Вы находитесь на странице: 1из 12

doi:10.1111/j.1440-1746.2008.05733.

G A S T R O E N T E R O L O G Y jgh_5733 830..840

Differential effect of oleic and palmitic acid on lipid


accumulation and apoptosis in cultured hepatocytes
Matteo Ricchi,* Maria Rosaria Odoardi,* Lucia Carulli,* Claudia Anzivino,* Stefano Ballestri,*
Adriano Pinetti,† Luca Isaia Fantoni,‡ Fabio Marra,§ Marco Bertolotti,* Sebastiano Banni,¶
Amedeo Lonardo,* Nicola Carulli* and Paola Loria*
*Dipartimento Integrato di Medicina Interna, Endocrinologia, Metabolismo & Geriatria – Università degli Studi di Modena e Reggio Emilia,

Dipartimento di Chimica, Università di Modena e Reggio Emilia, ‡Dipartimento di Scienze Biomediche, Sezione di Chimica Biologica, Università
degli Studi di Modena e Reggio Emilia, Modena, §Dipartimento di Medicina Interna and Center for Research, Transfer, and High Education
DENOTHE, Università di Firenze, Firenze, and ¶Dipartimento di Biologia Sperimentale, Università degli Studi di Cagliari, Cagliari, Italy

Key words Abstract


apoptosis, cell culture techniques, fatty acids,
saturated and insaturated, fatty liver, insulin
Background and Aim: Studies have shown monounsaturated oleic acid to be less toxic
resistance. than palmitic acid and to prevent/attenuate palmitic acid hepatocites toxicity in steatosis
models in vitro. However, to what degree these effects are mediated by steatosis extent is
Accepted for publication 21 October 2008. unknown.
Methods: We evaluated whether steatosis per se is associated with hepatocytes apoptosis
Correspondence and determined the role of oleic and palmitic acid, the most abundant fatty acids in western
Nicola Carulli and Paola Loria, Università degli diets, on triglyceride accumulation and apoptosis in an in vitro model of steatosis induced
Studi di Modena, Dipartimento di Medicina in three hepatocytic cell lines (HepG2, HuH7, WRL68). The impact of incubation for 24 h
Interna, Endocrinologia, Metabolismo e with oleic (0.66 and 1.32 mM) and palmitic acid (0.33 and 0.66 mM), alone or combined
Geriatria, Nuovo Ospedale Civile (molar ratio 2 : 1) on steatosis, apoptosis, and insulin signalling, was evaluated.
Sant’Agostino Estense di Baggiovara, Via Results: Concurrent with PPARg and SREBP-1 gene activation, steatosis extent was larger
Giardini Baggiovara, Modena 41100, Italy. when cells were treated with oleic than with palmitic acid; the latter fatty acid was
Email: paola.loria@unimore.it associated with increased PPARa expression. Cell apoptosis was inversely proportional to
steatosis deposition. Moreover, palmitic, but not oleic acid, impaired insulin signalling.
Partly supported by grants from MIUR
Ministero Istruzione Università e Ricerca
Despite the higher amount of fat resulting from incubation of the two fatty acids combined,
Scientifica -PRIN 2004061213_001.
the apoptosis rate and impaired insulin signalling were lower than in cells treated with
palmitic acid alone, indicating a protective effect of oleic acid.
Abbreviations: Conclusions: Oleic acid is more steatogenic but less apoptotic than palmitic acid in
DAPI, 4′,6-diamidino-2-phenylindole hepatocityc cell cultures. These data may provide a biological basis for clinical findings on
dihydrochloride; FA, Fatty Acid; FFA, Free dietary patterns and pathogenetic models of nonalcoholic fatty liver disease.
Fatty Acids; IR, Insulin Resistance; NEFA,
Non esterified fatty acids; OA, Oleic Acid;
PA, Palmitic Acid; T2D, Type 2 Diabetes;
TG, Triglycerides; VLDL, Very low density
lipoproteins
.

saturated fatty acid ratio in serum, fat and liver tissue might have a
Introduction pathogenetic role in the disease.10,11 Increased free fatty acids (FFA)
Fatty acids (FAs) are major components of biological cell mem- levels are linked with the pathogenesis of insulin resistance (IR),
branes that play important roles in intracellular signalling and as which is considered a major determinant in the pathogenesis of
precursors for ligands that bind to nuclear receptors.1,2 FAs repre- NAFLD. Peripheral IR results in increased concentrations of circu-
sent vital energy stores, but high-fat diets are associated with the lating FFAs due to unopposed antilipolytic insulin action. Addition-
development of obesity and type 2 diabetes (T2D).3,4 Several lines ally, accumulation of FFA results in an impaired post-receptor
of evidence indicate the importance of both quantitative and quali- insulin signalling12 contributing to IR and causing a further increase
tative (e.g. saturated vs unsaturated) changes in dietary FAs as in FFA.
relevant mechanisms for the development of nonalcoholic fatty The hepatocytes are not a physiological site of lipid storage and
liver disease (NAFLD) both in rodent models and in humans.5–9 development of steatosis is associated with cellular dysfunction
Finally, in obese NAFLD patients, the decreased unsaturated/ and apoptosis.13,14 This phenomenon, which also occurs in the

830 Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd
M Ricchi et al. Fatty acids and apoptosis in hepatocytes

kidney, pancreas and heart, is referred to as lipotoxicity and is Rutherford, NJ, USA), 1% bovine serum albumin (BSA), supple-
believed to play a significant role in the pathogenesis of tissue mented with FFA (oleic and palmitic acid alone or in association)
damage.15 In fact, the extent of apoptosis correlates with the sever- at the following final concentrations: a) PA: 0.33 mM and
ity of steatohepatitis and the stage of fibrosis in human NAFLD.16 0.66 mM; b) OA: 0.66 mM and 1.32 mM and c) mixtures of OA
Lipotoxicity also contributes to decreased insulin sensitivity, and PA (ratio 2:1) at two different final concentrations: PA
perhaps by promoting the accumulation of fat-derived metabolites 0.33 mM + OA 0.66 mM (final fatty acids concentration 1 mM)
that inhibit insulin signalling and action.3,17 and PA 0.66 mM + OA 1.32 mM (final fatty acids concentration
Fatty acids are chemically classified as saturated and unsatur- 2 mM). Control cell cultures were incubated with plain medium or
ated (monounsaturated and polyunsaturated) and their structure with medium added with the vehicle in which fatty acids were
affects their biological effects. This is of particular relevance for dissolved. After 24 h of incubation with PA and OA alone or in
the most abundant FAs present in the diet and in serum, namely association, the extent of steatosis, apoptosis and gene expression
palmitic acid (PA), a saturated FA, and oleic acid (OA), a monoun- were evaluated as detailed above. In agreement with previous
saturated FA.18 In spite of the amount of information available for studies,19,27 we found 24 h to be optimal incubation time. At 12 h
the pathogenesis of triglyceride accumulation, little is known on or less the TG accumulation was low and no detectable variations
the metabolic effects of PA and OA as determinants of the patho- in gene expression could be shown (data not shown). A more
physiology of NAFLD. prolonged incubation time (e.g. 48 h and 72 h) while not providing
Studies on the effect of FAs-induced steatosis on cellular apo- a significant advantage in terms of intracellular TG accumulation
ptosis have demonstrated that PA and OA mixtures-induced ste- was associated with a significant decrease in cell viability at higher
atosis is associated with apoptosis in hepatocyte cell cultures.19 FFA concentrations in our and others’ experience (data not
Moreover, similar to other cell lines, monounsaturated FAs were shown,28).
less toxic also in hepatocytes20 and were able to prevent/attenuate After fixation with formaldehyde, neutral lipids were stained
PA toxicity.21 However, it remains unknown to what extent these using 0.5% Oil-Red-O (Sigma-Aldrich) in isopropanol for 30 min
effects were mediated by degree/type of lipid accumulated in the and nuclei were stained with hematoxylin.
hepatocytes. For the electron microscopy pictures cells were detached from
Aim of the present study was to evaluate the role of OA and PA, the substrate with trypsin, fixed in 2.5% glutaraldeyde for 1 h and
the major FA present in western diets, on steatogenesis, cell sur- than post-fixed in tyrode 1% OsO4 (osmium tetraoxyde) for 1 h.
vival, FAs composition, gene expression and insulin signalling in Cells were then dehydrated in progressive concentrations of
an in vitro model of steatosis. Given that a priori it could not be ethanol, dried in propylene oxide and embedded in Durcupan’s
ruled out that specific hepatocytic cell lines might differ in their resin. Sections 50 nm were cut on an Ultratatome (Reichert-Jung,
susceptibility to steatosis/apoptosis, we carried out experiments in Wetzlar, Germany), placed on mesh copper grids (S162, ASSING,
three different cell lines. Rome, Italy) and stained with 7% uranyl acetate and 2.66% lead
citrate. Micro-photographs were taken by an electron microscope
(JEOL, model 2011, Peabody, MA, USA) at 200 kV.
Methods Intracellular triglyceride content was evaluated after lysis of the
cells with NaOH 0.3 N. Triglyceride concentration (mg/dL) was
Hepatocyte cell cultures
determined by standard technique with an automatic analyzer
Three cell lines with different characteristics were used: (i) HepG2 (Roche, Milan, Italy) and normalised by protein content (mg/mL).
cells are derived from a well differentiated human hepatoblastoma Total intracellular lipid content was evaluated by Nile Red stain-
cell line that retain many characteristics of normal differentiated ing (Adipored, Cambrex); briefly, cells were grown in 96 black-
quiescent hepatocytes, and are p53 wild-type;22,23 (ii) WRL-68 plates and treated with FAs. At the end of incubation cells were
cells, a fetal liver cell line;24 and (iii) p53-mutated HuH7 cells, washed twice with phosphate buffered saline (PBS) and incubated
derived from a differentiated hepatocellular carcinoma.25 HepG2 with Adipored for 10 min. Fluorescence was evaluated as previ-
and WRL68 cells were purchased from Istituto Zooprofilattico ously described.29
Sperimentale (Brescia, Italy) and HuH7 cells from Japanese
Cancer Research Resources Bank (Osaka, Japan).
Evaluation of apoptosis
Long-chain FAs, palmitic (16:0) and oleic (18:1) were provided
as sodium salts (Sigma-Aldrich, Milan, Italy). Palmitic acid and Apoptosis, assessed with DAPI (4’,6-diamidino-2-phenylindole
OA were dissolved in MetOH 99% (stock solution 100 mM). dihydrochloride, Sigma-Aldrich) staining and caspases 3/7 activ-
Stock solutions were kept at -20°C before the experiments. Solu- ity (Promega, Milan, Italy) were evaluated as previously
tions and reagents used for cell cultures were from GIBCO Life described.29
Technologies Ltd (Grand Island, NY, USA).
Measurement of the fatty acid composition
Protocol of the study—induction and
Lipids were extracted from cells using the method of Folch et al.30
evaluation of steatosis
Aliquots were mildly saponified as previously described31 in order
Steatosis was induced by a slight modification of previously to obtain free fatty acids for high pressure liquid chromatography
described methods.26,27 HepG2, WRL-68 and HuH-7 cell cultures (HPLC) analysis. Separation of FAs was carried out with a Agilent
were incubated with phenol red-free medium containing 10% of 1100 HPLC system (Agilent, Palo Alto, CA, USA) equipped with
charcoal stripped fetal bovine serum (FBS; Cambrex, East a diode array detector. A C-18 Inertsil 5 ODS-2 Chrompack

Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors 831
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd
Fatty acids and apoptosis in hepatocytes M Ricchi et al.

column (Chrompack International BV, Middleburg, The Nether- total cellular proteins were separated by sodium dodecyl sulphate-
lands), 5 mm particle size, 150 ¥ 4.6 mm, was used with a mobile polyacrylamide gel electrophoresis (SDS-PAGE) and analyzed by
phase of CH 3 CN/H2O/CH 3 COOH (70/30/0.12, v/v/v) at a flow Western blot using primary antibodies as indicated. Detection was
rate of 1.5 mL/min. Unsaturated fatty acids were detected at performed using a chemiluminescent substrate (ECL, Amersham,
200 nm. Spectra (195–315 nm) of the eluate were obtained every IL, USA). Primary antibodies were rabbit Phospho-Akt (Ser-473)
1.28 s and were electronically stored.32 These spectra were taken Antibody (Cell Signalling Technology, Danvers, MA, USA) and
to confirm the identification of the HPLC peaks. mouse Anti-b-Actin antibody (Sigma-Aldrich).
Analysis of saturated FAs and further confirmation of unsatur-
ated fatty acids was carried out by GC assay of FA methyl esters.
Free fatty acids obtained as described above were methylated by Statistical analysis
the addition of 14% BF3/CH3OH at room temperature and imme- Results were expressed as mean ⫾ standard error (SE). All data
diately extracted into a solvent consisting of n-hexane and water represent a minimum of three experiments conducted in triplicate,
(4:3 ratio). After centrifugation to separate the two phases, the unless otherwise specified. The significance of differences was
hexane phase was saved and the aqueous phase was further assessed by Student’s t-test for independent data. Linear regres-
extracted by another round of hexane. The two hexane collections sion analysis was performed by the least square method. The
were combined, dried, and redissolved in 500 mL of n-hexane.33 differences between slope values were evaluated by the analysis of
The gas chromatograph (Model 6890, Agilent) was equipped variance. Significance was accepted at the P < 0.05 level. Statisti-
with split ratio of 20:1 injection port, a flame ionization detector cal analysis was performed with the aid of SPSS statistical soft-
(FID), an autosampler (Model 7673, Agilent), a 100 m HP-88 ware (version 14.0 for Windows, SPSS Inc., Chicago, IL, USA).
fused capillary column (Agilent), and an Agilent ChemStation
software system. The injector and detector temperatures were set
at 250°C and 280°C respectively. Hydrogen served as carrier gas Results
(1 mL/min), and the FID gases were H2 (30 mL/min), N2 (30 mL/
min), and purified air (300 mL/min). The temperature program OA and PA differentially affect triglyceride
was as follows: initial temperature was 120°C, programmed at accumulation in hepatocytic cell lines
10°C/min to 210°C and 5°C/min to 230°C, then programmed at We first analyzed the development of lipid accumulation in an in
25°C/min to 250°C and held for 2 min. vitro model of hepatic steatosis. HepG2 were exposed to increas-
ing concentrations of OA or PA, or to a combination of the two
Determination of cellular mRNA level fatty acids at a 2:1 ratio in favour of OA. After 24 h, lipid accu-
mulation was evident in all cells exposed to FA, as indicated by
In HepG2 cell line total RNA was isolated using the RNeasy lipid staining with Nile Red staining (Fig. 1). The degree of fat accu-
tissue kit (Qiagen, Milan, Italy) and quantified/checked with RNA mulation was roughly proportional to the concentration of FA to
Nano LabChip (Agilent, Milan, Italy). About 1 mg of total RNA
was reverse-transcribed with the high capacity cDNA Archive Kit
(Applied Biosystems, Monza Italy). TaqMan polymerase chain
reactions (PCR) were performed on cDNA samples using the
TaqMan Universal PCR Master Mix (Applied Biosystems)
according to PRISM 7900 HT Sequence Detection Systems.
The TaqMan strategies for each gene have been developed as
Assay-on-Demand by Applied Biosystems. Gene expression pro-
filing was achieved using the comparative cycle threshold (CT)
method of relative quantification (the calibrator samples were non-
treated cells, with 18S RNA used as endogenous control). Data are
expressed as log2 of the relative quantity (RQ) defined also as ‘fold
induction versus the controls’.

Western blot analysis of cellular proteins


Confluent, serum-starved (12 h) HepG2 were treated with fatty
acid, after stimulation with 100 nM of insulin for 15′, the cells
were quickly placed on ice, and washed with ice-cold PBS. The
mono-layer was lysed in RIPA buffer (20 mM Tris·HCl, pH 7.4, Figure 1 Effect of different fatty acids on lipid accumulation in HepG2
150 mM NaCl, 5 mM EDTA, 1% Nonidet P-40, 1 mM Na3VO4, cells. After 24 h of incubation (for details see materials and methods)
1 mM phenyl methyl sulfonyl fluoride, 0.01% protease inhibitor intracellular lipid droplets were measured by Nile Red. Data are
cocktail [Protease Inhibitor Cocktail, Sigma-Aldrich]) and trans- expressed as means ⫾ standard error of Relative Fluorescence Units
ferred to micro-centrifuge tubes. Insoluble proteins were discarded (RFLU) per mg of protein. Three experiments conducted in duplicate.
by centrifugation at 12 000 rpm at 4°C. Protein concentration in *P < 0.01 vs control; **P < 0.01 vs palmitic acid (PA) 0.33 mM;
the supernatant was measured in duplicate using a commercially ***P < 0.01 vs PA 0.66 mM; ****P < 0.01 vs oleic acid (OA) 0.66 mM;
available assay (Pierce, Rockford, IL, USA). Equal amounts of *****P < 0.01 vs PO1 mM.

832 Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd
M Ricchi et al. Fatty acids and apoptosis in hepatocytes

a) b)

Figure 2 Representative electron microscopy photograph. HepG2


cells in the absence (Panel a) or presence (Panel b) of fatty acids 1 mM
(palmitic acid [PA] 0.33 mM + oleic acid [OA] 0.66 mM) for 24 h.

which cells were exposed. Similar findings were obtained for


WRL68 and HuH7 cell lines (data not shown). Finally, electron
microscopy pictures displayed a massive increase in the number
and size of lipid droplets into the cytosol of HepG2 cells incubated
with FAs as compared to controls (Fig. 2).
To evaluate quantitatively the relation between the type of FA
and the degree of triglyceride accumulation, we measured triglyc-
eride (TG) concentration in cells lysates (Fig. 3). All treatments
with FA caused a significant increase in triglyceride content of all
three cell lines tested in comparison to control cells. In addition,
there was a clear dose-dependency for all conditions tested.
However, OA induced a significantly higher triglyceride accumu-
lation than PA when used at equimolar concentrations (Fig. 3).
This effect was evident in all three lines, and was statistically
significant for HepG2 and WRL-68. Incubation of all cell cultures
with mixtures of both FAs showed a dose dependent increase in the
amount of triglycerides which was almost equivalent to the sum of
triglyceride levels induced by separate incubation with the two
different FA. Finally, the concentration of triglyceride in HepG2
cells was always greater than that found in WRL-68 cells but lower
than in HuH-7 treated with the same experimental schedule. To
sum up, the three different cell lines display a more pronounced
steatosis response in the order PA > PA + OA > OA; for each FA a
dose-response relationship was observed. However, the absolute Figure 3 Effect of different fatty acids on triglycerides accumulation.
ability to accumulate triglycerides was decreasing in the order HepG2 (Panel a), WRL-68 (Panel b) and HuH-7 (Panel c) cell cultures
HuH-7 > HepG2 > WRL-68. were incubated with palmitic acid (PA) (0.33 and 0.66 mM), oleic acid
(OA) (0.66 and 1.32 mM) or mixtures of the two fatty acids (1 mM or
2 mM) for 24 h. Triglyceride (TG) accumulation was evaluated as the
concentration of TGs in cell lysates after NaOH lysis. Columns represent
PA is a stronger apoptotic stimulus than OA
mean values ⫾ standard error of three different experiments conducted
We next analyzed the effects of the different FAs treatments on in triplicate. *P < 0.05 vs control; **P < 0.05 vs PA 0.66 mM.
programmed cell death in the three different lines of hepatocytic
cells, using the percentage of cells showing nuclear condensation
when stained with DAPI29 (Fig. 4). Palmitic acid at a concentration
of 0.33 mM increased significantly (P < 0.05) the percentage of indicate that PA, but not OA induces apoptosis in cultured
apoptotic cells only in HuH7 cell cultures while at 0.66 mM apo- hepatocytes.
ptosis was significantly increased in all cell lines tested. In con- Co-incubation with the two FAs at the lower concentrations
trast, OA, used at concentrations as high as 1.32 mM did not (1 mM) did not modify the rate of apoptosis. At the higher con-
induce any increase in the number of apoptotic cells as compared centrations (0.66 mM PA together with 1.32 mM OA) an increase
to untreated controls. Remarkably, comparison of the effects of the in the percentage of apoptotic cells was observed in all cell lines.
two FA used at equimolar concentrations showed a significantly However, in two out of three lines, the increase in apoptosis was
higher ability of PA to induce apoptosis in all cell lines. These data significantly lower in the presence of OA than when PA at a

Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors 833
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd
Fatty acids and apoptosis in hepatocytes M Ricchi et al.

Figure 5 Effect of fatty acids on caspases 3/7 activity. In HepG2 (Panel


Figure 4 Effect of fatty acids on apoptosis rate. In HepG2 (Panel a), a), WRL-68 (Panel b) and HuH-7 (Panel c) caspases 3/7 activity was
WRL-68 (Panel b) and HuH-7 (Panel c) apoptosis was quantified by quantified measuring the fluorescence from Z-DEVD-R110 cleavage and
assessing the characteristic nuclear changes of apoptosis using the normalised by protein content in parallel plates. Columns represent
nuclear binding dye 4’,6-diamidino-2-phenylindole dihydrochloride mean values ⫾ standard error of three different experiments conducted
(DAPI) and counting cell numbers under a fluorescent microscopy. in triplicate. *P < 0.05 vs control; **P < 0.05 vs palmitic acid (PA)
Columns represent mean values ⫾ standard error of three different 0.66 mM; ***P < 0.05 vs PA 0.33 mM.
experiments conducted in triplicate. *P < 0.05 vs control; **P < 0.05 vs
palmitic acid 0.66 mM.

activity except in HepG2, where a significant increase was


observed. However, it should be stressed that the increase in
concentration of 0.66 mM was used alone. These data are strongly response to OA was only 1–2 fold as compared to control, while
suggestive of a protective effect of OA on apoptosis induced PA induced an 8–16 fold increase. As a result, caspases 3/7 activity
by PA. in HepG2 exposed to OA was always significantly lower than in
To add further evidence to the differential effects of the two fatty cells treated with PA.
acids on apoptosis, we measured the activity of caspases 3/7, that In co-incubation experiments, the addition of OA to PA
belong to the group of the execution caspases, activated once the decreased caspases activity in comparison to cells treated with PA
cell death program is fully operative (Fig. 5). In cells exposed to alone in HepG2 and in WRL68 but not in HuH7 cell line. Thus,
PA, the activity of caspases 3/7 increased in a dose-dependent data on caspases activity are closely similar to those obtained with
fashion. In contrast, OA did not have any effects on caspase DAPI staining.

834 Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd
M Ricchi et al. Fatty acids and apoptosis in hepatocytes

PA and OA are associated with different FA

584.360*

578.460*

991.330*

662.510*

1058.980*
355.990
58.228
440.570
36.410

69.171

28.104

64.352

90.473

92.724
Total FAs
profiles in HepG2 cells
We hypothesized that the mechanisms of cell toxicity due to palm-
itic acid could be ascribed to different fatty acid profiles and to the
esterified/unesterified ratio. To test this hypothesis, we compared

1.674
0.018
1.646
0.388
1.786
0.289
1.772
0.195
2.303
0.172
1.806
0.209
2.154
0.202
22:0
individual and total fatty acid composition obtained by HPLC with
the results of TG content assessed enzymatically.
The absolute FAs composition in steatotic HepG2 cultures after

1.500
0.213
1.771
0.885
1.814
0.173
1.616
0.120
2.120
0.238
1.676
0.154
1.480
0.319
20:0
exposure to different treatments is shown in Table 1. All treatments
induced significant changes in the concentration of various FAs.
PA and OA concentration increased after incubation with the

37.363*

33.767*
23.949
4.812
25.694
4.244
28.533
3.536
22.165
1.211

1.287
27.514
3.065

4.272
respective FA. In addition, while PA incubation was associated

18:0
with an increase 16:1, 18:1c11 and 14:0, the incubation with OA
induced an increase of 18:0, 16:0 and 14:0. As expected, the

150.288*

223.582*

152.017*

169.168*

280.682*
incubation with the two FAs induced a combined pattern and was

99.095
20.159

20.891

40.311
104.335
8.484

5.296

18.811

27.117
also associated with a reduction in the concentration of 16:1 as

16:0
compared to PA alone. The putative metabolic fate of FAs added to
the medium based on results shown in Table 1 is depicted in

2.654
0.082
2.791
0.653
3.405
0.431
3.404
0.581
3.447
0.300
3.084
0.363
3.601
0.019
Figure 6.

15:0
Total FA content was proportional to the moles of fatty acids
added to the medium (data not shown). Contrasting with data on

15.323*

17.634*

16.687*
TG content, we found that OA and PA at equimolar doses induced

10.308
2.169
12.053
0.067

1.112
12.083
1.790

2.344
13.505
3.052

1.022
a comparable accumulation of total fatty acids. This strongly sug-
14:0
Fatty acid profile of HepG2 cells incubated with palmitic acid (PA), oleic acid (OA) or PA and OA combined
gests that after incubation with PA the concentration of TG is lower
(Fig. 1) than total FA concentration due to the presence of a con-

2.659*
sistent FFA not being detected in the total TG assay. It could be 1.567
0.158
1.584
0.178
1.859
0.441
1.314
0.504
1.542
0.181
1.625
0.070

0.154
12:0

indirectly estimated that the percentage of FFAs following incu-


bation with PA is in the order of magnitude of 15% (vs 0–1%
following incubation with OA).
18:1 c11

97.634*

86.541*
63.996
10.617
74.485
0.586

5.627
66.224
3.265
92.485
2.594
70.854
21.176

5.623
FAs structure and not TG content is the key
determinant of apoptosis
291.330*

594.991*

293.478*

532.038*
80.847
15.220
85.252
5.729
99.727
9.266

3.547

29.215

35.181

35.333
To better define the relationship between the degree of steatosis
18:1

and induction of apoptosis, we plotted the intracellular TG con-


centration with the activity of caspases 3/7 (Fig. 7). While these
7.276
0.345
6.696
0.036
8.037
0.353
8.441
1.174
11.683
3.459
8.433
1.808
9.594
0.610
parameters were significantly correlated for both FA, the slope of
22:4

the curve obtained with PA was significantly steeper than the one
observed for OA. It is of relevance that the regression for com-
bined treatments fall in between PA and OA, again suggesting a
16.400
0.207
17.143
0.207
21.924
0.833
18.484
3.109
20.588
3.826
19.636
2.014
23.122
6.802
18:2

protective effect of OA on PA induced cytotoxicity. Similar rela-


tionships were observed also plotting TG versus percentage of
apoptosis evaluated by DAPI staining (data not shown). These data
8.966
0.392
8.180
0.291
10.138
1.054
10.061
1.151
11.745
1.948
9.466
0.433
10.301
0.662

confirm that for a given amount of intracellular triglycerides, the


20:4

Data are expressed as nmoles/mg of proteins.

degree of apoptosis depends on the FA used for the induction of


steatosis being far higher for PA, followed by their association and
47.159*

68.936*

49.472*

by OA.
31.346
3.168

1.939

3.297
30.651
2.227
35.756
12.192
36.038
3.719

10.301
16:1

Gene expression
5.139
0.603
4.653
0.274
5.336
0.415
5.218
0.539
6.240
1.246
5.061
0.319
5.445
0.263
22:6

The expression of PPARalpha, gamma and SREBP-1 constantly


increased following exposure to FAs. However, as shown in
*P < 0.05 vs control.

Figure 8, PA but not OA induced a significant increase in PPARa


1.274
0.065
1.176
0.043
1.327
0.033
1.362
0.208
1.415
0.054
1.167
0.100
1.436
0.025

expression vs controls (P < 0.05). In contrast, incubation of


20:5

HepG2 with OA but not with PA was associated, at both concen-


trations used, with an increase in the expression of PPARg vs
Table 1

control

controls (P < 0.05). In addition OA at the highest concentration


O 0,6

O 1,3
P 0,3

P 0,6

PO 1

PO 2
SD

SD

SD

SD

SD

SD

SD

increased the expression of SREBP-1 vs controls (P < 0.05). When

Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors 835
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd
Fatty acids and apoptosis in hepatocytes M Ricchi et al.

Figure 6 Putative metabolic pathway compatible with changes in fatty


acid profile observed after adding palmitic acid (PA) and oleic acid (OA)
to cell cultures base on findings shown in Table 1. PA (16:0) increases
the concentration of 16:1, 18:1c11 presumably by stimulating delta 9
desaturation of PA yielding 16:1, further elongation (18:1c11). The
increase of 14:0 may be ascribed to peroxisomal beta oxidation of PA.
OA (18:1c9) increases 18:0, 16:0 and 14:0. 18:0 increase might be due
to a partial inhibition of SCD1 with a consequent increase of 16:0 and
14:0 as products of peroxisomal beta oxidation of 18:0.

Figure 7 Relationship between extent of triglyceride (TG) accumula-


tion and caspases 3/7 activity. Regression lines represent the relation-
ship between the two parameters after incubation of HepG2 cells
respectively with palmitic acid (PA; 䉲), PA + oleic acid (OA; 䊉) and OA
(䊏). Single points are the mean of three experiments that have in
parallel evaluated TGs content and caspases activity. The slope values
of PA vs PA + OA (t = 6.805; P < 0.0001)and OA vs PA + OA (t = -5.353;
P < 0.0001) regression lines are significantly different.

the effect of two equimolar doses of PA and OA were compared,


the difference did not reach statistical significance.

Figure 8 Effect of palmitic acid (PA) and oleic acid (OA) on PPARg,
PA inhibits signaling downstream of the SREBP-1 and PPARa gene expression. Expression was evaluated by
insulin receptor real time-polymerase chain reaction (PCR; for details see materials and
methods). Data are reported as mean ⫾ standard error of log2(RQ) nor-
Peripheral IR, is caused, at least in part, by blunted transmission
malized to the controls. *P < 0.05 vs control.
of the signal along the enzymatic cascade downstream of the
insulin receptor. A critical step in this pathway is represented by

836 Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd
M Ricchi et al. Fatty acids and apoptosis in hepatocytes

accumulation also in other cell types, such as islet b cells.35 It is


tempting to associate the more effective steatogenic property of
OA with the specific pattern of gene expression (PPAR-gamma
and SREBP-1) shown in the present study. In contrast, the lesser
steatosis extent and increased apoptosis are due to PPAR-alpha
activation which results in enhanced b-oxidation and thus oxida-
tive stress.36
The most intriguing finding of the present study is the dissocia-
tion between the effects on steatosis and apoptosis of the different
FAs. Steatosis per se induces cell apoptosis.14,15 Other studies have
indicated a greater pro-apoptotic effect of PA as compared to OA,
but few studies have examined the relationship between steatosis
extent and apoptosis in hepatic cell cultures.20,28 The finding that
Figure 9 Effect of fatty acids (FAs) on Akt phosphorylation. Akt phos- OA is less efficient in the induction of apoptosis is in agreement
phorylation was evaluated in confluent, serum-starved (12 h) HepG2 with the recent work by Mahli et al. who, however, at variance
were treated with FAs, after stimulation with 100 nM of insulin for 15′ with our study, found no differences in the steatogenic effect of
and analyzed by Western blot using Phospho-Akt (Ser-473) Antibody. OA and PA.20 This discrepancy might probably be accounted for
by differences in the experimental conditions notably including the
lower (0.2 mM in Mahli’s vs 0.66 mM in our study) FA concen-
phosphorylation and activation of the serine/threonine kinase trations used.19 In this study, we have shown a correlation between
Akt, a central regulator of glucose uptake, anabolic metabolism TG accumulation and apoptosis with both OA and PA. However,
and antiapoptotic signals.34 We compared the effects of steatosis the slope of this function was markedly different comparing the
induced by the different FAs, alone or in association, on insulin- two FAs indicating that, for a given amount of TG accumulation,
induced phosphorylation of Akt (Fig. 9). Insulin caused a rapid the effects on apoptosis were dramatically enhanced when cells
and robust phosphorylation of Akt on Ser473, an activation- were treated with PA (Fig. 6).
specific residue, in HepG2 cells. Steatosis induced by PA was A large number of molecular mechanisms have been implicated
associated with a marked decrease of insulin-induced Akt phos- in PA-associated apoptosis: ceramide production, NO synthesis,
phorylation, while in cells treated with OA Akt phosphorylation suppression of antiapoptotic factors such as Bcl-2,15 reactive
was maintained. Interestingly, in cells exposed to both FA, an oxygen species generation,37 endoplasmic reticulum stress;37,38
intermediate level of phosphorylation was detected. No differ- nuclear factor-kB activation39 and decreased synthesis of cardio-
ences were found among different fatty acid treatments in the lipin.40 In hepatocytes, steatosis may enhance the expression of
activity of ERK1-2 (data not shown). Taken together, these data inflammatory cytokines, such as TNF-a, and increase apoptosis
demonstrate that PA and OA differentially affect the early signal- via Fas receptor activation.16,19,27 A JNK stimulated mitochon-
ing pathways downstream of the insulin receptor. drial20 and a reticulum stress-mediated9,21 apoptotic pathways have
been also reported. Death receptors are cell surface proteins, which
can trigger apoptosis when bound by their ligands. Ribeiro &
Discussion Cortez-Pinto41 observed enhanced expression of the death receptor
In this in vitro model of steatogenesis, three different lines of Fas in alcoholic hepatitis and tumor necrosis factor (TNF)
hepatocytes were exposed to increasing doses of FAs for receptor-1 in NASH. Others have identified enhanced Fas expres-
comparison purposes. The effects of administering OA, PA or sion in alcoholic and nonalcoholic steatohepatitis16,42 Thus steato-
their combination on TG accumulation, cell survival, FAs hepatitis may sensitize hepatocytes to extracellular death ligands
composition, gene expression and insulin signalling were (e.g. Fas ligand, tumor necrosis factor-alpha) promoting an extrin-
explored. Data showed that while the extent of steatosis was more sic pathway to apoptosis.19,27 NF-kB is a transcription factor that
severe in cells treated with OA than in those exposed to PA, can upregulate both death receptors and ligands.43,44 NF-kB acti-
opposite effects of the two FAs were found on cell apoptosis. The vation in Kupffer cells or infiltrating monocytes is proinflamma-
greater steatosis extent observed following exposure to OA was tory and induces the expression of death ligands such as TNFa.
associated with prominent PPARg and SREBP-1 activation. In FFAs can directly activate the IKK-k/ NF-kB pathway in hepa-
contrast, PA supplementation was associated with PPARa activa- tocytes via a lysosomal, cathepsin B– dependent mechanism.27
tion. Moreover, PA, but not OA, impaired insulin signalling. This pathway involves the translocation of Bax to lysosomes with
Despite the higher steatosis extent resulting from incubation with subsequent lysosomal destabilization and release of the cysteine
the two FAs combined, apoptosis rate and insulin signalling protease, cathepsin B, into the cytosol. This subsequently leads to
impairment were lower than in cells treated with PA alone, indi- the activation of NF-kB, via IKK-k, and a subsequent increase in
cating a protective effect of OA. the expression of TNFa apoptosis has increasingly been linked to
As expected, in our study the accumulation of TGs was propor- inflammation and hepatic fibrogenesis.45–47
tional to the concentration of FAs in the culture medium. However, More recently, Gores’ group48 reported OA to sensitise hepato-
OA was a more powerful trigger for TG accumulation than PA cytes in vitro to to the death ligand TRAIL. Oleic acid led to
irrespective of the cell line we used. This is a novel finding for upregulation of the cognate TRAIL receptor death receptor 5
hepatocytes and may represent a general property of OA, which whose expression was enhanced in steatotic human liver samples.
has been previously reported to induce a more marked fat DR5 was responsible for FFA sensitisation to TRAIL killing medi-

Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors 837
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd
Fatty acids and apoptosis in hepatocytes M Ricchi et al.

ated by a JNK dependent mechanism. Moreover, as previously incorporated in hepatic TG are mostly derived from the circulating
reported, if Kupffer cells are activated they produce death ligands, NEFA pool and thus reflect the adipose tissue FAs composition.60
including Fas ligand, and TNFa which, in turn, induce apoptosis Individuals consuming western-type high-fat diets show a serum/
in hepatocytes expressing death receptors,49 this might be also a tissue predominance of PA with a relative reduction in OA.6,10,11 In
condition in NASH patients. Given that inhibiting apoptosis may principle, characterization of FAs profile in the serum/liver may
well prevent the feared consequences of chronic liver disease, assist in predicting those individuals with progressive/severe
cirrhosis, portal hypertension, and liver failure thus drugs are NAFLD.20 Our data also highlight that variations in the desaturase
being developed to block apoptosis.50 activity might have a role in the type of FAs stored in hepatic TG
The cells’ ability to readily incorporate FAs into cytoplasmic and thus in the pathogenesis of human NAFLD. Conversely, PA
TGs might serve as a protection against their pro-apoptotic deprivation and OA enrichment in diet might be a goal to prevent/
effects.51 The finding that OA resulted in a greater steatosis extent treat the disease.
implies that this FA is more readily incorporated into TG and In conclusion, while a limitation of the current study is the use
therefore is associated with less apoptosis than PA which was less of three hepatocyte cultures of different origin but no data on
steatogenic. Along these lines, the cytoprotective effect of OA in primary hepatocyte culture, our study showing that OA is more
vitro could thus be explained by this FA’s ability to promote steatogenic but less damaging than PA in hepatocyte cell cultures
channelling of PA into TG synthesis, as demonstrated in cultured may provide a biological clue useful to a better understanding of
fibroblasts.51 The toxic effect of PA on hepatocytic cell cultures animal models of NAFLD. As far as human NAFLD is concerned,
could be, at least, in part related to the amount of FFA associated our data might suggest two major clinical implications/research
with incubation with this FA. However, given that this explanation hypothesis. First, for the diagnosis of NASH, steatosis extent
is based on in vitro indirect data, our hypothesis needs to be evaluated histologically might not be as relevant as its chemical
verified by further studies given that a recent report disproves the composition. Second, in agreement with recent studies envisaging
paradigm that FFAs are increased in human NAFLD.52 In addition for saturated FAs a role of ‘second hit’,61 the chemical composition
to PA channelling into TG synthesis, changes in the composition of of steatosis might help differentiate those non-progressive (‘inert’)
the intracellular FAs pool may represent an alternative strategy to forms of NAFLD from those that, due to their enrichment in
protect cells from PA-dependent apoptosis. The addition of PA to saturated FAs, are at a substantial risk of progression. These
the medium resulted in an increased proportion of the less dam- hypotheses are worthy being tested in specific studies.
aging palmitoleic acid indicating the attempt to ‘detoxify’ PA. This
detoxification might be achieved through increased steroyl-CoA
desaturase 1 (D9) activity (Fig. 8), which protects from Acknowledgments
PA-induced apoptosis.53
We thank Dorval Ganazzi for the assistance in statistical analysis.
Our data provide in vitro evidence for the principle that TG
We are also grateful to Dr. Cristiana Bertolani and Nadia Navari,
accumulation might be a defence mechanism against the toxicity
coworkers of Prof. Marra, for their contribution.
of excess FFAs. Yamaguchi et al. recently reached the same con-
Part of these data have been presented in abstract form at the
clusion in the obese mouse model, wherein inhibited triglyceride
European Association for the Study of the Liver, Wien, April 2006.
synthesis was associated with improved hepatic steatosis though
to the expenses of worsened hepatic inflammatory and fibrotic
changes.54 Observations that simple steatosis is rarely progressive
in humans55–57 further confirm that FAs stored as inert triglycerides
References
are harmless unless associated to another injuring factor, such as 1 Chawla A, Repa JJ, Evans RM, Mangelsdorf DJ. Nuclear receptors
IR, or oxidative stress, that may switch on the inflammatory and lipid physiology: opening the x-files. Science 2001; 294:
cascade. Familial heterozygous hypobetalipoproteinemia repre- 1866–70.
sents a naturally occurring model of human simple steatosis due to 2 Clarke SD. The multi-dimensional regulation of gene expression by
genetically impaired capacity to export VLDL from the hepato- fatty acids: polyunsaturated fats as nutrient sensors. Curr. Opin.
Lipidol. 2004; 15: 13–18.
cytes into the bloodstream. Of interest, in the absence of IR,
3 McGarry JD. Banting lecture 2001: dysregulation of fatty acid
steatosis in these individuals, although massive, does not appear to metabolism in the etiology of type 2 diabetes. Diabetes 2002; 51:
be progressive.58 Studies have shown that PA impairs insulin sig- 7–18.
nalling via increased JNK activity leading, in turn, to phosphory- 4 Haslam DW, James WP. Obesity. Lancet 2005; 366: 1197–209.
lation of insulin receptor substrate-1 and 2 at the inhibitory sites 5 Nehra V, Angulo P, Buchman AL, Lindor KD. Nutritional and
eventually leading to IR.59 Increased circulating FFAs concentra- metabolic considerations in the etiology of non-alcoholic
tions represent an early correlate of peripheral IR, which may be steatohepatitis. Dig. Dis. Sci. 2001; 46: 2347–52.
demonstrated to occur before the development of obesity in 6 Musso G, Gambino R, De Michieli F et al. Dietary habits and their
experimental condition.12 In this connection, our data have shown relations to insulin resistance and postprandial lipemia in
that PA administration, in itself, impaired insulin signalling while nonalcoholic steatohepatitis. Hepatology 2003; 37: 909–16.
7 Hernandez R, Martinez-Lara E, Canuelo A et al. Steatosis recovery
OA partially restored it. This finding implies that the steatosis
after treatment with a balanced sunflower or olive oil-based diet:
extent is not the only, nor perhaps the major, determinant of involvement of perisinusoidal stellate cells. World J. Gastroenterol.
impaired insulin action. Indeed, rat models7,9 and human stud- 2005; 11: 7480–5.
ies5,6,8 consistently support the concentrations and the chemical 8 Cortez-Pinto H, Jesus L, Barros H, Lopes C, Moura MC,
structure of FAs to be fundamental players in the development Camilo ME. How different is the dietary pattern in non-alcoholic
and progression of NAFLD. In human NAFLD, FAs that are steatohepatitis patients? Clin. Nutr. 2006; 25: 816–23.

838 Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd
M Ricchi et al. Fatty acids and apoptosis in hepatocytes

9 Wang D, Wei Y, Pagliassotti MJ. Saturated fatty acids promote 29 Ricchi M, Bertolotti M, Anzivino C et al. 17Beta-estradiol prevents
endoplasmic reticulum stress and liver injury in rats with hepatic cytotoxicity from hydrophobic bile acids in HepG2 and WRL-68 cell
steatosis. Endocrinology 2006; 147: 943–51. cultures. J. Gastroenterol. Hepatol. 2006; 21: 894–901.
10 De Almeida IT, Cortez-Pinto H, Fidalgo G, Rodrigues D, 30 Folch J, Lees M, Sloane Stanley GH. A simple method for the
Camilo ME. Plasma total and free fatty acids composition in human isolation and purification of total lipides from animal tissues. J. Biol.
non-alcoholic steatohepatitis. Clin. Nutr. 2002; 21: 219–23. Chem. 1957; 226: 497–509.
11 Videla LA, Rodrigo R, Araya J, Poniachik J. Oxidative stress and 31 Banni S, Carta G, Contini MS et al. Characterization of conjugated
depletion of hepatic long-chain polyunsaturated fatty acids may diene fatty acids in milk, dairy products, and lamb tissues. J. Nutr.
contribute to nonalcoholic fatty liver disease. Free Radic. Biol. Med. Biochem. 1996; 7: 150–5.
2004; 37: 1499–507. 32 Melis MP, Angioni E, Carta G et al. Characterization of conjugated
12 Samuel VT, Liu ZX, Qu X et al. Mechanism of hepatic insulin linoleic acid and its metabolites by RP-HPLC with diode array
resistance in non-alcoholic fatty liver disease. J. Biol. Chem. 2004; detector. Eur. J. Lipid Sci. Technol. 2001; 103: 617–21.
279: 32345–53. 33 Ip C, Banni S, Angioni E et al. Conjugated linoleic acid-enriched
13 Browning JD, Horton JD. Molecular mediators of hepatic steatosis butter fat alters mammary gland morphogenesis and reduces cancer
and liver injury. J. Clin. Invest. 2004; 114: 147–52. risk in rats. J. Nutr. 1999; 129: 2135–42.
14 Canbay A, Gieseler RK, Gores GJ, Gerken G. The relationship 34 Whiteman EL, Cho H, Birnbaum MJ. Role of Akt/protein kinase B
between apoptosis and non-alcoholic fatty liver disease: an in metabolism. Trends Endocrinol. Metab. 2002; 13: 444–51.
evolutionary cornerstone turned pathogenic. Z. Gastroenterol. 2005; 35 Cnop M, Hannaert JC, Hoorens A, Eizirik DL, Pipeleers DG. Inverse
43: 211–17. relationship between cytotoxicity of free fatty acids in pancreatic
15 Unger RH, Orci L. Lipoapoptosis: its mechanism and its diseases. islet cells and cellular triglyceride accumulation. Diabetes 2001; 50:
Biochim. Biophys. Acta 2002; 1585: 202–12. 1771–7.
16 Feldstein AE, Canbay A, Angulo P et al. Hepatocyte apoptosis and 36 Li Z, Berk M, McIntyre TM, Gores GJ, Feldstein AE. The
fas expression are prominent features of human nonalcoholic lysosomal-mitochondrial axis in free fatty acid-induced hepatic
steatohepatitis. Gastroenterology 2003; 125: 437–43. lipotoxicity. Hepatology 2008; 47: 1495–503.
17 Schmitz-Peiffer C, Craig DL, Biden TJ. Ceramide generation is 37 Listenberger LL, Ory DS, Schaffer JE. Palmitate-induced apoptosis
sufficient to account for the inhibition of the insulin-stimulated PKB can occur through a ceramide-independent pathway. J. Biol. Chem.
pathway in C2C12 skeletal muscle cells pretreated with palmitate. J. 2001; 276: 14890–5.
Biol. Chem. 1999; 274: 24202–10. 38 Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola M, Volchuk A.
18 Baylin A, Kabagambe EK, Siles X, Campos H. Adipose tissue Chronic palmitate but not oleate exposure induces endoplasmic
biomarkers of fatty acid intake. Am. J. Clin. Nutr. 2002; 76: reticulum stress, which may contribute to INS-1 pancreatic beta-cell
750–7. apoptosis. Endocrinology 2006; 147: 3398–407.
19 Feldstein AE, Canbay A, Guicciardi ME, Higuchi H, Bronk SF, 39 Rakatzi I, Mueller H, Ritzeler O, Tennagels N, Eckel J.
Gores GJ. Diet associated hepatic steatosis sensitizes to Fas Adiponectin counteracts cytokine- and fatty acid-induced apoptosis
mediated liver injury in mice. J. Hepatol. 2003; 39: 978–83. in the pancreatic beta-cell line INS-1. Diabetologia 2004; 47:
20 Malhi H, Bronk SF, Werneburg NW, Gores GJ. Free fatty acids 249–58.
induce JNK-dependent hepatocyte lipoapoptosis. J. Biol. Chem. 40 Ostrander DB, Sparagna GC, Amoscato AA, McMillin JB,
2006; 281: 12093–101. Dowhan W. Decreased cardiolipin synthesis corresponds with
21 Wei Y, Wang D, Topczewski F, Pagliassotti MJ. Saturated fatty acids cytochrome c release in palmitate-induced cardiomyocyte apoptosis.
induce endoplasmic reticulum stress and apoptosis independently of J. Biol. Chem. 2001; 276: 38061–7.
ceramide in liver cells. Am. J. Physiol. Endocrinol. Metab. 2006; 41 Ribeiro PS, Cortez-Pinto H, Sola S et al. Hepatocyte apoptosis,
291: E275–81. expression of death receptors and activation of NfkappaB in the liver
22 Javitt NB. Hep G2 cells as a resource for metabolic studies: of non-alcoholic and alcoholic steatohepatitis patients. Am. J.
lipoprotein, cholesterol, and bile acids. FASEB J. 1990; 4: Gastroenterol. 2004; 99: 1718–19.
161–8. 42 Natori S, Rust C, Stadheim LM et al. Hepatocyte apoptosis is a
23 Dashti N, Wolfbauer G. Secretion of lipids, apolipoproteins, and pathologic feature of human alcoholic hepatitis. J. Hepatol. 2001;
lipoproteins by human hepatoma cell line, HepG2: effects of oleic 34: 248–53.
acid and insulin. J. Lipid Res. 1987; 28: 423–36. 43 Ghosh S, May MJ, Kopp EB. NF-kappa B and Rel proteins:
24 Gutierrez-Ruiz MC, Bucio L, Souza V, Gomez JJ, Campos C, evolutionarily conserved mediators of immune responses. Annu. Rev.
Carabez A. Expression of some hepatocyte-like functional properties Immunol. 1998; 16: 225–60.
of WRL-68 cells in culture. In Vitro Cell. Dev. Biol. Anim. 1994; 44 Green DR. Death and NF-kappaB in T cell activation: life at the
30A: 366–71. edge. Mol. Cell 2003; 11: 551–2.
25 Nakabayashi H, Taketa K, Yamane T, Miyazaki M, Miyano K, 45 Faouzi S, Burckhardt BE, Hanson JC et al. Anti-Fas induces hepatic
Sato J. Phenotypical stability of a human hepatoma cell line, HuH-7, chemokines and promotes inflammation by an Nfkappa
in long-term culture with chemically defined medium. Gann 1984; B-independent, caspase-3-dependent pathway. J. Biol. Chem. 2001;
75: 151–8. 276: 49077–82.
26 Okamoto Y, Tanaka S, Haga Y. Enhanced GLUT2 gene expression 46 Canbay A, Guicciardi ME, Higuchi H et al. Cathepsin B inactivation
in an oleic acid-induced in vitro fatty liver model. Hepatol. Res. attenuates hepatic injury and fibrosis during cholestasis. J. Clin.
2002; 23: 138–44. Invest. 2003; 112: 152–9.
27 Feldstein AE, Werneburg NW, Canbay A et al. Free fatty acids 47 Canbay A, Taimr P, Torok N et al. Apoptotic body engulfment by a
promote hepatic lipotoxicity by stimulating TNF-alpha expression human stellate cell line is profibrogenic. Lab. Invest. 2003; 83:
via a lysosomal pathway. Hepatology 2004; 40: 185–94. 655–63.
28 Gomez-Lechon MJ, Donato MT, Martinez-Romero A, Jimenez N, 48 Malhi H, Barreyro FJ, Isomoto H, Bronk SF, Gores GJ. Free fatty
Castell JV, O’Connor JE. A human hepatocellular in vitro model to acids sensitise hepatocytes TRAIL mediated cytotoxicity. Gut 2007;
investigate steatosis. Chem. Biol. Interact. 2007; 165: 106–16. 56: 1124–31.

Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors 839
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd
Fatty acids and apoptosis in hepatocytes M Ricchi et al.

49 Canbay A, Feldstein AE, Higuchi H et al. Kupffer cell engulfment 56 Harrison SA, Neuschwander-Tetri BA. Nonalcoholic fatty liver
of apoptotic bodies stimulates death ligand and cytokine expression. disease and nonalcoholic steatohepatitis. Clin. Liver Dis. 2004; 8:
Hepatology 2003; 38: 1188–98. 861–79.
50 Feldstein A, Gores GJ. Steatohepatitis and apoptosis: therapeutic 57 Adams LA, Lymp JF, St Sauver J et al. The natural history of
implications. Am. J. Gastroenterol. 2004; 99: 1718–19. nonalcoholic fatty liver disease: a population-based cohort study.
51 Listenberger LL, Han X, Lewis SE et al. Triglyceride accumulation Gastroenterology 2005; 129: 113–21.
protects against fatty acid-induced lipotoxicity. Proc. Natl. Acad. Sci. 58 Lonardo A, Lombardini S, Scaglioni F et al. Hepatic steatosis and
USA 2003; 100: 3077–82. insulin resistance: does etiology make a difference? J. Hepatol.
52 Puri P, Baillie RA, Wiest MM et al. A lipidomic analysis of 2006; 44: 190–6.
nonalcoholic fatty liver disease. Hepatology 2007; 46: 1081–90. 59 Solinas G, Naugler W, Galimi F, Lee MS, Karin M. Saturated fatty
53 Warensjo E, Ohrvall M, Vessby B. Fatty acid composition and acids inhibit induction of insulin gene transcription by JNK-mediated
estimated desaturase activities are associated with obesity and phosphorylation of insulin-receptor substrates. Proc. Natl. Acad. Sci.
lifestyle variables in men and women. Nutr. Metab. Cardiovasc. Dis. USA 2006; 103: 16454–9.
2006; 16: 128–36. 60 Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD,
54 Yamaguchi K, Yang L, McCall S et al. Inhibiting triglyceride Parks EJ. Sources of fatty acids stored in liver and secreted via
synthesis improves hepatic steatosis but exacerbates liver damage lipoproteins in patients with nonalcoholic fatty liver disease. J. Clin.
and fibrosis in obese mice with nonalcoholic steatohepatitis. Invest. 2005; 115: 1343–51.
Hepatology 2007; 45: 1366–74. 61 Gentile CL, Pagliassotti MJ. The role of fatty acids in the
55 Matteoni CA, Younossi ZM, Gramlich T, Boparai N, Liu YC, development and progression of nonalcoholic fatty liver disease.
McCullough AJ. Nonalcoholic fatty liver disease: a spectrum of J. Nutr. Biochem. 2008; 19: 567–76.
clinical and pathological severity. Gastroenterology 1999; 116:
1413–19.

840 Journal of Gastroenterology and Hepatology 24 (2009) 830–840 © 2009 The Authors
Journal compilation © 2009 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd

Вам также может понравиться