Sei sulla pagina 1di 9

Nutrition, Metabolism & Cardiovascular Diseases (2011) 21, 197e205

available at www.sciencedirect.com

journal homepage: www.elsevier.com/locate/nmcd

Insulin-mimetic action of conglutin-g, a lupin seed


protein, in mouse myoblasts
I. Terruzzi a, P. Senesi b, C. Magni c, A. Montesano a, A. Scarafoni c,
L. Luzi a,b,*, M. Duranti c

a
Nutrition-Metabolism Unit, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
b
Dipartimento di Sport, Nutrizione e Salute. Facolta` di Scienze motorie. Universita` degli Studi di Milano, Italy
c
Dipartimento di Scienze Molecolari Agroalimentari, Universita` degli Studi di Milano, Italy

Received 19 February 2009; received in revised form 7 July 2009; accepted 7 September 2009

KEYWORDS Abstract Background and aims: Lupin seed is referred to as an antidiabetic product in tradi-
Lupinus albus; tional medicine. Conglutin-g, a lupin seed glycoprotein, was found to cause a significant
Conglutin-g; plasma glucose reduction when orally administered to rats in glucose overload trials. Conglu-
Insulin signalling; tin-g was identified as being responsible for the claimed biological activity, and the aim of this
Insulin action work was to envisage its hypothetical insulin-mimetic cellular mechanism of action. Insulin is
responsible for proteosynthesis control through IRS/AKT/P70S6k/PHAS1 pathways modulation,
glucose homeostasis through PKC/Flotillin-2/caveolin-3/Cbl activation and muscle differentia-
tion/hypertrophy via muscle-specific MHC gene transcription control.
Methods and results: To assess whether conglutin-g modulates the same insulin-activated kinases,
myoblastic C2C12 cells were incubated after 72 h of differentiation with 100 nM insulin or 0.5 mg/
mL (w10 mM) conglutin-g. Metformin-stimulated cells were used as a positive control. The effect on
the above mentioned pathways was evaluated after 5, 10, 20 and 30 min. In the control cells medium
insulin, conglutin-g and metformin were not added. We demonstrated that insulin or conglutin-g
cell stimulation resulted in the persistent activation of protein synthetic pathway kinases and
increased glucose transport, glut4 translocation and muscle-specific gene transcription regulation.
Conclusions: Our results indicate that conglutin-g may regulate muscle energy metabolism, protein
synthesis and MHC gene transcription through the modulation of the same insulin signalling pathway,
suggesting the potential therapeutic use of this natural legume protein in the treatment of diabetes
and other insulin-resistant conditions, as well as the potential conglutin-g influence on muscle cells
differentiation and regulation of muscle growth.
2009 Elsevier B.V. All rights reserved.

* Corresponding author. Nutrition-Metabolism Unit, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy. Tel.: 39 02 2643
3738; fax: 39 02 2643 3407.
E-mail address: livio.luzi@unimi.it (L. Luzi).

0939-4753/$ - see front matter 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.numecd.2009.09.004
198 I. Terruzzi et al.

Introduction measurements at 280 nm were made. The extinction coef-


ficient of 1 for a solution of 1 mg/ml was used, according
White lupin seed conglutin-g is a mono-glycosylated protein to ref. [2].
consisting of two disulphide bonded subunits of 30 and
17 kDa (amino acid sequence available at TrEMBL, accession Experimental protocol
number: Q9FSH9). This 47 kDa monomeric unit undergoes
a pH-dependent reversible association to tetramer at C2C12 myoblasts were cultured at 37  C (5% CO2) in
neutral to slightly alkaline pH values [1,2]. Conglutin-g in a growth medium (GM) containing DMEM, 20% (v/v) fetal
its native conformation is unusually resistant to proteolysis bovine serum, 1% penicillinestreptomycin and 1% L-gluta-
by trypsin [3]. Conglutin-g was found to interact with mine. Seventy per cent confluent myoblasts (Ctu) were
insulin in vitro with a Kd around 7  105 M and, most differentiated in DMEM supplemented with 1% of horse
importantly, to significantly reduce plasma glucose in serum and antibiotics (DM) for 72 h. Cells used as control
rodents in doses ranging from 30 to 120 mg/kg body weight (Ct) were maintained in DM for the duration of the exper-
[4]. Therefore, this protein was identified as the respon- iment. Other three groups of cells were placed in DM with
sible molecule for the claimed anti-diabetic properties of the selective addition respectively of insulin (I; 100 nM),
lupin seeds in traditional medicine. Since insulin-binding to conglutin-g (Cg; 10 mM) and metformin (Mf; 400 mM) and
its own receptor causes a series of phosphorylation/de- lysed at 0, 5, 10, 20 and 30 min after the stimuli addition.
phosphorylation reactions, [5] which lead the insulin signal For immunofluorescence analysis differentiated C2C12
from the receptor to the final metabolic and myogenic myoblasts were stimulated with insulin, conglutin-g and
pathways (Fig. 1), we firstly hypothesize that the effect of metformin for 30 min. In differentiated cells immuno-
conglutin-g on blood glucose is due to an insulin-mimetic stained with anti-MHC, the DAPI stained nuclei per myo-
effect of the protein at the level of the intracellular tube, myotubes length and diameter were determined and
pathway insulin receptor/IRS-1/PI-3-kinase, eventually expressed as average.
leading to the recruitment and translocation of GLUT4
(Fig. 1). Secondly, as insulin promotes muscle anabolism,
we hypothesize the activation of a protein synthetic and
Electrophoretic techniques and immunoblotting
myogenic process by conglutin-g. analysis
The aim of this work was to test the effect of conglutin-g
in an in vitro model of mouse myoblasts, assessing the Conglutin-g SDS-PAGE was performed in NuPAGE Novex
modulation of muscle-specific genes and the phosphoryla- Bis-Tris 10% gels by using a XCell SureLock Mini-Cell
tion/activation of intracellular kinases involved in the (Invitrogen, Milan, Italy). SeeBlue Plus2 Prestained Stan-
insulin signalling cascade. Our results indicate that con- dard and SimplyBlue SafeStain (Invitrogen, Milan, Italy)
glutin-g shares, with insulin, common effects on the were used. For protein blot analysis, the gel was trans-
intracellular kinases tested in this work, suggesting ferred to nitrocellulose transfer membrane (Protran,
a possible therapeutic indication as an insulin-mimetic Whatman  Schleicher & Schuell) by blotting according to
agent. Towbin et al. [6] on a Transblot Electrophoretic Transfer
Cell (Bio-Rad, Milan, Italy). The membrane was blocked
with 3% fish gelatin for 2 h and washed three times with
Methods 0.25% fish gelatin solution both in PBS buffer (10 mM Na
phosphate, pH 7.4, containing 150 mM NaCl), then soaked
Materials for 2 h in PBS buffer containing rabbit anti-conglutin-g in
the ratio 1,500/1 (v/v). The antiserum was prepared and
Anti-actin (I-19), anti-AKT (C-20), antiecaveolin-3 (A-3), immuno-affinity purified as previously described [7]. The
anti4e (P-2), antieflotillin-2 (H-90), anti-IRS1 (H-165), bands were revealed by using horseradish peroxidase
anti-p-IRS1 (Tyr 632), anti-myogenin, anti-PI3-Kinase p85a conjugate with goat-antirabbit antiserum 2,000/1 (v/v)
(Z-8), anti-PKC (H-300), anti ea-tubulin (TU-16), anti- (Bio-Rad, Milan, Italy) and hydrogen peroxide with
phospho-ERK (E-4), anti-MHC, anti-phospho-Cbl (Tyr700) 4-chloronaphtol as substrate. C2C12 myofibers were
and anti-phosho-p70S6 kinase (Thr421/Ser 424), anti- homogenized as described [8]. Aliquots of 30 mg superna-
PHAS1(P-2), monoclonal or polyclonal primary antibodies tant proteins from the different samples were resolved by
and the peroxidase-conjugated secondary antibodies were 8% (p-IRS1 Tyr 632, IRS1, MHC, myogenin and p-CBL), 10%
purchased from Santa Cruz Biotechnology (Santa Cruz, CA, (PI3K p85, AKT1, p-p70 S6K and PKC), 12% (eIF-4E, p-ERK1,
U.S.A.). All other reagents were purchased from Sigma p-ERK2, caveolin-3 and flottilin-2) and 15% (PHAS1) SDS-
Chem. Co. (St. Louis, MO, U.S.A.). Mouse C2C12 myoblastic PAGE. Electrophoresed proteins were transferred as
cells were purchased from the European Collection of described [8] and the membranes incubated with species-
Animal Cell Cultures (ECACC). specific secondary antibodies. Immunoreactive bands
were visualized by an enhanced chemiluminescence
method (Amersham Pharmacia Biotech, Piscataway, NJ,
Purification of lupin conglutin-g USA). Total cell lysates of 1e2 mg of protein were immu-
noprecipitated with 10 ml antibodies against p-IRS (Tyr
Conglutin-g was purified using a combination of anion 632). The immunoprecipitated samples were subjected to
and cation exchange chromatography [1]. For the estima- SDS-PAGE and Western Blot analysis, which was conducted
tion of purified conglutin-g concentrations, 4 optical as described previously.
Conglutin-g and insulin action 199

Figure 1 Intracellular pathways of insulin signaling. Ins: insulin; IRS-1: insulin Receptor Substrate 1; PI3K: phosphatidylinositol
3-kinase; AKT also called PKB: protein kinase B; p70S6K: p70 ribosomal protein S6 kinase; PHAS1 also called 4EBP1: eukaryotic
initiation factor 4E-binding protein-1; eIF4E/4G/4A: eukaryotic initiation factor13 4E/4G/4A; PKC: protein kinase C; Flo-2: flotillin-
2; Glut4: glucose transporter 4; Cav-3: caveolin-3; CBL: tyrosine phosphorylated protein of the protooncogenes c-Cbl and Cbl-b;
CAP: Cbl associated protein; ERK1/2: extracellular signal-regulated kinases 1/2.

The membrane was stripped and reprobed with an lupin protein extract. Two main bands of Mr around 30 and
antibody to actin or tubulin to confirm equal protein 16e17 kDa were visible. The sizes of these bands fitted with
loading per sample. Quantitative measurement of immu- those of the large and small conglutin-g subunits. A minor
noreactive bands was performed by densitometric analysis band of 47 kDa was attributed to the uncleaved conglutin-g
using the Scion Image software (Scion Corporation, Fred- precursor (Duranti, unpublished results). This protein
erick, MD, USA). preparation was judged sufficiently homogenous to be
suitable for cell trials. The protein blot with specific anti-
Immunofluorescence analysis bodies (Fig. 2B) confirmed the identity of the proteins. The
lack of mobility differences between the purified conglutin-
For immunofluorescence, cells were fixed in 4% para- g and the one in the total protein extract suggested that no
formaldehyde, permeabilized with 0.2% Triton X-100, and modification to the covalent continuity of the protein had
blocked with PBS containing 1% bovine serum albumin. Cells occurred during the purification procedure.
were then immunostained with anti-MHC rhodamine
conjugated and nuclei revealed with DAPI staining. Analyses of insulin signaling proteins

Reproducibility and statistics analysis In our previous work, we focused our investigation on the
insulin-modulated pathways involved in protein anabolism,
All experiments were performed three times, with similar glucose homeostasis, gene expression and mRNA translation
results. Data are expressed as means  SE. The unpaired t- regulation, in different tissues [8e10]. We monitored the
test was used to compare AUC (area Under the Curve) ability of both insulin and conglutin-g to stimulate the same
calculated for the time courses. Differences between pathways in vitro. Metformin, the first-line oral anti-dia-
groups were considered statistically significant if P  0.05. betic drug, was used as a positive control. Fig. 3a and A
illustrate typical patterns of IRS-1observed on western blots
of C2C12 myotube at different times of stimulus by con-
Results glutin-g, insulin or metformin (time course). Compared to
the time course of control cells, insulin maximally stimu-
Homogeneity of lupin conglutin-g lated the IRS-1 protein content within 20 min by w32%, but
lupin protein stimulation was even greater (w35%) at
Fig. 2A shows the SDS-PAGE pattern under reducing condi- 10 min (p  0.007). Metformin exerted a gradual increase of
tions of the purified conglutin-g as compared to the total the amount of IRS-1, which was maximal at 30 min (w76%).
200 I. Terruzzi et al.

Metformin exerted an activation of p70S6K phosphorylation


within 10 min (w46%) then decreased until 30 min (w32%).
The activation of PHAS1 is associated with different
residues of phosphorylation that produce isoforms of the
enzymes slowly migrating on SDS-PAGE. The blots appear as
two distinct bands: a rapidly migrating band (a) and the
more slowly migrating bands (b) representing the more
active form of PHAS1. We evaluated the shift in the enzyme
mobility as indexes of insulin and conglutin-g effect on
PHAS1 phosphorylation. Fig. 3E and F show that the inten-
sities of a fast migrating (PHAS-1A) and b slowly migrating
(PHAS-1B) bands, were higher in conglutin-g, insulin and
metformin-stimulated cells with respect to the control.
(PHAS-1A conglutin-g vs control, p  0.02; PHAS-1A con-
glutin-g vs control , p  0.003). Phosphorylation of 4E-BP1
frees eIF-4E, allowing the assembly of the active complex
eIF-4F and the consequent binding of mRNA to the 40S
ribosomal subunit. Thus, the increment of 4E-BP1 phos-
phorylation or eIF-4E content indicates a stimulated
translation and an activation of the initial events in protein
biosynthesis.
Fig. 3G represents the amount of eIF4E after insulin or
conglutin-g stimulation in C2C12 myotube. Insulin
enhanced eIF4E protein concentration with a persistent
effect from 5 to 20 min (w45%). This effect decreased but
Figure 2 Panel A: SDS-PAGE pattern under reducing condi-
persisted until the end of the experiment (w30%). Con-
tions of the purified conglutin-g (lane: 1) as compared to the
glutin-g stimulated eIF4E protein concentration in 5 min
total lupin protein extract (lane: 2). M Z marker proteins.
(w21%) and the effect increased up to 30 min (w68%,
Panel B: protein blot with specific anti-conglutin-g antibodies
p  0.03). In contrast, metformin was not able to increase
on the purified conglutin-g (lane: 1) and the total protein
eIF4E enzyme concentration, except a weak increment at
extract (lane: 2). M Z marker proteins. See text for experi-
30 min.
mental details.
Glucose transport pathway
The tyrosine phosphorylation of IRS-1 generates docking PKC, a phosphatidylinositol (PI) 3-kinase downstream
sites for several SH2-containing proteins. Among these, the kinase, plays a very important role in activating glucose
predominant partner seems to be the p85 regulatory transport, but a separate PI3K-independent pathway,
subunit of the PI3K 3-kinase. We used anti-phospho-Tyr IRS- involving the insulin-stimulated CBL phosphorylation, is
1 antibody to measure IRS-1 phosphorylation and we found required to promote the GLUT4 translocation to the plasma
that, unlike control and metformin, both conglutin and membrane. Expression of flotillin-2 and caveolin-3 is
insulin are able to maintain Tyr IRS1 phosphorylation until necessary for the activation of this pathway. In our study
the end of the experiment as shown in Fig. 3a. conglutin-g increased the PKC concentration (Fig. 4A) at
Insulin enhanced p85-PI3 kinase concentration (Fig. 3B) approximately the same values at 10 (w28%) and 20 min
within 5 min (w62%), and the effect was persistent up to (w26%), then decreasing to w20% at the end of the
the end of the experiment (w56%) while the metformin experiment. These data, compared to control, were
effect was negligible. The effect of conglutin-g was slower significant with p  0.04. Insulin effect was earlier (5 min)
but robust, with a peak effect at 30 min (w40%). Fig. 3C than conglutin-g and reached a maximum at 20 min
shows the modulation of Akt-1, a downstream target of PI (w32%). Metformin was able to stimulate protein concen-
3-kinase involved in the insulin-signalling pathway leading tration more than the other two stimuli. Flotillin-2 protein
to glucose utilization, glycogen and protein synthesis, and concentration, increased from 5 to 30 min in all the tested
CAP dependent translation. Insulin and conglutin-g conditions, as represented in Fig. 4B, with a peak at 10 min
increased the concentration of Akt-1 protein at 20 min by insulin (w60%) and conglutin-g (w52%; p  0.03). The
(w13% and 33% respectively), while metformin reached the differences in protein content between the control group
top concentration at 5 min (w20%). The differences in Akt- and the conglutin-g group was significant with (p  0.03.).
1 protein content between the control and the conglutin-g Metformin did not exert any increase of flotillin-2 concen-
was significant with p  0.03. tration. Caveolin-3 protein concentration was increased by
both conglutin-g and insulin with a maximum stimulus at
Protein synthesis pathway 10 min (w22%) and 30 min (w34%) respectively, as repre-
As seen in Fig. 3D, insulin induced a p70S6 K phosphoryla- sented in Fig. 4C. Metformin exerted a greater stimulation
tion within 5 min (w40%) increasing up to w94% in 30 min. with respect to the other stimuli, which was up and above
Conglutin-g exerted a maximum stimulus on p70 S6 kinase 70% toward the end of the experiment. The data presented
phosphorylation within 5 min (67%), which persisted but in Fig. 4D show conglutin-g and insulin ability to maximally
gradually decreased until 30 min (w50%, p  0.02). phosphorylate Cbl at 10 min (w64%) and 30 min (w56%)
Conglutin-g and insulin action 201

Figure 3 a) representative blot of immunoprecipitated phospho Tyr IRS1. Densitometric analysis of A) insulin receptor substrate
1 protein (IRS-1), B) PI3K-p85 subunit, C) AKT-1, D) p70S6K phosphorylation, E) PHAS-1a phosphorylation, F) PHAS-1 b phosphory-
lation and G) eiF4-E protein content in C2C12 myofibers when not stimulated (Ct) or after insulin (I), conglutin-g (Cg) or metformin
(Mf) stimulation, at the times detailed in the Methods. The histograms depict the balance obtained by calculating the difference
between the densitometric value measured at specific experimental times in the stimulated cells and the densitometric value
measured at the same time in the control cells. The data of three independent experiments are expressed as a percentage in
relation to the amount of target protein at 0 min. Protein quantification was adjusted for the corresponding a-tubulin level and the
data expressed as mean S.D. The differences in IRS-1, AKT-1 and eiF4E protein content between the control and the conglutin-g
were significant, respectively, with *p  0.007, *p  0.03 and *p  0.03. The differences in p70S6 K phosphorylation, PHAS-1a and
PHASb between the control and the conglutin g stimulated cells were significant, respectively, with *p  0.02, *p  0.02 and
*p  0.003.

respectively. Differences among control and conglutin-g Metformin exerted a much greater activation on both ERK1
treated cells in Cbl phosphorylation level were significant and ERK2 with respect to both the other two stimuli.
(p  0.03). Metformin effect was barely detectable.
Analyses of muscle-specific gene expression
ERKs pathway
Neither insulin nor conglutin-g were able to significantly To evaluate whether conglutin-g, as insulin [11], is able to
modify ERK1 and ERK2 phosphorylation with respect to the trigger gene transcription control, we studied the content
control group, as shown in Fig. 4E and F. Conglutin-g of MHC muscle-specific protein, responsible for a tissue
increased the phosphorylation of ERK1 at 20 min, up to 16%, differentiation process. As shown in Fig. 5A, conglutin-g, as
and up to 27% ERK2 phosphorylation (p  0.03). Insulin well as insulin, was able to increase MHC protein content
caused a 30% increase of ERK1 activation at 20 min while (Fold Changes Z 4.5, p  0.05 and Fold Changes Z 10.2;
ERK2 insulin stimulation never exceeded 28% at 30 min. p  0.03 respectively) with respect to the unstimulated
202 I. Terruzzi et al.

Figure 4 Densitometric analysis of A) PKC protein content, B) flotillin-2 protein content, C) caveolin-3 protein content, D) Cbl
phosphorylation, E) ERK-1 phosphorylation and F) ERK-2 phosphorylation in C2C12 myofibers when not stimulated (Ct) or after
insulin (I), conglutin-g (Cg) or metformin (Mf) stimulation at the times described in the Methods. The histograms depict the balance
obtained by calculating the difference between the densitometric value measured at specific experimental times in the stimulated
cells and the densitometric value measured at the same time in the control cells. The data of three independent experiments are
expressed as a percentage in relation to the amount of target protein at 0 min. Protein quantification was adjusted for the cor-
responding a-tubulin level and the data expressed as mean S.D. The differences in PKC, flotillin-2 and caveolin-3 protein content
between the control and the conglutin-g were significant, respectively, with *p  0.004, *p  0.03 and *p  0.03. The differences in
Cbl, ERK1 and ERK2 phosphorylation between the control and the conglutin-g stimulated cells were significant with *p  0.03.

undifferentiated muscle cells (Ctu), but metformin was not myoblasts into myotubes, or myotubes hypertrophy, three-
able to induce the same gene expression. day differentiated C2C12 cells were stimulated with
Moreover, we studied the protein content of myogenin, conglutin-g, insulin and metformin for 30 min. Images of
a transcription factor playing a pivotal role in the activation MHC-positive myotubes, detected by immunofluorescence
of the MHC gene. Fig. 5B shows that myogenin concentra- (Fig. 5C), superimposed on DAPI stained nuclei (Fig. 5D),
tion by both insulin and conglutin-g was consistent with showed that in cells stimulated with metformin, conglutin-
that of MHC protein content and reached maximal peaks at g or insulin, as compared with the unstimulated differen-
30 min (w73% and 84% respectively), while, in this case too, tiated muscle cells, the number of nuclei present in
metformin did not show any effect. The myogenin protein MHC-positive myotubes (17.9 3.4, 23.5 6.1, 27.8 8.1
content between the control and the conglutin g was and 14.5 4.3 respectively as shown in Fig. 5E), myotubes
significant with p  0.02. diameter (cm1.3 0.4, cm1.9 0.4, cm2.3 0.7 and
To validate the effect of the stimuli on MHC protein cm1.2 0.4 respectively as shown in Fig. 5F), length
content and myoblasts differentiation and to investigate (cm17.7 3.7, cm18.4 3.0, cm21.6 4.0 and 13.7 2.5
whether those stimuli could improve the recruitment of respectively as shown in Fig. 5G) and fusion index
Conglutin-g and insulin action 203

Figure 5 Densitometric analysis of: A) MHC protein content and B) myogenin protein content in C2C12 myofibers when not
stimulated (Ct) or after insulin (I), conglutin-g (Cg) or metformin (Mf) stimulation at the times described in the methods. The
histograms depict the balance obtained by calculating the difference between the densitometric value measured at specific
experimental times in the stimulated cells and the densitometric value measured at the same time in the control differentiated
cells (Ct) for myogenin and in the control undifferentiated cells (Ctu) for MHC. The data of myogenin and MHC of three independent
experiments are expressed as a percentage in relation to the amount of target protein at 0 min and as Fold Changes respectively.
Protein quantification was adjusted for the corresponding a-tubulin level and the data expressed as mean S.D. The differences in
MHC and myogenin protein content between the control and the conglutin-g stimulated cells were significant., respectively , with
*p  0.05 and *p  0.02. C2C12 myoblasts were differentiated for 72 h and treated as described in the method section. C) MHC in
myotubes of control and stimulated with Mf, Cg and I respectively, was detected by immunofluorescence (red). D) Images of MHC-
positive myotubes were laid upon DAPI stained nuclei images. E) The nuclei present in MHC-positive myotubes were counted and
the average graphically represented. The difference between the control and the conglutin-g stimulated cells was significant with
*p  0.04. F) The diameters of MHC-positive myotubes were measured and the average, expressed in cm, were graphically rep-
resented. The difference between the control and the conglutin-g stimulated cells was significant with *p  0.03. G) The length of
MHC-positive myotubes was measured and the average, expressed in cm, is graphically represented. The difference between the
control and the conglutin-g stimulated cells was significant with *p  0.05. H) The number of DAPI stained nuclei present in MHC-
positive cells/number of total DAPI stained nuclei per microscopic field (fusion index) was determined and expressed as
percentage. The difference between the control and the conglutin-g stimulated cells was significant with *p  0.04. The data of
three independent experiments are expressed as mean S.D.

(57% 0.1, 68% 7.4, 74% 0.1 and 47% 0.1 respectively the active compound responsible for the blood glucose
as shown in Fig. 5H) gradually increased. lowering effect [4]. In the present work we have evaluated
conglutin-g effects on the activation of intracellular
kinases common to the insulin signaling cascade using in
Discussion vitro models of mouse myoblasts. We firstly observed that
incubation of cells with conglutin-g causes the activation of
Since the first findings at the beginning of the 30s, various the intracellular IRS-1/PI-3-kinase pathway eventually
authors have attempted the isolation of lupin components involved in glucose homeostasis [5] and protein synthesis
with glycemic lowering properties [12,13]. Our recent data stimulation. The eukaryotic translation initiation factor
support the conclusion that the lupin protein conglutin-g is eIF4E, released as the result of PHAS-1 phosphorylation,
204 I. Terruzzi et al.

acts on translation initiation and promotes the messenger potential treatment of type 1 and 2 diabetes. Moreover, the
RNA export of several genes involved in the cell cycle and present study on the mechanism of action makes conglutin-
growth [14]. Our present data show that incubation of g suitable for additional investigation as an insulin-sensi-
mouse myoblasts with conglutin-g stimulates p70 S6 kinase tizing compound, opening its possible applications both as
and eIF-4E activation as shown in Fig. 3D and G. Moreover, drug or food integrators in obesity and many other insulin-
conglutin-g is able to stimulate PHAS-1 activation (Fig. 3E resistant conditions such as the metabolic syndrome [20],
and F). Phosphorylation of AKT-1 is upstream of the phos- polycystic ovary [21] and HIV-lipodystrophy [22].
phorylation of p70 S6 kinase as well as of eIF-4E activation, In recent years there have been considerable advances
and both insulin and conglutin-g show comparable effects in the understanding of mechanisms by which insulin
on its phosphorylation with a modest stimulation above regulates, both in a positive and negative manner, the
basal at any lt (Fig. 3C). expression of genes encoding proteins involved in a wide
Taken together, these results suggest that conglutin-g variety of biologic activities. In particular, studying the
plays an important role in regulating the level of the impact of hyperinsulinemia on myosin heavy chain (MHC)
protein synthesis and modulating the activation of this gene regulation in human skeletal muscle [23], has shown
machinery in the cell. In skeletal muscle cells flotillin-2 that insulin exerts a rapid influence at the transcriptional
and caveolin-3 coordinately modulate the insulin-stimu- level for the MHC protein. In this present work we find
lated translocation of GLUT4, localized in flotillin-2-con- that insulin is able to increase, more than six hundred
taining perinuclear domains, to selective domains of times, the MHC gene expression with respect to the
sarcolemma. Upon insulin stimulation, caveolae micro- control. In addition, the expression of MHC gene results
domains containing caveolin-3 and the insulin receptor, positively regulated by conglutin-g with respect to the
move away from the plasma membrane toward the unstimulated differentiated muscle cells. Overall, our
cytoplasm and temporarily interact with flotillin-2/GLUT4- results demonstrate that MHC genes expressed in muscle
containing domains prior to reaching the sarcolemma. The tissues is transcriptionally regulated by both insulin and
insulin receptor moves from caveolin-3-to flotillin-2-con- conglutin-g, while metformin is unable to produce the
taining domains. GLUT4, together with flotillin-2, moves same effect.
to the sarcolemma, promoted by the insulin-stimulated The transcription factor myogenin plays a pivotal role in
tyrosine phosphorylation of CBL [15]. Our results show that the activation of muscle-specific gene MHC and is critical
conglutin-g, as insulin, increases both flottilin-2 and for skeletal muscle development [24e27]. Myogenin is
caveolin-3 concentrations (Fig. 4B and C) and CBL phos- absent in undifferentiated cells and is over expressed in
phorylation (Fig. 4D), playing an important role in the myoblasts committed to myogenic fate and in muscle
vesicular transport of GLUT4 and in the regulation of terminal differentiation. For these reasons we wanted to
muscle energy metabolism. assess whether the observed variation in MHC gene
The robust insulin-like activity of conglutin-g is difficult expression corresponded to an adequate expression of the
to explain since its primary structure (Swiss-Prot accession myogenic marker myogenin. In this work we provide
number: Q9FSH9), size and other molecular features are evidence that both conglutin-g and insulin are able to
totally dissimilar to those of insulin. Nonetheless, con- enhance mouse C2C12 myoblastic cells differentiation as
glutin-g shares with insulin some interaction properties: i. shown by an increase in the accumulation of differentiation
in vitro, conglutin-g binds to insulin itself; ii. like insulin, markers such as MHC and myogenin.
conglutin-g is capable of binding metal ions including ERKs are shown to negatively regulate skeletal muscle
Zn, Cu, Cd, Co in a decreasing order of cell differentiation [28,29] and ERKs inhibition is shown to
affinity [16]. How these findings can be related to the enhance myoblasts fusion and the expression of the late
effects monitored in the present work is not yet under- differentiation marker myosin heavy chain [30]. Consis-
stood. In two papers published in the 90s, an unusual tently in our study C2C12 cell differentiation and fusion to
insulin-binding activity of a soybean protein, basic globulin form multinucleated myotubes were accompanied by
7S (Bg7S), was also demonstrated. However no study on the a reduction in the activating phosphorylation levels of
potential bioactivity of this soybean protein was under- ERKs. It is reasonable to assume that the deprivation of
taken [17,18]. Lupin conglutin-g shares great amino acid serum mitogens or growth factors leads to inactivation
sequence identity with soybean Bg7S (Swiss-Prot Accession of the ERKs, but our previous data [8] showed that in human
number: Q8RVH5) [19] and other incompletely character- vascular endothelial cells insulin was able to activate ERKs
ized plant proteins, such as various endoglucanase inhibi- even in the absence of serum. Instead, in our cellular
tors (TrEMBL Accession numbers: B9VUU9, Q7XJE7, model, insulin and conglutin-g block ERK1/2 activity pre-
B9VUV0). The lupin seed hypoglycemic effects and the venting myoblast proliferation and promoting their differ-
presence of conglutin-g-related proteins in other seeds entiation, while metformin is not able to produce the same
makes it reasonable to speculate on the existence of effect. The present data indicate that conglutin-g, as
a family of plant proteins, which share some functional insulin, allows the recruitment of myoblasts, enhancing
property with mammalian insulin. A striking difference with their fusion into multinucleated myotubes and promoting
insulin is that conglutin-g is highly resistant to digestive increment of their length and diameter. Our present data
enzymes [3] and this might explain the maintenance of its provides elements to hypothesize that conglutin-g as
activity after absorption [4]. In this respect, further dedi- insulin could influence muscle cell differentiation and
cated studies are definitely needed to understand the contribute to the regulation of muscle growth. This last
metabolic fate of this protein. Nonetheless, the present possible effect of conglutin-g opens new posssbilities of
findings make this natural protein highly appealing for the marketing applications as food and sports integrators too.
Conglutin-g and insulin action 205

References [15] Fecchi K, Volonte D, Hezel MP, Schmeck K, Galbiati F. Spatial


and temporal regulation of GLUT4 translocation by flotillin-1
and caveolin-3 in skeletal muscle cells. FASEB J 2006;20:705e7.
[1] Duranti M, Scarafoni A, Gius C, Negri A, Faoro F. Heat-induced
[16] Duranti M, Scarafoni A, Di Cataldo A, Sessa F. Interaction of
synthesis and tunicamycinsensitive secretion of the putative
metal ions with lupin seed conglutin gamma. Phytochemistry
storage glycoprotein conglutin gamma from mature lupin
2001;56:529e33.
seeds. Eur J Biochem 1994;222:387e93.
[17] Komatsu S, Hirano H. Plant basic 7 S globulin-like proteins
[2] Duranti M, Sessa F, Scarafoni A, Bellini T, Dallocchio J.
have insulin and insulin-like growth factor binding activity.
Thermal stabilities of lupin seed conglutin gamma protomers
FEBS Lett 1991;294:210e2.
and tetramers. J Agric Food Chem 2000;48:1118e23.
[18] Watanabe Y, Barbashov SF, Komatsu S, Hemmings AM,
[3] Duranti M, Gius C, Sessa F, Vecchio G. The saccharide chain of
Miyagi M, Tsunasawa S, et al. A peptide that stimulates
lupin seed conglutin gamma is not responsible for the
phosphorylation of the plant insulin-binding protein. Isolation,
protection of the native protein from degradation by trypsin,
primary structure and cDNA cloning. Eur J Biochem 1994;224:
but facilitates the refolding of the acid-treated protein to the
167e72.
resistant conformation. Eur J Biochem 1995;230:886e91.
[19] Scarafoni A, Di Cataldo A, Vassilevskaia TD, Bekman EP,
[4] Magni C, Sessa F, Accardo E, Vanoni M, Morazzoni P,
Rodrigues-Pousada C, Ceciliani F, et al. Cloning, sequencing
Scarafoni A, et al. Conglutin gamma, a lupin seed protein,
and expression in the seeds and radicles of two Lupinus albus
binds insulin in vitro and reduces plasma glucose levels of
conglutin g genes. Biochim Biophys Acta 2001;1519:147e51.
hyperglycemic rats. J Nutr Biochem 2004;15:646e50.
[20] Grundy SM. Metabolic syndrome: a multiplex cardiovascular
[5] Saltiel AR, Kahn CR. Insulin signaling and the regulation of
risk factor. J Clin Endocrinol Metab 2007;92:399e404.
glucose and lipid metabolism. Nature 2001;414:799e806.
[21] Santana LF, de Sa MF, Ferriani RA, de Moura MD, Foss MC, dos
[6] Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of
Reis RM. Effect of metformin on the clinical and metabolic
proteins from polyacrylamide gels to nitrocellulose sheets:
assessment of women with polycystic ovary syndrome. Gyne-
procedure and some applications. Proc Natl Acad Sci U S A
col Endocrinol 2004;19:88e96.
1979;76:4350e4.
[22] Luzi L, Perseghin G, Tambussi G, Meneghini E, Scifo P,
[7] Magni C, Herndl A, Sironi E, Scarafoni A, Ballabio C, Restani P,
Pagliato E, et al. Intramyocellular lipid accumulation and
et al. One- and two-dimensional electrophoretic identification
reduced whole body lipid oxidation in HIV lipodystrophy. Am J
of IgE-binding polypeptides of Lupinus albus and other legume
Physiol Endocrinol Metab 2003;284:E274e80.
seeds. J Agric Food Chem 2005;53:4567e71.
[23] Houmard JA, ONeill DS, Zheng D, Hickey MS, Dohm GL.
[8]. Terruzzi I, Pellegatta F, Luzi L. Differential p70S6k and
Impact of hyperinsulinemia on myosin heavy chain gene
4E-BP1 regulation by insulin and amino acids in vascular
regulation. J Appl Phys 1999;86:1828e32.
endothelial and smooth muscle cells. Acta Diabetol 2005;42:
[24]. Olson E. Activation of muscle-specific transcription by
139e46.
myogenic helix-loop-helix proteins. Symp Soc Exp Biol 1992;
[9] Pellegatta F, Catapano AL, Luzi L, Terruzzi I. In human
46:331e41.
endothelial cells amino acids inhibit insulin-induced Akt and
[25] Dias P, Dilling M, Houghton P. The molecular basis of skeletal
ERK1/2 phosphorylation by an mTOR-dependent mechanism.
muscle differentiation. Semin Diagn Pathol 1994;11:3e14.
J Cardiovasc Pharmacol 2006;47:643e9.
[26] Buckingham M, Bajard L, Chang T, Daubas P, Hadchouel J,
[10] Terruzzi I, Allibardi S, Bendinelli P, Maroni P, Piccoletti R,
Meilhac S, et al. The formation of skeletal muscle: from
Vesco F, et al. Amino acid- and lipid-induced insulin resistance
somite to limb. J Anat 2003;202:59e68.
in rat heart: molecular mechanisms. Mol Cell Endocrinol 2002;
[27] Sabourin LA, Rudnicki MA. The molecular regulation of myo-
190:135e45.
genesis. Clin Genet 2000;57:16e25.
[11] Mounier C, Posner BI. Transcriptional regulation by insulin:
[28] Olson EN, Spizz G, Tainsky MA. The oncogenic forms of N-ras or
from the receptor to the gene. Can J Physiol Pharmacol 2006;
H-ras prevent skeletal myoblast differentiation. Mol Cell Biol.
84:713e24.
1987;7:2104e11.
[12] Ferranini A, Pirolli M. Lazione del decotto di semi di Lupinus
[29] Bennett AM, Tonks NK. Regulation of distinct stages of skeletal
albus sulla curva glicemica da carico di glucosio nei soggetti
muscle differentiation by mitogen-activated protein kinases.
normali e diabetici. Folia Med 1937;23:729e48.
Science 1997;278:1288e91.
[13] Orestano G. Sullazione ipoglicemica dei semi di Lupinus
[30] Tortorella LL, Milasincic DJ, Pilch PF. Critical proliferation-
albus. Arch Farmacol Sperim 1940;70:113e7.
independent window for basic fibroblast growth factor
[14] Culjkovic B, Topisirovic I, Skrabanek L, Ruiz-Gutierrez M,
repression of myogenesis via the p42/p44 MAPK signaling
Borden KL. eIF4E is a central node of an RNA regulation that
pathway. J Biol Chem 2001;276:13709e17.
governs cellular proliferation. J Cell Biol 2006;175:415e26.

Potrebbero piacerti anche