ArticlePDF AvailableLiterature Review

Current Perspectives on Akt Akt-ivation and Akt-ions

Authors:

Abstract and Figures

The serine/threonine kinase Akt is an effector of PI3K-generated phosphatidylinositol (3,4,5)-trisphosphate [PI(3,4,5)P3] and is a principle mediator of growth factor-induced signal transduction. Akt is activated through phosphorylation by specific kinases, and its activity is reduced directly by phosphorylation-site-specific phosphatases. In addition, Akt activity is effectively reduced by the action of phosphatases which dephosphorylate PI(3,4,5)P3, thereby reducing the levels of the essential lipid activators of PDK1 and Akt. The functions of Akt are pleiotropic and include regulation of cellular proliferation, differentiation, protein synthesis, and survival. Akt stimulates protein synthesis through actions on mTOR/p70S6K, and promotes survival by phosphorylating and inactivating pro-apoptotic molecules such as Ask1, Bad, Bax, and FoxO3a. Furthermore, loss of Akt decreases the intracellular ATP:AMP ratio, thus establishing a role for Akt in energy regulation. Three isoforms of Akt have been identified, and although redundant functions between isoforms exist, recent investigations have enumerated unique functions for each. Therefore, targeting specific Akt isozymes in a tissue- and context-specific fashion may lead to a greater understanding of Akt-mediated processes.
Content may be subject to copyright.
EBM
Experimental Biology and Medicine
Ronald W. Matheny, Jr. and Martin L. Adamo
Current Perspectives on Akt Akt-ivation and Akt-ions
doi: 10.3181/0904-MR-138
2009, 234:1264-1270.Experimental Biology and Medicine
http://ebm.rsmjournals.com/content/234/11/1264
Updated information and services can be found at:
http://ebm.rsmjournals.com/content/234/11/1264#BIBL
This article cites 78 articles, 40 of which can be accessed free at:
http://ebm.rsmjournals.com/content/234/11/1264#otherarticles
5 online articles that cite this articles can be accessed at:
© 2008 Society for Experimental Biology and Medicine
by guest on May 29, 2013http://ebm.rsmjournals.com/Downloaded from
MINIREVIEW
Current Perspectives on Akt Akt-ivation
and Akt-ions
RONALD W. MATHENY JR.AND MARTIN L. ADAMO
1
Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio,
Texas 78229; and The Sam and Ann Barshop Institute for Longevity and Aging Studies, University of
Texas Health Science Center at San Antonio, San Antonio, Texas 78229
The serine/threonine kinase Akt is an effector of PI3K-generated
phosphatidylinositol (3,4,5)-trisphosphate [PI(3,4,5)P3] and is a
principle mediator of growth factor-induced signal transduction.
Akt is activated through phosphorylation by specific kinases,
and its activity is reduced directly by phosphorylation-site-
specific phosphatases. In addition, Akt activity is effectively
reduced by the action of phosphatases which dephosphorylate
PI(3,4,5)P3, thereby reducing the levels of the essential lipid
activators of PDK1 and Akt. The functions of Akt are pleiotropic
and include regulation of cellular proliferation, differentiation,
protein synthesis, and survival. Akt stimulates protein synthesis
through actions on mTOR/p70S6K, and promotes survival by
phosphorylating and inactivating pro-apoptotic molecules such
as Ask1, Bad, Bax, and FoxO3a. Furthermore, loss of Akt
decreases the intracellular ATP:AMP ratio, thus establishing a
role for Akt in energy regulation. Three isoforms of Akt have
been identified, and although redundant functions between
isoforms exist, recent investigations have enumerated unique
functions for each. Therefore, targeting specific Akt isozymes in
a tissue- and context-specific fashion may lead to a greater
understanding of Akt-mediated processes. Exp Biol Med
234:1264–1270, 2009
Key words: Akt; growth; apoptosis
Introduction
Akt (also known as protein kinase B) is a serine/
threonine kinase downstream of phosphoinositide-39-OH
kinase (PI3K) and a member of the AGC family of protein
kinases. Since first discovered in 1977 as the oncogene in
the transforming retrovirus AKT8 (1), Akt has become the
subject of intense research aimed at defining its involvement
in cancer progression, metabolism, cellular growth and
differentiation, and survival. Much has been learned about
the regulation and actions of Akt since its cloning and
identification of human homologues (2), yet much still
remains to be elucidated. For example, there are three
isoforms of Akt in mammals, and the functions of each have
been shown to be redundant as well as unique depending on
the tissue examined, stimulus, outcome (e.g. differentiation)
or the conditions within the cellular milieu. Additionally,
our knowledge of Akt regulation in terms of both activation
and signal suppression is burgeoning, but further work must
be accomplished to apply these findings in practice in
humans; for instance, to attenuate cancer progression and
metastasis, or to delay onset or minimize age-related
sarcopenia. This review discusses factors involved in
regulating Akt activation, select substrates of Akt involved
in survival and growth, and isoform-specific functions of
Akt.
Activation of Akt
Members of the PI3K class of enzymes generate
phosphoinositol lipids that act as second-messengers in a
number of intracellular signaling cascades, including the
activation of Akt (3). PI3K catalytic enzymes are catego-
rized into three classes by their structure, substrate
specificity, and lipid products (4). Members of the Class
IA PI3Ks (a,b, and d) are heterodimers consisting of a 110
This work was supported by NIA grant R01AG026012 to MLA. RWM was supported
by pre-doctoral award from NIA training grant T32 AG021890-08.
1
To whom correspondence should be addressed at Department of Biochemistry MSC
7760, UT Health Science Center at San Antonio, 7703 Floyd Curl Drive, San
Antonio, TX 78229-3900. E-mail: adamo@biochem.uthscsa.edu
1264
DOI: 10.3181/0904-MR-138
1535-3702/09/23411-1264$15.00
Copyright Ó2009 by the Society for Experimental Biology and Medicine
by guest on May 29, 2013http://ebm.rsmjournals.com/Downloaded from
kDa catalytic subunit and an 85, 55, or 50 kDa regulatory
subunit (4, 5). PI3K activation leads to proliferation,
survival, cell migration, and growth; however, abnormally
increased PI3K signaling can promote aberrant proliferative
signals that lead to cellular transformation (6–8). When
bound to cognate ligands, receptor tyrosine kinases such as
the insulin-like growth factor-I receptor undergo autophos-
phorylation, resulting in the recruitment of adaptor mole-
cules including insulin receptor substrate (IRS) proteins (9,
10). PI3K regulatory subunits complexed to p110 catalytic
subunits then bind IRS and convert plasma membrane-
associated PI(4,5)P2 to PI(3,4,5)P3 (5). PI(3,4,5)P3 pro-
vides a phospholipid binding substrate for signaling effector
molecules such as phosphatidylinositol-dependent kinase 1
(PDK1) and Akt (11). At least two identified phosphatases
act to reduce PI(3,4,5)P3 levels: phosphatase and tensin
homologue deleted on chromosome 10 (PTEN) (12) and
SH2-containing inositol 59-phosphatase (SHIP) (13). PTEN
hydrolyzes the phosphate at the D3 position on the inositol
ring of PI(3,4,5)P3, thus generating PI(4,5)P2. SHIP
proteins remove phosphates from the D5 position, thus
generating PI(3,4)P2.
Akt exists as three isoforms in mammals (Akt1, Akt2,
and Akt3), and each is transcribed from separate genes (14–
17); additionally, a splice variant of Akt3 has been identified
(18). Akt contains an N-terminal pleckstrin homology (PH)
domain which interacts with PI(3,4)P2 and PI(3,4,5)P3 (19,
20), and a C-terminal domain that contains a hydrophobic
domain (HD) with homology to other AGC kinases (21).
Akt is activated by phosphorylation at two sites including
one in the activation loop (A-loop) (T308, T309, T305 in
Akt1, Akt2, and Akt3, respectively) in the catalytic domain,
and one in the C-terminal HD (S473, S474, S472 in Akt1,
Akt2, and Akt3, respectively) (21). The C-terminus of the
Akt3 splice variant, however, lacks the analogous portion of
the HD that contains S473, S474, or S472 present in the
other Akt isoforms (Fig. 1). Phosphorylation at the turn
motif (TM) at a highly conserved threonine residue in the
carboxy-terminus results in increased stability of the
molecule (22).
Phosphorylation at the A-loop is sufficient for activa-
tion of Akt, however full activation is achieved when both
the A-loop and HD sites are phosphorylated (21). Akt can
be activated through growth factor stimulation (21) and
oxidative stress (23, 24). Phosphorylation of the A-loop in
the catalytic domain is accomplished by PDK1 after
recruitment to the cell membrane and binding with
PI(3,4,5)P3 (11, 19, 25). HD phosphorylation has been
shown to be mediated, under certain conditions, through
actions of a number of factors including mammalian target
of rapamycin complex 2 (mTORC2) (26), DNA-PK (27),
lipid raft-associated elements (28), and autophosphorylation
of Akt itself (29). However, growth factor-stimulated
phosphorylation of Akt at the HD is considered to be
mediated principally by mTORC2.
Direct de-phosphorylation of Akt remained an enigma
until the discovery of PH domain leucine-rich repeat protein
phosphatase 1 (abbreviated as PHLPP1) (30), and more
recently, PHLPP2 (31). Both PHLPP1 and PHLPP2 were
found to de-phosphorylate the HD site of Akt, but they did
so in an Akt isoform-specific manner.
The mechanisms outlined above describe a general
scheme for full activation for Akt in response to growth
factor stimulation: generation of PI(3,4,5)P3 or PI(3,4)P2 by
PI3K results in membrane recruitment of Akt and co-
localization of PDK1. PDK1 phosphorylates Akt at the A-
loop which results in conformational change of Akt
allowing for phosphorylation of the HD by mTORC2. Akt
then acts on downstream targets to initiate signaling
cascades. Figure 2 presents these processes diagrammati-
cally.
Downstream of Akt: Survival, Growth,
and Energy Homeostasis
A number of known and putative Akt targets have been
identified thus far by virtue of their containing the essential
Akt consensus motif (R-X-R-X-X-S/T-B) where X is any
amino acid and B represents a bulky hydrophobic residue
(32, 33). Among these targets are several pro-apoptotic
molecules that are inactivated when phosphorylated by Akt,
Figure 1. Comparison of human Akt isoform domain structures.
Three isoforms of Akt exist in man that share approximately 80%
amino acid sequence homology. Additionally, an alternative splice
variant of Akt3 with a truncated hydrophobic domain (designated
‘‘Akt3-(c1)’’ in this figure) has been identified (see text). All Akt
isoforms possess an N-terminal PH domain responsible for
phospholipid binding that is tethered to a catalytic region containing
a threonine residue (T308 in Akt1) critical for activation of the
enzyme. A C-terminal hydrophobic domain (designated ‘‘HD’’ in this
figure) follows the catalytic domain and contains a serine residue
(S473 in Akt1) important for full activation. Phosphorylation of a
threonine residue in the turn motif (T450 in Akt1) by mTORC2
contributes to stability of the molecule. Numbers to the immediate left
of the images designate the first amino acid, and numbers to the
immediate right of the images represent the number of the most C-
terminal residue as determined by comparative sequence analysis. A
color version of this figure is available in the online journal.
AKT ACTIVATION AND ACTIONS 1265
by guest on May 29, 2013http://ebm.rsmjournals.com/Downloaded from
thus demonstrating that Akt can act as a survival factor.
There are many pro-apoptotic substrates of Akt including
apoptosis-signal regulating kinase 1 (Ask1) (34), Bad (35),
Bax (36), FoxO transcription factors (37–39), and human
caspase-9 (40). Ask1 is a mitogen-activated protein (MAP)
kinase kinase kinase that transmits pro-apoptotic signals
through activation of downstream stress-activated protein
kinases such as c-jun N-terminal kinase (JNK) and p38
MAPK (41). Phosphorylation of 14-3-3 by activated JNK
promotes release of 14-3-3-bound Bad, Bax, and FoxO
proteins, thus contributing to apoptosis through both
mitochondrial-dependent (Bad and Bax) and -independent
(FoxO3a) (42, 43). Each of these mechanisms will be
explained in the following sections.
Phosphorylation of Ask1 at serine 83 by Akt has been
shown to reduce Ask1 activation stimulated by oxidative
stress, as well as apoptosis induced by Ask1 overexpression
(34), thus providing evidence for a specific pro-survival role
of Akt acting on Ask1. Interaction of Bad with outer
mitochondrial membrane (OMM)-associated Bcl-2 and Bcl-
XL leads to disruption of OMM integrity and cytochrome c
release; hence, binding of Bad with Bcl-2 or Bcl-XL
promotes apoptosis through the mitochondrial intrinsic
death pathway. Phosphorylation of Bad at serine 136 by
Akt leads to dissociation from Bcl-2/Bcl-XL and promotes
association with cytosolic 14-3-3, thereby reducing pro-
death signals (35). Another protein that disrupts OMM
membrane integrity is Bax. Bax exists in the cytosol in a
conformation permissive for actions by JNK and p38
MAPK (44); once acted upon by apoptotic stimuli, Bax
undergoes a conformational change that permits insertion
into mitochondrial membranes and oligomerization. How-
ever, prior phosphorylation of cytosolic Bax at serine 184
by Akt prevents this conformational change and reduces
half-life, thereby impairing Bax-mediated apoptosis (36,
45).
Members of the FoxO family of transcription factors
are structurally related proteins whose name is derived from
the product of the fork head gene originally identified in D.
melanogaster (46). FoxO transcription factors belong to a
superfamily of Fox (Forkhead Box) proteins in a class
defined as O (Other) which reflects disparities in DNA-
binding domain sequences (47, 48). Among the FoxO
proteins, FoxO3a has been shown to possess pro-apoptotic
functions including regulation of FasL gene expression (49);
however, FoxO3a-mediated apoptosis can be attenuated by
Akt (39). FoxO3a is phosphorylated at three sites by Akt
(T32, S253, S315), although S315 is preferentially phos-
phorylated by serum- and glucocorticoid-inducible kinase
(SGK) (50). When phosphorylated by Akt at target residues,
nuclear FoxO3a associates with nuclear 14-3-3; this
association masks the nuclear localization sequence, and
the complex is exported to the cytosol in an inactive state
(51).
In addition to the target molecules described above, Akt
can phosphorylate a number of other pro-death molecules to
cause their inactivation, as well as oppose apoptosis
indirectly by acting on proteins involved in transcription
of pro-apoptotic genes (52). Altogether, it is clear that Akt
plays a vital role in cell survival and is positioned at a point
of convergence of a number of signaling pathways,
balancing both pro-survival and pro-death signals.
The mammalian target of rapamycin (mTOR) responds
to nutrient or growth factor inputs, depending on its
association with one of two adaptor molecules, either
Raptor or Rictor (53). One principle effector of mTOR
action is p70S6K, an AGC kinase family member whose
actions promote growth and survival (54, 55). Raptor-bound
mTOR, when also bound in complex with GbL (referred to
as mTOR complex 1, or mTORC1) is generally considered
the rapamycin-sensitive p70S6K kinase (56), but Rictor-
bound mTOR (the complex, in association with GbL and
SAPK interacting protein 1 (SIN1) (57), being referred to as
mTORC2) has been shown to phosphorylate rapamycin-
resistant mutants of p70S6K (58). Current models suggest
that Akt positively regulates mTOR by acting on mTOR-
inhibitory molecules such as proline-rich Akt substrate of 40
Figure 2. Regulation of Akt activation. Tyrosine kinase growth factor
receptor-induced activation of Akt is initiated upon growth factor (GF)
ligand binding to its cognate receptor. Recruitment of PI3K (p85
regulatory subunit complexed with p110 catalytic subunit) to the
receptor at the membrane occurs after binding of adaptor molecules,
including IRS docking proteins, to the intracellular subunits of
receptors. PI3K, in close proximity to PI(4,5)P2 (PIP
2
)then
phosphorylates the D3 position of the inositol ring generating
PI(3,4,5)P3 (PIP
3
). PIP
3
then recruits PH-domain-containing mole-
cules including PDK1 and Akt to the lipid bilayer (Akt recruitment not
shown for clarity), allowing for binding to PIP
3
, and phosphorylation of
Akt at the A-loop (T308 in Akt1) by PDK1. Reconversion of PIP
3
to
PIP
2
is promoted by PTEN, which reduces available PIP
3
for PDK1
and Akt binding, thus terminating growth factor signals. Phosphor-
ylation of Akt at the HD (S473 in Akt1) occurs through the actions of
the mTORC2 complex, which contains Rictor, SIN1, and mTOR.
Activated Akt can then act on substrates in the cytosol to promote
survival, growth and energy homeostasis, or translocate to the
nucleus and phosphorylate target molecules such as FoxO3a. A
color version of this figure is available in the online journal.
1266 MATHENY AND ADAMO
by guest on May 29, 2013http://ebm.rsmjournals.com/Downloaded from
kDa (PRAS40) and tuberous sclerosis complex 2 (TSC2)
(53, 59–61). mTORC2 is capable of phosphorylating Akt at
the HD site (26, 57), as well as controlling folding and
stability of the enzyme (22, 62); thus, there exists positive
feedback signaling between mTOR and Akt.
AMP-activated protein kinase (AMPK) acts as an
intracellular energy sensor (63) that is sensitive to Akt
signaling. In a recent study, decreased ATP levels were
observed in Akt1
/-
Akt2
/-
mouse embryonic fibroblasts
(MEFs) as compared to wild-type cells (64), suggesting that
depletion of Akt affects energy charge. This reduced energy
charge was associated with increased AMPK. Since AMPK
can inactivate mTOR through direct phosphorylation of
TSC2 (59), these findings suggest the existence of a
mechanism to reduce growth and translation during low-
energy states secondary to Akt depletion.
Isoform-Specific Actions of Akt
Three Akt isoforms have been identified, and murine
gene disruption models demonstrate that each isoform
possesses a distinct function (65). Mice lacking Akt1 have
reduced body size (66), mice lacking Akt2 show abnormal
glucose homeostasis and a diabetic phenotype (67, 68), and
mice lacking Akt3 have diminished brain size (69, 70). In
mammary tumor cells, Akt1 or Akt2 knockdown had
opposing effects on differentiation (71), while in melanoma
cells, Akt3 was found to be the principle Akt isoform
involved in tumor progression (72). Furthermore, in differ-
entiating skeletal muscle cells, Akt2 mRNA, protein, and
activity were found to be upregulated, and overexpression
of Akt2 prevented apoptosis in response to serum
deprivation, suggesting that Akt2 acts to reduce apoptosis
during myogenic differentiation (73). Although these data
demonstrate discrete functions for Akt isoforms, there is
also accumulating evidence for overlapping actions. For
example, each Akt isoform contributed to phosphorylation
of GSK3aand GSK3bin 3T3-L1 adipocytes (74). Addi-
tionally, knockdown of individual Akt isoforms in H157
cells demonstrated distinct roles for Akt3 acting on p27 and
Akt2 acting on GSK3a, but redundant roles for all three Akt
isoforms in regulating FoxO1, GSk3b, and TSC2 (31).
Moreover, in Akt1/Akt2 double knockout MEFs (DKO
MEFs), Akt3 alone was sufficient to prevent cell death until
reduced to only 20% of the initial level (75). Reduction of
Akt3 through increased concentrations of siRNA, however,
potentiated apoptosis in response to several pro-apoptotic
stimuli in DKO MEFs. This latter study underscored that
very limited amounts of total Akt, even of a single isoform,
can promote survival under non-stressed conditions; but that
under stress, cells require a higher threshold of Akt to
remain viable. Together, these studies illustrate the
complexity of Akt regulation and specificity, and demon-
strate that Akt isoforms possess both distinct and function-
ally redundant actions in growth, metabolism, and cell
survival.
Conclusions and Future Outlook
Much has been learned about Akt regulation and
function since it was first discovered, yet much still remains
to be elucidated. For instance, what elements control
intracellular localization of Akt? Akt activation depends
on proximity to PI(3,4)P2 and PI(3,4,5)P3 in cell mem-
branes and it is important to define those factors within the
cell that are responsible for membrane recruitment.
Interaction of the PH domain with phosphoinositide lipids
is essential for activation, but identification of the processes
and molecules involved in executing this specifically
targeted relocation is of importance. It then follows that
the location of Akt in unstimulated cells is of interest in
order to define movement patterns of Akt after stimuli.
Defining intracellular localization also extends to
isoform-specific locations within the cell, as well as
isoform-specific activation in response to stimuli. What
are the determining factors that contribute to activation of
Akt isoforms in response to growth factors? Why is one
isoform preferentially activated over another? One answer
may be that Akt isoforms are expressed at different levels in
a given cell-type. If availability of one isoform is greater
than another, then it may be a matter of proximity to
membrane-located signaling complexes. For example, if
Akt1 is the most highly expressed Akt isoform in a cell, then
it would logically follow that there would be more Akt1
available for recruitment to membranes than Akt2 or Akt3.
It is also possible that inactive Akt may be localized to
specific intracellular compartments or areas within the cell,
possibly near the membrane, that contain increased densities
of a particular isoform.
In addition to activation of Akt, it is important to further
characterize existing known phosphatases as well as to
discover new methods to de-activate the enzyme. The
identification of isoform-specific de-phosphorylation of Akt
by PHLPP1 and PHLPP2 was a groundbreaking discovery,
and further research in defining the mechanisms of action
and regulation of these two phosphatases is warranted.
An exciting and promising line of research involves the
development of Akt isoform-specific small molecule
inhibitors. Some inhibitors exert their effects by preventing
Akt from being activated due to changes in ternary structure.
For instance, an Akt-specific inhibitor commonly referred to
as ‘‘Akti-1/2’’ has recently been developed that can bind to
the PH domain and/or hinge region on Akt1 or Akt2 thereby
preventing activation (76). This inhibitor can also bind to
activated Akt and inhibit phosphorylation of substrates.
However, Akti-1/2 can also inhibit Akt3 in 3T3-L1
adipocytes and L6 myotubes (77), as well as in C2C12
myoblasts (Matheny and Adamo, unpublished observations)
at micromolar concentrations. Although Akti-1/2 is not
specific for Akt isoforms in some cell lines at specific
concentrations, this compound still possesses great potential
under conditions where all three Akt isoforms can act in a
redundant manner. In any case, intense research in the
AKT ACTIVATION AND ACTIONS 1267
by guest on May 29, 2013http://ebm.rsmjournals.com/Downloaded from
development of Akt isoform-specific inhibitory compounds
is ongoing (78).
In conclusion, Akt is at the crossroads of a number of
intracellular pathways and is a key signaling intermediate in
growth and survival. Much has been learned thus far with
respect to regulation and signaling of Akt, yet much remains
to be discovered; specifically, the localization of Akt
isoforms in response to various stimuli in different tissues,
as well as isoform-specific actions of Akt on target
substrates.
1. Staal SP, Hartley JW, Rowe WP. Isolation of transforming murine
leukemia viruses from mice with a high incidence of spontaneous
lymphoma. Proc Natl Acad Sci U S A 74:3065–3067, 1977.
2. Staal SP. Molecular cloning of the akt oncogene and its human
homologues AKT1 and AKT2: amplification of AKT1 in a primary
human gastric adenocarcinoma. Proc Natl Acad Sci U S A 84:5034–
5037, 1987.
3. Vanhaesebroeck B, Waterfield MD. Signaling by distinct classes of
phosphoinositide 3-kinases. Exp Cell Res 253:239–254, 1999.
4. Hirsch E, Costa C, Ciraolo E. Phosphoinositide 3-kinases as a common
platform for multi-hormone signaling. J Endocrinol 194:243–256,
2007.
5. Geering B, Cutillas PR, Nock G, Gharbi SI, Vanhaesebroeck B. Class
IA phosphoinositide 3-kinases are obligate p85-p110 heterodimers.
Proc Natl Acad Sci U S A 104:7809–7814, 2007.
6. Denley A, Kang S, Karst U, Vogt PK. Oncogenic signaling of class I
PI3K isoforms. Oncogene 27:2561–2574, 2008.
7. Kang S, Denley A, Vanhaesebroeck B, Vogt PK. Oncogenic
transformation induced by the p110beta, -gamma, and -delta isoforms
of class I phosphoinositide 3-kinase. Proc Natl Acad Sci U S A 103:
1289–1294, 2006.
8. Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H,
Gazdar A, Powell SM, Riggins GJ, Willson JK, Markowitz S, Kinzler
KW, Vogelstein B, Velculescu VE. High frequency of mutations of the
PIK3CA gene in human cancers. Science 304:554, 2004.
9. Jones JI, Clemmons DR. Insulin-like growth factors and their binding
proteins: biological actions. Endocr Rev 16:3–34, 1995.
10. Dey BR, Frick K, Lopaczynski W, Nissley SP, Furlanetto RW.
Evidence for the direct interaction of the insulin-like growth factor I
receptor with IRS-1, Shc, and Grb10. Mol Endocrinol 10:631–641,
1996.
11. Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PR, Reese
CB, Cohen P. Characterization of a 3-phosphoinositide-dependent
protein kinase which phosphorylates and activates protein kinase
Balpha. Curr Biol 7:261–269, 1997.
12. Cully M, You H, Levine AJ, Mak TW. Beyond PTEN mutations: the
PI3K pathway as an integrator of multiple inputs during tumorigenesis.
Nat Rev Cancer 6:184–192, 2006.
13. Kalesnikoff J, Sly LM, Hughes MR, Buchse T, Rauh MJ, Cao LP, Lam
V, Mui A, Huber M, Krystal G. The role of SHIP in cytokine-induced
signaling. Rev Physiol Biochem Pharmacol 149:87–103, 2003.
14. Jones PF, Jakubowicz T, Pitossi FJ, Maurer F, Hemmings BA.
Molecular cloning and identification of a serine/threonine protein
kinase of the second-messenger subfamily. Proc Natl Acad Sci U S A
88:4171–4175, 1991.
15. Jones PF, Jakubowicz T, Hemmings BA. Molecular cloning of a
second form of rac protein kinase. Cell Regul 2:1001–1009, 1991.
16. Konishi H, Kuroda S, Tanaka M, Matsuzaki H, Ono Y, Kameyama K,
Haga T, Kikkawa U. Molecular cloning and characterization of a new
member of the RAC protein kinase family: association of the pleckstrin
homology domain of three types of RAC protein kinase with protein
kinase C subspecies and beta gamma subunits of G proteins. Biochem
Biophys Res Commun 216:526–534, 1995.
17. Brodbeck D, Cron P, Hemmings BA. A human protein kinase Bgamma
with regulatory phosphorylation sites in the activation loop and in the
C-terminal hydrophobic domain. J Biol Chem 274:9133–9136, 1999.
18. Brodbeck D, Hill MM, Hemmings BA. Two splice variants of protein
kinase B gamma have different regulatory capacity depending on the
presence or absence of the regulatory phosphorylation site serine 472 in
the carboxyl-terminal hydrophobic domain. J Biol Chem 276:29550–
29558, 2001.
19. Andjelkovic M, Alessi DR, Meier R, Fernandez A, Lamb NJ, Frech M,
Cron P, Cohen P, Lucocq JM, Hemmings BA. Role of translocation in
the activation and function of protein kinase B. J Biol Chem 272:
31515–31524, 1997.
20. Franke TF, Kaplan DR, Cantley LC, Toker A. Direct regulation of the
Akt proto-oncogene product by phosphatidylinositol-3,4-bisphosphate.
Science 275:665–668, 1997.
21. Alessi DR, Andjelkovic M, Caudwell B, Cron P, Morrice N, Cohen P,
Hemmings BA. Mechanism of activation of protein kinase B by insulin
and IGF-1. EMBO J 15:6541–6551, 1996.
22. Facchinetti V, Ouyang W, Wei H, Soto N, Lazorchak A, Gould C,
Lowry C, Newton AC, Mao Y, Miao RQ, Sessa WC, Qin J, Zhang P,
Su B, Jacinto E. The mammalian target of rapamycin complex 2
controls folding and stability of Akt and protein kinase C. EMBO J 27:
1932–1943, 2008.
23. Van der Kaay J, Beck M, Gray A, Downes CP. Distinct phosphati-
dylinositol 3-kinase lipid products accumulate upon oxidative and
osmotic stress and lead to different cellular responses. J Biol Chem 274:
35963–35968, 1999.
24. Wang X, McCullough KD, Franke TF, Holbrook NJ. Epidermal growth
factor receptor-dependent Akt activation by oxidative stress enhances
cell survival. J Biol Chem 275:14624–14631, 2000.
25. Stokoe D, Stephens LR, Copeland T, Gaffney PR, Reese CB, Painter
GF, Holmes AB, McCormick F, Hawkins PT. Dual role of
phosphatidylinositol-3,4,5-trisphosphate in the activation of protein
kinase B. Science 277:567–570, 1997.
26. Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation
and regulation of Akt/PKB by the rictor-mTOR complex. Science 307:
1098–1101, 2005.
27. Bozulic L, Surucu B, Hynx D, Hemmings BA. PKBalpha/Akt1 acts
downstream of DNA-PK in the DNA double-strand break response and
promotes survival. Mol Cell 30:203–213, 2008.
28. Hill MM, Feng J, Hemmings BA. Identification of a plasma membrane
Raft-associated PKB Ser473 kinase activity that is distinct from ILK
and PDK1. Curr Biol 12:1251–1255, 2002.
29. Toker A, Newton AC. Akt/protein kinase B is regulated by
autophosphorylation at the hypothetical PDK-2 site. J Biol Chem
275:8271–8274, 2000.
30. Gao T, Furnari F, Newton AC. PHLPP: a phosphatase that directly
dephosphorylates Akt, promotes apoptosis, and suppresses tumor
growth. Mol Cell 18:13–24, 2005.
31. Brognard J, Sierecki E, Gao T, Newton AC. PHLPP and a second
isoform, PHLPP2, differentially attenuate the amplitude of Akt
signaling by regulating distinct Akt isoforms. Mol Cell 25:917–931,
2007.
32. Alessi DR, Caudwell FB, Andjelkovic M, Hemmings BA, Cohen P.
Molecular basis for the substrate specificity of protein kinase B;
comparison with MAPKAP kinase-1 and p70 S6 kinase. FEBS Lett
399:333–338, 1996.
33. Manning BD, Cantley LC. AKT/PKB signaling: navigating down-
stream. Cell 129:1261–1274, 2007.
34. Kim AH, Khursigara G, Sun X, Franke TF, Chao MV. Akt
phosphorylates and negatively regulates apoptosis signal-regulating
kinase 1. Mol Cell Biol 21:893–901, 2001.
35. Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, Greenberg ME.
1268 MATHENY AND ADAMO
by guest on May 29, 2013http://ebm.rsmjournals.com/Downloaded from
Akt phosphorylation of BAD couples survival signals to the cell-
intrinsic death machinery. Cell 91:231–241, 1997.
36. Gardai SJ, Hildeman DA, Frankel SK, Whitlock BB, Frasch SC,
Borregaard N, Marrack P, Bratton DL, Henson PM. Phosphorylation of
Bax Ser184 by Akt regulates its activity and apoptosis in neutrophils. J
Biol Chem 279:21085–21095, 2004.
37. Zheng WH, Kar S, Quirion R. Insulin-like growth factor-1-induced
phosphorylation of the forkhead family transcription factor FKHRL1 is
mediated by Akt kinase in PC12 cells. J Biol Chem 275:39152–39158,
2000.
38. Kops GJ, de Ruiter ND, De Vries-Smits AM, Powell DR, Bos JL,
Burgering BM. Direct control of the Forkhead transcription factor AFX
by protein kinase B. Nature 398:630–634, 1999.
39. Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, Anderson
MJ, Arden KC, Blenis J, Greenberg ME. Akt promotes cell survival by
phosphorylating and inhibiting a Forkhead transcription factor. Cell 96:
857–868, 1999.
40. Cardone MH, Roy N, Stennicke HR, Salvesen GS, Franke TF,
Stanbridge E, Frisch S, Reed JC. Regulation of cell death protease
caspase-9 by phosphorylation. Science 282:1318–1321, 1998.
41. Ichijo H, Nishida E, Irie K, ten Dijke P, Saitoh M, Moriguchi T, Takagi
M, Matsumoto K, Miyazono K, Gotoh Y. Induction of apoptosis by
ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38
signaling pathways. Science 275:90–94, 1997.
42. Tsuruta F, Sunayama J, Mori Y, Hattori S, Shimizu S, Tsujimoto Y,
Yoshioka K, Masuyama N, Gotoh Y. JNK promotes Bax translocation
to mitochondria through phosphorylation of 14-3-3 proteins. EMBO J
23:1889–1899, 2004.
43. Sunayama J, Tsuruta F, Masuyama N, Gotoh Y. JNK antagonizes Akt-
mediated survival signals by phosphorylating 14-3-3. J Cell Biol 170:
295–304, 2005.
44. Kim BJ, Ryu SW, Song BJ. JNK- and p38 kinase-mediated
phosphorylation of Bax leads to its activation and mitochondrial
translocation and to apoptosis of human hepatoma HepG2 cells. J Biol
Chem 281:21256–21265, 2006.
45. Xin M, Deng X. Nicotine inactivation of the proapoptotic function of
Bax through phosphorylation. J Biol Chem 280:10781–10789, 2005.
46. Weigel D, Jurgens G, Kuttner F, Seifert E, Jackle H. The homeotic
gene fork head encodes a nuclear protein and is expressed in the
terminal regions of the Drosophila embryo. Cell 57:645–658, 1989.
47. Huang H, Tindall DJ. Dynamic FoxO transcription factors. J Cell Sci
120:2479–2487, 2007.
48. Kaestner KH, Knochel W, Martinez DE. Unified nomenclature for the
winged helix/forkhead transcription factors. Genes Dev 14:142–146,
2000.
49. Wang X, Finegan KG, Robinson AC, Knowles L, Khosravi-Far R,
Hinchliffe KA, Boot-Handford RP, Tournier C. Activation of
extracellular signal-regulated protein kinase 5 downregulates FasL
upon osmotic stress. Cell Death Differ 3:2099–2108, 2006.
50. Brunet A, Park J, Tran H, Hu LS, Hemmings BA, Greenberg ME.
Protein kinase SGK mediates survival signals by phosphorylating the
forkhead transcription factor FKHRL1 (FOXO3a). Mol Cell Biol 21:
952–965, 2001.
51. Brunet A, Kanai F, Stehn J, Xu J, Sarbassova D, Frangioni JV, Dalal
SN, DeCaprio JA, Greenberg ME, Yaffe MB. 14-3-3 transits to the
nucleus and participates in dynamic nucleocytoplasmic transport. J Cell
Biol 156:817–828, 2002.
52. Parcellier A, Tintignac LA, Zhuravleva E, Hemmings BA. PKB and the
mitochondria: AKTing on apoptosis. Cell Signal 20:21–30, 2008.
53. Pende M. mTOR, Akt, S6 kinases and the control of skeletal muscle
growth. Bull Cancer 93:E39–43, 2006.
54. Harada H, Andersen JS, Mann M, Terada N, Korsmeyer SJ. p70S6
kinase signals cell survival as well as growth, inactivating the pro-
apoptotic molecule BAD. Proc Natl Acad Sci U S A 98:9666–9670,
2001.
55. Thomas G. The S6 kinase signaling pathway in the control of
development and growth. Biol Res 35:305–313, 2002.
56. Kim DH, Sarbassov DD, Ali SM, King JE, Latek RR, Erdjument-
Bromage H, Tempst P, Sabatini DM. mTOR interacts with raptor to
form a nutrient-sensitive complex that signals to the cell growth
machinery. Cell 110:163–175, 2002.
57. Jacinto E, Facchinetti V, Liu D, Soto N, Wei S, Jung SY, Huang Q, Qin
J, Su B. SIN1/MIP1 maintains rictor-mTOR complex integrity and
regulates Akt phosphorylation and substrate specificity. Cell 127:125–
137, 2006.
58. Ali SM, Sabatini DM. Structure of S6 kinase 1 determines whether
raptor-mTOR or rictor-mTOR phosphorylates its hydrophobic motif
site. J Biol Chem 280:19445–19448, 2005.
59. Inoki K, Li Y, Zhu T, Wu J, Guan KL. TSC2 is phosphorylated and
inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol 4:
648–657, 2002.
60. Sancak Y, Thoreen CC, Peterson TR, Lindquist RA, Kang SA, Spooner
E, Carr SA, Sabatini DM. PRAS40 is an insulin-regulated inhibitor of
the mTORC1 protein kinase. Mol Cell 25:903–915, 2007.
61. Vander Haar E, Lee SI, Bandhakavi S, Griffin TJ, Kim DH. Insulin
signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat
Cell Biol 9:316–323, 2007.
62. Ikenoue T, Inoki K, Yang Q, Zhou X, Guan KL. Essential function of
TORC2 in PKC and Akt turn motif phosphorylation, maturation and
signalling. EMBO J 27:1919–1931, 2008.
63. Carling D. The AMP-activated protein kinase cascade—a unifying
system for energy control. Trends Biochem Sci 29:18–24, 2004.
64. Hahn-Windgassen A, Nogueira V, Chen CC, Skeen JE, Sonenberg N,
Hay N. Akt activates the mammalian target of rapamycin by regulating
cellular ATP level and AMPK activity. J Biol Chem 280:32081–32089,
2005.
65. Dummler B, Hemmings BA. Physiological roles of PKB/Akt isoforms
in development and disease. Biochem Soc Trans 35:231–235, 2007.
66. Cho H, Thorvaldsen JL, Chu Q, Feng F, Birnbaum MJ. Akt1/PKBalpha
is required for normal growth but dispensable for maintenance of
glucose homeostasis in mice. J Biol Chem 276:38349–38352, 2001.
67. Cho H, Mu J, Kim JK, Thorvaldsen JL, Chu Q, Crenshaw EB 3rd,
Kaestner KH, Bartolomei MS, Shulman GI, Birnbaum MJ. Insulin
resistance and a diabetes mellitus-like syndrome in mice lacking the
protein kinase Akt2 (PKB beta). Science 292:1728–1731, 2001.
68. Garofalo RS, Orena SJ, Rafidi K, Torchia AJ, Stock JL, Hildebrandt
AL, Coskran T, Black SC, Brees DJ, Wicks JR, McNeish JD, Coleman
KG. Severe diabetes, age-dependent loss of adipose tissue, and mild
growth deficiency in mice lacking Akt2/PKB beta. J Clin Invest 112:
197–208, 2003.
69. Easton RM, Cho H, Roovers K, Shineman DW, Mizrahi M, Forman
MS, Lee VM, Szabolcs M, de Jong R, Oltersdorf T, Ludwig T,
Efstratiadis A, Birnbaum MJ. Role for Akt3/protein kinase Bgamma in
attainment of normal brain size. Mol Cell Biol 25:1869–1878, 2005.
70. Tschopp O, Yang ZZ, Brodbeck D, Dummler BA, Hemmings-
Mieszczak M, Watanabe T, Michaelis T, Frahm J, Hemmings BA.
Essential role of protein kinase B gamma (PKB gamma/Akt3) in
postnatal brain development but not in glucose homeostasis. Develop-
ment 132:2943–2954, 2005.
71. Maroulakou IG, Oemler W, Naber SP, Klebba I, Kuperwasser C,
Tsichlis PN. Distinct roles of the three Akt isoforms in lactogenic
differentiation and involution. J Cell Physiol 217:468–477, 2008.
72. Stahl JM, Sharma A, Cheung M, Zimmerman M, Cheng JQ, Bosenberg
MW, Kester M, Sandirasegarane L, Robertson GP. Deregulated Akt3
activity promotes development of malignant melanoma. Cancer Res 64:
7002–7010, 2004.
73. Fujio Y, Mitsuuchi Y, Testa JR, Walsh K. Activation of Akt2 inhibits
anoikis and apoptosis induced by myogenic differentiation. Cell Death
Differ 8:1207–1212, 2001.
74. Sale EM, Hodgkinson CP, Jones NP, Sale GJ. A new strategy for
AKT ACTIVATION AND ACTIONS 1269
by guest on May 29, 2013http://ebm.rsmjournals.com/Downloaded from
studying protein kinase B and its three isoforms. Role of protein kinase
B in phosphorylating glycogen synthase kinase-3, tuberin, WNK1, and
ATP citrate lyase. Biochemistry 45:213–223, 2006.
75. Liu X, Shi Y, Birnbaum MJ, Ye K, De Jong R, Oltersdorf T, Giranda
VL, Luo Y. Quantitative analysis of anti-apoptotic function of Akt in
Akt1 and Akt2 double knock-out mouse embryonic fibroblast cells
under normal and stressed conditions. J Biol Chem 281:31380–31388,
2006.
76. Barnett SF, Defeo-Jones D, Fu S, Hancock PJ, Haskell KM, Jones RE,
Kahana JA, Kral AM, Leander K, Lee LL, Malinowski J, McAvoy EM,
Nahas DD, Robinson RG, Huber HE. Identification and character-
ization of pleckstrin-homology-domain-dependent and isoenzyme-
specific Akt inhibitors. Biochem J 385:399–408, 2005.
77. Green CJ, Goransson O, Kular GS, Leslie NR, Gray A, Alessi DR,
Sakamoto K, Hundal HS. 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, 2008.
78. Wu Z, Hartnett JC, Neilson LA, Robinson RG, Fu S, Barnett SF,
Defeo-Jones D, Jones RE, Kral AM, Huber HE, Hartman GD, Bilodeau
MT. Development of pyridopyrimidines as potent Akt1/2 inhibitors.
Bioorg Med Chem Lett 18:1274–1279, 2008.
1270 MATHENY AND ADAMO
by guest on May 29, 2013http://ebm.rsmjournals.com/Downloaded from
... Based on the OPA1 His42Tyr gene expression signature, AKT was predicted to be inactivated (Blue shaded nodes; Fig. S2). The full activity of AKT relies on its total protein concentration and the phosphorylation levels of Thr308 and Ser473 residues, which ref lect the activity of upstream kinases PDK1 and mTORC2, respectively [50][51][52][53][54][55][56]. Our findings showed an increase in total AKT levels in mutant fibroblasts (about 57%). ...
Article
In several cases of mitochondrial diseases, the underlying genetic and bioenergetic causes of reduced oxidative phosphorylation (OxPhos) in mitochondrial dysfunction are well understood. However, there is still limited knowledge about the specific cellular outcomes and factors involved for each gene and mutation, which contributes to the lack of effective treatments for these disorders. This study focused on fibroblasts from a patient with Autosomal Dominant Optic Atrophy (ADOA) plus syndrome harboring a mutation in the Optic Atrophy 1 (OPA1) gene. By combining functional and transcriptomic approaches, we investigated the mitochondrial function and identified cellular phenotypes associated with the disease. Our findings revealed that fibroblasts with the OPA1 mutation exhibited a disrupted mitochondrial network and function, leading to altered mitochondrial dynamics and reduced autophagic response. Additionally, we observed a premature senescence phenotype in these cells, suggesting a previously unexplored role of the OPA1 gene in inducing senescence in ADOA plus patients. This study provides novel insights into the mechanisms underlying mitochondrial dysfunction in ADOA plus and highlights the potential importance of senescence in disease progression.
... By attaching to a protein complex made up of a regulatory subunit and a catalytic subunit, PIK3C3 regulates autophagy and macrophage phagocytosis ( Table 2) [114]. [96,106] Tumor proliferation and invasion [107] Class III Endosome fusion [114] Autophagy and phagosome formation [114] Macrophage phagocytosis [114] The PI3K pathway's main effector is AKT (a serine/threonine kinase), which has a number of downstream effectors that control important cellular functions [115,116]. AKT, also known as protein kinase B (PKB), exists in mammals in three different isoforms: AKT1, AKT2, and AKT3. While AKT3 is largely expressed in the brain and has relatively limited expression due to its tissue distribution, AKT1 and AKT2 are abundant in numerous tissues, including pancreatic tissue [117]. ...
Article
Full-text available
Renal cell carcinoma (RCC) represents 85–95% of kidney cancers and is the most frequent type of renal cancer in adult patients. It accounts for 3% of all cancer cases and is in 7th place among the most frequent histological types of cancer. Clear cell renal cell carcinoma (ccRCC), accounts for 75% of RCCs and has the most kidney cancer-related deaths. One-third of the patients with ccRCC develop metastases. Renal cancer presents cellular alterations in sugars, lipids, amino acids, and nucleic acid metabolism. RCC is characterized by several metabolic dysregulations including oxygen sensing (VHL/HIF pathway), glucose transporters (GLUT 1 and GLUT 4) energy sensing, and energy nutrient sensing cascade. Metabolic reprogramming represents an important characteristic of the cancer cells to survive in nutrient and oxygen-deprived environments, to proliferate and metastasize in different body sites. The phosphoinositide 3-kinase-AKT-mammalian target of the rapamycin (PI3K/AKT/mTOR) signaling pathway is usually dysregulated in various cancer types including renal cancer. This molecular pathway is frequently correlated with tumor growth and survival. The main aim of this review is to present renal cancer types, dysregulation of PI3K/AKT/mTOR signaling pathway members, crosstalk with VHL/HIF axis, and carbohydrates, lipids, and amino acid alterations.
... PI3-dependent kinase and TORC2-kinase phosphorylate Akt kinase at Thr 308 and Ser 473 , respectively, thereby activating the kinase [22,29]. Activated Akt kinases subsequently mediate many of the metabolic effects of insulin by phosphorylating its target proteins, for example glycogen synthase kinase 3β (GSK3β, disinhibition of glycogen synthesis), Akt target of 160 kDa (AS160, stimulation of Glut4 translocation), forkhead box protein O transcription factors (FoxO, inhibition of gluconeogenesis), phosphodiesterase (antagonize glucagon and adrenalin signaling), tuberous sclerosis complex 2 (TSC2, stimulation of TORC1-dependent protein synthesis and autophagy) and phosphoprotein phosphatases (activation of several covalently modified enzymes involved in energy storage) [30,31]. ...
Article
Full-text available
Metabolic derangement with poor glycemic control accompanying overweight and obesity is associated with chronic low-grade inflammation and hyperinsulinemia. Macrophages, which present a very heterogeneous population of cells, play a key role in the maintenance of normal tissue homeostasis, but functional alterations in the resident macrophage pool as well as newly recruited monocyte-derived macrophages are important drivers in the development of low-grade inflammation. While metabolic dysfunction, insulin resistance and tissue damage may trigger or advance pro-inflammatory responses in macrophages, the inflammation itself contributes to the development of insulin resistance and the resulting hyperinsulinemia. Macrophages express insulin receptors whose downstream signaling networks share a number of knots with the signaling pathways of pattern recognition and cytokine receptors, which shape macrophage polarity. The shared knots allow insulin to enhance or attenuate both pro-inflammatory and anti-inflammatory macrophage responses. This supposedly physiological function may be impaired by hyperinsulinemia or insulin resistance in macrophages. This review discusses the mutual ambiguous relationship of low-grade inflammation, insulin resistance, hyperinsulinemia and the insulin-dependent modulation of macrophage activity with a focus on adipose tissue and liver.
... В пользу участия nNOS в глюкозном транспорте свидетельствуют данные, что стимуляцию поглощения глюкозы в камбаловидной мышце крыс можно полностью заблокировать ингибитором nNOS, но детальные механизмы этого до конца не установлены [70][71][72][73][74][75]. Предполагают, что ключевую роль здесь играет изоформа μ, которая присутствует в скелетных и сердечной мышцах [72]. ...
Article
Full-text available
The study of the molecular mechanisms of metabolic syndrome (MS) and its complications are among the most acute problems of modern endocrinology. Functional changes in the expression, activity, and regulatory properties of neuronal NO synthase (nNOS), which catalyzes the formation of the most important secondary mediator, nitric oxide (NO), and its dependent NO/cGMP signaling pathways in the brain, myocardium, and skeletal muscles, play a key role among the molecular causes of MS. In the brain, nNOS is associated with NMDA receptors, the hyperactivation of which in MS leads to excessive stimulation of nNOS and hyperproduction of NO, which leads to NO-induced damage to neurons and disruption of the central regulation of physiological processes and neurodegeneration. In the myocardium with MS, there are changes in the expression and localization of nNOS, as well as its functional interaction with cytoskeletal proteins, which leads to disorders of myocardial contraction and hypertrophy. In skeletal muscles, nNOS controls their contraction, oxidative metabolism, is involved in the regulation of vascular relaxation, and also participates in the regulation of glucose transport. A decrease in the expression and activity of nNOS, as well as dysregulation of its activity in MS, cause disturbances of these processes and make a significant contribution to the development of insulin resistance and deterioration of glucose homeostasis. Thus, nNOS can be considered an important therapeutic target in the treatment of MS and other metabolic disorders, as well as to prevent their complications from the nervous and cardiovascular systems and the musculoskeletal system.
Article
Empty spiracles homeobox 2 (EMX2) is initially identified as a key transcription factor that plays an essential role in the regulation of neuronal development and some brain disorders. Recently, several studies emphasized that EMX2 could as a tumor suppressor, but its role in human clear cell renal cell carcinoma (ccRCC) remains unclear. In the present study, we investigated the role and underlying mechanism of EMX2 in the regulation of ccRCC progress. Our results demonstrated that EMX2 expression was markedly decreased in ccRCC tissues and cell lines, and low EMX2 expression predicted the poor prognosis of ccRCC patients. In addition, forced expression of EMX2 significantly inhibited the cell growth, migration, and invasion in vitro, as well as ccRCC tumor growth in nude mice, via, at least in part, regulating Akt/FOXO3a pathway. In detail, EMX2 could attenuate the phosphorylation levels of Akt and FOXO3a, and increase FOXO3a expression without affecting total Akt expression in vivo and in vitro. Meanwhile, shRNA‐mediated knockdown of FOXO3a expression could obviously attenuate the effects of EMX2 on cell growth, migration, invasion, and tumor growth. Furthermore, EMX2 could significantly attenuate the interaction between Akt and FOXO3a. Taken together, our results demonstrated that EMX2 could inhibit ccRCC progress through, at least in part, modulating Akt/FOXO3a signaling pathway, thus representing a novel role and underlying mechanism of EMX2 in the regulation of ccRCC progress.
Article
Full-text available
Due to the inherent radiation tolerance, patients who suffered from glioma frequently encounter tumor recurrence and malignant progression within the radiation target area, ultimately succumbing to treatment ineffectiveness. The precise mechanism underlying radiation tolerance remains elusive due to the dearth of in vitro models and the limitations associated with animal models. Therefore, a bioprinted glioma model is engineered, characterized the phenotypic traits in vitro, and the radiation tolerance compared to 2D ones when subjected to X‐ray radiation is assessed. By comparing the differential gene expression profiles between the 2D and 3D glioma model, identify functional genes, and analyze distinctions in gene expression patterns. Results showed that 3D glioma models exhibited substantial alterations in the expression of genes associated with the stromal microenvironment, notably a significant increase in the radiation tolerance gene ITGA2 (integrin subunit A2). In 3D glioma models, the knockdown of ITGA2 via shRNA resulted in reduced radiation tolerance in glioma cells and concomitant inhibition of the p‐AKT pathway. Overall, 3D bioprinted glioma model faithfully recapitulates the in vivo tumor microenvironment (TME) and exhibits enhanced resistance to radiation, mediated through the ITGA2/p‐AKT pathway. This model represents a superior in vitro platform for investigating glioma radiotherapy tolerance.
Article
AKT signaling plays a crucial role in muscle physiology, and is activated by stimuli, including insulin, growth factors, and exercise. Three AKT isoforms have been identified in mammals, and they possess both distinct and redundant functions. However, it is currently unknown what the predominant AKT isoform is in primary human skeletal myotubes, and very little is known regarding the effects of insulin and insulin‐like growth factor‐I (IGF‐I) on AKT isoforms activation in human myotubes. Thus, we sought to determine the abundances of each AKT isoform in primary human skeletal myotubes and their responses to insulin or IGF‐I. Analysis of protein lysates by liquid chromatography‐parallel reaction monitoring/mass spectrometry revealed that AKT1 was the most abundant AKT isoform and AKT3 was the least‐abundant isoform. Next, myotubes were treated with either 100 nM insulin or 10 nM IGF‐I for 5, 20, 45, or 60 min. In response to insulin, there was a significant treatment effect on phosphorylation of AKT1 and AKT2, but not AKT3 ( p < 0.01). In response to IGF‐I, there was a significant treatment effect on phosphorylation of pan‐AKT at all timepoints compared to control ( p < 0.01). Next, we determined how much of the total AKT isoform pool was phosphorylated. For insulin stimulation, AKT1 was significantly higher than AKT2 at 5 min and 60 min posttreatment ( p < 0.05 both) and significantly different than AKT3 at all timepoints ( p < 0.05). For IGF‐I stimulation, AKT1 was significantly higher than AKT2 at 45 and 60 min posttreatment ( p < 0.05 both) and significantly higher than AKT3 at all timepoints ( p < 0.05). Our findings reveal the differential phosphorylation patterns among the AKT isoforms and suggest a potential explanation for the regulatory role of AKT1 in skeletal muscle.
Article
Imidazo[4,5-b]pyridines are amongst the oldest known heteroaromatic derivatives. Their structural similarity with purine basis has however aroused the curiosity of biologists and resulted in the developments of innovative bioactive compounds. This review thus firstly describes the main synthetic ways currently used to produce imidazo[4,5-b]pyridine derivatives, and secondly gives examples of their potential, especially focusing on protein inhibition abilities, thus opening the way to applications as anti-- cancer or antimicrobial agents.
Article
In the present work, four new compounds based on the privileged structure acridone were efficiently synthesized following simple operational techniques and biologically tested on proliferative skeletal muscle cells (C2C12) and rhabdomyosarcoma cells (RD) showing no significant changes in the number of dead or viable cells at 1 µM during 24 or 48 h of treatment. Of relevance, acridone derivatives 3a-3d at 0.5 µM for 24 h effectively inhibited Akt activation in C2C12, while at 1 µM only compounds 3a and 3b have effect. RD cells showed a different response pattern. These cells treated with 3a (0.5 µM), 3b (0.5 µM) or 3d (0.5 or 1 µM) for 24 h shown significant Akt inhibition. In addition, 3a-3d assayed at 1 µM for 48 h were highly successful in inhibiting Akt phosphorylation. Finally, based on molecular docking and molecular dynamics simulations, we rationalize the experimental results mentioned above and propose that 3-phosphoinositide-dependent kinase-1 (PDK1) could be one of the molecular targets of this new series of 1, 3-dihydroxyacridone derivatives. Biological and in silico studies revealed that 3b could be considered as the most promising prototype for the development of new antitumor agents.
Article
Full-text available
Glucose homeostasis depends on insulin responsiveness in target tissues, most importantly, muscle and liver. The critical initial steps in insulin action include phosphorylation of scaffolding proteins and activation of phosphatidylinositol 3-kinase. These early events lead to activation of the serine-threonine protein kinase Akt, also known as protein kinase B. We show that mice deficient in Akt2 are impaired in the ability of insulin to lower blood glucose because of defects in the action of the hormone on liver and skeletal muscle. These data establish Akt2 as an essential gene in the maintenance of normal glucose homeostasis.
Article
Full-text available
Protein kinase B (PKB)/Akt has been strongly implicated in the insulin-dependent stimulation of GLUT4 translocation and glucose transport in skeletal muscle and fat cells. Recently an allosteric inhibitor of PKB (Akti) that selectively targets PKBalpha and -beta was reported, but as yet its precise mechanism of action or ability to suppress key insulin-regulated events such as glucose and amino acid uptake and glycogen synthesis in muscle cells has not been reported. We show here that Akti ablates the insulin-dependent regulation of these processes in L6 myotubes at submicromolar concentrations and that inhibition correlates tightly with loss of PKB activation/phosphorylation. Similar findings were obtained using 3T3-L1 adipocytes. Akti did not inhibit IRS1 tyrosine phosphorylation, phosphatidylinositol 3-kinase signaling, or activation of Erks, ribosomal S6 kinase, or atypical protein kinases C but significantly impaired regulation of downstream PKB targets glycogen synthase kinase-3 and AS160. Akti-mediated inhibition of PKB requires an intact kinase pleckstrin homology domain but does not involve suppression of 3-phosphoinositide binding to this domain. Importantly, we have discovered that Akti inhibition is critically dependent upon a solvent-exposed tryptophan residue (Trp-80) that is present within the pleckstrin homology domain of all three PKB isoforms and whose mutation to an alanine (PKB(W80A)) yields an Akti-resistant kinase. Cellular expression of PKB(W80A) antagonized the Akti-mediated inhibition of glucose and amino acid uptake. Our findings support a critical role for PKB in the hormonal regulation of glucose and system A amino acid uptake and indicate that use of Akti and expression of the drug-resistant kinase will be valuable tools in delineating cellular PKB functions.
Article
Full-text available
The region-specific homeotic gene fork head (fkh) promotes terminal as opposed to segmental development in the Drosophila embryo. We have cloned the fkh region by chromosomal walking. P element-mediated germ-line transformation and sequence comparison of wild-type and mutant alleles identify the fkh gene within the cloned region. fkh is expressed in the early embryo in the two terminal domains that are homeotically transformed in fkh mutant embryos. The nuclear localization of the fkh protein suggests that fkh regulates the transcription of other, subordinate, genes. The fkh gene product, however, does not contain a known protein motif, such as the homeodomain or the zinc fingers, nor is it similar in sequence to any other known protein.
Article
cDNA clones encoding the third member of the RAC protein kinase family, termed RAC-PK γ, were isolated from a rat brain cDNA library. The deduced amino acid sequence of RAC-PK γ was highly related to those of previously identified family members, RAC-PK α and β, that have a pleckstrin homology domain and a protein-serine/threonine kinase catalytic domain at the amino- and carboxyl-terminal regions, respectively. Northern blot analysis indicated that RAG-PK γ was expressed abundantly in brain and testis. Specific activities of RAC-PK α, β, and γ purified from transfected COS-7 cells were similar when measured by using myelin basic protein as a phosphate acceptor. Analysis using fusion proteins of glutathione S-transferase revealed that the pleckstrin homology domain of the three subtypes of RAC-PK associate with both protein kinase C subspecies and βγ subunits of G proteins. These results suggest that the pleckstrin homology domains of RAC protein kinase family could associate more than one protein to regulate the activity and/or intracellular distribution of this enzyme family by different ways.
Article
Serum- and glucocorticoid-inducible kinases (SGKs) form a novel family of serine/threonine kinases that are activated in response to a variety of extracellular stimuli. SGKs are related to Akt (also called PKB), a serine/threonine kinase that plays a crucial role in promoting cell survival. Like Akt, SGKs are activated by the phosphoinositide-3 kinase (PI3K) and translocate to the nucleus upon growth factor stimulation. However the physiological substrates and cellular functions of SGKs remained to be identified. We hypothesized that SGKs regulate cellular functions in concert with Akt by phosphorylating common targets within the nucleus. The best-characterized nuclear substrates of Akt are transcription factors of the Forkhead family. Akt phosphorylates Forkhead transcription factors such as FKHRL1, leading to FKHRL1's exit from the nucleus and the consequent shutoff of FKHRL1 target genes. We show here that SGK1, like Akt, promotes cell survival and that it does so in part by phosphorylating and inactivating FKHRL1. However, SGK and Akt display differences with respect to the efficacy with which they phosphorylate the three regulatory sites on FKHRL1. While both kinases can phosphorylate Thr-32, SGK displays a marked preference for Ser-315 whereas Akt favors Ser-253. These findings suggest that SGK and Akt may coordinately regulate the function of FKHRL1 by phosphorylating this transcription factor at distinct sites. The efficient phosphorylation of these three sites on FKHRL1 by SGK and Akt appears to be critical to the ability of growth factors to suppress FKHRL1-dependent transcription, thereby preventing FKHRL1 from inducing cell cycle arrest and apoptosis. These findings indicate that SGK acts in concert with Akt to propagate the effects of PI3K activation within the nucleus and to mediate the biological outputs of PI3K signaling, including cell survival and cell cycle progression.
Article
Murine leukemia viruses capable of malignant transformation of mink tissue culture cells have been isolated from an AKR thymoma cell line and from a spontaneous reticulum cell sarcoma in an NIH Swiss mouse partially congenic for the AKR ecotropic virus-inducing locus Akv-2. In contrast to the recently described mink cell focus-inducing strains of murine leukemia virus, at least one of the two transforming strains is replication defective. Nonproducer mink cells carrying the genome of the transforming virus of AKR origin have been isolated, and pseudotype transforming viruses generated.
Article
A novel serine/threonine protein kinase (termed rac-PK) has recently been identified and cloned from cDNA libraries derived from the human cell lines MCF-7 and WI38. A second form of this protein kinase, termed rac protein kinase beta, has been identified from cDNAs derived from the same cell lines. These two closely related forms show 90% homology, although the beta form with a predicted Mr 60,200 has a carboxyl terminal extension of 40 amino acids in comparison to the alpha form. This extension has a high serine content with 11 serine residues in the last 30 amino acids. The beta form of the protein has been shown by both in vitro translation and bacterial expression to be approximately 5000 Da larger than the alpha form. rac protein kinase beta is encoded by a 3.4-kb transcript and the alpha form is encoded by a 3.2-kb mRNA. Using gene-specific probes both transcripts were detected in all cell types analyzed, although levels of expression were different for the two forms. The catalytic domain of rac protein kinase beta shows a high degree of homology to both the protein kinase C and cyclic AMP-dependent protein kinase families, and hence rac protein kinases appear to represent a new subfamily of the second messenger serine/threonine protein kinases.
Article
A partial cDNA was isolated that encoded a protein kinase, termed rac (related to the A and C kinases). This cDNA was subsequently used to screen libraries derived from the human cell lines MCF-7 and WI38 and led to the isolation of full-length cDNA clones. DNA sequence analysis identified an open reading frame of 1440 base pairs encoding a protein of 480 amino acids (Mr, 55,716). This result was supported by the synthesis of a Mr 58,000 protein in an in vitro translation system that used RNA transcribed from cloned cDNAs with SP6 RNA polymerase. The predicted protein contains consensus sequences characteristic of a protein kinase catalytic domain and shows 73% and 68% similarity to protein kinase C and the cAMP-dependent protein kinase, respectively. Northern (RNA) analysis revealed a single mRNA transcript of 3.2 kilobases that varied up to 300-fold between different cell lines. Specific antisera directed towards the carboxyl terminal of the rac protein kinase were prepared and used to identify that phosphorylated several substrates in immunoprecipitates prepared with the rac-specific antisera.
Article
A previous report described the isolation of a directly transforming retrovirus, AKT8, from a spontaneous thymoma of an AKR mouse. The AKT8 provirus has now been molecularly cloned from a transformed, nonproducer cell line. The virus genome contains both viral and nonviral, cell-related sequences; the nonviral sequence has been designated v-akt, the presumed viral oncogene of the AKT8 virus. This gene lacks homology to the 16 other oncogenes tested. The cloned provirus has undergone a partial deletion, during cell passage in vitro, that prevents direct demonstration of the transforming ability of this molecular clone. Two human homologues of the v-akt oncogene, AKT1 and AKT2, were cloned. A survey of 225 human tumors for changes involving AKT1 led to the discovery of a 20-fold amplification of this gene in one of the five gastric adenocarcinomas tested. The results demonstrate that AKT8 has the characteristic structure of a directly transforming retrovirus and that it contains a gene derived from highly conserved cellular sequences that may be involved in the pathogenesis of some human malignancies.