ArticlePDF AvailableLiterature Review

JNK2: A Negative Regulator of Cellular Proliferation

Taylor & Francis
Cell Cycle
Authors:

Abstract and Figures

The three different c-Jun N-terminal kinases (JNKs) are activated in multiple cell types by both apoptotic and mitogenic signals, and in turn regulate the activity of transcription factors such as c-Jun. Being highly homologous and ubiquitously expressed, the JNK1 and JNK2 proteins have been thought to perform redundant functions in many physiological process. However, our data from Jnk1-/- or Jnk2-/- cells and mice suggest that both JNK isozymes perform distinct functions in regulating cellular proliferation via differential regulation of c-Jun, which is a critical regulator of cell-cycle progression. Absence of JNK1, the positive regulator of c-Jun, leads to decreased fibroblast proliferation. In contrast, JNK2 deficiency leads to reduced c-Jun degradation, thereby augmenting c-Jun levels and cellular proliferation. Various cell types including fibroblasts, erythroblasts and hepatocytes from Jnk2-/- mice exhibit increased proliferation rates compared to their wild-type counterparts. These data therefore suggests that JNK2, in contrast to JNK1, is a negative regulator of cellular proliferation in multiple cell types.
Content may be subject to copyright.
©2005 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
[Cell Cycle 3:12, 1520-1523; December 2004]; ©2004 Landes Bioscience
1520
Cell C
y
cle 2004; Vol. 3 Issue 12
Kanaga Sabapathy
1,
*
Erwin F. Wagner
2
1
National Cancer Centre; Singapore , Singapore
2
R
esearch Institute of Molecular Pathology; Vienna, Austria
*Correspondence to: Dr. Kanaga Sabapathy; National Cancer Centre; 11, Hospital
Drive; Singapore 169610 Singapore; Tel.: +65.6436.8349; Fax: + 65.6226.5694;
Email: cmrksb@nccs.com.sg
Received 10/15/04; Accepted 10/18/04
P
reviously published online as a
Cell
Cycle
E-publication:
http://www
.landesbioscience.com/journals/cc/abstract.php?id=1315
KEY WORDS
JNK, c-Jun, proliferation, cyclin D, erythropoiesis
ACKNOWLEDGEMENTS
We thank Wolfram Jochum for help with the
immunostaining.
This work was supported by the National
Medical Research Council of Singapore (K.S.) and
the Austrian Federal Ministry of Science, Transport
and the Arts (E.F.W.). The IMP is supported by
Boehringer Ingelheim.
Perspective
JNK2
A Negative Regulator of Cellular Proliferation
ABSTRACT
The three different c-Jun N-terminal kinases (JNKs) are activated in multiple cell types
by both apoptotic and mitogenic signals, and in turn regulate the activity of transcription
factors such as c-Jun. Being highly homologous and ubiquitously expressed, the JNK1
and JNK2 proteins have been thought to perform redundant functions in many physio-
logical process. However, our data from
Jnk1
-
/-
or Jnk2
-
/-
cells and mice suggest that both
JNK isozymes perform distinct functions in regulating cellular proliferation via differential
regulation of c-Jun, which is a critical regulator of cell-cycle progression. Absence of
JNK1, the positive regulator of c
-Jun, leads to decreased fibroblast proliferation. In
contrast, JNK2 deficiency leads to reduced c-Jun degradation, thereby augmenting c-Jun
levels and cellular proliferation. Various cell types including fibroblasts, erythroblasts and
hepatocytes from
Jnk2
-
/-
mice exhibit increased proliferation rates compared to their
wild-type counterparts. These data therefore suggests that JNK2, in contrast to JNK1, is
a negative regulator of cellular proliferation in multiple cell types.
JNK FUNCTIONS
Structural and functional similarities are frequently found among many proteins, and
such resemblance often suggests common modes of action or regulation. Many compo-
nents of cellular signaling modules, including kinases, are phylogenically grouped together
as families based on such homologies, the degree of which can sometimes be extremely
high. However, the question that remains unresolved is if such similar molecules are
evolutionarily conserved and carried through to perform exactly similar and redundant
functions, or if they have specific and different biological roles that have yet to be uncovered.
One such family of proteins is the c-Jun-N-terminal (JNK) kinases, that are present
ubiquitously in the organism. JNKs signal stress responses, thereby leading to apoptosis
induction, as well as pr
oinflammator
y and some form of mitogenic signals.
1-3
The JNK
subgr
oup consists of three members—JNK1, JNK2 and JNK3—which are highly homol-
ogous, and are present as multiple isoforms generated through alternate splicing at the
carboxy-terminus.
4-6
Among the three members of the JNK family, JNK3 is expressed pre-
dominantly in the brain whereas both JNK1 and JNK2 are expressed ubiquitously.
6
Combinatorial use of the various JNKs and their upstream kinases are thought to lead to
differ
ential regulation of substrate proteins in response to multiple stimuli, thereby estab-
lishing signal-specificity. Nevertheless, the specific roles of the different JNK proteins have
not been fully addressed, although the different JNK isozymes may have evolved for specific
biological functions, pr
obably depending on the activ
ating stimuli and r
esponding cell
type.
Upon stimulation, the activated JNKs phosphorylate transcription factors like c-Jun
and ATF-2, which participate in the activation and formation of the AP-1 complex.
7
H
ither
to, most in vitro studies have used c-Jun as the model substrate to investigate the
natur
e of JNK
-
substrate binding activity and specificity
. All thr
ee JNKs hav
e been sho
wn
to bind to the delta domain of c-Jun and phosphorylate it on serines 63 and 73, leading
to its activation.
5
Of the two, JNK2 has been shown to have a twenty-five fold higher
binding affinity for c-Jun than JNK1.
5
Moreover, although the various isoforms of both
JNK1 and JNK2 w
er
e sho
wn to bind and phosphor
ylate the substrates to various extents
in in vitro experiments, the JNK1 isoforms appear to be slightly more efficient in phospho-
r
ylating c
-Jun.
8
Besides being involved in phosphorylation and subsequent activation of substrates
upon signal stimulation, the JNKs hav
e been suggested to cause degradation of the same
substrates under nonstressed conditions.
9
The inactive JNKs have
been shown to cause degradation of transcription factors such as
c
-Jun, ATF2 and p53—a function that was thought to be dependent
on binding of JNKs to the substrates, and occurs in the absence of
phosphor
ylation of the substrates.
9
D
egradation of the substrates is
inhibited in the presence of activating stimuli, which now signal
through the activated JNKs and stabilize the substrates by phoshory-
lating them.
10
Thus, the dual activity of the JNKs in targeting c-Jun
and other substrates for ubiquitin-mediated degradation or in
protecting them from entering the degradation pathway via phospho-
rylation points to an intricate role of JNK in regulating c-Jun stability.
Nonetheless, the specific role of the different JNK proteins in regu-
lating substrate metabolism was hitherto unknown.
JNKS AND PROLIFERATION CONTROL
The biological effects of the JNK proteins on cellular proliferation
have been alluded to indirectly in many previous studies. Oncogenic
Ha-ras has been shown to activate the JNK pathway.
11
In addition,
the transforming small GTP binding proteins Rac1 and Cdc42 have
been shown to activate the JNKs.
12,13
Moreover, it was recently
shown that the JNK pathway is activated in some tumor cell types,
indicating that the JNK pathway might be essential for tumorigene-
sis.
14
In support of these indirect studies, we and others have
demonstrated a causal role for the JNKs in regulating cellular prolif-
eration using fibroblasts lacking both JNK1 and JNK2.
15,16
The
Jnk1
-/-
Jnk2
-/-
fibroblasts were found to exhibit a proliferation defect
and were also defective in c-Jun phosphorylation, which has been
shown to be essential for cellular proliferation.
17,18
In vivo studies
using cells from knockout and transgenic mice have indicated that
c-Jun is essential for efficient transition of the G
1
-S phase of the cell
cycle, and cells lacking c-Jun have a severe proliferation defect.
17
In
addition, it was demonstrated that c-Jun N-terminal phosphorylation
is important for efficient cellular proliferation.
19
Fibroblasts from
mice carrying a mutant c-jun allele having the JNK phosphoacceptor
serines 63 and 73 changed to alanines (junAA) proliferate slower
than wild-type cells, though this defect was not as severe as found
with c-
jun
-/-
fibroblasts.
19
SPECIFIC ROLES OF JNK1 AND JNK2 IN REGULATING
C-JUN-DEPENDENT PROLIFERATION
In our recent findings published in Molecular Cell, we demon-
strated that JNK1 and JNK2 perform distinct functions in regulating
cellular pr
oliferation.
16
Consistent with the notion that c
-
J
un phos
-
phor
ylation is r
equir
ed for efficient pr
oliferation, w
e sho
w
ed that
Jnk1
-/-
fibr
oblasts, in which c
-
J
un phosphor
ylation is reduced, exhib-
ited a pr
oliferation defect. H
o
w
ever,
Jnk2
-/-
fibr
oblasts w
er
e found to
hav
e incr
eased proliferation rates, and a concomitant increase in the
levels of phosphorylated c-Jun. Moreover, JNK2 deficiency also led
to increased proliferation of erthyroblasts suggesting that the negative
effect of JNK2 on cellular pr
oliferation was not r
estricted to fibr
ob
-
lasts. O
ur r
esults indicate that despite their biochemical and str
uc
tural
similarities and earlier expectations, JNK1 and JNK2 ex
er
t differ
ent
effects on c-Jun turnover and activity, which correlates with their
different effects on cell proliferation. JNK1 appears to be a positive
regulator of cellular proliferation, being required for phosphorylation
and activation of c-Jun. On the contrary, JNK2 appears to primarily
function as a negative regulator of c-Jun stability, correlating with its
negative effect on cell pr
oliferation.
At a molecular level, our findings indicate that JNK1 contributes
to c-Jun phosphorylation more than JNK2, especially after cellular
stimulation. Very little JNK1 was associated with c-Jun in unstimu-
lated cells, but the interaction between the two is increased dramat-
ically upon cell stimulation.
16
As JNK1 is also the major c-Jun
kinase present in the cells, this resulted in increased c-Jun N-terminal
phosphorylation and elevated AP-1 activity. By contrast, c-Jun was
mostly bound to JNK2 in unstimulated cells, although this interac-
tion did not result in c-Jun phosphorylation. Instead, JNK2 appeared
to be responsible for targeting c-Jun to degradation, correlating with
previous reports that inactive JNKs can target c-Jun and other sub-
strates for degradation.
9
These findings made under physiological
conditions, correlate with findings based on in vitro experiments
showing that inactive JNK2 has 25x more affinity for c-Jun binding
than JNK1.
5
In addition to decreasing the amount of JNK2 bound
c-Jun and increasing the binding of JNK1 to c-Jun, cellular stimula-
tion also resulted in JNK1-mediated c-Jun phosphorylation, activa-
tion and stabilization. Previously, transient transfection experiments
demonstrated that both JNK1 and JNK2 could phosphorylate c-Jun
and other targets,
8
giving rise to the expectation that there are no
major differences between the kinase activities of JNK1 and JNK2.
We also obtained similar results in transient transfections,
16
but
under physiological conditions, there were clear differences regarding
the relative roles of JNK1 and JNK2 in c-Jun regulation.
Other recent reports have also focused on c-Jun turnover. Nateri
et al. demonstrated that c-Jun is degraded by the E3 ligase
Fbw7-containing Skp/Cullin/F-box protein complex (SCF
Fbw7
),
only when it is phosphor
ylated on the JNK phosphor
ylation sites
(i.e., serines 63 and 73).
20
SCF
Fbw7
-dependent c-Jun degradation
was shown to occur in neuronal cells, where SCF
Fb
w7
is highly
expressed, suggesting that this could be a cell-type dependent pathway
(B
ehrens A, personal communication). Moreover, Goa et al. have
recently showed that the E3 ligase Itch, controlled c-Jun turnover in
T cells.
21
I
tch was sho
wn to be efficiently phosphor
ylated b
y JNK1,
rather than JNK2, that led to activ
ation of its E3 ligase activity
.
Itch-mediated c-Jun degradation was found to be independent of
JNK-mediated c-Jun phosphorylation.
21
Furthermore, human
De-Etiolated-1 gene product, whose expression was highest in the
ovary and some lymphoid organs was shown to regulate c-Jun stability
www.landesbioscience.com
Cell C
ycle 1521
JNK2
Figure 1. Increased hepatocyte proliferation after partial hepatactomy in
JNK2 deficient livers. H&E staining of Jnk2
+/+
and Jnk2
-
/-
livers 30 h after
partial hepatectomy (top panel). BrdU-immunostaining of wild-type and
JNK2 deficient livers 30 h after hepatectomy are shown in the lower panel.
Arrows point to some BrdU-positive hepatocytes. n = 3; Magnification: 40x.
JNK2 NEGATIVELY REGULATES PROLIFERATION
OF HEPATOCYTES AND KERATINOCYTES
Similar differences in the control of cellular
pr
oliferation in various other cell types have also
been noted. S
ince it was shown that c-Jun was
r
equired for proper hepatocyte proliferation after
partial hepatectomy (PH),
23
we have also evaluated
the effect of JNK2 loss during PH by determining
the number of S-phase cells by BrdU incorporation.
We noticed that liver regeneration after partial
hepatectomy occurred much faster in
Jnk2
-/-
mice
compared to the wild-type counterparts (Sabapathy
an Wagner, unpublished data). Compared with
controls, the rate of BrdU incorporation by hepato-
cytes was distinctly increased in
Jnk2
-/-
mice (Fig. 1),
suggesting that the absence of JNK2 leads to accel
-
erated hepatocyte proliferation. Moreover, recent
data from Weston et al. suggested that keratinocyte
proliferation was also differentially altered by JNK1
and JNK2.
24
Absence of JNK1 led to thinner epi-
dermis compared to wild-type mice, due to a
marked reduction of the proliferating cells in the
stratum basale. In contrast, skin from
Jnk2
-/-
mice
revealed keratinocyte hyperplasia, resulting in
increased number of epithelial cell layers, which was
attributed to enhanced proliferation. In addition to
increased proliferation due to lack of JNK2, a
recent report also suggested that specific inhibition
of JNK2 resulted in aneuploidy in human cells.
25
This was proposed to be due to defects in chromo-
some segregation and central spindle formation
during anaphase, which eventually resulted in poly-
ploidy. Together, the emerging trend points to
increased proliferation in multiple cell types in the
absence of JNK2 (Table 1).
OUTLOOK
Hitherto, there are no reports identifying JNK2
mutations in human cancers, though the c-Jun/JNK
pathway has been sho
wn to be activated in many
cancers.
14
Thus, the possibility exists that JNK2’
s
degradation inducing activity is reduced in cancer
cells by specific activation of a JNK2-specific
inhibitor(s), or vice-versa. Further investigations are
r
equired to explore these possibilities. Nevertheless,
the genetic data suggest that JNK2 has specifically
evolved to keep a check on total JNK activity within
the cell, which if left uncontrolled, could lead to
pathological conditions.
Ther
efore, one could envis-
age a model, as depicted in F
igur
e 2, wher
eby both
JNK1 and JNK2 regulates substrate turnover and
function in a r
ecipr
ocal fashion, thereby creating a
regulatory circuit that controls JNK activity. The
data also imply that JNK2-specific pharmacological
activ
ators that specifically incr
ease its degradation
activity or JNK1-specific inhibitors that reduce JNK1 kinase activity
would be useful clinically in human disease, where JNK activity is
elev
ated, such as cancers and neur
onal disor
ders.
Figure 2. Proposed model of JNK1 and JNK2 action. The model shows that when wild-type
cells are in a quiescent state, c-Jun is predominantly bound to inactive JNK2. This probably
results in c-Jun being targeted for degradation (dashed arrows), thereby maintaining low basal
levels of c-Jun. JNK1 bound to c-Jun phosphorylates c-Jun at a reduced rate (solid arrows). After
cell stimulation, JNK2 dissociates from c-Jun, giving access for activated JNK1 to interact with
c-Jun. This results in increased JNK1-mediated c-Jun phosphorylation. The degradative signal
by JNK2 is also inhibited, as less JNK2 is c-
Jun bound, while activated JNK2 makes a minor
contribution to c-Jun phosphorylation. The net effect is increased c-Jun stability and activity. In
Jnk1
-/-
cells, absence of JNK1 leads to loss of basal c-Jun phosphorylation and increased
targeting by JNK2 of c-Jun to degradation, resulting in reduced basal c-Jun levels and activity.
Upon stimulation, c-Jun is only weakly phosphorylated and partially stabilized. By contrast,
absence of JNK2 results in c
-
Jun being less degraded in the absence of stimulating signals,
thereby resulting in higher basal levels. Upon stimulation, the stable c-Jun is further phospho-
rylated by JNK1, resulting in increased activity and stability. Represents phosphorylation
sig
nals;
dashed line represents degradation signal.
T
able 1
ABSENCE
OF
JNK2 RESUL
TS
IN
INCREASED PROLIFERA
TION
IN VARIOUS CELL-TYPES
Cell-type Jnk1
-/-
Jnk2
-/-
R
eference
Fibroblasts Reduced proliferation Increased proliferation 16
Erythroblasts No difference Increased proliferation 16
Hepatocytes
Not tested
Increased proliferation
This repor
t
Keratinocytes Reduced proliferation Increased proliferation 24
1522 Cell Cycle 2004; Vol. 3 Issue 12
by assembling a CUL4A ubiquitin ligase.
22
T
ogether with our findings,
it is evident that c-Jun stability is controlled by several distinct mech-
anisms that appear to be cell-type specific.
JNK2
www.landesbioscience.com
Cell C
ycle 1523
Altogether, it is evident that structurally and functionally similar
pr
oteins can be evolutionarily selected to perform distinct and oppo-
site functions, which can be unrav
eled by genetic studies. The JNK
family of pr
oteins would thus serve as a paradigm for other functional
studies aiming to elucidate the specific functions of highly similar
proteins.
References
1.
Weston CR, Davis RJ. The JNK signal transduction pathway. Curr Opin Genet Dev 2002;
12:14
-21.
2. Manning AM, Davis RJ. Targeting JNK for therapeutic benefit: From junk to gold? Nat
Rev Drug Discov 2003; 2:554-65.
3. Lin A, Dibling B. The true face of JNK activation in apoptosis. Aging Cell 2002; 1:112-6.
4. Dérijard B, Hibi M, Wu IH, Barrett T, Su B, Deng T, Karin M, Davis RJ. JNK1: A
pr
otein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun
activ
ation domain. Cell 1994; 76:1025-37.
5.
Kallunki T, Su B, Tsigelny I, Sluss HK, Dérijard B, Moore G, Davis RJ, Karin M. JNK2
contains a specificity-determining region responsible for efficient c-Jun binding and phos-
phorylation. Genes Dev 1994; 8:2996-3007.
6. Mohit AA, Martin JH, Miller CA. p49{I3F12} kinase: A novel MAP kinase expressed in a
subset of neur
ons in the human nervous system. Neuron 1995; 14:67-78.
7.
Eferl R, Wagner EF. AP-1: A double-edged sword in tumorigenesis. Nat Rev Cancer 2003;
3:859
-68.
8. Gupta S, Barrett T, Whitmarsh AJ, Cavanagh J, Sluss HK, Dérijard B, Davis RJ. Selective
interaction of JNK protein kinase isoforms with transcription factors. EMBO J 1996;
15:2760
-70.
9. Fuchs SY, Fried VA, Ronai Z. Stress-activated kinases regulate protein stability. Oncogene
1998; 17:1483-90.
10. Musti AM, Treier M, Bohmann D. Reduced ubiquitin-dependent degradation of c-Jun
after phosphorylation by MAP kinases. Science 1997; 275:400-2.
11. Smeal T, Binétruy B, Mercola D, Birrer M, Karin M. Phosphorylation of cJun on Serines
63 and 73 is required for oncogenic and transcriptional cooperation with Ha-Ras. Nature
1991; 354:494-6.
12. Coso OA, Chiariello M, Yu JC, Teramoto H, Crespo P, Xu N, Miki T, Gutkind JS. The
small GTP-binding proteins Rac1 and Cdc42 regulate the activity of the JNK/SAPK
signaling pathway. Cell 1995; 81:1137-46.
13. Hill CS, Wynne J, Treisman R. The Rho family GTPases RhoA, Rac1, and CDC42Hs
regulate transcriptional activation by SRF. Cell 1995; 81:1159-70.
14. Zoumpourlis V, Papassava P, Linardopoulos S, Gillespie D, Balmain A, Pintzas A. High
levels of phosphorylated c-Jun, Fra-1, Fra-2 and ATF-2 proteins correlate with malignant
phenotypes in the multistage mouse skin carcinogenesis model. Oncogene 2000;
19:4011-21.
15. Tournier C, Hess P, Yang DD, Xu J, Turner TK, Nimnual A, Bar-Sagi D, Jones SN, Flavell
RA, Davis RJ. Requirement of JNK for stress-induced activation of the cytochrome
c-mediated death pathway. Science 2000; 288:870-4.
16.
Sabapathy K, Hochedlinger K, Nam SY, Bauer A, Karin M, Wagner EF. Distinct roles for
JNK1 and JNK2 in regulating JNK activity and c-Jun-dependent cell proliferation. Mol
Cell 2004; 15:713-25.
17.
Schr
eiber M, Kolbus A, Piu F, Szabowski A, Mohle-Steinlein U, Tian J, Karin M, Angel P,
Wagner EF. Control of cell cycle progression by c-Jun is p53 dependent. Genes Dev 1999;
13:607
-
19.
18. Wisdom R, Johnson RS, Moore C. c-Jun regulates cell cycle progression and apoptosis by
distinct mechanisms. EMBO J 1999; 18:188
-
97.
19. Behrens A, Sibilia M, Wagner EF. Amino-terminal phosphorylation of c-Jun regulates
str
ess
-induced apoptosis and cellular proliferation. Nat Genet 1999; 21:326-29.
20. Nateri AS, Riera-Sans L, Da Costa C, Behrens A. The ubiquitin ligase SCFFbw7 antago-
niz
es apoptotic JNK signaling. Science 2004; 303:1374-8.
21. Gao M, Labuda T, Xia Y, Gallagher E, Fang D, Liu YC, Karin M. Jun turnover is controlled
thr
ough JNK-dependent phosphorylation of the E3 ligase itch. Science 2004; 306:271-5.
22. Wertz IE, O'Rourke KM, Zhang Z, Dornan D, Arnott D, Deshaies RJ, Dixit VM. Human
D
e-etiolated-1 regulates c-Jun by assembling a CUL4A ubiquitin ligase. Science 2004;
303:1371-4.
23.
Behrens A, Sibilia M, David JP, Möhle-Steinlein U, Tronche F, Schütz G, Wagner EF.
Impaired postnatal hepatocyte proliferation and liver regeneration in mice lacking c-
jun in
the liv
er
. EMBO J 2002; 21:1782–90.
24. Weston CR, Wong A, Hall JP, Goad ME, Flavell RA, Davis RJ. The c-Jun NH2-terminal
kinase is essential for epidermal gr
o
wth factor expr
ession during epidermal morphogenesis.
P
r
oc N
atl A
cad Sci USA 2004; 101:14114-9.
25. MacCorkle RA, Tan TH. Inhibition of JNK2 disrupts anaphase and produces aneuploidy
in mammalian cells. J B
iol Chem 2004; 279:40112
-
21.
JNK2
... In the diabetic state, testosterone can phosphorylate and activate C-jun (a molecule that functions in renal inflammation) (112)(113)(114). Activated C-jun may upregulate monocyte chemoattractant protein-1 (MCP-1) expression. ...
Article
Full-text available
Diabetic nephropathy (DN) is the most common microvascular complication in diabetes and one of the leading causes of end-stage renal disease. The standard treatments for patients with classic DN focus on blood glucose and blood pressure control, but these treatments can only slow the progression of DN instead of stopping or reversing the disease. In recent years, new drugs targeting the pathological mechanisms of DN (e.g., blocking oxidative stress or inflammation) have emerged, and new therapeutic strategies targeting pathological mechanisms are gaining increasing attention. A growing number of epidemiological and clinical studies suggest that sex hormones play an important role in the onset and progression of DN. Testosterone is the main sex hormone in males and is thought to accelerate the occurrence and progression of DN. Estrogen is the main sex hormone in females and is thought to have renoprotective effects. However, the underlying molecular mechanism by which sex hormones regulate DN has not been fully elucidated and summarized. This review aims to summarize the correlation between sex hormones and DN and evaluate the value of hormonotherapy in DN.
... As a member of MAPK protein family, MAPK9 plays a part in multiples signals of mammalian cells [24] and is found improperly activated or inactivated in cells of different cancer types. For example, MAPK9 is a contributor to the development and progression of glioma, prostate carcinoma or lung carcinoma [25], but acts as a negative regulator in cellular proliferation [26]. Our findings verified the negatively regulatory function of MAPK9 mediated by TMED3 in EC cells. ...
Article
Full-text available
Background As one of gynecologic tumors, endometrial carcinoma (EC) has been characterized by high incidence rate, but its molecular pathogenesis has remained unclear. TMED3 is a membrane protein and has been indicated to implicate several tumor-related diseases. In the current study, we aimed to explore the physiological function of TMED3 in EC progression. Methods Through bioinformatic analysis using The Cancer Genome Atlas database and immunohistochemistry assay on tissue microarray, we examined whether TMED3 was upregulated in EC tissues. After constructing TMED3-knockdown cell models via lentiviral transfection, qPCR and western blot were employed to determine the expression levels of TMED3 mRNA and protein. Then, Celigo cell counting assay, CCK8 assay, flow cytometry, wound-healing assay and Transwell assay were used to detect cell proliferation, cell cycle, cell apoptosis and cell migration, respectively. Results As a result, it was found that TMED3 was upregulated in EC cells, which was also verified in clinical samples. We then found that downregulation of TMED3 considerably restrained cell cycle, cell growth and migration but promoted apoptosis of EC cells. The following in-vivo experiments also verified that tumor growth was inhibited after TMED3 knockdown. The exploration in molecular mechanisms showed that TMED3 deletion may weaken cellular viability through upregulating pro-apoptotic proteins and targeting PI3K/AKT signaling pathways. Conclusions This study suggested that knocking down TMED3 affected the malignant phenotype of EC cells and thus limited tumor progression, which provided insights to the development of targeted drugs for EC treatment.
... In this study, vaginal LPS preconditioning of the fetal brain activated the P38 pathway, whereas JNK was activated by amniotic LPS (Figure 4A,B). JNK2 has been considered as a negative regulator of cell proliferation signaling [53] and mainly a response to LPS in microglia [54]. Our data showed that VAF more reduced the JNK2 activity than AF ( Figure 4A-C). ...
Article
Full-text available
Chorioamnionitis (CAM) is an increasingly common disease affecting pregnant women which derives from bacterial vaginosis. In different clinical cases, it has been shown that CAM can cause multiple risk factors for fetal brain damage, such as infection, and intra-uterine asphyxia. However, the molecular mechanism remains unknown. In this study, we established a novel CAM mouse model by exposing pregnant mice to a combination of three risk factors: vaginal lipopolysaccharides (LPS), amniotic LPS, and ischemic reperfusion. We found amniotic LPS caused Parkinson’s disease-like fetal brain damage, in a dose and time-dependent manner. Moreover, the mechanism of this fetal brain damage is apoptosis induced by amniotic LPS but it was inhibited by being pretreated with a vaginal LPS challenge before amniotic LPS injection. In contrast, amniotic LPS with continuous ischemic reperfusion caused a higher level of apoptotic cell death than amniotic LPS alone. In particular, a potential neuroprotective biomarker phosphorylation (p)-CREB (ser133) appeared in only vaginal LPS preconditioned before amniotic LPS, whereas ischemic reperfusion triggered IKK phosphorylation after amniotic LPS. Despite the need for many future investigations, this study also discussed a developed understanding of the molecular mechanism of how these phenotypes occurred.
... The expression of the target genes MAPK9, CREBBP and EGLN3 could be regulated by the miRNAs chi-miR-15b-5p, chi-miR-16b-5p, chi-miR-17-5p, chi-miR-20a-5p, chi-miR-93-5p, and miR-142-x, which were highly expressed in the SMGs of 1-month-old kids and expressed at low levels in the SMGs of 12-month-old unbred adolesent goats and 24month-old adult goats. JNK2 (encoded by MAPK9) is implicated in apoptosis (Sabapathy and Wagner, 2004;Dhanasekaran and Reddy, 2008;Wang et al., 2010). EGLN3 and CREBBP also play important roles in the occurrence and development of disease, and EGLN3 can promote cell apoptosis (Lee et al., 2005). ...
Article
Full-text available
Submandibular glands (SMGs) are one of the primary components of salivary glands in goats. The proteins and biologically active substances secreted by the SMGs change with growth and development. Our previous studies showed that most of the differentially expressed genes in the SMGs of goats at different developmental stages are involved in immune-related signaling pathways, but the miRNA expression patterns in the same tissues are unknown. The aim of this study was to reveal the expression profile of miRNAs at three different developmental stages, detect differentially expressed miRNAs (DE miRNAs) and predict disease-related DE miRNAs. SMG tissue samples were collected from groups of 1-month-old kids, 12-month-old maiden goats and 24-month-old adult goats (three samples from each group), and high-throughout transcriptome sequencing was conducted. A total of 178, 241 and 7 DE miRNAs were discovered between 1-month-old kids and 12-month-old maiden goats, between 1-month-old kids and 24-month-old adult goats, and between 12-month-old maiden goats and 24-month-old adult goats, respectively. Among these DE miRNAs, 88 DE miRNAs with medium or high expression levels (TPM ≥50) were classified into five expression pattern clusters. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses indicated that some of the predicted target genes of the DE miRNAs in the five clusters were enriched in disease-related GO terms and pathways. MiRNA target genes in significant pathways were significantly enriched in Hepatitis B (FDR = 9.03E-10) and Pathways in cancer (FDR = 4.2E-10). Further analysis was performed with a PPI network, and 10 miRNAs were predicted to play an important role in the occurrence and prevention of diseases during the growth and development of goats.
... PHD1 has further been shown to undergo phosphorylation in response to JNK2 activation [97]. JNK2 is also a regulator of the cell cycle [98]. Phosphorylation of serine 74 and serine 162 was detected in breast cancer cells by mass spectrometry. ...
Article
Full-text available
The cell cycle is an important cellular process whereby the cell attempts to replicate its genome in an error-free manner. As such, mechanisms must exist for the cell cycle to respond to stress signals such as those elicited by hypoxia or reduced oxygen availability. This review focuses on the role of transcriptional and post-transcriptional mechanisms initiated in hypoxia that interface with cell cycle control. In addition, we discuss how the cell cycle can alter the hypoxia response. Overall, the cellular response to hypoxia and the cell cycle are linked through a variety of mechanisms, allowing cells to respond to hypoxia in a manner that ensures survival and minimal errors throughout cell division.
... MAPK9 was found to be abnormally activated in multiple caner types, including glioma, lung carcinoma, lymphoblastic leukemia and prostate carcinoma, which indicated that MAPK9 critically contributes to cancer progression and development (42). Conversely, in certain cancers, it has reported that MAPK9 plays a role as a negative regulator in cell proliferation via activation of the JNK pathway (43,44). Fan et al (45), for example, showed that specifically inhibiting JNK1/2 expression suppressed the vinblastine-induced phosphorylation of the anti-apoptotic proteins Bcl-2 and Bcl-X L , leading to inactivation of Bcl-2 and Bcl-X L , which may explain why Bcl-2 protein was significantly downregulated in shTMED3 cells in the present study. ...
Article
Full-text available
Chordoma is a rare low‑grade tumor of the axial skeleton. Over previous decades, a range of targeted drugs have been used for treating chordoma, with more specific and effective therapies under investigation. Transmembrane Emp24 protein transport domain containing 3 (TMED3) is a novel gene reported to be a regulator of oncogenesis, cancer development and metastasis; however, its role in chordoma remains unclear. In the present study, the expression of TMED3 was investigated in chordoma cells, and the effect of TMED3 knockdown on chordoma development was examined in vitro and in vivo, followed by exploration of differentially expressed proteins in TMED3‑silenced chordoma cells via an apoptosis antibody array. Reverse transcription‑quantitative PCR and western blot assays were performed to determine the expression levels. It was revealed that TMED3 was highly expressed in chordoma, and that knockdown of TMED3 inhibited cell viability and migration, and enhanced the apoptosis of chordoma cells. Additionally, knockdown of TMED3 inhibited the expression of Bcl‑2, heat shock protein 27, insulin‑like growth factor (IGF)‑I, IGF‑II, IGF binding protein‑2, Livin, Akt, CDK6 and cyclin D1 proteins, whereas MAPK9 was upregulated. Furthermore, a xenograft nude mice model demonstrated that TMED3 expression promoted tumor growth. Collectively, the present findings suggested that knockdown of TMED3 inhibited cell viability and migration, and enhanced apoptosis in chordoma cells, and that TMED3 may be a novel target for chordoma therapy.
... JNK1 and JNK2 are expressed ubiquitously throughout the entire body. It is well known that isoforms JNK1 and JNK2 play different roles in regulating c-Jun expression and cell proliferation [19,20]. By contrast, the expression pattern of JNK3 is limited and observed mainly in the brain, heart, and testes [18]. ...
Article
Full-text available
JNK is a protein kinase, which induces transactivation of c-jun. The three isoforms of JNK, JNK1, JNK2, and JNK3, are encoded by three distinct genes. JNK1 and JNK2 are expressed ubiquitously throughout the body. By contrast, the expression of JNK3 is limited and observed mainly in the brain, heart, and testes. Concerning the biological properties of JNKs, the contribution of upstream regulators and scaffold proteins plays an important role in the activation of JNKs. Since JNK signaling has been described as a form of stress-response signaling, the contribution of JNK3 to pathophysiological events, such as stress response or cell death including apoptosis, has been well studied. However, JNK3 also regulates the physiological functions of neurons and non-neuronal cells, such as development, regeneration, and differentiation/reprogramming. In this review, we shed light on the physiological functions of JNK3. In addition, we summarize recent advances in the knowledge regarding interactions between JNK3 and cellular reprogramming.
... However, in the current stage, our results could not distinguish between the effects of glycofullerene treatment on the individual regulatory roles of ERK1 and 2 in PM-exposed HaCaT cells, due to the similarity between the regulation patterns of ERK1 and 2 after PM and glycofullerenes treatment. Additionally, JNK-1 and -2 were also shown to play different roles in various conditions [53]. Under normal conditions, in several cells and animal models (such as the proliferation of keratinocytes in the mouse skin), JNK1 acts as a positive regulator, while JNK2 exhibits a negative or downregulatory function [54]. ...
Article
Full-text available
Exposure to particulate matter (PM) has been linked to pulmonary and cardiovascular dysfunctions, as well as skin diseases, etc. PM impairs the skin barrier functions and is also involved in the initiation or exacerbation of skin inflammation, which is linked to the activation of reactive oxygen species (ROS) pathways. Fullerene is a single C60 molecule which has been reported to act as a good radical scavenger. However, its poor water solubility limits its biological applications. The glyco-modification of fullerenes increases their water solubility and anti-bacterial and anti-virus functions. However, it is still unclear whether it affects their anti-inflammatory function against PM-induced skin diseases. Hence, glycofullerenes were synthesized to investigate their effects on PM-exposed HaCaT human keratinocytes. Our results showed that glycofullerenes could reduce the rate of PM-induced apoptosis and ROS production, as well as decrease the expression of downstream mitogen-activated protein kinase and Akt pathways. Moreover, PM-induced increases in inflammatory-related signals, such as cyclooxygenase-2, heme oxygenase-1, and prostaglandin E2, were also suppressed by glycofullerenes. Notably, our results suggested that PM-induced impairment of skin barrier proteins, such as filaggrin, involucrin, repetin, and loricrin, could be reduced by pre-treatment with glycofullerenes. The results of this study indicate that glycofullerenes could be potential candidates for treatments against PM-induced skin diseases and that they exert their protective effects via ROS scavenging, anti-inflammation, and maintenance of the expression of barrier proteins.
... However, a previous study suggests that JNK2 might be more important than the JNK1 in skin diseases such as human squamous cell carcinoma [58]. Furthermore, JNK2 −/− mice showed a greater resistance to chemically-induced skin cancer than JNK1 −/− mice [59], which might imply that JNK2 is more sensitive to some chemical agents-induced skin damage, but the details need further investigation. In addition, the functions of Akt are associated with skin aging [60] and are also known to play a prominent role in the healing of wounds and tissue regeneration [61,62]. ...
Article
Full-text available
Particulate matter (PM), a widespread air pollutant, consists of a complex mixture of solid and liquid particles suspended in air. Many diseases have been linked to PM exposure, which induces an imbalance in reactive oxygen species (ROS) generated in cells, and might result in skin diseases (such as aging and atopic dermatitis). New techniques involving nanomedicine and nano-delivery systems are being rapidly developed in the medicinal field. Fullerene, a kind of nanomaterial, acts as a super radical scavenger. Lower water solubility levels limit the bio-applications of fullerene. Hence, to improve the water solubility of fullerene, while retaining its radical scavenger functions, a fullerene derivative, fullerenol C60(OH)36, was synthesized, to examine its biofunctions in PM-exposed human keratinocyte (HaCaT) cells. The PM-induced increase in ROS levels and expression of phosphorylated mitogen-activated protein kinase and Akt could be inhibited via fullerenol pre-treatment. Furthermore, the expression of inflammation-related proteins, cyclooxygenase-2, heme oxygenase-1, and prostaglandin E2 was also suppressed. Fullerenol could preserve the impaired state of skin barrier proteins (filaggrin, involucrin, repetin, and loricrin), which was attributable to PM exposure. These results suggest that fullerenol could act against PM-induced cytotoxicity via ROS scavenging and anti-inflammatory mechanisms, and the maintenance of expression of barrier proteins, and is a potential candidate compound for the treatment of skin diseases.
Chapter
Injury and growth stimulation both remarkably increase the hepatic expression of Gadd45β. This contrasts with expression in liver cancer, where promoter methylation frequently silences Gadd45β, due to a suppressive function that is often proapoptotic. In normal hepatocytes, Gadd45β facilitates cell survival, growth, and proliferation. Gadd45β binds MKK7-downstream of TNFα and its receptors-to prevent this kinase from activating JNK2. Hence, the Gadd45β-/- genotype increases cell injury and decreases cell proliferation during liver regeneration (compensatory growth and proliferation). Liver hyperplasia (de novo growth and proliferation) is an alternate form of growth, caused by drugs that activate the nuclear receptor, CAR. As in regeneration, the Gadd45β-/- genotype considerably slows growth during hyperplasia. However, there is no injury and the slowing occurs because Gadd45β normally binds to CAR and activates its transcriptional stimulation. Thus, Gadd45β protects the liver through two entirely different processes: Binding MKK7 to block damaging signal transduction, or binding CAR to coactivate anabolic transcription.
Article
Full-text available
The transcriptional activity of c-Jun is augmented through phosphorylation at two sites by a c-Jun amino-terminal kinase (JNK). All cells express two distinct JNK activities, 46 and 55 kD in size. It is not clear which of them is the more important c-Jun kinase and how they specifically recognize c-Jun. The 46-kD form of JNK was identified as a new member of the MAP kinase group of signal-transducing enzymes, JNK1. Here, we report the molecular cloning of the 55-kD form of JNK, JNK2, which exhibits 83% identity and similar regulation to JNK1. Despite this close similarity, the two JNKs differ greatly in their ability to interact with c-Jun. JNK2 binds c-Jun approximately 25 times more efficiently than JNK1, and as a result has a lower Km toward c-Jun than JNK1. The structural basis for this difference was investigated and traced to a small beta-strand-like region near the catalytic pocket of the enzyme. Modeling suggests that this region is solvent exposed and therefore is likely to serve as a docking site that increases the effective concentration of c-Jun near JNK2. These results explain how two closely related MAP kinases can differ in their ability to recognize specific substrates and thereby elicit different biological responses.
Article
Full-text available
The JNK protein kinase is a member of the MAP kinase group that is activated in response to dual phosphorylation on threonine and tyrosine. Ten JNK isoforms were identified in human brain by molecular cloning. These protein kinases correspond to alternatively spliced isoforms derived from the JNK1, JNK2 and JNK3 genes. The protein kinase activity of these JNK isoforms was measured using the transcription factors ATF2, Elk-1 and members of the Jun family as substrates. Treatment of cells with interleukin-1 (IL-1) caused activation of the JNK isoforms. This activation was blocked by expression of the MAP kinase phosphatase MKP-1. Comparison of the binding activity of the JNK isoforms demonstrated that the JNK proteins differ in their interaction with ATF2, Elk-1 and Jun transcription factors. Individual members of the JNK group may therefore selectively target specific transcription factors in vivo.
Article
The c-Jun NH2-terminal kinase (JNK) is activated when cells are exposed to ultraviolet (UV) radiation. However, the functional consequence of JNK activation in UV-irradiated cells has not been established. It is shown here that JNK is required for UV-induced apoptosis in primary murine embryonic fibroblasts. Fibroblasts with simultaneous targeted disruptions of all the functional Jnk genes were protected against UV-stimulated apoptosis. The absence of JNK caused a defect in the mitochondrial death signaling pathway, including the failure to release cytochrome c. These data indicate that mitochondria are influenced by proapoptotic signal transduction through the JNK pathway.
Article
The c-Jun NH2-terminal kinase (JNK) is a member of an evolutionarily conserved sub-family of mitogen-activated protein (MAP) kinases. Recent studies have led to progress towards understanding the physiological function of the JNK signaling pathway, including the analysis of the phenotype of knockout mice. An important role for JNK in the non-canonical Wnt-signaling pathway has been established. Insight into the role of scaffold proteins that may assemble functional JNK modules has been achieved. In addition, a small molecule pharmacological inhibitor of JNK has been described and it is likely that this drug will facilitate future studies of JNK function.
Article
Recent advances indicate a link between tumour promoters, transformation, and AP-1 activity. Protein kinase C activation increases AP-1 DNA-binding activity independently of new protein synthesis. AP-1 is also stimulated by transforming oncoproteins and growth factors. These proteins are thought to participate in a signalling cascade affecting the nuclear AP-1 complex composed of the Jun and Fos proteins. Because c-Jun is the most potent transactivator in the AP-1 complex and is elevated in Ha-ras-transformed cells, in which c-Fos is downregulated, we focused on it as a potential target. c-Jun could convert input from an oncogenic signalling cascade into changes in gene expression. Indeed, transformation of rat embryo fibroblasts by c-Jun requires an intact transcriptional activation domain and cooperation with oncogenic Ha-ras. Expression of oncogenic Ha-ras augments transactivation by c-Jun and stimulates its phosphorylation. Here we describe the mapping of the Ha-ras-responsive phosphorylation sites to serines 63 and 73 of c-Jun. Site-directed mutagenesis indicates that phosphorylation of these serines is essential for stimulation of c-Jun activity and for cooperation with Ha-ras in ocogenic transformation.
Article
The c-fos serum response element (SRE) forms a ternary complex with the transcription factors SRF (serum response factor) and TCF (ternary complex factor). By itself, SRF can mediate transcriptional activation induced by serum, lysophosphatidic acid, or intracellular activation of heterotrimeric G proteins. Activated forms of the Rho family GTPases RhoA, Rac1, and CDC42Hs also activate transcription via SRF and act synergistically at the SRE with signals that activate TCF. Functional Rho is required for signaling to SRF by several stimuli, but not by activated CDC42Hs or Rac1. Activation of the SRF-linked signaling pathway does not correlate with activation of the MAP kinases ERK, SAPK/JNK, or MPK2/p38. Functional Rho is required for regulated activity of the c-fos promoter. These results establish SRF as a nuclear target of a novel Rho-mediated signaling pathway.
Article
c-Jun amino-terminal kinases (JNKs) and mitogen-activated protein kinases (MAPKs) are closely related; however, they are independently regulated by a variety of environmental stimuli. Although molecules linking growth factor receptors to MAPKs have been recently identified, little is known about pathways controlling JNK activation. Here, we show that in COS-7 cells, activated Ras effectively stimulates MAPK but poorly induces JNK activity. In contrast, mutationally activated Rac1 and Cdc42 GTPases potently activate JNK without affecting MAPK, and oncogenic guanine nucleotide exchange factors for these Rho-like proteins selectively stimulate JNK activity. Furthermore, expression of inhibitory molecules for Rho-related GTPases and dominant negative mutants of Rac1 and Cdc42 block JNK activation by oncogenic exchange factors or after induction by inflammatory cytokines and growth factors. Taken together, these findings strongly support a critical role for Rac1 and Cdc42 in controlling the JNK signaling pathway.
Article
Monoclonal antibody 3F12 identifies a cytoplasmic antigen of 49 kDa in human hippocampus and neocortex. The distribution of 3F12 immunoreactive neurons closely matches that of Alzheimer's disease (AD) targeted neurons in these areas. In some hippocampal neurons of AD patients, this antigen colocalizes with ALZ-50, indicating the presence of AD pathology in these neurons. Molecular characterization of the 3F12 cDNA revealed it to be a member of the MAP kinase family, showing 43% amino acid sequence identity to human extracellular related kinase 2 (p42mapk). We have confirmed that p493F12 kinase autophosphorylates both threonine and tyrosine residues, as expected for a MAP kinase. The p49 mRNA is expressed exclusively in the nervous system. In the brain, the distribution of these neurons closely corresponds to 3F12 antigen-bearing neurons. The p493F12 gene maps to the human chromosome 21q21 region, a region that may be important in the pathogenesis of AD and Down's syndrome.
Article
The ultraviolet (UV) response of mammalian cells is characterized by a rapid and selective increase in gene expression mediated by AP-1 and NF-kappa B. The effect on AP-1 transcriptional activity results, in part, from enhanced phosphorylation of the c-Jun NH2-terminal activation domain. Here, we describe the molecular cloning and characterization of JNK1, a distant relative of the MAP kinase group that is activated by dual phosphorylation at Thr and Tyr during the UV response. Significantly, Ha-Ras partially activates JNK1 and potentiates the activation caused by UV. JNK1 binds to the c-Jun transactivation domain and phosphorylates it on Ser-63 and Ser-73. Thus, JNK1 is a component of a novel signal transduction pathway that is activated by oncoproteins and UV irradiation. These properties indicate that JNK1 activation may play an important role in tumor promotion.