)
The stimulus-induced tyrosine phosphorylation of Munc18c facilitates vesicle exocytosis
.
J. Biol. Chem.
281
,
17624
17634
89
Kioumourtzoglou
,
D.
,
Gould
,
G.W.
and
Bryant
,
N.J.
(
2018
)
Proximity ligation assay to study the GLUT4 membrane trafficking machinery
.
Methods Mol. Biol.
1713
,
217
227
90
Jewell
,
J.L.
,
Oh
,
E.
,
Bennett
,
S.M.
,
Meroueh
,
S.O.
and
Thurmond
,
D.C.
(
2008
)
The tyrosine phosphorylation of Munc18c induces a switch in binding specificity from syntaxin 4 to Doc2beta
.
J. Biol. Chem.
283
,
21734
21746
91
Aran
,
V.
,
Bryant
,
N.J.
and
Gould
,
G.W.
(
2011
)
Tyrosine phosphorylation of Munc18c on residue 521 abrogates binding to Syntaxin 4
.
BMC Biochem.
12
,
19
92
Sano
,
H.
,
Peck
,
G.R.
,
Kettenbach
,
A.N.
,
Gerber
,
S.A.
and
Lienhard
,
G.E.
(
2011
)
Insulin-stimulated GLUT4 protein translocation in adipocytes requires the Rab10 guanine nucleotide exchange factor Dennd4C
.
J. Biol. Chem.
286
,
16541
16545
93
Weber
,
T.
,
Parlati
,
F.
,
McNew
,
J.A.
,
Johnston
,
R.J.
,
Westermann
,
B.
,
Sollner
,
T.H.
et al (
2000
)
SNAREpins are functionally resistant to disruption by NSF and alphaSNAP
.
J. Cell Biol.
149
,
1063
1072
94
Shen
,
J.
,
Tareste
,
D.C.
,
Paumet
,
F.
,
Rothman
,
J.E.
and
Melia
,
T.J.
(
2007
)
Selective activation of cognate SNAREpins by Sec1/Munc18 proteins
.
Cell
128
,
183
195
95
Zhao
,
M.
,
Wu
,
S.
,
Zhou
,
Q.
,
Vivona
,
S.
,
Cipriano
,
D.J.
,
Cheng
,
Y.
et al (
2015
)
Mechanistic insights into the recycling machine of the SNARE complex
.
Nature
518
,
61
67
96
Wang
,
S.
,
Crisman
,
L.
,
Miller
,
J.
,
Datta
,
I.
,
Gulbranson
,
D.R.
,
Tian
,
Y.
et al (
2019
)
Inducible Exoc7/Exo70 knockout reveals a critical role of the exocyst in insulin-regulated GLUT4 exocytosis
.
J. Biol. Chem.
294
,
19988
19996
97
Wang
,
S.
,
Liu
,
Y.
,
Crisman
,
L.
,
Wan
,
C.
,
Miller
,
J.
,
Yu
,
H.
et al (
2020
)
Genetic evidence for an inhibitory role of tomosyn in insulin-stimulated GLUT4 exocytosis
.
Traffic
21
,
636
646
98
Robinson
,
L.J.
,
Pang
,
S.
,
Harris
,
D.S.
,
Heuser
,
J.
and
James
,
D.E.
(
1992
)
Translocation of the glucose transporter (GLUT4) to the cell surface in permeabilized 3T3-L1 adipocytes: effects of ATP insulin, and GTP gamma S and localization of GLUT4 to clathrin lattices
.
J. Cell Biol.
117
,
1181
1196
99
Lienhard
,
G.E.
(
2008
)
Non-functional phosphorylations?
Trends Biochem. Sci.
33
,
351
352
100
Eickelschulte
,
S.
,
Hartwig
,
S.
,
Leiser
,
B.
,
Lehr
,
S.
,
Joschko
,
V.
,
Chokkalingam
,
M.
et al (
2021
)
AKT/AMPK-mediated phosphorylation of TBC1D4 disrupts the interaction with insulin-regulated aminopeptidase
.
J. Biol. Chem.
296
,
100637
101
Blot
,
V.
and
McGraw
,
T.E.
(
2006
)
GLUT4 is internalized by a cholesterol-dependent nystatin-sensitive mechanism inhibited by insulin
.
EMBO J.
25
,
5648
5658
102
Carvalho
,
E.
,
Schellhorn
,
S.E.
,
Zabolotny
,
J.M.
,
Martin
,
S.
,
Tozzo
,
E.
,
Peroni
,
O.D.
et al (
2004
)
GLUT4 overexpression or deficiency in adipocytes of transgenic mice alters the composition of GLUT4 vesicles and the subcellular localization of GLUT4 and insulin-responsive aminopeptidase
.
J. Biol. Chem.
279
,
21598
21605
103
Lizunov
,
V.A.
,
Lee
,
J.P.
,
Skarulis
,
M.C.
,
Zimmerberg
,
J.
,
Cushman
,
S.W.
and
Stenkula
,
K.G.
(
2013
)
Impaired tethering and fusion of GLUT4 vesicles in insulin-resistant human adipose cells
.
Diabetes
62
,
3114
3119
104
Cushman
,
S.W.
,
Goodyear
,
L.J.
,
Pilch
,
P.F.
,
Ralston
,
E.
,
Galbo
,
H.
,
Ploug
,
T.
et al (
1998
)
Molecular mechanisms involved in GLUT4 translocation in muscle during insulin and contraction stimulation
.
Adv. Exp. Med. Biol.
441
,
63
71
105
Lauritzen
,
H.P.
,
Galbo
,
H.
,
Brandauer
,
J.
,
Goodyear
,
L.J.
and
Ploug
,
T.
(
2008
)
Large GLUT4 vesicles are stationary while locally and reversibly depleted during transient insulin stimulation of skeletal muscle of living mice: imaging analysis of GLUT4-enhanced Green fluorescent protein vesicle dynamics
.
Diabetes
57
,
315
324
106
Schmelzle
,
K.
,
Kane
,
S.
,
Gridley
,
S.
,
Lienhard
,
G.E.
and
White
,
F.M.
(
2006
)
Temporal dynamics of tyrosine phosphorylation in insulin signaling
.
Diabetes
55
,
2171
2179
107
Brannmark
,
C.
,
Nyman
,
E.
,
Fagerholm
,
S.
,
Bergenholm
,
L.
,
Ekstrand
,
E.M.
,
Cedersund
,
G.
et al (
2013
)
Insulin signaling in type 2 diabetes: experimental and modeling analyses reveal mechanisms of insulin resistance in human adipocytes
.
J. Biol. Chem.
288
,
9867
9880
108
Nyman
,
E.
,
Cedersund
,
G.
and
Stralfors
,
P.
(
2012
)
Insulin signaling - mathematical modeling comes of age
.
Trends Endocrinol. Metab.
23
,
107
115
109
Giorgetti
,
S.
,
Ballotti
,
R.
,
Kowalski-Chauvel
,
A.
,
Cormont
,
M.
and
Van Obberghen
,
E.
(
1992
)
Insulin stimulates phosphatidylinositol-3-kinase activity in rat adipocytes
.
Eur. J. Biochem.
207
,
599
606
110
Sedaghat
,
A.R.
,
Sherman
,
A.
and
Quon
,
M.J.
(
2002
)
A mathematical model of metabolic insulin signaling pathways
.
Am. J. Physiol. Endocrinol. Metab.
283
,
E1084
E1101
111
Oatey
,
P.B.
,
Venkateswarlu
,
K.
,
Williams
,
A.G.
,
Fletcher
,
L.M.
,
Foulstone
,
E.J.
,
Cullen
,
P.J.
et al (
1999
)
Confocal imaging of the subcellular distribution of phosphatidylinositol 3,4,5-trisphosphate in insulin- and PDGF-stimulated 3T3-L1 adipocytes
.
Biochem J.
344
Pt 2
,
511
518
112
Gray
,
A.
,
Van Der Kaay
,
J.
and
Downes
,
C.P.
(
1999
)
The pleckstrin homology domains of protein kinase B and GRP1 (general receptor for phosphoinositides-1) are sensitive and selective probes for the cellular detection of phosphatidylinositol 3,4-bisphosphate and/or phosphatidylinositol 3,4,5-trisphosphate in vivo
.
Biochem J.
344
,
929
936
113
Guilherme
,
A.
,
Soriano
,
N.A.
,
Furcinitti
,
P.S.
and
Czech
,
M.P.
(
2004
)
Role of EHD1 and EHBP1 in perinuclear sorting and insulin-regulated GLUT4 recycling in 3T3-L1 adipocytes
.
J. Biol. Chem.
279
,
40062
40075
114
Guilherme
,
A.
,
Soriano
,
N.A.
,
Bose
,
S.
,
Holik
,
J.
,
Bose
,
A.
,
Pomerleau
,
D.P.
et al (
2004
)
EHD2 and the novel EH domain binding protein EHBP1 couple endocytosis to the actin cytoskeleton
.
J. Biol. Chem.
279
,
10593
10605
115
Chang
,
L.
,
Chiang
,
S.H.
and
Saltiel
,
A.R.
(
2007
)
TC10alpha is required for insulin-stimulated glucose uptake in adipocytes
.
Endocrinology
148
,
27
33
116
Baumann
,
C.A.
,
Ribon
,
V.
,
Kanzaki
,
M.
,
Thurmond
,
D.C.
,
Mora
,
S.
,
Shigematsu
,
S.
et al (
2000
)
CAP defines a second signalling pathway required for insulin-stimulated glucose transport
.
Nature
407
,
202
207
117
Lim
,
C.Y.
,
Bi
,
X.
,
Wu
,
D.
,
Kim
,
J.B.
,
Gunning
,
P.W.
,
Hong
,
W.
et al (
2015
)
Tropomodulin3 is a novel Akt2 effector regulating insulin-stimulated GLUT4 exocytosis through cortical actin remodeling
.
Nat. Commun.
6
,
5951
118
Kee
,
A.J.
,
Yang
,
L.
,
Lucas
,
C.A.
,
Greenberg
,
M.J.
,
Martel
,
N.
,
Leong
,
G.M.
et al (
2015
)
An actin filament population defined by the tropomyosin Tpm3.1 regulates glucose uptake
.
Traffic
16
,
691
711
119
Kruse
,
R.
,
Krantz
,
J.
,
Barker
,
N.
,
Coletta
,
R.L.
,
Rafikov
,
R.
,
Luo
,
M.
et al (
2017
)
Characterization of the CLASP2 protein interaction network identifies SOGA1 as a microtubule-associated protein
.
Mol. Cell Proteom.
16
,
1718
1735
120
Langlais
,
P.
,
Dillon
,
J.L.
,
Mengos
,
A.
,
Baluch
,
D.P.
,
Ardebili
,
R.
,
Miranda
,
D.N.
et al (
2012
)
Identification of a role for CLASP2 in insulin action
.
J. Biol. Chem.
287
,
39245
39253
121
Chiu
,
T.T.
,
Jensen
,
T.E.
,
Sylow
,
L.
,
Richter
,
E.A.
and
Klip
,
A.
(
2011
)
Rac1 signalling towards GLUT4/glucose uptake in skeletal muscle
.
Cell Signal.
23
,
1546
1554
122
Sylow
,
L.
,
Kleinert
,
M.
,
Pehmoller
,
C.
,
Prats
,
C.
,
Chiu
,
T.T.
,
Klip
,
A.
et al (
2014
)
Akt and Rac1 signaling are jointly required for insulin-stimulated glucose uptake in skeletal muscle and downregulated in insulin resistance
.
Cell Signal.
26
,
323
331
123
Sun
,
Y.
,
Jaldin-Fincati
,
J.
,
Liu
,
Z.
,
Bilan
,
P.J.
and
Klip
,
A.
(
2016
)
A complex of Rab13 with MICAL-L2 and alpha-actinin-4 is essential for insulin-dependent GLUT4 exocytosis
.
Mol. Biol. Cell
27
,
75
89
124
Kane
,
S.
,
Sano
,
H.
,
Liu
,
S.C.
,
Asara
,
J.M.
,
Lane
,
W.S.
,
Garner
,
C.C.
et al (
2002
)
A method to identify serine kinase substrates. Akt phosphorylates a novel adipocyte protein with a Rab GTPase-activating protein (GAP) domain
.
J. Biol. Chem.
277
,
22115
22118
125
Ramm
,
G.
,
Larance
,
M.
,
Guilhaus
,
M.
and
James
,
D.E.
(
2006
)
A role for 14-3-3 in insulin-stimulated GLUT4 translocation through its interaction with the RabGAP AS160
.
J. Biol. Chem.
281
,
29174
29180
126
Stockli
,
J.
,
Davey
,
J.R.
,
Hohnen-Behrens
,
C.
,
Xu
,
A.
,
James
,
D.E.
and
Ramm
,
G.
(
2008
)
Regulation of glucose transporter 4 translocation by the Rab guanosine triphosphatase-activating protein AS160/TBC1D4: role of phosphorylation and membrane association
.
Mol. Endocrinol.
22
,
2703
2715
127
Chen
,
S.
,
Murphy
,
J.
,
Toth
,
R.
,
Campbell
,
D.G.
,
Morrice
,
N.A.
and
Mackintosh
,
C.
(
2008
)
Complementary regulation of TBC1D1 and AS160 by growth factors, insulin and AMPK activators
.
Biochem. J.
409
,
449
459
128
Larance
,
M.
,
Ramm
,
G.
,
Stockli
,
J.
,
van Dam
,
E.M.
,
Winata
,
S.
,
Wasinger
,
V.
et al (
2005
)
Characterization of the role of the Rab GTPase-activating protein AS160 in insulin-regulated GLUT4 trafficking
.
J. Biol. Chem.
280
,
37803
37813
129
Bruno
,
J.
,
Brumfield
,
A.
,
Chaudhary
,
N.
,
Iaea
,
D.
and
McGraw
,
T.E.
(
2016
)
SEC 16a is a RAB 10 effector required for insulin-stimulated GLUT4 trafficking in adipocytes
.
J. Cell Biol.
214
,
61
76
130
Bruno
,
J.H.
,
Brumfeld
,
A.
,
Iaea
,
D.
and
Mcgraw
,
T.E.
(
2015
)
The RAB10 effector protein SEC16A regulates GLUT4 trafficking in adipocytes
.
Diabetes
64
,
A489
A489
131
Peck
,
G.R.
,
Chavez
,
J.A.
,
Roach
,
W.G.
,
Budnik
,
B.A.
,
Lane
,
W.S.
,
Karlsson
,
H.K.
et al (
2009
)
Insulin-stimulated phosphorylation of the Rab GTPase-activating protein TBC1D1 regulates GLUT4 translocation
.
J. Biol. Chem.
284
,
30016
30023
132
Mafakheri
,
S.
,
Florke
,
R.R.
,
Kanngiesser
,
S.
,
Hartwig
,
S.
,
Espelage
,
L.
,
De Wendt
,
C.
et al (
2018
)
AKT and AMP-activated protein kinase regulate TBC1D1 through phosphorylation and its interaction with the cytosolic tail of insulin-regulated aminopeptidase IRAP
.
J. Biol. Chem.
293
,
17853
17862
133
Chadt
,
A.
,
Leicht
,
K.
,
Deshmukh
,
A.
,
Jiang
,
L.Q.
,
Scherneck
,
S.
,
Bernhardt
,
U.
et al (
2008
)
Tbc1d1 mutation in lean mouse strain confers leanness and protects from diet-induced obesity
.
Nat. Genet.
40
,
1354
1359
134
Sun
,
Y.
,
Chiu
,
T.T.
,
Foley
,
K.P.
,
Bilan
,
P.J.
and
Klip
,
A.
(
2014
)
Myosin Va mediates Rab8A-regulated GLUT4 vesicle exocytosis in insulin-stimulated muscle cells
.
Mol. Biol. Cell
25
,
1159
1170
135
Chen
,
Y.
,
Wang
,
Y.
,
Zhang
,
J.
,
Deng
,
Y.
,
Jiang
,
L.
,
Song
,
E.
et al (
2012
)
Rab10 and myosin-Va mediate insulin-stimulated GLUT4 storage vesicle translocation in adipocytes
.
J. Cell Biol.
198
,
545
560
136
Inoue
,
M.
,
Chang
,
L.
,
Hwang
,
J.
,
Chiang
,
S.H.
and
Saltiel
,
A.R.
(
2003
)
The exocyst complex is required for targeting of Glut4 to the plasma membrane by insulin
.
Nature
422
,
629
633
137
Sano
,
H.
,
Peck
,
G.R.
,
Blachon
,
S.
and
Lienhard
,
G.E.
(
2015
)
A potential link between insulin signaling and GLUT4 translocation: association of Rab10-GTP with the exocyst subunit Exoc6/6b
.
Biochem. Biophys. Res. Commun.
465
,
601
605
138
Ewart
,
M.A.
,
Clarke
,
M.
,
Kane
,
S.
,
Chamberlain
,
L.H.
and
Gould
,
G.W.
(
2005
)
Evidence for a role of the exocyst in insulin-stimulated Glut4 trafficking in 3T3-L1 adipocytes
.
J. Biol. Chem.
280
,
3812
3816
139
Bose
,
A.
,
Guilherme
,
A.
,
Robida
,
S.I.
,
Nicoloro
,
S.M.
,
Zhou
,
Q.L.
,
Jiang
,
Z.Y.
et al (
2002
)
Glucose transporter recycling in response to insulin is facilitated by myosin Myo1c
.
Nature
420
,
821
824
140
Bose
,
A.
,
Robida
,
S.
,
Furcinitti
,
P.S.
,
Chawla
,
A.
,
Fogarty
,
K.
,
Corvera
,
S.
et al (
2004
)
Unconventional myosin Myo1c promotes membrane fusion in a regulated exocytic pathway
.
Mol. Cell. Biol.
24
,
5447
5458
141
Yip
,
M.F.
,
Ramm
,
G.
,
Larance
,
M.
,
Hoehn
,
K.L.
,
Wagner
,
M.C.
,
Guilhaus
,
M.
et al (
2008
)
CaMKII-mediated phosphorylation of the myosin motor Myo1c is required for insulin-stimulated GLUT4 translocation in adipocytes
.
Cell Metab.
8
,
384
398
142
Boguslavsky
,
S.
,
Chiu
,
T.
,
Foley
,
K.P.
,
Osorio-Fuentealba
,
C.
,
Antonescu
,
C.N.
,
Bayer
,
K.U.
et al (
2012
)
Myo1c binding to submembrane actin mediates insulin-induced tethering of GLUT4 vesicles
.
Mol. Biol. Cell
23
,
4065
4078
143
Fukuda
,
N.
,
Emoto
,
M.
,
Nakamori
,
Y.
,
Taguchi
,
A.
,
Miyamoto
,
S.
,
Uraki
,
S.
et al (
2009
)
DOC2B: a novel syntaxin-4 binding protein mediating insulin-regulated GLUT4 vesicle fusion in adipocytes
.
Diabetes
58
,
377
384
144
Widberg
,
C.H.
,
Bryant
,
N.J.
,
Girotti
,
M.
,
Rea
,
S.
and
James
,
D.E.
(
2003
)
Tomosyn interacts with the t-SNAREs syntaxin4 and SNAP23 and plays a role in insulin-stimulated GLUT4 translocation
.
J. Biol. Chem.
278
,
35093
35101
145
Yu
,
H.
,
Rathore
,
S.S.
,
Gulbranson
,
D.R.
and
Shen
,
J.
(
2014
)
The N- and C-terminal domains of tomosyn play distinct roles in soluble N-ethylmaleimide-sensitive factor attachment protein receptor binding and fusion regulation
.
J. Biol. Chem.
289
,
25571
25580
146
Timmers
,
K.I.
,
Clark
,
A.E.
,
Omatsu-Kanbe
,
M.
,
Whiteheart
,
S.W.
,
Bennett
,
M.K.
,
Holman
,
G.D.
et al (
1996
)
Identification of SNAP receptors in rat adipose cell membrane fractions and in SNARE complexes co-immunoprecipitated with epitope-tagged N-ethylmaleimide-sensitive fusion protein
.
Biochem. J.
320
(
Pt 2
),
429
436
147
Cain
,
C.C.
,
Trimble
,
W.S.
and
Lienhard
,
G.E.
(
1992
)
Members of the VAMP family of synaptic vesicle proteins are components of glucose transporter-containing vesicles from rat adipocytes
.
J. Biol. Chem.
267
,
11681
11684
148
Pavarotti
,
M.A.
,
Tokarz
,
V.
,
Frendo-Cumbo
,
S.
,
Bilan
,
P.J.
,
Liu
,
Z.
,
Zanni-Ruiz
,
E.
et al (
2021
)
Complexin-2 redistributes to the membrane of muscle cells in response to insulin and contributes to GLUT4 translocation
.
Biochem. J.
478
,
407
422
149
Sollner
,
T.
,
Bennett
,
M.K.
,
Whiteheart
,
S.W.
,
Scheller
,
R.H.
and
Rothman
,
J.E.
(
1993
)
A protein assembly-disassembly pathway in vitro that may correspond to sequential steps of synaptic vesicle docking, activation, and fusion
.
Cell
75
,
409
418
150
Song
,
H.
,
Orr
,
A.
,
Duan
,
M.
,
Merz
,
A.J.
and
Wickner
,
W.
(
2017
)
Sec17/Sec18 act twice, enhancing membrane fusion and then disassembling cis-SNARE complexes
.
eLlife
6
,
e26646
151
Chakrabarti
,
R.
,
Buxton
,
J.
,
Joly
,
M.
and
Corvera
,
S.
(
1994
)
Insulin-sensitive association of GLUT-4 with endocytic clathrin-coated vesicles revealed with the use of brefeldin A
.
J. Biol. Chem.
269
,
7926
7933
152
Al-Hasani
,
H.
,
Hinck
,
C.S.
and
Cushman
,
S.W.
(
1998
)
Endocytosis of the glucose transporter GLUT4 is mediated by the GTPase dynamin
.
J. Biol. Chem.
273
,
17504
17510
153
Kao
,
A.W.
,
Ceresa
,
B.P.
,
Santeler
,
S.R.
and
Pessin
,
J.E.
(
1998
)
Expression of a dominant interfering dynamin mutant in 3T3L1 adipocytes inhibits GLUT4 endocytosis without affecting insulin signaling
.
J. Biol. Chem.
273
,
25450
25457
154
Volchuk
,
A.
,
Narine
,
S.
,
Foster
,
L.J.
,
Grabs
,
D.
,
De Camilli
,
P.
and
Klip
,
A.
(
1998
)
Perturbation of dynamin II with an amphiphysin SH3 domain increases GLUT4 glucose transporters at the plasma membrane in 3T3-L1 adipocytes. Dynamin II participates in GLUT4 endocytosis
.
J. Biol. Chem.
273
,
8169
8176
155
Hartig
,
S.M.
,
Ishikura
,
S.
,
Hicklen
,
R.S.
,
Feng
,
Y.
,
Blanchard
,
E.G.
Voelker
,
K.A.
et al (
2009
)
The F-BAR protein CIP4 promotes GLUT4 endocytosis through bidirectional interactions with N-WASp and Dynamin-2
.
J. Cell Sci.
122
,
2283
2291
156
Lodhi
,
I.J.
,
Chiang
,
S.H.
,
Chang
,
L.
,
Vollenweider
,
D.
,
Watson
,
R.T.
,
Inoue
,
M.
et al (
2007
)
Gapex-5, a Rab31 guanine nucleotide exchange factor that regulates Glut4 trafficking in adipocytes
.
Cell Metab.
5
,
59
72
157
Lodhi
,
I.J.
,
Bridges
,
D.
,
Chiang
,
S.H.
,
Zhang
,
Y.
,
Cheng
,
A.
,
Geletka
,
L.M.
et al (
2008
)
Insulin stimulates phosphatidylinositol 3-phosphate production via the activation of Rab5
.
Mol. Biol. Cell
19
,
2718
2728
158
Su
,
X.
,
Lodhi
,
I.J.
,
Saltiel
,
A.R.
and
Stahl
,
P.D.
(
2006
)
Insulin-stimulated interaction between insulin receptor substrate 1 and p85alpha and activation of protein kinase B/Akt require Rab5
.
J. Biol. Chem.
281
,
27982
27990
159
Lamb
,
C.A.
,
McCann
,
R.K.
,
Stockli
,
J.
,
James
,
D.E.
and
Bryant
,
N.J.
(
2010
)
Insulin-regulated trafficking of GLUT4 requires ubiquitination
.
Traffic
11
,
1445
1454
160
Sadler
,
J.B.A.
,
Lamb
,
C.A.
,
Welburn
,
C.R.
,
Adamson
,
I.S.
,
Kioumourtzoglou
,
D.
,
Chi
,
N.W.
et al (
2019
)
The deubiquitinating enzyme USP25 binds tankyrase and regulates trafficking of the facilitative glucose transporter GLUT4 in adipocytes
.
Sci. Rep.
9
,
4710
161
Gustavsson
,
J.
,
Parpal
,
S.
,
Karlsson
,
M.
,
Ramsing
,
C.
,
Thorn
,
H.
,
Borg
,
M.
et al (
1999
)
Localization of the insulin receptor in caveolae of adipocyte plasma membrane
.
FASEB J.
13
,
1961
1971
162
Karlsson
,
M.
,
Thorn
,
H.
,
Parpal
,
S.
,
Stralfors
,
P.
and
Gustavsson
,
J.
(
2002
)
Insulin induces translocation of glucose transporter GLUT4 to plasma membrane caveolae in adipocytes
.
FASEB J.
16
,
249
251
163
Scherer
,
P.E.
,
Lisanti
,
M.P.
,
Baldini
,
G.
,
Sargiacomo
,
M.
,
Mastick
,
C.C.
and
Lodish
,
H.F.
(
1994
)
Induction of caveolin during adipogenesis and association of GLUT4 with caveolin-rich vesicles
.
J. Cell Biol.
127
,
1233
1243
164
Jedrychowski
,
M.P.
,
Gartner
,
C.A.
,
Gygi
,
S.P.
,
Zhou
,
L.
,
Herz
,
J.
,
Kandror
,
K.V.
et al (
2010
)
Proteomic analysis of GLUT4 storage vesicles reveals LRP1 to be an important vesicle component and target of insulin signaling
.
J. Biol. Chem.
285
,
104
114
165
Jordens
,
I.
,
Molle
,
D.
,
Xiong
,
W.
,
Keller
,
S.R.
and
McGraw
,
T.E.
(
2010
)
Insulin-regulated aminopeptidase is a key regulator of GLUT4 trafficking by controlling the sorting of GLUT4 from endosomes to specialized insulin-regulated vesicles
.
Mol. Biol. Cell
21
,
2034
2044
166
Pan
,
X.
,
Zaarur
,
N.
,
Singh
,
M.
,
Morin
,
P.
and
Kandror
,
K.V.
(
2017
)
Sortilin and retromer mediate retrograde transport of Glut4 in 3T3-L1 adipocytes
.
Mol. Biol. Cell
28
,
1667
1675
167
Kandror
,
K.V.
and
Pilch
,
P.F.
(
1994
)
Gp160, a tissue-specific marker for insulin-activated glucose transport
.
Proc. Natl Acad. Sci. U.S.A.
91
,
8017
8021
168
Ross
,
S.A.
,
Herbst
,
J.J.
,
Keller
,
S.R.
and
Lienhard
,
G.E.
(
1997
)
Trafficking kinetics of the insulin-regulated membrane aminopeptidase in 3T3-L1 adipocytes
.
Biochem. Biophys. Res. Commun.
239
,
247
251
169
Ross
,
S.A.
,
Scott
,
H.M.
,
Morris
,
N.J.
,
Leung
,
W.Y.
,
Mao
,
F.
,
Lienhard
,
G.E.
et al (
1996
)
Characterization of the insulin-regulated membrane aminopeptidase in 3T3-L1 adipocytes
.
J. Biol. Chem.
271
,
3328
3332
170
Hatakeyama
,
H.
and
Kanzaki
,
M.
(
2011
)
Molecular basis of insulin-responsive GLUT4 trafficking systems revealed by single molecule imaging
.
Traffic
12
,
1805
1820
171
Shi
,
J.
and
Kandror
,
K.V.
(
2005
)
Sortilin is essential and sufficient for the formation of Glut4 storage vesicles in 3T3-L1 adipocytes
.
Dev. Cell
9
,
99
108
172
Ariga
,
M.
,
Nedachi
,
T.
,
Katagiri
,
H.
and
Kanzaki
,
M.
(
2008
)
Functional role of sortilin in myogenesis and development of insulin-responsive glucose transport system in C2C12 myocytes
.
J. Biol. Chem.
283
,
10208
10220
173
Morris
,
N.J.
,
Ross
,
S.A.
,
Lane
,
W.S.
,
Moestrup
,
S.K.
,
Petersen
,
C.M.
,
Keller
,
S.R.
et al (
1998
)
Sortilin is the major 110-kDa protein in GLUT4 vesicles from adipocytes
.
J. Biol. Chem.
273
,
3582
3587
174
Watson
,
R.T.
,
Khan
,
A.H.
,
Furukawa
,
M.
,
Hou
,
J.C.
,
Li
,
L.
,
Kanzaki
,
M.
et al (
2004
)
Entry of newly synthesized GLUT4 into the insulin-responsive storage compartment is GGA dependent
.
EMBO J.
23
,
2059
2070
175
Gillingham
,
A.K.
,
Koumanov
,
F.
,
Pryor
,
P.R.
,
Reaves
,
B.J.
and
Holman
,
G.D.
(
1999
)
Association of AP1 adaptor complexes with GLUT4 vesicles
.
J. Cell Sci.
112
(
Pt 24
),
4793
4800
176
Bernhardt
,
U.
,
Carlotti
,
F.
,
Hoeben
,
R.C.
,
Joost
,
H.G.
and
Al-Hasani
,
H.
(
2009
)
A dual role of the N-terminal FQQI motif in GLUT4 trafficking
.
Biol. Chem.
390
,
883
892
177
Li
,
L. V.
and
Kandror
,
K. V
. (
2005
)
Golgi-localized, gamma-ear-containing, Arf-binding protein adaptors mediate insulin-responsive trafficking of glucose transporter 4 in 3T3-L1 adipocytes
.
Mol. Endocrinol.
19
,
2145
2153
178
Koumanov
,
F.
,
Pereira
,
V.J.
,
Whitley
,
P.R.
and
Holman
,
G.D.
(
2012
)
GLUT4 traffic through an ESCRT-III-dependent sorting compartment in adipocytes
.
PLoS One
7
,
e44141
179
Ma
,
J.
,
Nakagawa
,
Y.
,
Kojima
,
I.
and
Shibata
,
H.
(
2014
)
Prolonged insulin stimulation down-regulates GLUT4 through oxidative stress-mediated retromer inhibition by a protein kinase CK2-dependent mechanism in 3T3-L1 adipocytes
.
J. Biol. Chem.
289
,
133
142
180
Yang
,
Z.
,
Hong
,
L.K.
,
Follett
,
J.
,
Wabitsch
,
M.
,
Hamilton
,
N.A.
,
Collins
,
B.M.
et al (
2016
)
Functional characterization of retromer in GLUT4 storage vesicle formation and adipocyte differentiation
.
FASEB J.
30
,
1037
1050
181
Perera
,
H.K.
,
Clarke
,
M.
,
Morris
,
N.J.
,
Hong
,
W.
,
Chamberlain
,
L.H.
and
Gould
,
G.W.
(
2003
)
Syntaxin 6 regulates Glut4 trafficking in 3T3-L1 adipocytes
.
Mol. Biol. Cell
14
,
2946
2958
182
Proctor
,
K.M.
,
Miller
,
S.C.
,
Bryant
,
N.J.
and
Gould
,
G.W.
(
2006
)
Syntaxin 16 controls the intracellular sequestration of GLUT4 in 3T3-L1 adipocytes
.
Biochem. Biophys. Res. Commun.
347
,
433
438
183
Roccisana
,
J.
,
Sadler
,
J.B.
,
Bryant
,
N.J.
and
Gould
,
G.W.
(
2013
)
Sorting of GLUT4 into its insulin-sensitive store requires the Sec1/Munc18 protein mVps45
.
Mol. Biol. Cell
24
,
2389
2397
184
Boal
,
F.
,
Hodgson
,
L.R.
,
Reed
,
S.E.
,
Yarwood
,
S.E.
,
Just
,
V.J.
,
Stephens
,
D.J.
et al (
2016
)
Insulin promotes Rip11 accumulation at the plasma membrane by inhibiting a dynamin- and PI3-kinase-dependent, but Akt-independent, internalisation event
.
Cell Signal.
28
,
74
82
185
Welsh
,
G.I.
,
Leney
,
S.E.
,
Lloyd-Lewis
,
B.
,
Wherlock
,
M.
,
Lindsay
,
A.J.
,
McCaffrey
,
M.W.
et al (
2007
)
Rip11 is a Rab11- and AS160-RabGAP-binding protein required for insulin-stimulated glucose uptake in adipocytes
.
J. Cell Sci.
120
,
4197
4208
186
Cormont
,
M.
,
Bortoluzzi
,
M.N.
,
Gautier
,
N.
,
Mari
,
M.
,
van Obberghen
,
E.
and
Le Marchand-Brustel
,
Y.
(
1996
)
Potential role of Rab4 in the regulation of subcellular localization of Glut4 in adipocytes
.
Mol. Cell. Biol.
16
,
6879
6886
187
Imamura
,
T.
,
Huang
,
J.
,
Usui
,
I.
,
Satoh
,
H.
,
Bever
,
J.
and
Olefsky
,
J.M.
(
2003
)
Insulin-induced GLUT4 translocation involves protein kinase C-lambda-mediated functional coupling between Rab4 and the motor protein kinesin
.
Mol. Cell. Biol.
23
,
4892
4900
188
Shibata
,
H.
,
Omata
,
W.
and
Kojima
,
I.
(
1997
)
Insulin stimulates guanine nucleotide exchange on Rab4 via a wortmannin-sensitive signaling pathway in rat adipocytes
.
J. Biol. Chem.
272
,
14542
14546
189
Reed
,
S.E.
,
Hodgson
,
L.R.
,
Song
,
S.
,
May
,
M.T.
,
Kelly
,
E.E.
,
McCaffrey
,
M.W.
et al (
2013
)
A role for Rab14 in the endocytic trafficking of GLUT4 in 3T3-L1 adipocytes
.
J. Cell Sci.
126
,
1931
1941
190
Bogan
,
J.S.
,
Rubin
,
B.R.
,
Yu
,
C.
,
Loffler
,
M.G.
,
Orme
,
C.M.
,
Belman
,
J.P.
et al (
2012
)
Endoproteolytic cleavage of TUG protein regulates GLUT4 glucose transporter translocation
.
J. Biol. Chem.
287
,
23932
23947
191
Yu
,
C.
,
Cresswell
,
J.
,
Loffler
,
M.G.
and
Bogan
,
J.S.
(
2007
)
The glucose transporter 4-regulating protein TUG is essential for highly insulin-responsive glucose uptake in 3T3-L1 adipocytes
.
J. Biol. Chem.
282
,
7710
7722
192
Habtemichael
,
E.N.
,
Li
,
D.T.
,
Alcazar-Roman
,
A.
,
Westergaard
,
X.O.
,
Li
,
M.
,
Petersen
,
M.C.
et al (
2018
)
Usp25m protease regulates ubiquitin-like processing of TUG proteins to control GLUT4 glucose transporter translocation in adipocytes
.
J. Biol. Chem.
293
,
10466
10486
193
Fazakerley
,
D.J.
,
Naghiloo
,
S.
,
Chaudhuri
,
R.
,
Koumanov
,
F.
,
Burchfield
,
J.G.
,
Thomas
,
K.C.
et al (
2015
)
Proteomic analysis of GLUT4 storage vesicles reveals tumor suppressor candidate 5 (TUSC5) as a novel regulator of insulin action in adipocytes
.
J. Biol. Chem.
290
,
23528
23542
194
Beaton
,
N.
,
Rudigier
,
C.
,
Moest
,
H.
,
Muller
,
S.
,
Mrosek
,
N.
,
Roder
,
E.
et al (
2015
)
TUSC5 regulates insulin-mediated adipose tissue glucose uptake by modulation of GLUT4 recycling
.
Mol. Metab.
4
,
795
810
195
Davey
,
J.R.
,
Humphrey
,
S.J.
,
Junutula
,
J.R.
,
Mishra
,
A.K.
,
Lambright
,
D.G.
,
James
,
D.E.
et al (
2012
)
TBC1D13 is a RAB35 specific GAP that plays an important role in GLUT4 trafficking in adipocytes
.
Traffic
13
,
1429
1441
196
Camus
,
S.M.
,
Camus
,
M.D.
,
Figueras-Novoa
,
C.
,
Boncompain
,
G.
,
Sadacca
,
L.A.
,
Esk
,
C.
et al (
2020
)
CHC22 clathrin mediates traffic from early secretory compartments for human GLUT4 pathway biogenesis
.
J. Cell Biol.
219
,
e201812135
197
Li
,
J.
,
Peters
,
P.J.
,
Bai
,
M.
,
Dai
,
J.
,
Bos
,
E.
,
Kirchhausen
,
T.
et al (
2007
)
An ACAP1-containing clathrin coat complex for endocytic recycling
.
J. Cell Biol.
178
,
453
464
198
Williams
,
D.
and
Pessin
,
J.E.
(
2008
)
Mapping of R-SNARE function at distinct intracellular GLUT4 trafficking steps in adipocytes
.
J. Cell Biol.
180
,
375
387
199
Guo
,
H.L.
,
Zhang
,
C.
,
Liu
,
Q.
,
Li
,
Q.
,
Lian
,
G.
,
Wu
,
D.
et al (
2012
)
The Axin/TNKS complex interacts with KIF3A and is required for insulin-stimulated GLUT4 translocation
.
Cell Res.
22
,
1246
1257
200
Chi
,
N.W.
and
Lodish
,
H.F.
(
2000
)
Tankyrase is a Golgi-associated mitogen-activated protein kinase substrate that interacts with IRAP in GLUT4 vesicles
.
J. Biol. Chem.
275
,
38437
38444
201
Yeh
,
T.Y.
,
Sbodio
,
J.I.
,
Tsun
,
Z.Y.
,
Luo
,
B.
and
Chi
,
N.W.
(
2007
)
Insulin-stimulated exocytosis of GLUT4 is enhanced by IRAP and its partner tankyrase
.
Biochem. J.
402
,
279
290
202
Hosaka
,
T.
,
Brooks
,
C.C.
,
Presman
,
E.
,
Kim
,
S.K.
,
Zhang
,
Z.
,
Breen
,
M.
et al (
2005
)
P115 interacts with the GLUT4 vesicle protein, IRAP, and plays a critical role in insulin-stimulated GLUT4 translocation
.
Mol. Biol. Cell
16
,
2882
2890
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY). Open access for this article was enabled by the participation of University of Bath in an all-inclusive Read & Publish pilot with Portland Press and the Biochemical Society under a transformative agreement with JISC.

Supplementary data