The maintenance of energetic homeostasis in the face of limited available nutrients is a complex problem faced by all organisms. One important mechanism to maintain energetic homeostasis involves the activation of the energy sensor AMP-activated protein kinase (AMPK). AMPK is a cell-autonomous energy sensor that is highly sensitive to and regulated by the ATP to ADP and ATP to AMP ratios. However, the genetic analysis of AMPK signaling in vertebrates has been complicated by the existence of multiple redundant AMPK subunits. Here, we describe the identification of mutations in the single Drosophila melanogaster AMPK catalytic subunit (AMPKα) and their implications for neural maintenance and integrity. This article provides a citation replacement for previously published ampkα alleles, transgenes and neuronal phenotypes, which remain accurate; however, they were used in a previously published study that has subsequently been retracted (Mirouse et al., 2013).

AMP-activated protein kinase (AMPK) acts as a cellular energy sensor and is activated by ADP and AMP, which accumulate when ATP levels are low (Braco et al., 2012; Johnson et al., 2010; Kahn et al., 2005). AMPK then mediates the cellular response to energetic stress by activating energy-producing activities, while inhibiting energy-consuming ones, such as protein translation and cell growth/division. AMPK is a heterotrimeric protein with a 63-kDa catalytic α subunit and two regulatory β and γ subunits (38 and 36 kDa, respectively), each of which is encoded by distinct genes (α1, α2; β1, β2; γ1, γ2, γ3 in mammals; Davies et al., 1994; Gao et al., 1996; Mitchelhill et al., 1994; Nielsen et al., 2003; Stapleton et al., 1996). AMPK is also implicated in a number of signaling pathways (Hardie, 2004; Hardie, 2007; Shaw, 2009).

AMPK can be activated upstream by the tumor suppressor liver kinase B1 (LKB1; Amin et al., 2009; Shaw et al., 2004) and Ca2+/calmodulin-dependent kinase kinase β (Hawley et al., 2005; Hurley et al., 2005). AMPK signaling acts downstream to inhibit protein and lipid synthesis, for example, activating elongation factor-2 kinase, which causes inhibition of the elongation step of translation (Horman et al., 2002; Winder et al., 1997); inhibiting the target-of-rapamycin (TOR) pathway, which stimulates the initiation step of protein synthesis by the phosphorylation of multiple targets (Proud, 2004); and phosphorylating and inhibiting the fatty acid synthesis and rate-limiting enzyme ACC, which subsequently lowers malonyl-CoA levels and increases fatty acid uptake into mitochondria (Hardie and Hawley, 2001; Merrill et al., 1997; Winder et al., 1997).

We have further investigated the role of AMPK in vivo and show here that AMPK is required to maintain cellular integrity in neurons within Drosophila melanogaster.

AMPK contains three protein subunits, α, β, and γ, which form a heterotrimer. The α subunit (AMPKα) encodes for a highly conserved serine/threonine kinase, whereas the other subunits are regulatory. From a Drosophila melanogaster forward genetic screen for mutants affecting larval neuronal dendrite development and maintenance (Medina et al., 2006), we identified several lethal mutations in AMPKα. The ethyl methane sulfonate (EMS) mutants, ampkα1 and ampkα2, contain a single amino acid change (S211L, completely conserved) and a premature stop codon (Q295 STOP), respectively, whereas ampkα3 has a 16-bp deletion creating a stop codon (Y141 STOP; Fig. 1A). All ampkα mutants, whether homozygous or in trans with a deletion covering the locus, displayed a completely penetrant and nearly identical phenotype, with significantly enlarged plasma membrane domains in dendrites, but not in axonal compartments (Fig. 1C; unpublished data). In addition, ampkα1 and ampkα3 could be rescued to viability with either a chromosomal duplication carrying a wild-type ampkα gene, a wild-type AMPKα transgene, or a transgene that is tagged with the red fluorescent protein mCherry (Fig. 1D; see Materials and Methods). The requirement for ampkα is cell autonomous because transgene expression within only neurons rescues the phenotype (Fig. 1D). Therefore, these mutations represent genetic disruptions of the single AMPKα catalytic subunit in the D. melanogaster genome. (These data and these alleles were previously published as a part of a retracted study (Mirouse et al., 2013) but accurately allow the genetic analysis of AMPK function in vivo.)

Identification of mutations within the single Drosophila melanogaster ampkα gene.

Fig. 1.
Identification of mutations within the single Drosophila melanogaster ampkα gene.

(A) Schematic domain representation of AMPKα and corresponding genetic lesions in mutants. The serine/threonine kinase domain (black, aa 39–280) and T-Loop (gray, aa 167–194) are shown with the sites of mutations, S211L, Q295STOP, and Y141STOP, for ampkα1, ampkα2, and ampkα3, respectively. (B) Representative image of wild-type da neurons expressing an Actin::GFP fusion transgene in a second instar larva. (C) ampkα mutants display enlarged plasma membrane domains (arrows) in sensory neuron dendrites, but not axons. (D) A wild-type ampkα transgene expressed autonomously within da neurons completely rescues the dendrite phenotype. (B–D) Background genotypes are w; Gal4109(2)80, UAS-actin::GFP. Anterior toward the left and dorsal toward the top. Bars, 20 µm.

Fig. 1.
Identification of mutations within the single Drosophila melanogaster ampkα gene.

(A) Schematic domain representation of AMPKα and corresponding genetic lesions in mutants. The serine/threonine kinase domain (black, aa 39–280) and T-Loop (gray, aa 167–194) are shown with the sites of mutations, S211L, Q295STOP, and Y141STOP, for ampkα1, ampkα2, and ampkα3, respectively. (B) Representative image of wild-type da neurons expressing an Actin::GFP fusion transgene in a second instar larva. (C) ampkα mutants display enlarged plasma membrane domains (arrows) in sensory neuron dendrites, but not axons. (D) A wild-type ampkα transgene expressed autonomously within da neurons completely rescues the dendrite phenotype. (B–D) Background genotypes are w; Gal4109(2)80, UAS-actin::GFP. Anterior toward the left and dorsal toward the top. Bars, 20 µm.

The recovery of null mutations in ampkα has allowed for in vivo analysis of AMPK function in a multicellular organism, which has revealed an unexpected role for the kinase in the maintenance of cell integrity. This implies that at least one of the pathways that normally maintain cell integrity cannot function without AMPK activity. These results are in agreement with previous studies that have investigated the role of both AMPKβ and AMPKγ in neural maintenance and neurodegeneration in Drosophila, and found that both of these AMPK regulatory subunits are also essential to the maintenance of neural integrity (Andersen et al., 2012; Spasić et al., 2008; Tschäpe et al., 2002).

Mutant AMPKα allele isolation and characterization

An ethyl methane sulfonate mutagenesis screen on the X chromosome was performed as previously described (Medina et al., 2006). Early second instar larvae were visually screened for dendritic defects using fluorescent microscopy. The ampkα mutants, lethal at late second/third instar stages, were mapped to ∼150 kb on the X chromosome using a molecularly defined deficiency (Df[1]Exel6227), an undefined deficiency (Df[1]AD11), and a duplication of the Y chromosome (Dp[1;Y]/Df[1]svr). Predicted coding regions for genes in the region were sequenced using PCR amplicons made from mutant genomic DNA, and one gene (AMPKα; CG3051; NM_057965) was discovered that had significant mutations in all three alleles. Alleles have been contributed to the Bloomington Stock Center.

Construction of AMPKα transgenes

The wild-type AMPKα transgene was cloned into the pUAST vector (Brand and Perrimon, 1993) as an EcoRI–BglII fragment of an EST, corresponding to an AMPKα-RA transcript (http://flybase.org). The mCherry-AMPKα fusion protein was made using a mCherry construct (provided by R. Tsien, University of California, San Diego, San Diego, CA) at the N terminus fused in-frame to AMPKα into the pUAST vector. The UAS-mCherry-AMPKα transgene rescues viability and fertility when expressed by Ubiquitin-Gal4 in either ampkα1 or ampkα3 mutants. The phosphomimetic-activated form of AMPKα (AMPKα T184D) was made by PCR-based, site-directed mutagenesis converting base C549 to G549. The transgenes were introduced into a w1118 stock by P element-mediated transformation. Transgenes have been contributed to the Bloomington Stock Center.

Neuron visualization technique

Larvae were covered in a glycerol solution at 22°C and gently covered with a coverslip (22×50 mm; Fisher Scientific) to restrict movement, but not cause bursting of the body wall. Images were quickly taken using a Pan-Neofluar 40×/1.3 NA oil immersion lens with a 2-µm optical slice and LSM Imaging software (Carl Zeiss MicroImaging, Inc.). Images were resized and cropped with Photoshop (Adobe), and imported into Illustrator (Adobe) for labels and arrangement.

This research received no specific grant from any funding agency in the public, commercial or not-for-profit sectors.

Amin
N.
,
Khan
A.
,
St Johnston
D.
,
Tomlinson
I.
,
Martin
S.
,
Brenman
J.
,
McNeill
H.
(
2009
).
LKB1 regulates polarity remodeling and adherens junction formation in the Drosophila eye.
Proc. Natl. Acad. Sci. USA
106
,
8941
8946
.
Andersen
R. O.
,
Turnbull
D. W.
,
Johnson
E. A.
,
Doe
C. Q.
(
2012
).
Sgt1 acts via an LKB1/AMPK pathway to establish cortical polarity in larval neuroblasts.
Dev. Biol.
363
,
258
265
.
Braco
J. T.
,
Gillespie
E. L.
,
Alberto
G. E.
,
Brenman
J. E.
,
Johnson
E. C.
(
2012
).
Energy-dependent modulation of glucagon-like signaling in Drosophila via the AMP-activated protein kinase.
Genetics
192
,
457
466
.
Brand
A. H.
,
Perrimon
N.
(
1993
).
Targeted gene expression as a means of altering cell fates and generating dominant phenotypes.
Development
118
,
401
415
.
Davies
S. P.
,
Hawley
S. A.
,
Woods
A.
,
Carling
D.
,
Haystead
T. A.
,
Hardie
D. G.
(
1994
).
Purification of the AMP-activated protein kinase on ATP-gamma-sepharose and analysis of its subunit structure.
Eur. J. Biochem.
223
,
351
357
.
Gao
G.
,
Fernandez
C. S.
,
Stapleton
D.
,
Auster
A. S.
,
Widmer
J.
,
Dyck
J. R.
,
Kemp
B. E.
,
Witters
L. A.
(
1996
).
Non-catalytic beta- and gamma-subunit isoforms of the 5′-AMP-activated protein kinase.
J. Biol. Chem.
271
,
8675
8681
.
Hardie
D. G.
(
2004
).
The AMP-activated protein kinase pathway – new players upstream and downstream.
J. Cell Sci.
117
,
5479
5487
.
Hardie
D. G.
(
2007
).
AMP-activated/SNF1 protein kinases: conserved guardians of cellular energy.
Nat. Rev. Mol. Cell Biol.
8
,
774
785
.
Hardie
D. G.
,
Hawley
S. A.
(
2001
).
AMP-activated protein kinase: the energy charge hypothesis revisited.
Bioessays
23
,
1112
1119
.
Hawley
S. A.
,
Pan
D. A.
,
Mustard
K. J.
,
Ross
L.
,
Bain
J.
,
Edelman
A. M.
,
Frenguelli
B. G.
,
Hardie
D. G.
(
2005
).
Calmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinase.
Cell Metab.
2
,
9
19
.
Horman
S.
,
Browne
G.
,
Krause
U.
,
Patel
J.
,
Vertommen
D.
,
Bertrand
L.
,
Lavoinne
A.
,
Hue
L.
,
Proud
C.
,
Rider
M.
(
2002
).
Activation of AMP-activated protein kinase leads to the phosphorylation of elongation factor 2 and an inhibition of protein synthesis.
Curr. Biol.
12
,
1419
1423
.
Hurley
R. L.
,
Anderson
K. A.
,
Franzone
J. M.
,
Kemp
B. E.
,
Means
A. R.
,
Witters
L. A.
(
2005
).
The Ca2+/calmodulin-dependent protein kinase kinases are AMP-activated protein kinase kinases.
J. Biol. Chem.
280
,
29060
29066
.
Johnson
E. C.
,
Kazgan
N.
,
Bretz
C. A.
,
Forsberg
L. J.
,
Hector
C. E.
,
Worthen
R. J.
,
Onyenwoke
R.
,
Brenman
J. E.
(
2010
).
Altered metabolism and persistent starvation behaviors caused by reduced AMPK function in Drosophila.
PLoS ONE
5
,
e12799
.
Kahn
B. B.
,
Alquier
T.
,
Carling
D.
,
Hardie
D. G.
(
2005
).
AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism.
Cell Metab.
1
,
15
25
.
Medina
P. M.
,
Swick
L. L.
,
Andersen
R.
,
Blalock
Z.
,
Brenman
J. E.
(
2006
).
A novel forward genetic screen for identifying mutations affecting larval neuronal dendrite development in Drosophila melanogaster.
Genetics
172
,
2325
2335
.
Merrill
G. F.
,
Kurth
E. J.
,
Hardie
D. G.
,
Winder
W. W.
(
1997
).
AICA riboside increases AMP-activated protein kinase, fatty acid oxidation, and glucose uptake in rat muscle.
Am. J. Physiol.
273
,
E1107
E1112
.
Mirouse
V.
,
Swick
L. L.
,
Kazgan
N.
,
St Johnston
D.
,
Brenman
J. E.
(
2013
).
LKB1 and AMPK maintain epithelial cell polarity under energetic stress.
J. Cell Biol.
[Retraction] doi: 10.1083/jcb.20070205310112013r
Mitchelhill
K. I.
,
Stapleton
D.
,
Gao
G.
,
House
C.
,
Michell
B.
,
Katsis
F.
,
Witters
L. A.
,
Kemp
B. E.
(
1994
).
Mammalian AMP-activated protein kinase shares structural and functional homology with the catalytic domain of yeast Snf1 protein kinase.
J. Biol. Chem.
269
,
2361
2364
.
Nielsen
J. N.
,
Mustard
K. J.
,
Graham
D. A.
,
Yu
H.
,
MacDonald
C. S.
,
Pilegaard
H.
,
Goodyear
L. J.
,
Hardie
D. G.
,
Richter
E. A.
,
Wojtaszewski
J. F.
(
2003
).
5′-AMP-activated protein kinase activity and subunit expression in exercise-trained human skeletal muscle.
J. Appl. Physiol.
94
,
631
641
.
Proud
C. G.
(
2004
).
Role of mTOR signalling in the control of translation initiation and elongation by nutrients.
Curr. Top. Microbiol. Immunol.
279
,
215
244
.
Shaw
R. J.
(
2009
).
LKB1 and AMP-activated protein kinase control of mTOR signalling and growth.
Acta Physiol. (Oxf.)
196
,
65
80
.
Shaw
R. J.
,
Kosmatka
M.
,
Bardeesy
N.
,
Hurley
R. L.
,
Witters
L. A.
,
DePinho
R. A.
,
Cantley
L. C.
(
2004
).
The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress.
Proc. Natl. Acad. Sci. USA
101
,
3329
3335
.
Spasić
M. R.
,
Callaerts
P.
,
Norga
K. K.
(
2008
).
Drosophila alicorn is a neuronal maintenance factor protecting against activity-induced retinal degeneration.
J. Neurosci.
28
,
6419
6429
.
Stapleton
D.
,
Mitchelhill
K. I.
,
Gao
G.
,
Widmer
J.
,
Michell
B. J.
,
Teh
T.
,
House
C. M.
,
Fernandez
C. S.
,
Cox
T.
,
Witters
L. A.
et al.  (
1996
).
Mammalian AMP-activated protein kinase subfamily.
J. Biol. Chem.
271
,
611
614
.
Tschäpe
J. A.
,
Hammerschmied
C.
,
Mühlig-Versen
M.
,
Athenstaedt
K.
,
Daum
G.
,
Kretzschmar
D.
(
2002
).
The neurodegeneration mutant löchrig interferes with cholesterol homeostasis and Appl processing.
EMBO J.
21
,
6367
6376
.
Winder
W. W.
,
Wilson
H. A.
,
Hardie
D. G.
,
Rasmussen
B. B.
,
Hutber
C. A.
,
Call
G. B.
,
Clayton
R. D.
,
Conley
L. M.
,
Yoon
S.
,
Zhou
B.
(
1997
).
Phosphorylation of rat muscle acetyl-CoA carboxylase by AMP-activated protein kinase and protein kinase A.
J. Appl. Physiol.
82
,
219
225
.

Competing interests

The authors have no competing interests to declare.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.