ArticlePDF Available

IRAS, the human homologue of Nischarin, prolongs survival of transfected PC12 cells

Authors:

Figures

Western blotting of hIRAS-transfected PC12 cell membrane and cytosolic fractions. Immunorevelation was performed using 1201 antiserum specific for human IRAS1 with enhanced chemiluminescence. Memb, membranes; cyt, cytosol. (b) Effect of hIRAS transfection on caspase-3 activity in serum-deprived cells. PC12 cells transfected with empty vector (pcDNA3.1) and hIRAS-transfected cells (IE10 subclone) were grown in complete (open symbols) or serum-free (closed symbols) medium. At the indicated times, cell lysates were prepared and used to determine caspase enzymatic activities by measuring the release of the para-nitroaniline chromophore from peptide substrate (DEVD) selective for caspase-3-like protease. Background levels were determined by parallel incubations of samples in the presence of caspase-3-like inhibitor, Ac-DEVD-CHO (10 M) and subtracted for each individual value. Enzymatic activity is expressed in arbitrary units as O.D/g protein for each sample. Data are the meanS.E.M. of triplicate determinations from three experiments. *P<0.05 compared to hIRAS-transfected cells. (c) PC12 cells either expressing hIRAS (IE10, 7D5, and 6E7 sublines) or non-expressing hIRAS (pcDNA3.1 and 7D5a sublines) were grown in serum-free medium for 6 and 24 h. Caspase-3 activities were measured as in (b) Results are expressed as percent activity recorded in pcDNA3.1 cells for each experiment. MeanS.E.M. of three to five independent experiments. *P<0.03 as compared to pcDNA3.1 cell line. (d) hIRAS-mediated protection of apoptosis measured by Annexin V-FITC staining and flow cytometry. Upper two panels, control cells or hIRAS-expressing cells were grown for 48 h in serum-free medium before harvesting and analysis. Percentage values represent late apoptosis (upper right quadrant) and early apoptosis (lower right quadrant) populations of cells. In each case, a representative experiment out of three is shown. Lower panel: results are meanS.E.M. of three independent experiments. The late and early apoptotic cells were taken together for this purpose. (e) Survival curves of empty vector and hIRAS-transfected cells. PC12 cells stably transfected with empty vector (pcDNA3.1 subclone) or hIRAS (IE10 subclone) were recovered after 24, 48, 72, and 96 h in serum-free medium and viable cells excluding trypan blue were counted. Results are expressed as percent of control at time 0 h for each cell line. Data shown are meanS.E.M. of triplicate determinations from three to six independent experiments. *P<0.05 as compared to pcDNA3.1 cells. (f) Upper graph: Effect of hIRAS transfection on staurosporine-induced caspase-3 activation. PC12 cells not expressing hIRAS (pcDNA3.1 and 7D5a sublines) or expressing hIRAS (IE10 and 7D5 sublines) were grown in complete medium (15% serum) in the absence (open bars) or presence (closed bars) of staurosporine (1 M) for 6 h. Results are expressed as the meanS.E.M. of three independent experiments each performed in triplicate. Lower graph: Effect of hIRAS transfection on thapsigargin-induced caspase-3 activation. PC12 cells transfected with empty vector (pcDNA3.1 subline) and IRAS-transfected cells (IE10 subline) were grown in 5% serum-containing medium with thapsigargin (100 nM) for 6 h. Results are expressed as the meanS.E.M. of two independent experiments each performed in sextuplicate. *P<0.01 as compared to pcDNA3.1 cells.
… 
Content may be subject to copyright.
Letter to the Editor
IRAS, the human homologue of Nischarin, prolongs
survival of transfected PC12 cells
Cell Death and Differentiation (2003) 10, 933–935. doi:10.1038/sj.cdd.4401275
Dear Editor,
IRAS was discovered only recently
1
and is a highly unique
protein with little structural similarity to known proteins listed in
sequence databases. Nischarin was cloned as the mouse
homologue of IRAS.
2
The amino-acid sequence of Nischarin
is about 80% homologous with IRAS, with a main difference
being that Nischarin lacks the N-terminal 244 amino acids of
IRAS.
1,2
Nischarin was reported
2
to interact with the
cytoplasmic tail of the integrin a5subunit of the fibronectin
receptor, and to inhibit cell migration and lamellipodia
formation in transfected 3T3 cells. The human protein IRAS
(hIRAS) was also found to interact with insulin receptor
substrates (IRS) in HEK 293 cells and to enhance IRS4-
dependent insulin activation of extracellularly regulated
kinase, ERK1/2.
3
Herein, we report a new cellular role for
IRAS, which is to allow PC12 cells to experience markedly
less caspase-3 enzyme activity induced by apoptotic stimuli
such as serum deprivation, thapsigargin, or staurosporine
treatment. This decrease in apoptotic effector enzyme activity
was accompanied by a decrease in the proportion of apoptotic
cells presumably due to an increase in the delay to initiate
apoptosis.
IRAS and Nischarin have been shown to exist in multiple
tissues and cell lines.
1-4
Rat IRAS is endogenously expressed
in PC12 cells (Genbank EST 106159) as a 210 kDa
immunoreactive protein (Figure 1a), which is similar in size
to the full-length form of Nischarin already detected in
neuronal mouse cells.
2
Three clonal sublines (IE10, 6E7,
and 7D5) were selected by transfecting hIRAS cDNA into
PC12 cells, isolating single cells, and propagating under
geneticin selection. In addition, two empty-vector clonal
sublines (pcDNA3.1 and 7D5a) were isolated in parallel by
the same procedure. Cytologically, the cell lines were
indistinguishable under phase contrast microscopy. Transfec-
tion of hIRAS into PC12 cells led to the appearance of the
167 kDa human protein that was predominantly localized in
the cytosol of cells (Figure 1a). The exact relation between the
210 kDa rat IRAS (rIRAS) and the 167 kDa hIRAS protein is
not fully known, since the rat homologue has only been
partially sequenced (EST 106159). Based on rat and human
tissue Northern blot analysis,
4
it has been suggested that
alternative splicing may be responsible for two molecular
weight forms of this protein. If we assume that the two proteins
play the same role in PC12 cells, then hIRAS-expressing PC12
cells constitute a moderate overexpression model of IRAS.
IRAS and Nischarin exhibit 100% amino-acid homology
over an amino-acid sequence domain identified to bind a5-
integrin.
2
The integrins, particularly the fibronectin receptor,
have been linked to cell survival pathways. Overexpression of
the a5 subunit protects cell lines against apoptotic stimuli, in
part by modulating the expression of the antiapoptotic protein
Bcl2 by activating the PI3kinase/Akt pathway.
5,6
In this
context, we hypothesized that IRAS might also modulate the
survival of cells. An increase in caspase-3 activity is an index
of apoptosis. We therefore measured the activities of
caspase-3-like proteases after serum deprivation and com-
pared the hIRAS-expressing and -nonexpressing cell lines.
No differences in caspase-3 basal activities could be
observed between the five cell lines grown in 15% serum-
containing medium. However, by 3 h of serum starvation,
caspase-3-like activities had increased dramatically in the
control cells, attaining their plateau by 6 h (5.9-fold increase;
Figure 1b). By comparison, caspase-3 activities in hIRAS-
transfected cells (i.e., IE10 clone) were increased four-fold by
6 h of serum starvation, but remained statistically less than
those of control cells (Figure 1b) at least until 24 h in serum-
free medium. As shown in Figure 1c, after 6 and 24 h of serum
deprivation, higher levels of enzymatic activity were found in
both control cell lines (pcDNA3.1 and 7D5a) compared to all
hIRAS-expressing cell lines (IE10, 7D5 and 6E7 lines). Assay
of Annexin V-FITC binding followed by flow cytometric
measurements was next performed to evaluate quantitatively
the apoptotic population in hIRAS-expressing and -nonex-
pressing cell lines. As shown in Figure 1d, about 60% of
control cells (pcDNA3.1 and 7D5a clones) appeared apoptotic
(Annexin V-FITC positive, lower right quadrant) after 48 h in
serum-free medium. Under the same conditions, hIRAS-
expressing sublines (IE10, 7D5, and 6E7) showed a marked
reduction in apoptotic populations (Figure 1d). Thus, reduced
caspase-3 activation in serum-deprived hIRAS-expressing
cells seemed to correlate with a reduction in the apoptotic cell
population. Next, cell growth rate and survival in serum-
containing medium or in serum-free medium, respectively,
were assessed microscopically. In this set of experiments, the
number of viable cells were recorded by counting trypan blue
excluding cells with a hemocytometer after different periods of
time. The growth curves indicated that hIRAS-expressing
cells behave similarly as control cells in serum-containing
medium (data not shown). However, serum deprivation
resulted in the death of 45% control cells within 24 h versus
that of 0% hIRAS-expressing IE10 cells (Figure 1e). After 72 h
serum deprivation, all the hIRAS-expressing clones (IE10,
7D5, and 6E7) showed a greater proportion of viable cells as
assessed by trypan blue exclusion than control cell lines
(pcDNA3.1 and 7D5a) (data not shown). Thus, another main
Cell Death and Differentiation (2003) 10, 933–935
&
2003 Nature Publishing Group All rights reserved 1350-9047/03
$
25.00
www.nature.com/cdd
Figure 1 (a) Western blotting of hIRAS-transfected PC12 cell membrane and cytosolic fractions. Immunorevelation was performed using 1201 antiserum specific for
human IRAS
1
with enhanced chemiluminescence. Memb, membranes; cyt, cytosol. (b) Effect of hIRAS transfection on caspase-3 activity in serum-deprived cells. PC12
cells transfected with empty vector (pcDNA3.1) and hIRAS-transfected cells (IE10 subclone) were grown in complete (open symbols) or serum-free (closed symbols)
medium. At the indicated times, cell lysates were prepared and used to determine caspase enzymatic activities by measuring the release of the para-nitroaniline
chromophore from peptide substrate (DEVD) selective for caspase-3-like protease. Background levels were determined by parallel incubations of samples in the
presence of caspase-3-like inhibitor, Ac-DEVD-CHO (10 mM) and subtracted for each individual value. Enzymatic activity is expressed in arbitrary units as O.D/mg protein
for each sample. Data are the mean7S.E.M. of triplicate determinations from three experiments. *Po0.05 compared to hIRAS-transfected cells. (c) PC12 cells either
expressing hIRAS (IE10, 7D5, and 6E7 sublines) or non-expressing hIRAS (pcDNA3.1 and 7D5a sublines) were grown in serum-free medium for 6 and 24 h. Caspase-3
activities were measured as in (b) Results are expressed as percent activity recorded in pcDNA3.1 cells for each experiment. Mean7S.E.M. of three to five independent
experiments. *Po0.03 as compared to pcDNA3.1 cell line. (d) hIRAS-mediated protection of apoptosis measured by Annexin V-FITC staining and flow cytometry. Upper
two panels, control cells or hIRAS-expressing cells were grown for 48 h in serum-free medium before harvesting and analysis. Percentage values represent late
apoptosis (upper right quadrant) and early apoptosis (lower right quadrant) populations of cells. In each case, a representative experiment out of three is shown. Lower
panel: results are mean7S.E.M. of three independent experiments. The late and early apoptotic cells were taken together for this purpose. (e) Survival curves of empty
vector and hIRAS-transfected cells. PC12 cells stably transfected with empty vector (pcDNA3.1 subclone) or hIRAS (IE10 subclone) were recovered after 24, 48, 72, and
96 h in serum-free medium and viable cells excluding trypan blue were counted. Results are expressed as percent of control at time 0 h for each cell line. Data shown are
mean7S.E.M. of triplicate determinations from three to six independent experiments. *Po0.05 as compared to pcDNA3.1 cells. (f) Upper graph: Effect of hIRAS
transfection on staurosporine-induced caspase-3 activation. PC12 cells not expressing hIRAS (pcDNA3.1 and 7D5a sublines) or expressing hIRAS (IE10 and 7D5
sublines) were grown in complete medium (15% serum) in the absence (open bars) or presence (closed bars) of staurosporine (1 mM) for 6 h. Results are expressed as
the mean7S.E.M. of three independent experiments each performed in triplicate. Lower graph: Effect of hIRAS transfection on thapsigargin-induced caspase-3
activation. PC12 cells transfected with empty vector (pcDNA3.1 subline) and IRAS-transfected cells (IE10 subline) were grown in 5% serum-containing medium with
thapsigargin (100 nM) for 6 h. Results are expressed as the mean7S.E.M. of two independent experiments each performed in sextuplicate. *Po0.01 as compared to
pcDNA3.1 cells.
Letter to the Editor
934
Cell Death and Differentiation
difference between control cells and hIRAS-expressing cell
lines is prolonged survival of the latter in serum-free
conditions, which may be related to the decrease in
caspase-3 activity and apoptotic cell population observed
previously. When serum is readded after 24 h serum depriva-
tion, we observed that hIRAS-expressing cell lines proved
able to proliferate again in marked contrast with control cells
that continued to die (56715% and 250767% of trypan blue
excluding cells after 72 h in serum-containing medium for
control and hIRAS-expressing cells, respectively; 100% refers
to the viable cells recorded after 24 h serum deprivation in
each cell line).
In an attempt to generalize that transfection of hIRAS
delays PC12 cells apoptosis, treatments were next performed
with two different cytotoxic agents, thapsigargin and staur-
osporine. Thapsigargin, a Ca
2 þ
-ATPase inhibitor that in-
creases intracellular calcium by inhibiting the uptake of
calcium into the endoplasmic reticulum, is known to induce
apoptosis in many cell types including PC12 cells.
7
Staur-
osporine, a protein kinase inhibitor, also induces apoptosis in
PC12 cells.
8
Staurosporine treatment of PC12 cells induces a
sustained elevation of intracellular calcium and accumulation
of reactive oxygen species.
8
Both of these apoptotic stimuli
are known to trigger caspase-3 activation.
7,8
. Thapsigargin
was applied to the cells in 5% serum-containing medium, and
then caspase-3 activities were recorded. As shown in
Figure 1f, thapsigargin (100 nM) enhanced caspase-3 activity
in control cells (pcDNA3.1 clone). In contrast, no enhance-
ment by thapsigargin of caspase-3 activity was observed in
hIRAS-expressing cells (IE10 clone). Staurosporine (1 mM)
was added to complete medium (containing 15% serum), thus
excluding any impact of low serum concentration on caspase-
3 activity. In this paradigm, the sublines stably expressing
hIRAS (1E10 and 7D5) showed lower caspase-3 activation
after 6 h treatment with 1 mM staurosporine compared to
control cells (pcDNA3.1 and 7D5a clones) (Figure 1f).
Caspase-3 activity after 6 h was increased 3.6- and 8.8-fold
in control sublines (pcDNA3.1 and 7D5a, respectively) versus
2.3- and 2.8-fold in hIRAS-expressing cells (IE10 and 7D5
clones respectively). We thus have confirmed the activation of
this apoptosis effector enzyme by these two drugs, and in the
process shown that hIRAS also inhibits the increase in
caspase-3 activity in these conditions.
In summation, IRAS appears to impinge on a mechanism
central to apoptosis induced by distinct stimuli and which lies
upstream of the caspase-3 activation resulting at least in a
delayed cell death. According to the results of Figure 1e, it
seems that a continued proapoptotic stimulus (96 h serum
deprivation, for example) eventually overrides the IRAS block
of apoptosis. As readdition of serum at an earlier time point of
apoptosis triggering (after 24 h serum deprivation) allowed the
hIRAS-expressing cells to divide again, we speculate that
hIRAS lowers the probability of initiation of the apoptotic
program rather than prolonging each individual apoptotic
event as described for caspase-3 inhibitors.
9
Thus, hIRAS
blocks apoptosis before destruction of membranes and loss of
phosphatidylserine asymmetry and seems to protect the cells
from the loss of the proliferative potential. Intriguingly, it
appears from the literature that expression of the a5 integrin
subunit delays serum-deprivation-induced as well as staur-
osporine-induced apoptosis in a manner very similar to the
one obtained by the transfection of hIRAS.
10
The mechanism of action of IRAS remains unresolved at
this time and is currently under investigation. Hypotheses
have been tested according to the proteins that are known to
interact with IRAS.
2,3
IRAS may be implicated either in the
insulin or the integrin dependent survival pathway, both of
which imply PI3-kinase activation.
11,6
However, our assays to
inhibit the protective effect of IRAS with LY294002 or
wortmannin (two PI3-kinase inhibitors) were unsuccessful;
although a little enhancement of caspase-3 activity in serum-
deprived hIRAS-expressing clones was recorded, no sig-
nificant increase in the proportion of apoptotic cells could be
reached with these inhibitors (data not shown). Likewise,
PD98059, an inhibitor of the ERK1/2 pathway, was unable to
induce an increase in hIRAS-expressing apoptotic cells (data
not shown).
Our study has thus identified a new cellular role for the
recently cloned human protein IRAS. We demonstrated that
hIRAS expression in PC12 cells results in protection against
apoptosis over a 2–3-day period of time. Although the
mechanism of action of IRAS is not resolved, it may represent
a previously unknown class of protein modulating the fine
regulation of cell death and growth.
Acknowledgements
We thank Dr. Peter Eickelman (Solvay Pharmaceuticals, Hannover,
Germany) for advice with the transfection protocol, and Mary Elise Lutrick
(University of Mississippi Medical Center) for carefully selecting and
maintaining the sublines. This work was partially supported by a grant from
Solvay Pharma (Hanover, Germany) and Grant MH49248 from the
National Institute of Mental Health (USA) for J Piletz’s group.
M Dontenwill
1*
, G Pascal
2
, JE Piletz
3
, M Chen
3
,
J Baldwin
3
, P Ronde
´
1
, L Dupuy
2
, D Urosevic
2
,
H Greney
2
, K Takeda
1
and P Bousquet
2
1
Pharmacologie et Physicochimie des Interactions Cellulaires et Mole
´
culaires,
UMR 7034 CNRS, Faculte
´
de Pharmacie, Universite
´
Louis Pasteur de
Strasbourg, Illkirch, France
2
Laboratoire de Neurobiologie et Pharmacologie Cardiovasculaire, Faculte
´
de
Medecine, Strasbourg, France
3
Department of Psychiatry, Division of Neurobiology and Behavior Research,
University of Mississippi Medical Center, Jackson, MS, USA
* Corresponding author: M Dontenwill, Pharmacologie et Physicochimie des
Interactions Cellulaires et Mole
´
culaires, UMR CNRS 7034, Faculte
´
de
Pharmacie, Universite
´
Louis Pasteur de Strasbourg, Illkirch, France. Tel: þ 33
3 90244267; Fax: þ 33 3 90244313; E-mail: mdontenwill@aspirine.u-
strasbg.fr
1. Piletz et al. (2000). DNA Cell Biol 19: 319–329
2. Alahari et al. (2000). J. Cell. Biol. 151: 1141–1154
3. Sano et al. (2002). J. Biol. Chem. 277: 19439–19447
4. Ivanov et al. (1998). J. Auton. Nerv. Sys. 7: 98–110
5. Matter and Ruoslahti (2001). J Biol. Chem. 276: 27757–27763
6. Lee and Juliano (2000). Mol. Biol. Cell 11: 1973–1987
7. Takadera and Ohyashiki (1998). Biochim. Biophys. Acta 1401: 63–71
8. Kruman et al. (1998). J. Neurosci. Res. 51: 293–308
9. McCarthy et al. (1997). J. Cell. Biol. 136: 215–227
10. O’Brien et al. (1996). Exp. Cell. Res. 224: 208–213
11. Barber et al. (2001). J. Biol. Chem. 276: 32814–32821
Letter to the Editor
935
Cell Death and Differentiation
... Research interest in Nischarin/IRAS in the CNS has steadily increased since 2003 [7][8] (Fig. 1 and Table S1). Related research on Nischarin mainly focuses on the influence of CNS development [6] and the regulation of CNS diseases [9][10][11]. ...
... Nischarin up-regulation can promote neuronal apoptosis caused by oxidative stress injury [10][11], however, it has a protective effect on neurons injured by inflammatory stimuli [9]. Similarly, IRAS overexpression has also been found to have a protective effect on neuronal damage caused by starvation or pro-apoptotic factors [8,13]. In addition, Nischarin in the rostral ventrolateral medulla (RVLM) of rats negatively regulates blood pressure [14]. ...
... Nischarin/IRAS and neuronal apoptosis The relationship between Nischarin/IRAS and neuronal apoptosis was first studied in 2003 [8,13]. But unlike the pro-apoptotic effect of Nischarin/ IRAS in cancer [35], the effect of Nischarin/IRAS on neuronal apoptosis is inconclusive. ...
Article
Full-text available
Background: Murine Nischarin and its human homolog IRAS are scaffold proteins highly expressed in the central nervous system (CNS). Nischarin was initially discovered as a tumor suppressor protein, and recent studies have also explored its potential value in the CNS. Research on IRAS has largely focused on its effect on opioid dependence. Although the role of Nischarin/IRAS in the physiological function and pathological process of the CNS has gradually attracted attention and the related research results are expected to be applied in clinical practice, there is no systematic review of the role and mechanisms of Nischarin/IRAS in the CNS so far. Aim: of review: This review will systematically analyze the role and mechanism of Nischarin/IRAS in the CNS, and provide necessary references and possible targets for the treatment of neurological diseases, thereby broadening the direction of Nischarin/IRAS research and facilitating clinical translation. Key scientific concepts of review: The pathophysiological processes affected by dysregulation of Nischarin/IRAS expression in the CNS are mainly introduced, including spinal cord injury (SCI), opioid dependence, anxiety, depression, and autism. The molecular mechanisms such as factors regulating Nischarin/IRAS expression and signal transduction pathways regulated by Nischarin/IRAS are systematically summarized. Finally, the clinical application of Nischarin/IRAS has been prospected.
... In contrast to the findings that exogenous expression of NISCH in breast cancer cells suppresses cell survival, in vitro studies from the early 2000s on the function of IRAS (then considered a human homologue of mouse nischarin) support the opposite claim. Overexpression of NISCH delayed apoptosis induced by a variety of stimuli [92,93], partially through activation of the PI3 kinase pathway. Nischarin was also shown to bind insulin receptor substrate protein, activate ERK and promote survival [94]. ...
Article
Full-text available
Nischarin was reported to be a tumor suppressor that plays a critical role in breast cancer initiation and progression, and a positive prognostic marker in breast, ovarian and lung cancers. Our group has found that nischarin had positive prognostic value in female melanoma patients, but negative in males. This opened up a question whether nischarin has tumor type-specific and sex-dependent roles in cancer progression. In this study, we systematically examined in the public databases the prognostic value of nischarin in solid tumors, regulation of its expression and associated signaling pathways. We also tested the effects of a nischarin agonist rilmenidine on cancer cell viability in vitro. Nischarin expression was decreased in tumors compared to the respective healthy tissues, most commonly due to the deletions of the nischarin gene and promoter methylation. Unlike in healthy tissues where it was located in the cytoplasm and at the membrane, in tumor tissues nischarin could also be observed in the nuclei, implying that nuclear translocation may also account for its cancer-specific role. Surprisingly, in several cancer types high nischarin expression was a negative prognostic marker. Gene set enrichment analysis showed that in tumors in which high nischarin expression was a negative prognostic marker, signaling pathways that regulate stemness were enriched. In concordance with the findings that nischarin expression was negatively associated with pathways that control cancer growth and progression, nischarin agonist rilmenidine decreased the viability of cancer cells in vitro. Taken together, our study lays a ground for functional studies of nischarin in a context-dependent manner and, given that nischarin has several clinically approved agonists, provides rationale for their repurposing, at least in tumors in which nischarin is predicted to be a positive prognostic marker.
... Based on biochemical, pharmacological, and functional characterizations, imidazoline receptors were subdivided into three subtypes: I1, I2, and non-I1/I2 (I3). Imidazoline receptor antisera-selected (IRAS) protein, cloned from a human hippocampus cDNA expression library by Piletz et al. (2000), shared similarity with natural I1R in localization, physiological function, and signal transduction, so it was considered functional I1R candidate proteins (Dontenwill et al. 2003;Li et al. 2006;Piletz et al. 2000;Piletz et al. 2003;Abdel-Rahman 2006, 2008). Agmatine is the endogenous ligand of I1R and a decarboxylic product of L-arginine and has been proven to play a role in analgesia, anti-depression, anti-convulsion, neuroprotection, and so on (Uzbay 2012;Xu et al. 2018). ...
Article
Full-text available
Dexmedetomidine has been used as a sedative drug in the clinic for a long time. Many studies demonstrated that the sedative mechanism of dexmedetomidine might be related to the activation of α2-adrenoceptor (α2AR). In addition, it was reported that dexmedetomidine had some affinity for the I1-imidazoline receptor (I1R); however, the role of I1R in dexmedetomidine-induced sedative effects and its possible mechanism are poorly studied. In the present study, we found that agmatine, an I1R agonist, was able to enhance the sedative effect of dexmedetomidine in mice. Efaroxan, an α2AR and I1R antagonist, could prevent and rescue the sedative action of dexmedetomidine in mice, and its preventive effect was better than atipamezole, the specific α2AR antagonist. Knockout of imidazoline receptor antisera-selected (IRAS), the functional I1R candidate protein, suppressed the dexmedetomidine-induced sedation. Moreover, IRAS knockout led to the inhibition of agmatine and efaroxan in regulating dexmedetomidine-induced sedative effects in mice, but not of atipamezole. We then used CHO cell lines that stably expressed α2AR and IRAS to investigate the possible molecular mechanism of IRAS in regulating the dexmedetomidine-induced sedative effect. The results showed that IRAS expression significantly up-regulated dexmedetomidine-induced ERK phosphorylation, which was enhanced by agmatine and inhibited by efaroxan at low concentrations. Therefore, by taking advantage of pharmacological and genetic approaches, our finding revealed the evidence that IRAS plays an important role in the sedative effects of dexmedetomidine, and the ERK signal pathway may be involved in the mechanism of IRAS in regulating dexmedetomidine-induced sedation. This study may offer valuable insights for the advancement of novel anesthetic adjuvants.
... Rilmenidine acts via a novel mechanism wherein the newly identified imidazoline receptor, nish-1 (previously f13e9.1) in C. elegans, mediates signaling to the genetic pathways relating to CR to extend lifespan. Although the imidazoline receptor signaling had been implicated to interact with alpha/beta-integrin or insulin-dependent PI3 kinase/Akt pro-survival pathways (Dontenwill et al. 2003), the connection of the imidazoline receptor to CR or longevity had not been studied. We showed that rilmenidine-induced lifespan and healthspan extension are dependent upon the imidazoline receptor, nish-1. ...
Preprint
Full-text available
Caloric restriction increases lifespan across species and has health benefits in humans. Because complying with a low-calorie diet is challenging, here we investigated pharmacological interventions mimicking the benefits of caloric restriction. Searching for compounds that elicit a similar gene expression signature to caloric restriction, we identified rilmenidine, an I1-imidazoline receptor agonist and prescription medication for the treatment of hypertension. We then show that treating C. elegans with rilmenidine at young and older ages increases lifespan. We also demonstrate that the stress-resilience, healthspan, and lifespan benefits upon rilmenidine treatment in worms are mediated by the I1-imidazoline receptor nish-1, implicating this receptor as a potential longevity target. Furthermore, we show that rilmenidine treatment increased ERK phosphorylation via NISH-1. Consistent with the shared caloric-restriction-mimicking gene signature, supplementing rilmenidine to caloric restricted C. elegans, genetic reduction of TORC1 function, or rapamycin treatment did not further increase lifespan. The rilmenidine-induced longevity required the transcription factors FOXO/DAF-16 and NRF1,2,3/SKN-1, both important for caloric restriction-mediated longevity. Furthermore, we find that autophagy, but not AMPK signaling, was needed for rilmenidine-induced longevity. Lastly, we find that treating mice with rilmenidine showed transcriptional changes in liver and kidney similar to caloric restriction. Overall, our findings reveal rilmenidine as a caloric restriction mimetic and as a novel geroprotective compound.
... IRAS are 100% identical ( Fig. 2; ref. 14). In humans, Nischarin/ IRAS was first discovered as an I1-imidazoline receptor, which are expressed in both neurons and astrocytes (14,15). Human and mouse Nischarin differ in the alanine/proline rich region, which is removed in human Nischarin (Fig. 2). ...
Article
Tumor suppressor genes regulate cell growth and prevent spontaneous proliferation that could lead to aberrant tissue function. Deletions and mutations of these genes typically lead to progression through the cell-cycle checkpoints, as well as increased cell migration. Studies of these proteins are important as they may provide potential treatments for breast cancers. In this review, we discuss a comprehensive overview on Nischarin, a novel protein discovered by our laboratory. Nischarin, or imidazoline receptor antisera-selected protein, is a protein involved in a vast number of cellular processes, including neuronal protection and hypotension. The NISCH promoter experiences hypermethylation in several cancers, whereas some highly aggressive breast cancer cells exhibit genomic loss of the NISCH locus. Furthermore, we discuss data illustrating a novel role of Nischarin as a tumor suppressor in breast cancer. Analysis of this new paradigm may shed light on various clinical questions. Finally, the therapeutic potential of Nischarin is discussed. Cancer Res; 75(20); 1-8. ©2015 AACR.
... Nischarin was found to bind to the α5 subunit of integrins, and inhibited Rac-mediated cell motility and invasion in breast and colon epithelial cells (18)(19)(20)(21). Notably, IRAS, the human homolog of Nischarin, was described as an imidazoline receptor (22) with anti-apoptotic activity (23,24). Nischarin mRNA expression levels have been reported to be significantly higher in the brain and kidney compared with those in the heart, liver, lung and skeletal muscle (19). ...
Article
Full-text available
The present study aimed to investigate the expression of Nischarin protein in primary breast cancer (PBC), and to evaluate its role in tumor metastasis. Paired specimens of breast cancer tissues and adjacent normal tissues were surgically obtained from 60 patients with PBC at the Zhejiang Cancer Hospital (Hangzhou, China). Nischarin protein concentrations were determined by an ELISA assay. Breast cancer tissues exhibited a significantly lower concentration of Nischarin (5.86±3.19 ng/ml) compared with that of the adjacent noncancerous tissues (9.25±3.65 ng/ml; P<0.001). Furthermore, cancer tissue from patients with lymph node metastasis had significantly lower levels of Nischarin protein (4.69±2.40 ng/ml) than those of patients without lymph node metastasis (7.04±3.47 ng/ml; P=0.004). There was no significant difference in Nischarin protein expression levels between patients with grade I, II or III PBC (grade I, 5.44±3.57 ng/ml; grade II, 6.42±3.85 ng/ml and grade III, 5.10±1.18 ng/ml; P=0.765). The significant differences in the expression of Nischarin between: i) Cancer tissue and noncancerous tissue and ii) patients with and without lymph node metastasis, suggested that Nischarin may have a significant role in tumor occurrence and metastasis of breast cancer. Nischarin expression may therefore be used as a marker to predict the invasiveness and metastasis of PBC.
Preprint
Full-text available
Background: Scaffolding protein nischarin (NISCH) was reported to be a tumor suppressor that plays a critical role in breast cancer initiation and progression through regulation of the cytoskeleton dynamics. NISCH expression was reported to be a positive prognostic marker in breast, ovarian and lung cancers. Our group has found that in melanoma, NISCH had positive prognostic value in female patients, but negative in males. These findings opened up a question whether NISCH has tumor type-specific and sex-dependent roles in cancer progression. Results: In this study, we systematically examined in the public databases the prognostic value of NISCH in solid tumors, regulation of its expression and associated signaling pathways with the special emphasis on the possible differences between male and female cancer patients. We found that NISCH expression was decreased in tumor compared to the respective healthy tissues, and that this was most commonly due to the deletions of the NISCH gene and promoter methylation. We also report that, unlike in healthy tissues where it was located in the cytoplasm and at the membrane, NISCH could be observed in the nuclei in tumor tissues. Surprisingly, we found that in many cancer types – colon, liver, skin, ovarian, prostate, and kidney – high NISCH expression was a negative prognostic marker. Gene set enrichment analysis showed that, while there were common pathways associated with NISCH expression in all the examined cancer types, in tumors in which high NISCHexpression was a negative prognostic marker Wnt-Notch-Hedgehog signaling gene networks were enriched. Conclusions: Our study questions the current tumor suppressor status of nischarin and lays a ground for functional studies in a context-dependent manner in cancer.
Article
5-isothiocyanato-2-benzofuranyl-2-imidazoline (BU99006) was shown to selectively and irreversibly bind to I2-imidazoline receptors (IRs) in vitro and in vivo, but cell signalling consequences of I2-IR alkylation have not been reported previously. This study assessed by Western blot the effects of BU99006 on nischarin (candidate I1-IR), μ-OR (regulated by nischarin) and associated signalling mediators in the mouse brain. Acute treatment with BU99006 (20 mg/kg, i.p., 1–3 h) led to fast (peak at 1 h) and shortlasting (decline up to 3 h) upregulation of nischarin and μ-OR contents in the hippocampus (less or non-significant effects in cortex) and altered the expression of cytoskeletal β-actin (reduced contents at 3 h). In the same hippocampal samples of BU99006-treated mice, an inhibition of the MAPK species MEK, ERK and JNK was detected at 1 and/or 2 h after drug administration, which was paralleled by enhanced calpain activity (increased contents of p25 and spectrin breakdown products). Correlation analysis indicated the involvement of cdk5/p25 in MEK/ERK inhibition. These neurochemical effects of I2-alkylating BU99006 show a close relation between I1- and I2-IRs expressed in the mouse brain and between these receptors and the μ-OR, accompanied by cytoskeletal alterations and differential effects on multifunctional MAPK and cdk5 signalling pathways.
Article
Nischarin, a novel integrin binding protein, has been demonstrated its negative effects on cell migration and invasion. However, the biological role of Nischarin in breast cancer has not been fully elucidated yet. Our study aimed to analyze the association between Nischarin expression and clinical features of breast cancer patients, and further investigate the role of Nischarin in breast cancer cells apoptosis, migration and invasion. Results showed that Nischarin expression was significantly lower in breast cancer tissues (37.8%, 23/67) than in normal tissues (61.8%, 21/34; P < 0.05), and the expression of Nischarin significantly negatively correlated with estrogen receptor status. Similarly, Nischarin expression was highest in normal breast cell line HBL-100 while triple-negative breast cancer cell line MDA-MB-231 had the lowest expression of Nischarin. Further experiments demonstrated that overexpression of Nischarin may induce apoptosis, and inhibit cell migration and invasion. The present data confirmed that Nishcharin might be a novel tumor suppressor and plays an important role in breast cancer cell apoptosis and metastasis, which can be used as a potential therapeutic target for breast cancer treatment.
Article
Various imidazoline receptor (IR) proteins have been proposed to mediate the effects of selective I1- and I2-IR drugs. However, the association of these IR-binding proteins with classic I1- and I2-radioligand binding sites remains somewhat controversial. In this study, three IR antibodies (anti-NISCH and anti-nischarin for I1-IRs; and anti-IRBP for I1/I2-IRs) were used to immunodetect, characterize and compare IR protein patterns in brain (mouse and human; total homogenate, subcellular fractionation, grey and white matter) and some cell systems (neurones, astrocytes, human platelets). Various immunoreactive IRs (specific molecular weight bands coincidently detected with the different antibodies) were related to I1-IR (167 kDa, 105/115 kDa and 85 kDa proteins) or I2-IR (66 kDa, 45 kDa and 30 kDa proteins) types. The biochemical characterization of cortical 167 kDa protein, localized in the membrane/cytosol but not in the nucleus, indicated that this I1-IR also forms part of higher order nischarin-related complexes. The contents of I1-IR (167 kDa, 105/115 kDa, and 85 kDa) proteins in mouse brain cortex were upregulated by treatment with I1-drugs (moxonidine, efaroxan) but not with I2-drugs (BU-224, LSL 61122). Conversely, the contents of I2-IR (66 kDa, 45 kDa and 30 kDa) proteins in mouse brain cortex were modulated by treatment with I2-drugs (decreases after BU-224 and LSL 61122, and increases after idazoxan) but not with I1-drugs (with the exception of moxonidine). These findings further indicate that brain immunoreactive IR proteins exist in multiple forms that can be grouped in the already known I1- and I2-IR types, which are expressed both in neurones and astrocytes. © The Author(s) 2015.
Article
Full-text available
Renewal of the gastrointestinal epithelium involves a coordinated process of terminal differenti- ation and programmed cell death. Integrins have been implicated in the control of apoptotic processes in various cell types. Here we examine the role of integrins in the regulation of apoptosis in gastrointestinal epithelial cells with the use of a rat small intestinal epithelial cell line (RIE1) as a model. Overexpression of the integrin a5 subunit in RIE1 cells conferred protection against several proapoptotic stimuli. In contrast, overexpression of the integrin a2 subunit had no effect on cell survival. The antiapoptotic effect of the a5 subunit was partially retained by a mutated version that had a truncation of the cytoplasmic domain. The antiapoptotic effects of the full- length or truncated a5 subunit were reversed upon treatment with inhibitors of phosphatidylino- sitol 3-kinase (PI-3-kinase), suggesting that the a5b1 integrin might interact with the PI-3-kinase/ Akt survival pathway. When cells overexpressing a5 were allowed to adhere to fibronectin, there was a moderate activation of protein kinase B (PKB)/Akt, whereas no such effect was seen in a2-overexpressing cells adhering to collagen. Furthermore, in cells overexpressing a5 and adher- ing to fibronectin, there was a dramatic enhancement of the ability of growth factors to stimulate PKB/Akt; again, this was not seen in cells overexpressing a2 subunit and adhering to collagen or fibronectin. Expression of a dominant negative version of PKB/Akt in RIE cells blocked to ability of a5 to enhance cell survival. Thus, the a5b1 integrin seems to protect intestinal epithelial cells against proapoptotic stimuli by selectively enhancing the activity of the PI-3-kinase/Akt survival pathway.
Article
Full-text available
There is increasing evidence for a central role in mammalian apoptosis of the interleukin-1 beta-converting enzyme (ICE) family of cysteine proteases, homologues of the product of the nematode "death" gene, ced-3. Ced-3 is thought to act as an executor rather than a regulator of programmed cell death in the nematode. However, it is not known whether mammalian ICE-related proteases (IRPs) are involved in the execution or the regulation of mammalian apoptosis. Moreover, an absolute requirement for one or more IRPs for mammalian apoptosis has yet to be established. We have used two cell-permeable inhibitors of IRPs, Z-Val-Ala-Asp.fluoromethylketone (ZVAD.fmk) and t-butoxy carbonyl-Asp.fluoromethylketone (BD.fmk), to demonstrate a critical role for IRPs in mammalian apoptosis induced by several disparate mechanisms (deregulated oncogene expression, ectopic expression of the Bcl-2 relative Bak, and DNA damage-induced cell death). In all instances, ZVAD.fmk and BD.fmk treatment inhibits characteristic biochemical and morphological events associated with apoptosis, including cleavage of nuclear lamins and poly-(ADP-ribose) polymerase, chromatin condensation and nucleosome laddering, and external display of phosphatidylserine. However, neither ZVAD.fmk nor BD.fmk inhibits the onset of apoptosis, as characterized by the onset of surface blebbing; rather, both act to delay completion of the program once initiated. In complete contrast, IGF-I and Bcl-2 delay the onset of apoptosis but have no effect on the kinetics of the program once initiated. Our data indicate that IRPs constitute part of the execution machinery of mammalian apoptosis induced by deregulated oncogenes, DNA damage, or Bak but that they act after the point at which cells become committed to apoptosis or can be rescued by survival factors. Moreover, all such blocked cells have lost proliferative potential and all eventually die by a process involving cytoplasmic blebbing.
Article
Full-text available
The imidazoline-1 receptor (IR1) is considered a novel target for drug discovery. Toward cloning an IR1, a truncated cDNA clone was isolated from a human hippocampal lambda gt11 cDNA expression library by relying on the selectivity of two antisera directed against candidate IR proteins. Amplification reactions were performed to extend the 5' and 3' ends of this cDNA, followed by end-to-end PCR and conventional cloning. The resultant 5131-basepair molecule, designated imidazoline receptor-antisera-selected (IRAS) cDNA, was shown to encode a 1504-amino acid protein (IRAS-1). No relation exists between the amino acid sequence of IRAS-1 and proteins known to bind imidazolines (e.g., it is not an alpha2-adrenoceptor or monoamine oxidase subtype). However, certain sequences within IRAS-1 are consistent with signaling motifs found in cytokine receptors, as previously suggested for an IR1. An acidic region in IRAS-1 having an amino acid sequence nearly identical to that of ryanodine receptors led to the demonstration that ruthenium red, a dye that binds the acidic region in ryanodine receptors, also stained IRAS-1 as a 167-kD band on SDS gels and inhibited radioligand binding of native I1 sites in untransfected PC-12 cells (a source of authentic I1 binding sites). Two epitope-selective antisera were also generated against IRAS-1, and both reacted with the same 167-kD band on Western blots. In a host-cell-specific manner, transfection of IRAS cDNA into Chinese hamster ovary cells led to high-affinity I1 binding sites by criteria of nanomolar affinity for moxonidine and rilmenidine. Thus, IRAS-1 is the first protein discovered with characteristics of an IR1.
Article
Full-text available
Integrins have been implicated in key cellular functions, including cytoskeletal organization, motility, growth, survival, and control of gene expression. The plethora of integrin alpha and beta subunits suggests that individual integrins have unique biological roles, implying specific molecular connections between integrins and intracellular signaling or regulatory pathways. Here, we have used a yeast two-hybrid screen to identify a novel protein, termed Nischarin, that binds preferentially to the cytoplasmic domain of the integrin alpha5 subunit, inhibits cell motility, and alters actin filament organization. Nischarin is primarily a cytosolic protein, but clearly associates with alpha5beta1, as demonstrated by coimmunoprecipitation. Overexpression of Nischarin markedly reduces alpha5beta1-dependent cell migration in several cell types. Rat embryo fibroblasts transfected with Nischarin constructs have "basket-like" networks of peripheral actin filaments, rather than typical stress fibers. These observations suggest that Nischarin might affect signaling to the cytoskeleton regulated by Rho-family GTPases. In support of this, Nischarin expression reverses the effect of Rac on lamellipodia formation and selectively inhibits Rac-mediated activation of the c-fos promoter. Thus, Nischarin may play a negative role in cell migration by antagonizing the actions of Rac on cytoskeletal organization and cell movement.
Article
Full-text available
Integrin-mediated cell adhesion is necessary for the survival of many cell types, and loss of adhesion causes apoptosis. We have previously shown that the α5β1 integrin supports cell survival on fibronectin and increases Bcl-2 protein expression. Here we show that bcl-2 transcription is elevated in cells that attach to fibronectin through αvβ1 or to vitronectin through αvβ3 but is not elevated in cells attaching through the αvβ1 integrin. Bcl-2 protein expression and protection from apoptosis under serum-free conditions correlated withbcl-2 transcription. This integrin-mediated regulation ofbcl-2 is Shc- and FAK-dependent, and activation of Ras by FAK is required. Furthermore, Ras mediates this up-regulation of bcl-2 by activating the phosphatidylinositol 3-kinase-AKT pathway. Mitogen-activated protein kinase did not appear to be necessary for the activation of bcl-2 transcription. Therefore, our work characterizes the pathway that mediates the effect of integrins on bcl-2 transcription and cell survival.
Article
Full-text available
The ability of insulin to protect neurons from apoptosis was examined in differentiated R28 cells, a neural cell line derived from the neonatal rat retina. Apoptosis was induced by serum deprivation, and the number of pyknotic cells was counted. p53 and Akt were examined by immunoblotting after serum deprivation and insulin treatment, and caspase-3 activation was examined by immunocytochemistry. Serum deprivation for 24 h caused ∼20% of R28 cells to undergo apoptosis, detected by both pyknosis and activation of caspase-3. 10 nm insulin maximally reduced the amount of apoptosis with a similar potency as 1.3 nm (10 ng/ml) insulin-like growth factor 1, which acted as a positive control. Insulin induced serine phosphorylation of Akt, through the phosphatidylinositol (PI) 3-kinase pathway. Inhibition of PI 3-kinase with wortmannin or LY294002 blocked the ability of insulin to rescue the cells from apoptosis. SN50, a peptide inhibitor of NF-κB nuclear translocation, blocked the rescue effect of insulin, but neither insulin or serum deprivation induced phosphorylation of IκB. These results suggest that insulin is a survival factor for retinal neurons by activating the PI 3-kinase/Akt pathway and by reducing caspase-3 activation. The rescue effect of insulin does not appear to be mediated by NF-κB or p53. These data suggest that insulin provides trophic support for retinal neurons through a PI 3-kinase/Akt-dependent pathway.
Article
Full-text available
The insulin receptor substrates (IRSs) are key components in signaling from the insulin receptor, and consequently any proteins that interact with them are expected to participate in insulin signaling. In this study we have searched for proteins that interact with IRS-4 by identifying the proteins that coimmunoprecipitated with IRS-4 from human embryonic kidney 293 cells by microsequencing through mass spectrometry. A group of proteins was found. These included phosphatidylinositol 3-kinase, a protein previously identified as an IRS-4 interactor, and several proteins for which there was no previous evidence of IRS-4 association. One of these proteins, named IRAS, that had been found earlier in another context was examined in detail. The results from the overexpression of IRAS, where its amount was about the same as that of IRS-4, indicated that IRAS associated directly with IRS-4 and showed that the increased complexation of IRS-4 with IRAS did not alter the insulin-stimulated tyrosine phosphorylation of IRS-4 or the association of IRS-4 with phosphatidylinositol 3-kinase or Grb2. On the other hand, overexpression of IRAS enhanced IRS-4-dependent insulin stimulation of the extracellularly regulated kinase. The domains of IRAS and IRS-4 responsible for the association of these two proteins were identified, and it was shown that IRAS also associates with IRS-1, IRS-2, and IRS-3.
Article
It is clear that certain integrins can regulate the growth of tumors, probably by contributing to signal transduction processes. In the present study we have used HT29 human colon carcinoma cells stably transfected with human cDNA for the integrin alpha 5 subunit and studied the effects of alpha 5 expression on the induction of apoptosis. We observe that apoptosis can be triggered in HT29 cells by removal of serum and that this process can be suppressed by the stable expression of full-length integrin alpha 5 subunits. While the mechanism underlying this effect is still unclear, these observations suggest that the alpha 5 beta 1 integrin plays an important role in modulating tumor cell responses to growth factors and nutrients.
Article
Thapsigargin, an endoplasmic reticular Ca2+-ATPase inhibitor, induced apoptotic cell death (chromatin condensation and DNA fragmentation) accompanied by the activation of CPP32-like protease, a member of the interleukin-1beta converting enzyme protease (ICE) family, but not the activation of ICE-like protease. Nerve growth factor (NGF) completely inhibited the cell death and CPP32-like activation induced by thapsigargin while Ac-Asp-Glu-Val-Asp-CHO, an inhibitor of CPP32-like protease, reduced the cell death. PD98059, a specific inhibitor of Map kinase kinase, did not reduce the protective effect of NGF on thapsigargin-induced cell death. These results suggest that calcium ion-induced apoptotic cell death was mediated by CPP32-like, but not ICE-like, protease and was regulated by a neurotrophic factor possibly, through the Map kinase cascade independent pathway.
Article
The bacterial alkaloid staurosporine is widely employed as an inducer of apoptosis in many cell types including neurons. The intracellular cascades that mediate staurosporine-induced apoptosis are largely unknown. Exposure of cultured PC12 cells to staurosporine resulted in a rapid (min) and prolonged (1-6 hr) elevation of intracellular free calcium levels [Ca2+]i, accumulation of mitochondrial reactive oxygen species (ROS), and decreased mitochondrial 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction (1-4 hr). These early events were followed by membrane lipid peroxidation, loss of mitochondrial transmembrane potential, and nuclear apoptotic changes. Treatment of cells with serum or nerve growth factor within 1-2 hr of staurosporine exposure resulted in recovery of [Ca2+]i and ROS levels, and rescued the cells from apoptosis. The increased [Ca2+]i and ROS production were required for staurosporine-induced apoptosis because the intracellular calcium chelator BAPTA and uric acid (an agent that scavenges peroxynitrite) each protected cells against apoptosis. The caspase inhibitor zVAD-fmk and the anti-apoptotic gene product Bcl-2 prevented the sustained [Ca2+]i increase and ROS accumulation induced by staurosporine indicating that caspases act very early in the apoptotic process. Our data indicate that a [Ca2+]i increase is an early and critical event in staurosporine-induced apoptosis that engages a cell death pathway involving ROS production, oxidative stress, and mitochondrial dysfunction.