ArticlePDF Available

Targeting AMP-activated protein kinase in adipocytes to modulate obesity-related adipokine production associated with insulin resistance and breast cancer cell proliferation

Authors:
  • Idorsia Pharmaceuticals Ltd

Abstract and Figures

Adipokines, e.g. TNFα, IL-6 and leptin increase insulin resistance, and consequent hyperinsulinaemia influences breast cancer progression. Beside its mitogenic effects, insulin may influence adipokine production from adipocyte stromal cells and paracrine enhancement of breast cancer cell growth. In contrast, adiponectin, another adipokine is protective against breast cancer cell proliferation and insulin resistance.AMP-activated protein kinase (AMPK) activity has been found decreased in visceral adipose tissue of insulin-resistant patients. Lipopolysaccharides (LPS) link systemic inflammation to high fat diet-induced insulin resistance. Modulation of LPS-induced adipokine production by metformin and AMPK activation might represent an alternative way to treat both, insulin resistance and breast cancer. Human preadipocytes obtained from surgical biopsies were expanded and differentiated in vitro into adipocytes, and incubated with siRNA targeting AMPKalpha1 (72 h), LPS (24 h, 100 μg/ml) and/or metformin (24 h, 1 mM) followed by mRNA extraction and analyses. Additionally, the supernatant of preadipocytes or derived-adipocytes in culture for 24 h was used as conditioned media to evaluate MCF-7 breast cancer cell proliferation. Conditioned media from preadipocyte-derived adipocytes, but not from undifferentiated preadipocytes, increased MCF-7 cell proliferation (p < 0.01). Induction of IL-6 mRNA by LPS was reduced by metformin (p < 0.01), while the LPS-induced mRNA expression of the naturally occurring anti-inflammatory cytokine interleukin 1 receptor antagonist was increased (p < 0.01). Silencing of AMPKalpha1 enhanced LPS-induced IL-6 and IL-8 mRNA expression (p < 0.05). Adipocyte-secreted factors enhance breast cancer cell proliferation, while AMPK and metformin improve the LPS-induced adipokine imbalance. Possibly, AMPK activation may provide a new way not only to improve the obesity-related adipokine profile and insulin resistance, but also to prevent obesity-related breast cancer development and progression.
Content may be subject to copyright.
SHOR T REPOR T Open Access
Targeting AMP-activated protein kinase in
adipocytes to modulate obesity-related adipokine
production associated with insulin resistance and
breast cancer cell proliferation
Jean Grisouard
1*
, Kaethi Dembinski
1
, Doris Mayer
2
, Ulrich Keller
1,3
, Beat Müller
4
and Mirjam Christ-Crain
1,3
Abstract
Background: Adipokines, e.g. TNFa, IL-6 and leptin increase insulin resistance, and consequent hyperinsulinaemia
influences breast cancer progression. Beside its mitogenic effects, insulin may influence adipokine production from
adipocyte stromal cells and paracrine enhancement of breast cancer cell growth. In contrast, adiponectin, another
adipokine is protective against breast cancer cell proliferation and insulin resistance.
AMP-activated protein kinase (AMPK) activity has been found decreased in visceral adipose tissue of insulin-
resistant patients. Lipopolysaccharides (LPS) link systemic inflammation to high fat diet-induced insulin resistance.
Modulation of LPS-induced adipokine production by metformin and AMPK activation might represent an
alternative way to treat both, insulin resistance and breast cancer.
Methods: Human preadipocytes obtained from surgical biopsies were expanded and differentiated in vitro into
adipocytes, and incubated with siRNA targeting AMPKalpha1 (72 h), LPS (24 h, 100 μg/ml) and/or metformin (24 h,
1 mM) followed by mRNA extraction and analyses. Additionally, the supernatant of preadipocytes or derived-
adipocytes in culture for 24 h was used as conditioned media to evaluate MCF-7 breast cancer cell proliferation.
Results: Conditioned media from preadipocyte-derived adipocytes, but not from undifferentiated preadipocytes,
increased MCF-7 cell proliferation (p < 0.01). Induction of IL-6 mRNA by LPS was reduced by metformin (p < 0.01),
while the LPS-induced mRNA expression of the naturally occurring anti-inflammatory cytokine interleukin 1
receptor antagonist was increased (p < 0.01). Silencing of AMPKalpha1 enhanced LPS-induced IL-6 and IL-8 mRNA
expression (p < 0.05).
Conclusions: Adipocyte-secreted factors enhance breast cancer cell proliferation, while AMPK and metformin
improve the LPS-induced adipokine imbalance. Possibly, AMPK activation may provide a new way not only to
improve the obesity-related adipokine profile and insulin resistance, but also to prevent obesity-related breast
cancer development and progression.
Introduction
Breast cancer is the most frequent cancer type among
women worldwide, and 21% of all breast cancer deaths
worldwide are estimated to be attributable to obesity
and physical inactivity [1,2]. The incidence of type 2 dia-
betes mellitus (T2DM) is presumed to be a direct result
of the obesity epidemic [3]. The breast cancer risk is
increased in diabetic women independently from obesity
[4,5]. Thus, both, obesity and diabetes are risk factors
for breast cancer and the prevalence of each of these
diseases will continue to rise worldwide [6].
Adipokines are polypeptides produced and secreted by
the adipose tissue and dysregulation of their production
and secretion in the obese state leads to obesity-related
complications [7,8]. Invasive breast tumours break
through the basement membrane and infiltrate fibrous
* Correspondence: jean.grisouard@unibas.ch
1
Department of Biomedicine, University Hospital Basel, Basel, CH-4031,
Switzerland
Full list of author information is available at the end of the article
Grisouard et al.Diabetology & Metabolic Syndrome 2011, 3:16
http://www.dmsjournal.com/content/3/1/16 METABOLIC SYNDROME
DIABETOLOGY &
© 2011 Grisouard et al; licensee BioMed Cen tral Ltd. This is an Open Access article distributed under the terms of the Creative
Commons Attri bution License (http://creativecommons.org /licenses/by/2.0), which permits unrestricted use, distribution, and
reproductio n in any medium, provided the original work is properly cited.
tissue barriers, resulting in an immediate juxtaposition
of adipocytes and breast cancer cells, thus allowing para-
crine interactions between the two cell types [9]. Insulin
resistance and consequent hyperinsulinaemia may be a
common factor linking T2DM and cancer [10]. Beside
its stimulating effects on breast cancer cell proliferation
[11], insulin may influence adipokine production from
adipose tissue and adipocyte stromal cells and may
indirectly reinforce cancer cell growth. Similarly, insulin
resistance-induced hyperglycaemia and hyperlipidaemia
may alter adipokine production in adipocytes and
enhance breast cancer development [12].
Chronic low grade systemic inflammation is associated
with obesity and insulin resistance. Lipopolysaccharide
(LPS) from the gut microbiota is a triggering factor link-
ing inflammation to high fat diet-induced metabolic syn-
drome [13,14]. Metformin is the first line oral
antidiabetic drug for patients with T2DM [15]. AMP-
activated protein kinase (AMPK) targets cytokine secre-
tion from the adipose tissue and the activation of this
kinase by metformin could explain the beneficial effects
of this drug on inflammation [16].
In view of these relationships, we aimed to assess the
effect of low grade inflammation induced by LPS and of
metformin on adipokine production in human adipo-
cytes and to observe the stimulatory effect of adipocyte-
secreted factors on breast cancer cell proliferation.
Materials and methods
The study was approved by the local Ethics Committee
and informed consent was obtained from patients. Sub-
cutaneous fat tissue samples were obtained from obese
donors (BMI > 30 kg/m
2
, males and females, mean age
47 years) during elective abdominal surgery performed
for various non-malignant conditions.
Preadipocytes were isolated, expanded in vitro until
confluence and subjected to adipogenic differentiation
medium for 14 days as previously described [17,18].
After differentiation, adipocytes were washed twice with
warm phosphate buffered saline (PBS) and were kept for
48 h in low glucose (5 mM) medium. Then, adipocytes
were pre-incubated with 1 mM metformin (Sigma-
Aldrich, Buchs, Switzerland) for 1 h followed by treat-
ment with 100 ng/ml LPS (Sigma-Aldrich). AMPKa1
silencing was performed as previously described [18].
Low glucose DMEM medium with 2% dextran-coated
charcoal-treated heat-inactivated FCS (DCC-FCS, Lubio
Science, Luzern, Switzerland) was added to confluent
preadipocytes or preadipocyte-derived adipocytes. After
24 h in culture, the supernatant or conditioned medium
was collected, filtered with a 0.2 μm syringe filter and
stored at -20°C.
MCF-7 breast cancer cells were maintained in DMEM
(25 mM glucose) containing 10% FCS (Lubio Science).
Then, cells were grown in medium supplemented with
10% DCC-FCS for 72 h. 1 × 10
4
cancer cells/well were
plated in a 96-well plate in medium containing 2%
DCC-FCS for 24 h and stimulated every 24 h for a total
of 72 h with low glucose DMEM + 2% DCC-FCS or
conditioned medium. At the end of incubation time,
cells were washed twice with PBS, fixed for 5 min with
100 μl of 3% paraformaldehyde and stained for 10 min
with 100 μl of 1% crystal violet dye dissolved in 10%
ethanol. Plates were extensively washed with water to
remove traces of unbound crystal violet dye. After air
drying, the bound dye was dissolved in 100 μl of 10%
acetic acid. Optical density was read at 595 nm using a
plate reader (Bucher biotec, Basel, CH) [11].
For quantitative analysis of adipokine expression, RNA
was isolated and 1 μg total RNA was subjected to
reverse transcription-PCR. cDNA was subjected to
quantitative real-time PCR analysis using the power
Sybr
®
-Green PCR master mix (Applied Biosystems) and
the ABI 7500 Sequence detection system [19]. Primers
were designed as follows: IL-6 sense primer, 5-
TCTTCAGAACGAATTGACAAACAAA-3, IL-6 anti-
sense primer, 5-GCTGCTTTCACACATGTTACTC
TTG-3, IL-8 sense primer, 5-GCCATAAAGT-
CAAATTTAGCTGGAA-3, IL-8 antisense primer, 5-
GTGCTTCCACATGTCCTCACA-3; interleukin 1
receptor antagonist (IL-1RA) sense primer, 5-TGC
CTGTCCTGTGTCAAGTC-3and IL-1RA antisense
primer, 5-TCTCGCTCAGGTCAGTGATG-3.Hypox-
anthine-guanine phosphoribosyltransferase (HPRT) pri-
mers were used as loading control (HPRT sense primer,
5-TCAGGCAGTATAATCCAAAGATGGT-3and
HPRT antisense primer, 5-AGTCTGGCTTATATC-
CAACACTTC-3.
Data are presented as mean ± standard deviation (SD)
from a minimum of three independent experiments.
One-way analysis of variance was performed and the
Tukeys posthoc multiple comparison test was applied.
Overall, a Pvalue < 0.05 was considered significant.
Results
Conditioned medium (CM) from preadipocytes and pre-
adipocyte-derived adipocytes (14 days after differentia-
tion) was collected and breast cancer MCF-7 cell
proliferation was evaluated after 72 h in these media (5
mM glucose) (Figure 1). DMEM medium (5 mM glu-
cose) was used as basal. CM from preadipocytes did not
increase MCF-7 cell proliferation (p = n.s.). However,
CM from preadipocyte-derived adipocytes increased
MCF-7 cell proliferation to 1.28 ± 0.02 fold (p < 0.001
vs. basal and p < 0.01 vs. CM preadipocytes).
Incubation of preadipocyte-derived adipocytes with
100 ng/ml LPS increased IL-6 mRNA expression to 10.5
± 1.9 fold (p < 0.001 vs. basal)(Figure 2A) and induction
Grisouard et al.Diabetology & Metabolic Syndrome 2011, 3:16
http://www.dmsjournal.com/content/3/1/16
Page 2 of 7
of IL-6 mRNA by LPS was significantly reduced by 1
mM metformin to 5.3 ± 0.8 fold (p < 0.01 vs. LPS).
Additionally, 1 mM metformin enhanced LPS-induced
mRNA expression of IL-1RA, a naturally occurring anti-
inflammatory cytokine (p < 0.01 vs. LPS) (Figure 2B).
We silenced AMPKa1 subunit in preadipocyte-derived
adipocytes to check its involvement in the effects of LPS
on adipocytokine mRNA expression. Using siRNA pool
specifically targeting AMPKa1, AMPKa1 mRNA expres-
sion and protein content were decreased to 0.28 ± 0.03
and 0.50 ± 0.08 fold, respectively (p < 0.001 vs. control
siRNA)(data not shown). AMPKa1 silencing enhanced
LPS-induced IL-6 and IL-8 mRNA expression (Figure 3,
p < 0.05 vs. LPS in control siRNA cells).
Discussion
Decreased AMPK activity has been found in visceral
adipose tissue of patients with central obesity due to
Cushings syndrome [20] and of obese insulin-resistant
individuals [21]. This suggests a central role for this
enzyme in obesity and related insulin resistance. We
therefore aimed to investigate how AMPK modulates
adipokine production triggered by obesity- and type 2
diabetes mellitus (T2DM)-related factors and how such
a modulation may prevent insulin resistance and breast
tumour cell proliferation. In turn, adipokines alter
AMPK activity and might play a crucial role in adipo-
kine-altered insulin sensitivity and breast tumour cell
growth (Figure 4). Our findings suggest that conditioned
medium from human adipocytes increase breast cancer
cell proliferation. Our data also show that metformin
and AMPK alter LPS-induced adipokine expression,
favoring the anti-inflammatory adipokine expression and
decreasing pro-inflammatory adipokine expression in
human adipocytes.
Interestingly, CM obtained from murine 3T3-L1 fully
differentiated adipocytes induced growth of MCF-7 cells
Figure 1 Effect of adipocyte conditioned medium (CM) on
breast cancer cell proliferation. MCF-7 breast cancer cells were
incubated for 72 h with low glucose DMEM medium (basal), low
glucose DMEM conditioned medium from preadipocytes (CM
Preadipocytes) or low glucose DMEM conditioned medium from
preadipocyte-derived adipoytes (CM Adipocytes). Then, proliferation
of MCF-7 cells was measured by colorimetric method using crystal
violet dye. Results are expressed as fold of basal (**, p < 0.01 vs. CM
Preadipocytes; ***, p < 0.001 vs. basal; n.s., not significant).
Figure 2 Metformin modulates LPS-induced adipocytokine expression. Human subcutaneous preadipocyte-derived adipocytes were pre-
incubated for 1 h with 1 mM metformin followed by stimulation with 100 ng/ml LPS for 24 h. mRNA expression of IL-6 (A) and interleukin-1
receptor antagonist (IL1RA) (B) were analysed by quantitative real-time PCR. (*, p < 0.05 vs. basal; **, p < 0.01 vs. LPS; ***, p < 0.001 vs. basal; n.s.,
not significant).
Grisouard et al.Diabetology & Metabolic Syndrome 2011, 3:16
http://www.dmsjournal.com/content/3/1/16
Page 3 of 7
Figure 3 AMPKalpha1 prevents the LPS-induced IL-6 and IL-8 adipocytokine expression. Human subcutaneous preadipocyte-derived
adipocytes were transfected for 72 h with control non-targeting siRNA pool (Control siRNA) or siRNA pool specifically targeting AMPKalpha1
(AMPKa1 siRNA) and incubated for 24 h with 100 ng/ml LPS. mRNA expression of IL-6 (A) and IL-8 (B) was analysed by quantitative real-time
PCR. (*, p < 0.05 vs. LPS in control siRNA cells; ***, p < 0.001 vs. basal in control siRNA cells).
Figure 4 Summary of our research aims and hypotheses. Our preliminary results suggest that AMPK modulates the adipokine expression
and secretion profiles from human adipocytes upon stimulation by inflammatory and/or metabolic factors. Imbalance in the adipokine profile
triggers insulin resistance and breast cancer cell growth. Moreover, adipokines alter AMPK activity and modulation of this activity might inhibit
the downstream effects of pro-inflammatory and pro-proliferative adipokines (TNFa, IL-6, IL-8, leptin) and improve the effects of anti-
inflammatory and anti-proliferative adipokines (adiponectin, IL1-RA) in insulin resistant tissues and breast tumours. AMPK: AMP-dependent protein
kinase, LPS: lipopolysaccharides, FFA: free fatty acids, TNFa: tumour necrosis factor a, IL-6: interleukin 6, IL-8: interleukin-8, IL1RA: interleukin-1
receptor antagonist).
Grisouard et al.Diabetology & Metabolic Syndrome 2011, 3:16
http://www.dmsjournal.com/content/3/1/16
Page 4 of 7
and such changes were attributed to reduced apoptosis
rather than increased proliferation [22]. In our study,
apoptosis was not analysed by specific assays. However,
floating dead cells were removed by washing of the cell
layer with PBS to eliminate most of the dead cells before
fixation and staining. Nevertheless, we cannot exclude
that apoptotic cells still attached to the culture dish
were also stained by crystal violet under our experimen-
tal conditions. Further experiments will be needed to
expand our findings in other parameters which are
important for tumour progression such as apoptosis,
invasion and migration.
Additionally, this doctoral thesis reported that condi-
tioned media from murine preadipocytes was less effi-
cient on MCF-7 cell proliferation and that low glucose
medium reduced the effect of murine adipocyte condi-
tioned media on MCF7 growth [22]. Since incubation
with low glucose medium reduced IGF1mRNA levels in
adipocytes and since adipocyte conditioned media prolif-
erative effect was reverted by inhibiting the IGF-1 path-
way, the author suggests that adipocyte-produced IGF1
has a crucial role in promoting cancer cell growth [22].
Similar results were obtained regarding human preadi-
pocyte conditioned media in our study. To avoid the
possible effects of high glucose concentration on adipo-
cyte secretion such as IGF in the culture supernatant,
our human adipocytes were incubated in low glucose (5
mM) medium before collecting the conditioned media.
Low glucose medium might prevent the IGF release and
this suggests that other adipokines are involved in breast
cancer cell growth.
To obtain our data, both male and female adipose tis-
sues were considered since the number of tissue avail-
able from females was not sufficient. The small size of
the studied cohort of patients does not allow drawing
conclusions about any gender specific differences in pro-
liferation. Further work should be performed and adi-
pose tissues from females are certainly more relevant
regarding breast cancer proliferation. Ultimately, differ-
ence in a variety of parameters between fat depots is
important and fat tissue isolated from the breast should
be considered. Also, discrepancies between adipocytes
from different donors were observed and the results
were not consistent for both LPS-induced IL-1RA
mRNA expression after AMPKa1-silencing and LPS-
induced IL-8 mRNA expression upon metformin incu-
bation (data not shown). This suggests that metformin
and AMPK might alter the expression of some adipo-
kines (e.g. IL-6) in a similar way and expression of some
other adipokines in a different manner. Therefore, met-
formin might have some AMPK-dependent and -inde-
pendent effects, which could be unraveled if AMPK
expression is silenced upon metformin incubation.
Further experiments also need to be performed to check
the effects of metformin and AMPK modulation of adi-
pokine expression on breast cancer cell proliferation.
Recent studies demonstrated that metformin treat-
ment increased adiponectin plasma levels in women
with polycystic ovary syndrome (PCOS) [23] and T2DM
patients [24] while metformin treatment tends to
decrease IL-6 plasma levels [23] as well as circulating
levels of leptin [25] in PCOS patients, and significantly
reduces resistin and TNF-aplasma levels [24] in T2DM
patients. Moreover, metformin inhibition of TNF-a-
induced IL-6 secretion was abolished after silencing of
AMPKa1 in human umbilical vein endothelial cells [26].
Together with our data, this suggests that metformin,
via AMPK activation, improves the adipokine profile
from human adipocytes in overweight and obese
patients. This might contribute to a faster and better
improvement of the chronic low grade inflammatory
state and therefore insulin resistance in these patients.
In turn, adiponectin-induced AMPK activity in skeletal
muscles is involved in the regulation of mitochondrial
function and oxidative stress, glucose and lipid metabo-
lism, and exercise endurance [27]. The same study
further suggests that agonism of adiponectin signaling in
muscles provides a new treatment modality for insulin
resistance.
In patients with breast cancer, the effects of metfor-
min and AMPK on the adipokine imbalance might
diminish tumor progression since the present study
showed that adipocyte conditioned medium promoted
breast cancer cell proliferation. Decreasing pro-inflam-
matory adipokine secretion from adipocyte stromal cells
will reduce breast cancer cell growth and proliferation.
Increasing anti-inflammatory adipokines such as adipo-
nectin will inhibit invasion and migration of breast can-
cer cells. Indeed, adiponectin targets AMPK activity in
breast cancer cells [28]. This leads to an inhibition of
the mammalian target of rapamycin (mTOR) pathway
and a decrease in protein synthesis, inhibition of adhe-
sion, migration and invasion of breast cancer cells.
These results correlate with the activation of AMPK by
metformin in breast cancer cells [29]. However and
unlike adiponectin, other adipokines (e.g. leptin) might
differently alter AMPK activity in various tissues [30].
Therefore, it will be crucial to study the effect of each
adipokine on AMPK activity in breast cancer cells.
In conclusion, the present study shows some prelimin-
ary but encouraging data to further unravel the specific
role of AMPK and adipokines to alleviate obesity-related
insulin resistance and breast cancer complications. Acti-
vation of AMPK might improve adipokine production
triggered by obesity- and diabetes-related factors in
human adipocytes. In turn, some adipokines might acti-
vate AMPK and increase insulin sensitivity and inhibits
breast tumor development.
Grisouard et al.Diabetology & Metabolic Syndrome 2011, 3:16
http://www.dmsjournal.com/content/3/1/16
Page 5 of 7
Acknowledgements and funding
This study was supported by grants from the University
of Basel (Forschungsfonds-Förderung für Nachwuchs-
forschende der Universität Basel) and the Stiftung der
Diabetes-Gesellschaft Region Basel to Jean Grisouard, by
grants from the Novartis Stiftung für Medizinisch-Biolo-
gische Forschung and the Swiss National Research
Foundation (PP00P3_123346/1) to Mirjam Christ-Crain
and a grant from Sanofi-Aventis to Beat Müller.
We thank Dr. Daniel M. Frey (Department of Surgery,
Div. of General Surgery and Surgical Research, Univer-
sity Hospital Basel) and Dr. Ralph Peterli (Department
of Surgery, Claraspital Basel) for providing us with
human adipose tissue samples.
Author details
1
Department of Biomedicine, University Hospital Basel, Basel, CH-4031,
Switzerland.
2
Hormones and Signal Transduction, German Cancer Research
Centre, DKFZ-ZMBH Alliance, Heidelberg, D-69120, Germany.
3
Division of
Endocrinology, Diabetes and Clinical Nutrition, University Hospital Basel,
Basel, CH-4031, Switzerland.
4
Medical University Clinic, Kantonsspital Aarau,
Aarau, CH-5001, Switzerland.
Authorscontributions
JG conceived of the study, participated in its design and coordination,
carried out the breast cancer cell proliferation assays and the AMPKalpha1
silencing of the adipocytes, analysed the results and drafted the manuscript.
KD carried out the adipocyte and breast cancer cell culture work and the
mRNA analyses. DM, UK, BM and MCC participated in the design of the
study and coordination, and helped to draft the manuscript. All authors read
and approved the final manuscript.
Competing interests
The authors declare that they have no competing interests.
Received: 19 January 2011 Accepted: 20 July 2011
Published: 20 July 2011
References
1. Danaei G, Vander Hoorn S, Lopez AD, Murray CJ, Ezzati M: Causes of
cancer in the world: comparative risk assessment of nine behavioural
and environmental risk factors. Lancet 2005, 366:1784-1793.
2. Carmichael AR: Obesity and prognosis of breast cancer. Obes Rev 2006,
7:333-340.
3. Mokdad AH, Ford ES, Bowman BA, Dietz WH, Vinicor F, Bales VS, Marks JS:
Prevalence of obesity, diabetes, and obesity-related health risk factors,
2001. Jama 2003, 289:76-79.
4. Larsson SC, Mantzoros CS, Wolk A: Diabetes mellitus and risk of breast
cancer: a meta-analysis. International journal of cancer 2007, 121:856-862.
5. Vigneri P, Frasca F, Sciacca L, Pandini G, Vigneri R: Diabetes and cancer.
Endocr Relat Cancer 2009, 16:1103-1123.
6. Obesity and overweight/Diabetes/Breast cancer. [http://www.who.int/].
7. Kershaw EE, Flier JS: Adipose tissue as an endocrine organ. J Clin
Endocrinol Metab 2004, 89:2548-2556.
8. Calle EE, Kaaks R: Overweight, obesity and cancer: epidemiological
evidence and proposed mechanisms. Nat Rev Cancer 2004, 4:579-591.
9. Lorincz AM, Sukumar S: Molecular links between obesity and breast
cancer. Endocr Relat Cancer 2006, 13:279-292.
10. Maccio A, Madeddu C, Mantovani G: Adipose tissue as target organ in the
treatment of hormone-dependent breast cancer: new therapeutic
perspectives. Obes Rev 2009, 10:660-670.
11. Shukla A, Grisouard J, Ehemann V, Hermani A, Enzmann H, Mayer D:
Analysis of signaling pathways related to cell proliferation stimulated by
insulin analogs in human mammary epithelial cell lines. Endocr Relat
Cancer 2009, 16:429-441.
12. Wellen KE, Hotamisligil GS: Inflammation, stress, and diabetes. J Clin Invest
2005, 115:1111-1119.
13. Erridge C, Attina T, Spickett CM, Webb DJ A: High-fat meal induces low-
grade endotoxemia: evidence of a novel mechanism of postprandial
inflammation. Am J Clin Nutr 2007, 86:1286-1292.
14. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM,
Fava F, Tuohy KM, Chabo C, Waget A, Delmee E, Cousin B, Sulpice T,
Chamontin B, Ferrieres J, Tanti JF, Gibson GR, Casteilla L, Delzenne NM,
Alessi MC, Burcelin R: Metabolic endotoxemia initiates obesity and insulin
resistance. Diabetes 2007, 56:1761-1772.
15. Bosi E: Metformin, the gold standard in type 2 diabetes: what does the
evidence tell us? Diabetes Obes Metab 2009, 11(Suppl 2):3-8.
16. Daval M, Foufelle F, Ferre P: Functions of AMP-activated protein kinase in
adipose tissue. J Physiol 2006, 574:55-62.
17. Grisouard J, Bouillet E, Timper K, Radimerski T, Dembinski K, Frey DM,
Peterli R, Zulewski H, Keller U, Müller B, Christ-Crain1 M: Both inflammatory
and classical lipolytic pathways are involved in lipopolysaccharides
(LPS)-induced lipolysis in human adipocytes. Innate Immunity 2010.
18. Grisouard J, Timper K, Radimerski TM, Dembinski K, Frey DM, Kola B,
Korbonits M, Herrmann P, Krähenbühl S, Zulewski H, Keller U, Müller B,
Christ-Crain M: Mechanisms of metformin action on glucose transport
and metabolism in human adipocytes. Biochem Pharmacol 2010,
80:1736-1745.
19. Linscheid P, Seboek D, Zulewski H, Keller U, Muller B: Autocrine/paracrine
role of inflammation-mediated Calcitonin Gene-Related Peptide and
Adrenomedullin expression in human adipose tissue. Endocrinology 2005,
146:2699-708.
20. Christ-Crain M, Kola B, Lolli F, Fekete C, Seboek D, Wittmann G, Feltrin D,
Igreja SC, Ajodha S, Harvey-White J, Kunos G, Muller B, Pralong F, Aubert G,
Arnaldi G, Giacchetti G, Boscaro M, Grossman AB, Korbonits M: AMP-
activated protein kinase mediates glucocorticoid induced metabolic
changes: a novel mechanism in Cushings syndrome. Faseb J 2008,
22:1672-1683.
21. Gauthier MS, Mott M, Gokce N, Apovian C, Ruderman NB: Decreased AMP-
activated protein kinase activity is associated with markers of
inflammation and infiltration of immune cells in visceral and
subcutaneous adipose tissue, and with whole-body insulin resistance in
obese patients [abstract]. Obesity 2009, 17:s93.
22. DEsposito V: Control of breast cancer cell growth by adipocyte-released
factors. [PhD Thesis] 2009.
23. Jakubowska J, Bohdanowicz-Pawlak A, Milewicz A, Szymczak J, Bednarek-
Tupikowska G, Demissie M: Plasma cytokines in obese women with
polycystic ovary syndrome, before and after metformin treatment.
Gynecological Endocrinol 2008, 24:378-384.
24. Derosa G, Maffiolia P, Salvadeoa SAT, Ferraria I, Ragonesib PD, Quercic F,
Franzettid IG, Gadaletae G, Ciccarellif L, Piccinnig MN, DAngeloa A,
Ciceroh AFG: Effects of sitagliptin or metformin added to pioglitazone
monotherapy in poorly controlled type 2 diabetes mellitus patients.
Metabolism 2010, 59:887-895.
25. Marciniak A, Nawrocka-Rutkowska J, Brodowska A, Sienkiewicz R,
Szydlowska I, Starczewski A: Leptin concentrations in patients with
polycystic ovary syndrome before and after met-formin treatment
depending on insulin resistance, body mass index and androgen con-
centrations-introductory report. Folia Histochemica et Cytobiologica 2009,
47:323-328.
26. Huang NL, Chiang SH, Hsueh CH, Liang YJ, Chen YJ, Lai LP: Metformin
inhibits TNF-α-induced IκB kinase phosphorylation, IκB-αdegradation
and IL-6 production in endothelial cells through PI3K-dependent AMPK
phosphorylation. Int J Cardiol 2009, 134:169-175.
27. Iwabu M, Yamauchi T, Okada-Iwabu M, Sato K, Nakagawa T, Funata1 M,
Yamaguchi1 M, Namiki S, Nakayama R, Tabata M, Ogata H, Kubota1 N,
Takamoto I, Hayashi YK, Yamauchi N, Waki H, Fukayama M, Nishino I,
Tokuyama K, Ueki K, Oike Y, Ishii S, Hirose K, Shimizu T, Touhara K,
Kadowaki T: Adiponectin and AdipoR1 regulate PGC-1a and
mitochondria by Ca2+ andAMPK/SIRT1. Nature 2010, 464:1313-1319.
28. Taliaferro-Smith L, Nagalingam A, Zhong D, Zhou W, Saxena NK, Sharma D:
LKB1 is required for adiponectin-mediated modulation of AMPK-S6K axis
and inhibition of migration and invasion of breast cancer cells. Oncogene
2009, 28:2621-2633.
29. Pollak M: Insulin and insulin-like growth factor signalling in neoplasia.
Nature Rev Cancer 2008, 8:915-928.
Grisouard et al.Diabetology & Metabolic Syndrome 2011, 3:16
http://www.dmsjournal.com/content/3/1/16
Page 6 of 7
30. Lim CT, Kola B, Korbonits M: AMPK as a mediator of hormonal signalling.
J Mol Endocrinol 2010, 44:87-97.
doi:10.1186/1758-5996-3-16
Cite this article as: Grisouard et al.: Targeting AMP-activated protein
kinase in adipocytes to modulate obesity-related adipokine production
associated with insulin resistance and breast cancer cell proliferation.
Diabetology & Metabolic Syndrome 2011 3:16.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Grisouard et al.Diabetology & Metabolic Syndrome 2011, 3:16
http://www.dmsjournal.com/content/3/1/16
Page 7 of 7
... AMPK contributes to insulin sensitivity due to an increase in glucose uptake in insulin sensitive organs, but, unlike leptin, adiponectin is reduced in obese subjects, thus its anti-inflammatory, anti-proliferative and pro-apoptotic effects are compromised in this population [127]. Solid tumours present adiponectin receptors [128] and studies with MCF-7 human breast cancer have indicated positive apoptotic response effects via p53 are modulated by AMPK phosphorylation induced by adiponectin [129,130] specially in ERα negative cancer [130][131][132]. Besides that, adiponectin is able to inhibit the activity of an enzyme responsible for estrogen production, aromatase, one of the major risk factors for breast cancer development in postmenopausal women [90]. ...
... Regards to energy metabolism, the hyperglycaemia and hyperinsulinemia observed in insulin resistant patients are also associated to elevated risk of cancer and a widely known mechanism of increasing insulin sensitivity and glucose uptake is upregulating AMPK activity [181]. It was demonstrated that in MCF-7 breast cancer cultured with serum from human pre adipocytes led to cell proliferation yet AMPK and metformin treatments recovered the adipokine imbalance caused by LPS stimulation [129]. These findings reassure the importance of AMPK activation on obesity-related energy disturb, insulin resistance and cancer growth. ...
Article
Full-text available
Adipose tissue is considered an endocrine organ whose complex biology can be explained by the diversity of cell types that compose this tissue. The immune cells found in the stromal portion of adipose tissue play an important role on the modulation of inflammation by adipocytokines secretion. The interactions between metabolic active tissues and immune cells, called immunometabolism, is an important field for discovering new pathways and approaches to treat immunometabolic diseases, such as obesity and cancer. Moreover, physical exercise is widely known as a tool for prevention and adjuvant treatment on metabolic diseases. More specifically, aerobic exercise training is able to increase the energy expenditure, reduce the nutrition overload and modify the profile of adipocytokines and myokines with paracrine and endocrine effects. Therefore, our aim in this review was to cover the effects of aerobic exercise training on the immunometabolism of adipose tissue in obesity and cancer, focusing on the exercise-related modification on adipose tissue or immune cells isolated as well as their interaction.
... Adipocyte-derived soluble factors called adipokines including adiponectin, leptin, IL-6, IL1β, and TNFα have known to be involved in the breast cancer progression through their receptors [6,8]. AMPK is noticed to regulate the expression of IL-6 and IL-8 in adipocytes, suggesting that targeting AMPK in adipocytes could be a novel way to modulate obesity-related adipokine production associated with insulin resistance and breast cancer progression [9]. In addition, adipocytes, as well as obesity in breast cancer patients, are closely related with the increased resistance to chemotherapy including trastuzumab and docetaxel leading to the challenges for the clinical management of the tumor [10]. ...
... Recent studies have noticed that the adipocyte-derived tumor microenvironment plays crucial roles in tumor progression of various types of human cancer including breast cancer [5,7,9,23]. We in the present study also notice that adipocyte-derived CM stimulate the proliferation and motility of breast cancer cells. ...
Article
Full-text available
Objective: Although it is well known that adipocyte significantly affects breast cancer progression, its mechanism has not been fully understood. Here, we analyzed the effect of adipocytes on breast cancer progression including cell proliferation and migration. Materials and Methods: We treated the conditioned media obtained from mouse 3T3-L1-derived or human adipose tissue-derived mesenchymal stem cells (hAMSC)-derived adipocytes to breast cancer cells, MCF-7 and MDA-MB-231. And then, cells viability and proliferation were analyzed using MTT assays and colony forming assays, respectively. Also mRNA expression of inflammatory cytokines and proteins expression in main signal pathway were analyzed by RT-qPCR and immunoblotting, respectively. Results: Adipocyte-derived conditioned media increased the proliferation and migration of MCF-7 and MDA-MB-231 cells while little effects in a human normal immortalized mammary epithelial cell line MCF10A. In addition, adipocyte-derived conditioned media induced phosphorylation of AKT and mTOR and upregulated the expression of target genes of the PI3K-AKT-mTOR pathway including IL6, IL1β, IL1α and TNFα in breast cancer cells. Furthermore, BEZ235 a dual inhibitor of PI3K and mTOR significantly decreased the adipocyte-mediated the proliferation and migration of breast cancer cells. Conclusion: Adipocyte-derived conditioned media enhance the proliferation and migration of breast cancer cells through the PI3K-AKT-mTOR pathway, supporting the importance of heterotypic interactions between breast cancer cells and adipocytes in the tumor microenvironment.
... These findings indicate that FA also has the ability to improve the impaired glucose homeostasis through enhancing AMPK phosphorylation in skeletal muscle. It has been reported that leptin activates AMPK which leads to phosphorylation of ACC and subsequent stimulation of fatty acid oxidation [23,24]. Since fatty acid plays a central role in the state of insulin resistance, the potentiality of leptin to stimulate fatty acid oxidation in skeletal muscle may result in prevention of lipotoxicity and consequent insulin resistance [23,24]. ...
... It has been reported that leptin activates AMPK which leads to phosphorylation of ACC and subsequent stimulation of fatty acid oxidation [23,24]. Since fatty acid plays a central role in the state of insulin resistance, the potentiality of leptin to stimulate fatty acid oxidation in skeletal muscle may result in prevention of lipotoxicity and consequent insulin resistance [23,24]. The present study showed that FA could increase phosphorylation of ACC in muscle. ...
Article
Full-text available
Objective: To evaluate the insulin sensitivity action of ferulic acid (FA) in skeletal muscle and hypothalamus of high-fat diet (HFD)-induced obese mice. Methods: Obese mouse model was induced by HFD (45 kcal% lard fat) for 16 weeks. After 8 weeks of HFD feeding, these obese mice were orally treated with FA at doses of 25 and 50 mg/kg/day for 8 weeks. At the end of all treatments, the epididymal fat, pancreas, skeletal muscle and hypothalamus were removed for biochemical parameter and protein expression examinations. Results: FA treatment significantly decreased leptin level in fat tissue and insulin level in pancreas (P < 0.05). Interestingly, obese mice treated with FA increased the protein expressions of insulin receptor substrate-1, phosphatidylinositol 3-kinase, and phosphorylated-protein kinase B in both muscle and brain (P < 0.05). The phosphorylations of adenosine monophosphate-activated protein kinase and acetyl-CoA carboxylase in muscle, and leptin receptor protein in hypothalamus were also increased (P < 0.05). The pancreatic islets histology showed smaller size in obese mice treated with FA compared to untreated obese mice. Conclusions: These findings indicate the beneficial effect of FA in improving insulin resistance in HFD-induced obese mice. These effects are probably mediated via modulating the insulin receptor substrate/phosphatidylinositol 3-kinase/protein kinase B or adenosine monophosphate-activated protein kinase pathways.
... The Michigan Cancer Foundation-7 (MCF-7) cell line, derived from a human breast adenocarcinoma, is the most extensively used experimental tool to study ER + tumor endocrine resistance in vitro (4,5). Indeed, long-term estrogen starvation of MCF-7 cells eventually promotes proliferation via estrogen-independent mechanisms, such as those under the control of the adipokines leptin (6), adiponectin (7,8) or resistin (9). Interestingly, a common regulatory mechanism of leptin and adiponectin-mediated growth of MCF-7 cells is autophagy (10,11) , an intracellular process leading to the degradation of damaged or dysfunctional subcellular organelles (12). ...
Article
Full-text available
Apelin-13 is an adipokine known for its growth-inducing effects on human breast cancer cells in an estrogen-containing environment. However, the response of these cells to apelin-13 in the absence of estrogen and its association with the expression of the apelin receptor (APLNR) has not yet been investigated. In the present study, we show that the breast cancer cell line MCF-7 expresses the APLNR as shown by immunofluorescence and flow cytometry, under conditions of ER starvation and that culture of these cells in the presence of apelin-13 results in an increased growth rate and a diminished autophagy flux. Moreover, the binding of APLNR by apelin-13 resulted in an increased growth rate (assayed by AlamarBlue) and a diminished autophagy flux (monitored by Lysotracker Green). The latter observations were reversed in the presence of exogenous estrogen. Finally, apelin-13 induces the deactivation of the apoptotic kinase AMPK. Taken together, our results show that APLNR signaling in breast cancer cells is functional and prevents tumor growth under conditions of estrogen starvation. They furthermore suggest an alternative mechanism of estrogen-independent tumor growth thereby positioning the APLNR-AMPK axis as a novel pathway and a possible therapeutic target in endocrine resistance of breast cancer cells.
... Nevertheless, the current approach will guide our future studies to build targeted treatment models that focus on suppressing adipocytes and IL6. There are already studies targeting specific proteins or signaling patterns by adipocytes to control cancer [153,154]. Also, high levels of adipocytes lead to upregulated IL6, which build resistance to anti-VEGF therapy in breast cancer [155]. These suggest attractive therapy models with resistance terms for our future work. ...
Article
Full-text available
The most common kind of cancer among women is breast cancer. Understanding the tumor microenvironment and the interactions between individual cells and cytokines assists us in arriving at more effective treatments. Here, we develop a data-driven mathematical model to investigate the dynamics of key cell types and cytokines involved in breast cancer development. We use time-course gene expression profiles of a mouse model to estimate the relative abundance of cells and cytokines. We then employ a least-squares optimization method to evaluate the model's parameters based on the mice data. The resulting dynamics of the cells and cytokines obtained from the optimal set of parameters exhibit a decent agreement between the data and predictions. We perform a sensitivity analysis to identify the crucial parameters of the model and then perform a local bifurcation on them. The results reveal a strong connection between adipocytes, IL6, and the cancer population, suggesting them as potential targets for therapies. Author summary One of the outstanding challenges of the mathematical modeling of cancer progression is the existence of many unknown parameters. In this work, we develop a data-driven mathematical model of breast cancer progression by deriving a system of ordinary differential equations for the interaction networks of key cell types and molecules in breast tumors. To overcome the limitations of unknown parameters, we utilize a time course data of a PyMT mice model of breast cancer and estimate parameters using an optimization method. Although the predicted dynamics of cancer and necrotic cells using the obtained values of parameters are in good agreement with the data, the predicted values for a few other variables do not match the data. This might indicate that there are some other key PLOS COMPUTATIONAL BIOLOGY PLOS Computational Biology | https://doi.
... In vitro studies using conditioned media from 3T3-L1 derived murine adipocytes showed upregulation of anti-apoptotic transcriptional programs in MCF-7 breast cancer cells (5). Similarly, conditioned media from breast adipocytes induced migration in non-cancerous normal breast epithelial cells (6), and increased migration, proliferation, viability and invasion in a variety of oestrogen receptor (ER)-positive and ER-negative breast cancer cell lines (7)(8)(9)(10)(11)(12)(13). ...
Article
Full-text available
Background/aim: Recent research highlights the role of cancer-associated adipocytes (CAA) in promoting breast cancer cell migration, invasion and resistance to therapy. This study aimed at identifying cellular proteins differentially regulated in breast cancer cells co-cultured with CAA. Materials and methods: Adipocytes isolated from human breast adipose tissue were co-cultured with hormone receptor-positive (MCF-7) or -negative (MDA-MB-231) breast cancer cells using a transwell co-culture system. Proteomes of co-cultured and control breast cancer cells were compared quantitatively using iTRAQ labelling and tandem mass spectrometry, and the results were validated by western blotting. Results: A total of 1,126 and 1,218 proteins were identified in MCF-7 and MDA-MB-231 cells, respectively. Among these, 85 (MCF-7) and 63 (MDA-MB-231) had an average fold change >1.5 following co-culture. Pathway analysis revealed that CAA-induced enrichment of proteins involved in metabolism, the ubiquitin proteasome, and purine synthesis. Conclusion: This study provides a proteomic platform for investigating the paracrine role of CAA in promoting breast cancer cell metastasis and resistance to therapy.
... Blocking TNFα significantly reduces breast tumor size [153]. In addition, TNFα regulates interleukin synthesis, adiponectin secretion, and aromatase expression in adipose tissue [154][155][156]. ...
Article
Full-text available
Obesity is a global pandemic and it is well evident that obesity is associated with the development of many disorders including many cancer types. Breast cancer is one of that associated with a high mortality rate. Adipocytes, a major cellular component in adipose tissue, are dysfunctional during obesity and also known to promote breast cancer development both in vitro and in vivo. Dysfunctional adipocytes can release metabolic substrates, adipokines, and cytokines, which promote proliferation, progression, invasion, and migration of breast cancer cells. The secretion of adipocytes can alter gene expression profile, induce inflammation and hypoxia, as well as inhibit apoptosis. It is known that excessive free fatty acids, cholesterol, triglycerides, hormones, leptin, interleukins, and chemokines upregulate breast cancer development. Interestingly, adiponectin is the only adipokine that has anti-tumor properties. Moreover, adipocytes are also related to chemotherapeutic resistance, resulting in the poorer outcome of treatment and advanced stages in breast cancer. Evaluation of the adipocyte secretion levels in the circulation can be useful for prognosis and evaluation of the effectiveness of cancer therapy in the patients. Therefore, understanding about functions of adipocytes as well as obesity in breast cancer may reveal novel targets that support the development of new anti-tumor therapy. In this systemic review, we summarize and update the effects of secreted factors by adipocytes on the regulation of breast cancer in the tumor microenvironment.
... AMPKα1/sirtuin 1 (SIRT1) signaling pathway exerts its antiinflammatory effects both in endothelial cells [32], adipocytes [188] and macrophages [189], and AMPKα1 inhibits the activation of the NF-κB system [190]. Activation of AMPKα1 suppresses the synthesis of pro-inflammatory cytokines, such as IL-6 and IL-8 in adipocytes [191]. A combination of resveratrol (120 mg/kg/day) and quercetin (240 mg/ kg/day) attenuates high fat diet-induced circulating inflammatory markers such as TNF-α, IL-6, and MCP-1 through regulating AMPKα1/ SIRT1 signaling pathway in rats [70]. ...
Chapter
Full-text available
The tumor microenvironment increases the growth and invasion of cancer cells, makes classical chemotherapy applications inadequate, and is associated with a poor cancer prognosis. Recent studies reveal that cancer stroma supports tumor growth and metastasis and develops resistance to chemotherapy. In vitro co-culture techniques are widely used to study cross-talk between tumor microenvironment cells such as adipocytes, endothelial cells, fibroblasts, macrophages, and cancer cells. Co-culture techniques are classified into two main categories: indirect and direct methods. Transwell (indirect) co-culture of mature adipocytes with cancer cells has shown cancer cell viability, growth, proliferation, invasion, and metastases. This chapter covers the general methods of co-culture studies and will emphasize the results obtained on the co-culture of adipocytes and cancer cells.
Chapter
The microenvironment depends and generates dependence on all the cells and structures that share the same niche, the biotope. The contemporaneous view of the tumor microenvironment (TME) agrees with this idea. The cells that make up the tumor, whether malignant or not, behave similarly to classes of elements within a living community. These elements inhabit, modify and benefit from all the facilities the microenvironment has to offer and that will contribute to the survival and growth of the tumor and the progression of the disease.
Conference Paper
Full-text available
Parental self-efficacy: Questionnaire development and testing Childhood overweight and obesity is an epidemic in the United States (US). Parents are primary agents of change in children and should be targeted for interventions regarding healthy lifestyle promotion. However, parents need to possess self-efficacy to engender a family ethos espousing a healthy lifestyle for their children. There has been little research in this area and no instrument to assess it. The purpose of this study was to develop a reliable and valid questionnaire for the assessment of parental beliefs in this regard for their children aged 6-11 years. A 35-item questionnaire was developed, based on US Department of Agriculture guidelines on diet and physical activity and a review of the literature. Content validity was assessed and deemed acceptable by eight experts in relevant fields, with a content validity index of 0.97. A pilot sample of 15 eligible participants was used to assess initial reliability (α=0.95). A new convenience sample of 146 English reading and writing parents of children aged 6-11 years who have access to a computer and the internet were recruited. This questionnaire, a demographics information sheet and two surveys for testing concurrent validity, the Self-efficacy for Eating and Exercise (SEB-Ex) Behaviors Surveys, were given to the participants, who were mostly: married (84%), female (88%), White (82%) non-Hispanic or Latino (91%) with at least some college education (97%). A subsample of 25 participants completed the questionnaire again in 5-10 days to check test-retest reliability. After data collection, exploratory factor analysis confirmed the two hypothesized subscales: diet behaviors (DB) and physical activity behaviors (PAB), which also demonstrated ample convergent and discriminant validity, except for a single item, which was dropped from the questionnaire. Internal reliability of the 34-item total questionnaire (α=0.94), as well as the DB (α=0.93) and PAB (α=0.94) subscales, were excellent. Test-retest reliability and concurrent validity of the total score and DB were respectable. Overall, this questionnaire appears to be a reliable and valid measure. However, further psychometric testing in a broader sample of the target population and with parents of children in other age groups is warranted.
Article
Full-text available
Insulin and insulin-like growth factors (IGFs) are well known as key regulators of energy metabolism and growth. There is now considerable evidence that these hormones and the signal transduction networks they regulate have important roles in neoplasia. Epidermiological, clinical and laboratory research methods are being used to investigate novel cancer prevention and treatment strategies related to insulin and IGF signalling. Pharmacological strategies under study include the use of novel receptor-specific antibodies, receptor kinase inhibitors and AMP-activated protein kinase activators such as metformin. There is evidence that insulin and IGF signalling may also be relevant to dietary and lifestyle factors that influence cancer risk and cancer prognosis. Recent results are encouraging and have justified the expansion of many translational research programmes.
Article
Full-text available
High fat diet-induced endotoxaemia triggers low-grade inflammation and lipid release from adipose tissue. This study aims to unravel the cellular mechanisms leading to the lipopolysaccharide (LPS) effects in human adipocytes. Subcutaneous pre-adipocytes surgically isolated from patients were differentiated into mature adipocytes in vitro. Lipolysis was assessed by measurement of glycerol release and mRNA expression of pro-inflammatory cytokines were evaluated by real-time PCR. Treatment with LPS for 24 h induced a dose-dependent increase in interleukin (IL)-6 and IL-8 mRNA expression. At 1 µg/ml LPS, IL-6 and IL-8 were induced to 19.5 ± 1.8-fold and 662.7 ± 91.5-fold (P < 0.01 vs basal), respectively. From 100 ng/ml to 1 µg/ml, LPS-induced lipolysis increased to a plateau of 3.1-fold above basal level (P < 0.001 vs basal). Co-treatment with inhibitors of inhibitory kappa B kinase kinase beta (IKKβ) or NF-κB inhibited LPS-induced glycerol release. Co-treatment with the protein kinase A (PKA) inhibitor H-89, the lipase inhibitor orlistat or the hormone-sensitive lipase (HSL) inhibitor CAY10499 abolished the lipolytic effects of LPS. Co-treatment with the MAPK inhibitor, U0126 also reduced LPS-induced glycerol release. Inhibition of lipolysis by orlistat or CAY10499 reduced LPS-induced IL-6 and IL-8 mRNA expression. Induction of lipolysis by the synthetic catecholamine isoproterenol or the phosphodiesterase type III inhibitor milrinone did not alter basal IL-6 and IL-8 mRNA expression after 24 treatments whereas these compounds enhanced LPS-induced IL-6 and IL-8 mRNA expression. Both the inflammatory IKKβ/NF-κB pathway and the lipolytic PKA/HSL pathways mediate LPS-induced lipolysis. In turn, LPS-induced lipolysis reinforces the expression of pro-inflammatory cytokines and, thereby, triggers its own lipolytic activity.
Article
The discovery of leptin in the mid-1990s has focused attention on the role of proteins secreted by adipose tissue. Leptin has profound effects on appetite and energy balance, and is also involved in the regulation of neuroendocrine and immune function. Sex steroid and glucocorticoid metabolism in adipose tissue has been implicated as a determinant of body fat distribution and cardiovascular risk. Other adipose products, for example, proinflammatory cytokines, complement factors and components of the coagulation/fibrinolytic cascade, may mediate the metabolic and cardiovascular complications associated with obesity.
Article
Most research confirms that metformin therapy has a positive influence on cardiovascular risk factors (CVRF) such as dyslipidemia, insulin resistance and hyperandrogenism in polycystic ovary syndrome (PCOS). The aims of the present study were to establish other CVRF, such as plasma adiponectin, tumor necrosis factor-alpha (TNFalpha), interleukin-6 (IL-6) and C-reactive protein (CRP) levels, in obese premenopausal women with PCOS and to investigate the effect of metformin treatment on these factors. The study group consisted of 29 PCOS woman with body mass index (BMI) >25 kg/m(2). They were treated over 6 months with 500 mg metformin twice daily. Twenty-nine healthy individuals matched for age and BMI were controls. Adiponectin, TNFalpha, IL-6 and CRP levels were examined in all PCOS (before and after treatment) and control women. In the PCOS group significantly lower plasma adiponectin and TNFalpha levels were observed, whereas there were no differences in plasma IL-6 and CRP levels between PCOS and control groups. Plasma adiponectin increased significantly after metformin treatment, but levels of inflammatory factors did not change.
Article
Diabetes mellitus has been associated with an increased risk of several types of cancers, but its relationship with breast cancer remains unclear. We conducted a meta-analysis of case-control and cohort studies to assess the evidence regarding the association between diabetes and risk of breast cancer. Studies were identified by searching MEDLINE (1966-February 2007) and the references of retrieved articles. We identified 20 studies (5 case-control and 15 cohort studies) that reported relative risk (RR) estimates (odds ratio, rate ratio/hazard ratio, or standardized incidence ratio) with 95% confidence intervals (CIs) for the relation between diabetes (largely Type II diabetes) and breast cancer incidence. Summary RRs were calculated using a random-effects model. Analysis of all 20 studies showed that women with (versus without) diabetes had a statistically significant 20% increased risk of breast cancer (RR, 1.20; 95% CI, 1.12-1.28). The summary estimates were similar for case-control studies (RR, 1.18; 95% CI, 1.05-1.32) and cohort studies (RR, 1.20; 95% CI, 1.11-1.30). Meta-analysis of 5 cohort studies on diabetes and mortality from breast cancer yielded a summary RR of 1.24 (95% CI, 0.95-1.62) for women with (versus without) diabetes. Findings from this meta-analysis indicate that diabetes is associated with an increased risk of breast cancer.
Article
The mechanisms of metformin effects on glucose transport and metabolism were investigated in human adipocytes. Human preadipocytes obtained from surgical biopsies were differentiated in vitro into adipocytes and the effects of metformin on glucose uptake, glucose oxidation and the involved signaling pathways were analyzed. Metformin (1mM, 24h) increased glucose uptake 2.3±0.2-fold (p<0.001 vs. basal) in human adipocytes, without altering cell viability and oxygen consumption. Metformin did not alter GLUT-1 mRNA expression and protein content but increased GLUT-4 mRNA expression and cellular protein content, leading to increased GLUT-4 protein content in the plasma membrane. Neither basal nor insulin-induced phosphorylation of Akt at Ser-473 and AS160 (Akt substrate of 160kDa) at Thr-642 were enhanced by metformin. Suppression of metformin-induced AMP-activated protein kinase (AMPK) activity by AMPKα1 silencing, however, reduced metformin-associated GLUT-4 expression and stimulation of glucose uptake. In addition, metformin induced glucose oxidation. In conclusion, activation of AMPKα1 without impairment of cell respiration is crucial for metformin-mediated increase in GLUT-4 protein content and glucose uptake in human adipocytes.