Skip to main content
Erschienen in: Diabetologia 9/2013

Open Access 01.09.2013 | Article

Human adipose microRNA-221 is upregulated in obesity and affects fat metabolism downstream of leptin and TNF-α

verfasst von: A. Meerson, M. Traurig, V. Ossowski, J. M. Fleming, M. Mullins, L. J. Baier

Erschienen in: Diabetologia | Ausgabe 9/2013

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Aims/hypothesis

MicroRNAs (miRNAs) are short endogenous RNAs that regulate multiple biological processes including adipogenesis and fat metabolism. We sought to identify miRNAs that correlate with BMI and to elucidate their upstream regulation and downstream targets.

Methods

Microarray-based expression profiling of 233 miRNAs was performed on subcutaneous abdominal adipose tissue biopsies from 29 non-diabetic Pima Indian participants. Correlation of the expression levels of eight miRNAs with BMI was assessed by quantitative reverse transcription (QRT) PCR in adipose samples from 80 non-diabetic Pima Indians with a BMI of 21.6–54.0 kg/m2. The upstream regulation of one of these miRNAs, miR-221, was tested by treating cultured human pre-adipocytes with leptin, TNF-α and insulin. Predicted targets of miR-221 were validated using QRT-PCR, immunoblots and luciferase assays. The downstream effects of miR-221 overexpression were assayed by proteomic analysis.

Results

Expression levels of miR-221 were positively correlated with BMI (particularly in women) and fasting insulin concentrations, while the levels of miR-193a-3p and miR-193b-5p were negatively correlated with BMI; other miRNAs did not show significant associations in the 80 samples. miR-221 was downregulated by leptin and TNF-α treatment in cultured human pre-adipocytes. Conversely, miR-221 overexpression upregulated several proteins involved in fat metabolism, mimicking peroxisome proliferator-activated receptor (PPAR) activation. Furthermore, miR-221 directly downregulated the adiponectin receptor 1 (ADIPOR1) and the transcription factor v-ets erythroblastosis virus E26 oncogene homolog 1 (ETS1). Adiponectin signalling is known to promote insulin sensitivity, and ETS1 is crucial for angiogenesis.

Conclusions/interpretation

Our data suggest that miR-221 may contribute to the development of the insulin resistance that typically accompanies obesity, by affecting PPAR signalling pathways and by directly downregulating ADIPOR1 and ETS1.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00125-013-2950-9) contains peer-reviewed but unedited supplementary material, which is available to authorised users.
Abkürzungen
ADIPOR1
Adiponectin receptor 1
AKR
Aldoketoreductase
ETS1
v-ets Erythroblastosis virus E26 oncogene homologue 1
FASN
Fatty acid synthase
HOMA1-IR
Homeostasis model assessment 1 of insulin resistance
HRP
Horseradish peroxidase
miRNA
MicroRNA
MMP
Matrix metalloproteinase
NIDDK
National Institute of Diabetes and Digestive and Kidney Diseases
NIH
National Institutes of Health
PPAR
Peroxisome proliferator-activated receptor
pri-miRNA
Primary miRNA
QRT
Quantitative reverse transcription
SAT
Subcutaneous adipose tissue
UTR
Untranslated region
VEGF
Vascular endothelial growth factor

Introduction

Recent studies report the functional involvement of microRNAs (miRNAs) in metabolic and endocrine pathways [13] and adipogenesis [48]. Differential expression of miRNAs has been reported in the tissues of obese vs non-obese human individuals [2, 6, 9, 10] and in humans and animals with diabetes [11, 12]. However, the mechanisms of action of most miRNAs that are deregulated in obesity are unknown.
We used a hypothesis-free approach to identify miRNAs in adipose tissue with expression levels that correlated with BMI in Pima Indian individuals. We identified miR-221 as a highly expressed candidate miRNA that was upregulated in individuals with the highest BMIs, in agreement with previous studies [6, 13]. We then investigated the cause of miR-221 upregulation in obesity and assessed possible molecular mechanisms mediated by downstream targets of miR-221.

Methods

Participants and biopsy collection
Participants were predominately of Pima Indian heritage and all were free of diabetes. Biopsies and clinical data were available for 80 healthy individuals who were not taking any medication (see electronic supplementary material [ESM] Table 1). Participants were admitted to the Clinical Research Unit, NIDDK, Phoenix, AZ, USA, where they consumed a weight-maintaining diet (containing 50% of calories as carbohydrates, 30% as fat and 20% as protein) for 2–3 days prior to clinical testing. A 3 h 75 g OGTT and abdominal subcutaneous adipose tissue (SAT) needle biopsies under local anaesthesia with 1% lidocaine were performed after a 12 h overnight fast. Variants in the miR-221 promoter were analysed for association with BMI by genotyping in a population-based sample of 3,500 Pima Indians (including 80 who underwent adipose tissue biopsies). Informed consent was obtained for all participants, and the study was approved by the institutional review board of the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH).
RNA extraction, microRNA expression profiling and validation
Total RNA was extracted from 80 frozen SAT biopsies using the miRNeasy kit (Qiagen, Venlo, the Netherlands), quantified with a NanoDrop spectrophotometer (Thermo Scientific, Wilmington, DE, USA), and its integrity assayed on an Agilent 2100 BioAnalyzer (Agilent Technologies, Santa Clara, CA, USA). Samples (n = 29) showing the highest quality RNA were labelled with the 3DNA FlashTag Biotin RNA Labeling kit (Genisphere, Hatfield, PA, USA) and hybridised with GeneChip miRNA arrays (Affymetrix, Santa Clara, CA, USA) for expression profiling. Three random samples were rehybridised to new arrays to ascertain reproducibility. The array cartridges were processed on an Affymetrix Fluidics Station 450 and scanned on an Affymetrix GeneChip 3000 7G with AutoLoader. The results were normalised using the Affymetrix miRNA QC tool and analysed with MeV software v4.3 [14] (Dana Farber Cancer Institute, Boston, MA, USA) and Excel 2003 (Microsoft, Redmond, WA, USA).
Expression levels of specific miRNAs were validated in all 80 RNA samples by quantitative reverse transcription (QRT) PCR using TaqMan assays (Life Technologies, Carlsbad, CA, USA) on an ABI 7900HT Fast QPCR system (Life Technologies). U6 and U44 small nuclear RNAs were used for normalisation. The differential expression of specific miRNAs was correlated to individuals’ BMI and insulin and glucose levels during the OGTT. The homeostasis model assessment 1 of insulin resistance (HOMA1-IR) was calculated as previously described [15]. miRNA target prediction used the TargetScan algorithm (for a list of predicted miR-221 targets, see www.​targetscan.​org/​cgi-bin/​targetscan/​vert_​61/​targetscan.​cgi?​species=​Human&​gid=​&​mir_​sc=​miR-221/​222/​222ab/​1928&​mir_​c=​&​mir_​nc=​&​mirg=​&​sortType=​cs&​allTxs=​&​incl_​nc=​All) [16, 17].
Cell culture, treatment and transfection
Primary human pre-adipocytes (Zen-Bio, Research Triangle, NC, USA) and SH-SY5Y human neuroblastoma-derived cells (American Type Culture Collection (ATCC), Manassas, VA, USA) were cultured in MEM/F-12 1:1 containing 10% FBS (ATCC). Cells were serum-starved for 24 h and then treated with recombinant human leptin (L4146; Sigma, St Louis, MO, USA) at concentrations of 0, 50, 200 and 500 ng/ml, or TNF-α (H8916; Sigma) at concentrations of 5–50 ng/ml, for 24 h prior to harvesting. Pre-adipocyte differentiation was induced in DM-2 medium (Zen-Bio) for more than 10 days. TNF-α treatment of differentiated adipocytes was for 10 or 16 days, at 10 or 100 ng/ml [18]. Insulin treatment was at 100 nmol/l for 1 h and 24 h. Total RNA was extracted using the miRNeasy kit (Qiagen).
For 3′ untranslated (UTR) reporter assays, JetPrime (Polyplus-transfection SA, Illkirch, France) was used to co-transfect HEK 293 cells (ATCC) with miRNA 3′ UTR target expression vectors for human ETS1, ADIPOR1, ADIPOR2 or control (Genecopoeia, Germantown, MD, USA), and human miR-221 mimic (Dharmacon, Lafayette, CO, USA) or scrambled control oligonucleotide, in 24-well plates. Cells were lysed 48 h post-transfection, and dual-luciferase readings were performed using the Genecopoeia Luc-Pair miR Luciferase Assay Kit on a TD-20/20 Luminometer (Turner Designs, Sunnyvale, CA, USA). Transfection of pre-adipocytes for immunoblotting and proteomic analysis was in six-well plates and 75 cm2 flasks, respectively.
Immunoblots
We used Bio-Rad (Hercules, CA, USA) equipment and reagents for immunoblots, and the following antibodies: adiponectin receptor 1 (ADIPOR1; sc-99183; Santa Cruz Biotechnology, Santa Cruz, CA, USA), and from Novus Bio (Littleton, CO, USA), v-ets erythroblastosis virus E26 oncogene homolog1 (ETS1) (NBP1-47474), β-actin (NB600-532), anti-mouse horseradish peroxidase (HRP;NB7539) and anti-rabbit HRP (NB730-H).
Proteomic analysis
Relative quantification of digested peptides was performed by stable isotope labelling of primary amines as previously described [19]. Dimethylated peptides were separated by nanoflow liquid chromatography using a Waters (Milford, MA, USA) nanoAcquity LC system. Data dependent acquisition was used in the mass spectrometry analysis on Thermo LTQ-Orbitrap Velos (Waltham, MA, USA). Proteins were identified using Proteome Discoverer 1.3 (Thermo Scientific). Quantification was performed by integrating the extracted ion chromatogram of the differentially labelled peptides.
QRT-PCR for mRNAs
This was performed on an ABI 7900HT Fast QPCR system using PerfeCTa FastMix (Quanta BioSciences, Gaithersburg, MD, USA). Primer sequences are given in ESM Table 2. GAPDH expression was used for normalisation.
Statistical analysis
The statistical significance between groups was determined using SigmaStat (SPSS, Chicago, IL, USA) using two-way ANOVA, the Wilcoxon rank-sum test for two independent samples, the Mann–Whitney U test or the t test as appropriate.

Results

To identify miRNAs that might be relevant to studies of obesity, miRNA array analyses were performed using the GeneChip platform (Affymetrix) on RNA isolated from 29 abdominal SAT biopsies (ESM Fig. 1a). A comparison of miRNA expression levels between individuals in the highest tertile vs the lowest tertile of BMI (ten vs nine individuals, respectively) identified several miRNAs that were differentially expressed between the two groups (Fig. 1a). QRT-PCR was used to validate both the mature and primary (pri)-miRNA for these eight miRNAs in a larger sample of 80 individuals.
Three of the miRNAs, miR-221, miR-193a-3p and miR193b-5p, were significantly correlated with BMI after adjusting for age and sex (Fig. 1b–g, Table 1), at both the mature and primary transcript levels; the other miRNAs assayed in the larger sample did not show significant correlations with BMI that were consistent between the mature and the primary transcript forms. Sex differences were also analysed. For the mature miR-221, the correlation with BMI was stronger when the analysis was restricted to women (p < 0.0001). Although miR-221 maps to the X chromosome, there was no significant difference in miR-221 expression between males and females (data not shown). Expression levels for miR-193a-3p, miR-193b-5p and miR-221 also showed nominal correlations with plasma glucose levels or insulin responses to a 75 g OGTT (Table 1). Since miR-221 displayed the highest expression levels in adipose tissue, we focused on this miRNA and examined potential causes for its upregulation in obesity and the effects of miR-221 on predicted downstream targets.
Table 1
Summary of associations between BMI, glucose and insulin responses to an OGTT (all traits adjusted for age and sex) and the expression of mature and primary transcript (pri-) forms of miR-193a, miR-193b-5p and miR-221 in 80 SAT biopsies
miRNA type
 
BMI
2 h glucose
Fasting insulin
2 h insulin
HOMA1-IR
(n = 80)
(n = 80)
(n = 64)
(n = 64)
(n = 68)
miR-193a-3p
R
−0.004
  
−0.323
 
p
0.060
NS
NS
0.009
NS
pri-miR-193a
R
−0.009
 
−0.440
  
p
0.010
NS
0.018
NS
NS
miR-193b-5p
R
−0.006
−0.269
−0.267
−0.355
0.077
p
0.047
0.016
0.053
0.004
0.056
pri-miR-193b
R
−0.008
−0.273
−0.383
−0.37
 
p
0.015
0.014
0.014
0.0026
NS
miR-221
R
0.015
 
0.302
 
0.135
p
<0.0001
NS
0.005
NS
0.066
miR-221 (women only, n = 35)
R
0.644
 
0.736
 
0.506
p
<0.0001
NS
0.042
NS
0.004
pri-miR-221
R
0.007
    
p
0.035
NS
NS
NS
NS
The HOMA1-IR and insulin correlations were no longer significant when adjusted for BMI. R and p represent linear regression R and p values
The upregulation of miR-221 in obesity could either be caused by a cis-acting variation at the miR-221 locus or by a secondary effect. To determine whether a variation in genomic sequence was the primary cause, the promoter region of miR-221 was sequenced using DNA from 24 unrelated Pima Indian individuals. Two variants, rs2858060 and rs2745709, were identified, and subsequent genotyping in a population-based sample of Pima Indians (n = 3,500) revealed no significant association between either variant and BMI or miR-221 expression levels in the 80 biopsies (data not shown). This suggests that the positive correlation observed between miR-221 expression and BMI is probably a consequence of obesity.
It is well known that obesity is associated with higher levels of leptin and TNF-α [20, 21], and also with a decrease in insulin sensitivity accompanied by hyperinsulinaemia [21]. Therefore, we examined whether leptin, TNF-α or insulin could potentially affect the expression of miR-221. Cultured human pre-adipocytes were treated with increasing concentrations of leptin (50, 200 and 500 ng/ml) for 24 h. Expression levels of miR-221 showed a dose-dependent downregulation, the largest decrease occurring with 500 ng/ml of leptin (Fig. 2a). Leptin signalling affects the brain as well as peripheral tissues; therefore, we tested whether leptin could regulate miR-221 levels in a neuronal cell model as well as in adipocytes. Treating SH-SY5Y human neuroblastoma-derived cells with leptin (100 and 500 ng/ml) caused a trend of downregulation of miR-221 (Fig. 2b).
To assess the effects of TNF-α on miR-221 expression, human pre-adipocytes were treated with 5, 10, 25 or 50 ng/ml of TNF-α for 24 h. Exposure to TNF-α for 24 h resulted in a decrease in miR-221 expression levels at all the concentrations tested (Fig. 2c). To evaluate the effects of chronic exposure to TNF-α on miR-221 expression in differentiated adipocytes, human pre-adipocytes were treated with standard differentiation media and allowed to differentiate for over 10 days. The adipocytes were then treated with 10 or 100 ng/ml of TNF-α for 10 or 16 days. miR-221 expression levels were similarly decreased (Fig. 2d). In contrast to leptin and TNF-α, no significant change in miR-221 expression was observed following treatment with 100 nmol/l insulin (data not shown).
To find direct downstream targets of miR-221, the TargetScan prediction program was used to identify genes containing binding sites for the miRNA paralogs miR-221 and miR-222 (see Methods for the link containing the list of predicted gene targets). Two of the predicted target genes, ADIPOR1 (Fig. 3a) and ETS1 (Fig. 3b), could have a potential role in causing insulin resistance as a consequence of obesity and were therefore selected for further study.
To test whether miR-221 could downregulate ADOPOR1 and/or ETS1 expression, luciferase assays were performed using ADOPOR1 and ETS1 3′ UTR reporter plasmids. To assess binding in this artificial system, HEK 293 cells (selected for rapid growth and ease of transfection) were co-transfected with a miR-221 mimic or scrambled non-specific oligonucleotide (negative control) along with an ADIPOR1 3′ UTR reporter plasmid, or with one of two ETS1 3′ UTR reporter plasmids. The 3′ UTR of ETS1 is 3.6 kb in size and was therefore cloned into two separate constructs with overlapping sequence. A reporter plasmid containing the 3′ UTR of ADIPOR2, which also functions as an adiponectin receptor but has no predicted binding site for miR-221, was also tested. The miR-221 mimic caused a significant reduction in luciferase activity for the ADIPOR1 and both ETS1 3′ UTR reporter plasmids, but not for the ADIPOR2 3′ UTR plasmid (Fig. 3c,d). In human pre-adipocytes, transfection with the miR-221 mimic reduced the levels of endogenous ETS1 mRNA levels (Fig. 3e) but not ADIPOR1 mRNA (data not shown). In contrast, both ETS1 and ADIPOR1 were reduced at the protein level (Fig. 3f,g).
Since we had demonstrated that both leptin and TNF-α could downregulate the expression of miRNA-221 and that miRNA-221 could regulate ETS1 expression, we next determined whether leptin and/or TNF-α treatment would result in a downstream effect on ETS1 expression. Human pre-adipocytes were treated with either leptin (50, 200 and 500 ng/ml) or TNF-α (10 ng/ml) for 24 h. ETS1 mRNA expression levels were increased during treatment with both leptin (200 and 500 ng/ml) and TNF-α (Fig. 4a,b). To examine chronic exposure to TNF-α in adipocytes, differentiated human adipocytes were treated with 10 and 100 ng/ml of TNF-α for 10 or 16 days. As with the pre-adipocytes, ETS1 mRNA levels were increased in adipocytes treated with TNF-α (Fig. 4c).
Specific analysis of ETS1 and ADIPOR1 as targets of miR-221 was hypothesis-driven. To examine the effects of miR-221 on protein expression with a hypothesis-free approach, mass spectrometry was used to perform a proteomic analysis on human adipocytes transfected with miR-221. Human pre-adipocytes were transfected with either a miR-221 mimic or a scrambled non-specific siRNA control and then treated with differentiation media for 10 days, before total protein was harvested. Out of approximately 2,200 detectable proteins, 251 (115 known and 136 uncharacterised) were significantly (p < 0.05) upregulated or downregulated in the miR-221 mimic transfected cells compared with controls (ESM Table 3). Only five of the 251 differentially regulated proteins (upregulated: eukaryotic translation initiation factor 5 [EIF5]; member of the RAS oncogene family [RAB1A]; downregulated: guanine nucleotide binding protein alpha inhibiting 2 [GNAI2]; thrombospondin 1 [THBS1]; tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, gamma polypeptide [YWHAG]) were predicted miR-221 targets, suggesting that most observed changes in protein levels were a downstream effect or that some miR-221 target genes were missed by the TargetScan prediction program.
Ingenuity software was used to place the 115 known differentially expressed proteins into physiological interaction networks. The most significantly upregulated interaction network (with a score of 43 and 20 focus molecules) was that of energy production and fatty acid metabolism (ESM Fig. 1b). This network included fatty acid synthase (FASN), which was upregulated about fivefold in the miR-221-transfected cells, and members of the aldoketoreductase (AKR) family (AKR1C1–AKR1C4). The most significantly downregulated network (with a score of 44 and 22 focus molecules) was that of cellular assembly and maintenance and tissue development (ESM Fig. 1c).
To determine whether proteins upregulated by miR-221 in the proteomic analysis were also upregulated at the mRNA level, QRT-PCR was used to measure the mRNA levels of seven of them (AKR1C1–AKR1C4, FASN, HADHA and HADHB) following miR-221 overexpression. Among these targets, AKR1C4 and HADHA were significantly upregulated by miR-221, while several others showed a non-significant trend (Fig. 4d). Other affected proteins may be regulated post-transcriptionally by downstream effectors of miR-221.
Prediction of upstream regulators that could potentially control several of the differentially expressed proteins yielded four molecules with high activation scores (Table 2). Two of these predicted common regulators (peroxisome proliferator-activated receptor (PPAR)α and PPARγ) belong to the PPAR family, while the other two (pirinixic acid and bezafibrate) are PPAR agonists. Therefore, overexpression of miR-221 in cultured adipocytes appears to affect the proteome in a manner that mimics PPAR activation.
Table 2
Top common regulators predicted to effect the proteomic changes observed in cultured adipocytes overexpressing miR-221, using Ingenuity pathway analysis software
Regulator
Molecule type
Predicted state
z score
p values of overlap
MYCN
Transcription regulator
 
1.913
1.05 × 10−10
INSR
Kinase
 
1.782
6.24 × 10−10
Pirinixic acid
Chemical toxicant
Activated
3.268
2.57 × 10−9
Bezafibrate
Chemical drug
Activated
2.184
1.19 × 10−8
TP53
Transcription regulator
 
0.692
4.05 × 10−8
PPARα
Ligand-dependent nuclear receptor
Activated
3.108
6.44 × 10−8
Dinoprost
Chemical – endogenous
 
0.254
7.10 × 10−8
PPARγ
Ligand-dependent nuclear receptor
Activated
2.601
1.62 × 10−7
Mannose
Chemical – endogenous
 
1.000
2.52 × 10−7
ACOX1
Enzyme
 
0.113
4.57 × 10−7
MYCN, v-myc myelocytomatosis viral related oncogene, neuroblastoma derived (avian); INSR, insulin receptor; ACOX1, acyl-coenzyme A oxidase 1, palmitoyl

Discussion

Our results show that adipose miR-221 expression is positively correlated with increasing BMI in the Pima Indian population. This correlation is in agreement with previous reports also showing that adipose miR-221 is upregulated in obesity [6, 13]; however, the mechanism(s) for the upregulation in our study are unclear. Both leptin and TNF-α, whose levels are known to be increased in the obese state, were found to downregulate miR-221 in cultured cells. This suggests a more complex mechanism in vivo, which may include desensitisation to the effects of leptin and TNF-α in the chronically obese state [2225].
In our proteomic analysis, we sought to identify the downstream consequences of miR-221 upregulation. The most significantly upregulated physiological network in adipocytes transfected with miR-221 consists of proteins involved in fatty acid metabolism, including FASN. It is thought that increased levels of fatty acids and increased expression of FASN may play a role in the development of insulin resistance [2629]. Prediction of upstream regulators that can activate several of the differentially expressed proteins yielded PPARα, PPARγ and two PPAR agonists, which may again indicate that upregulation of miR-221 in the obese state activates pathways involved in insulin sensitivity [30, 31].
We have further shown that miR-221 directly downregulates ADIPOR1 and ETS1 expression; therefore, it may be possible that obesity-associated upregulation of miR-221 can result in a decrease in ADIPOR1 and/or ETS1, which may in turn lead to the development of obesity-related metabolic consequences such as insulin resistance or type 2 diabetes. ADIPOR1 functions as a receptor for adiponectin and mediates a signalling pathway that promotes insulin sensitivity and is suppressed in insulin resistance and type 2 diabetes caused by obesity [32]. ETS1 is a transcription factor that regulates the expression of a wide spectrum of genes including cytokines and chemokines [33], and it has been proposed that obesity-associated inflammation may also play a significant role in the development of type 2 diabetes [34].
In addition, the previously described role of ETS1 in vascular endothelial growth factor (VEGF)-induced angiogenesis through its regulation of matrix metalloproteinases (MMPs) [35] that is subject to regulation by other miRNAs, e.g. miR-199a-5p [36], and the importance of VEGF signalling for adipose function [37] suggest that higher miR-221 expression in obesity may lead to decreased vascularisation and increased hypoxia and inflammation in adipose tissue. Supporting this notion is our group’s previous report that MMP3, a target of ETS1 [38], is downregulated in pre-adipocytes and stromal vascular cells from obese individuals [39], and that genetic variation in the MMP3 locus is only partially responsible for this downregulation. Additionally, downregulation of ETS1 may be essential to PPAR signalling [40], in line with the observed proteomic effects in which miR-221 overexpression mimicked PPAR activation. ETS1 and ADIPOR1 were absent from the list of proteins detected in our mass spectrometry analysis, possibly due to an overall lower expression level of these proteins, as well as a bias against membrane (such as ADIPOR1) and nuclear (such as ETS1) proteins inherent in the purification method employed.
miR-221 is a known player in oncogenesis, affecting the proliferation and behaviour of various cell types [4147]. Deregulation of ETS1 in a variety of cancers suggests that ETS1 could mediate the effects of miR-221 in oncogenesis. Recent studies have shown that an ETS1-mediated transcription of members of the MMP family is responsible for remodelling of the extracellular matrix in both angiogenesis and invasion [48]. Resistance to TNF-α promotes tumourigenesis [49, 50], and elevated miR-221 may have a role in mediating these effects via its suppression of ETS1 [51]. Thus, chronic upregulation of miR-221 in obesity may potentially increase cancer risk. However, additional studies are required to demonstrate that miR-221 is a valid link between obesity and obesity-associated diseases such as the metabolic syndrome, type 2 diabetes and cancers.

Acknowledgements

The authors thank C. Bogardus for assistance with data analysis and critically reviewing this manuscript, C. Mason for assistance with data analysis and S. Parrington for maintaining the human biopsies (all from the Phoenix Epidemiology and Clinical Research Branch, NIDDK, NIH, USA). The authors also thank the volunteers from the Gila River Indian Community in Arizona. Some of the results in this manuscript were presented in posters at the Keystone Symposia on Obesity (J2, 2011) and Adipose Tissue Biology (J5, 2013) at Keystone, CO, and at the Gordon Research Conference on the Biology of Post-Transcriptional Gene Regulation (2012), Newport, RI.

Funding

This research was funded by the intramural programme of NIDDK.

Duality of interest

The authors declare that there is no duality of interest associated with this manuscript.

Contribution statement

AM, MT, VO and LJB contributed to the conception and design of the study. AM, MT, VO, JMF and MM contributed to the acquisition of data. AM, MT and LJB contributed to analysis and interpretation of the data. All authors contributed to drafting the article or critically revising its intellectual content, and all authors approved the final version.
Open Access This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Literatur
1.
Zurück zum Zitat Xie H, Sun L, Lodish HF (2009) Targeting microRNAs in obesity. Expert Opin Ther Targets 13:1227–1238PubMedCrossRef Xie H, Sun L, Lodish HF (2009) Targeting microRNAs in obesity. Expert Opin Ther Targets 13:1227–1238PubMedCrossRef
2.
Zurück zum Zitat Heneghan HM, Miller N, Kerin MJ (2010) Role of microRNAs in obesity and the metabolic syndrome. Obes Rev 11:354–361PubMedCrossRef Heneghan HM, Miller N, Kerin MJ (2010) Role of microRNAs in obesity and the metabolic syndrome. Obes Rev 11:354–361PubMedCrossRef
3.
Zurück zum Zitat Guay C, Roggli E, Nesca V, Jacovetti C, Regazzi R (2011) Diabetes mellitus, a microRNA-related disease? Transl Res 157:253–264PubMedCrossRef Guay C, Roggli E, Nesca V, Jacovetti C, Regazzi R (2011) Diabetes mellitus, a microRNA-related disease? Transl Res 157:253–264PubMedCrossRef
4.
Zurück zum Zitat Karbiener M, Fischer C, Nowitsch S et al (2009) microRNA miR-27b impairs human adipocyte differentiation and targets PPARγ. Biochem Biophys Res Commun 390:247–251PubMedCrossRef Karbiener M, Fischer C, Nowitsch S et al (2009) microRNA miR-27b impairs human adipocyte differentiation and targets PPARγ. Biochem Biophys Res Commun 390:247–251PubMedCrossRef
5.
Zurück zum Zitat Kim SY, Kim AY, Lee HW et al (2010) miR-27a is a negative regulator of adipocyte differentiation via suppressing PPARγ expression. Biochem Biophys Res Commun 392:323–328PubMedCrossRef Kim SY, Kim AY, Lee HW et al (2010) miR-27a is a negative regulator of adipocyte differentiation via suppressing PPARγ expression. Biochem Biophys Res Commun 392:323–328PubMedCrossRef
6.
Zurück zum Zitat Ortega FJ, Moreno-Navarrete JM, Pardo G et al (2010) MiRNA expression profile of human subcutaneous adipose and during adipocyte differentiation. PLoS One 5:e9022PubMedCrossRef Ortega FJ, Moreno-Navarrete JM, Pardo G et al (2010) MiRNA expression profile of human subcutaneous adipose and during adipocyte differentiation. PLoS One 5:e9022PubMedCrossRef
7.
Zurück zum Zitat Sun L, Xie H, Mori MA et al (2011) Mir193b–365 is essential for brown fat differentiation. Nat Cell Biol 13:958–965PubMedCrossRef Sun L, Xie H, Mori MA et al (2011) Mir193b–365 is essential for brown fat differentiation. Nat Cell Biol 13:958–965PubMedCrossRef
8.
Zurück zum Zitat Zaragosi L-E, Wdziekonski B, Brigand K et al (2011) Small RNA sequencing reveals miR-642a-3p as a novel adipocyte-specific microRNA and miR-30 as a key regulator of human adipogenesis. Genome Biol 12:R64PubMedCrossRef Zaragosi L-E, Wdziekonski B, Brigand K et al (2011) Small RNA sequencing reveals miR-642a-3p as a novel adipocyte-specific microRNA and miR-30 as a key regulator of human adipogenesis. Genome Biol 12:R64PubMedCrossRef
9.
Zurück zum Zitat Martinelli R, Nardelli C, Pilone V et al (2010) miR-519d overexpression is associated with human obesity. Obesity 18:2170–2176PubMedCrossRef Martinelli R, Nardelli C, Pilone V et al (2010) miR-519d overexpression is associated with human obesity. Obesity 18:2170–2176PubMedCrossRef
10.
Zurück zum Zitat Fu T, Choi S-E, Kim D-H et al (2012) Aberrantly elevated microRNA-34a in obesity attenuates hepatic responses to FGF19 by targeting a membrane coreceptor β-Klotho. PNAS 109:16137–16142PubMedCrossRef Fu T, Choi S-E, Kim D-H et al (2012) Aberrantly elevated microRNA-34a in obesity attenuates hepatic responses to FGF19 by targeting a membrane coreceptor β-Klotho. PNAS 109:16137–16142PubMedCrossRef
11.
Zurück zum Zitat Klöting N, Berthold S, Kovacs P et al (2009) MicroRNA expression in human omental and subcutaneous adipose tissue. PLoS One 4:e4699PubMedCrossRef Klöting N, Berthold S, Kovacs P et al (2009) MicroRNA expression in human omental and subcutaneous adipose tissue. PLoS One 4:e4699PubMedCrossRef
12.
Zurück zum Zitat Herrera BM, Lockstone HE, Taylor JM et al (2010) Global microRNA expression profiles in insulin target tissues in a spontaneous rat model of type 2 diabetes. Diabetologia 53:1099–1109PubMedCrossRef Herrera BM, Lockstone HE, Taylor JM et al (2010) Global microRNA expression profiles in insulin target tissues in a spontaneous rat model of type 2 diabetes. Diabetologia 53:1099–1109PubMedCrossRef
13.
Zurück zum Zitat Xie H, Lim B, Lodish HF (2009) MicroRNAs induced during adipogenesis that accelerate fat cell development are downregulated in obesity. Diabetes 58:1050–1057PubMedCrossRef Xie H, Lim B, Lodish HF (2009) MicroRNAs induced during adipogenesis that accelerate fat cell development are downregulated in obesity. Diabetes 58:1050–1057PubMedCrossRef
14.
Zurück zum Zitat (2003) TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34:374–378PubMed (2003) TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34:374–378PubMed
15.
Zurück zum Zitat Matthews DR, Hosker JP, Rudenski AS et al (1985) Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 28:412–419PubMedCrossRef Matthews DR, Hosker JP, Rudenski AS et al (1985) Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 28:412–419PubMedCrossRef
16.
Zurück zum Zitat Lewis BP, Burge CB, Bartel DP (2005) Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120:15–20PubMedCrossRef Lewis BP, Burge CB, Bartel DP (2005) Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120:15–20PubMedCrossRef
17.
Zurück zum Zitat Grimson A, Farh KK-H, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP (2007) MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell 27:91–105PubMedCrossRef Grimson A, Farh KK-H, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP (2007) MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell 27:91–105PubMedCrossRef
18.
Zurück zum Zitat Xing H, Northrop JP, Grove JR, Kilpatrick KE, Su J-L, Ringold GM (1997) TNFα-mediated inhibition and reversal of adipocyte differentiation is accompanied by suppressed expression of PPARγ without effects on Pref-1 expression. Endocrinology 138:2776–2783PubMedCrossRef Xing H, Northrop JP, Grove JR, Kilpatrick KE, Su J-L, Ringold GM (1997) TNFα-mediated inhibition and reversal of adipocyte differentiation is accompanied by suppressed expression of PPARγ without effects on Pref-1 expression. Endocrinology 138:2776–2783PubMedCrossRef
19.
Zurück zum Zitat Boersema PJ, Raijmakers R, Lemeer S, Mohammed S, Heck AJR (2009) Multiplex peptide stable isotope dimethyl labeling for quantitative proteomics. Nat Protoc 4:484–494PubMedCrossRef Boersema PJ, Raijmakers R, Lemeer S, Mohammed S, Heck AJR (2009) Multiplex peptide stable isotope dimethyl labeling for quantitative proteomics. Nat Protoc 4:484–494PubMedCrossRef
20.
Zurück zum Zitat Bjørbaek C, Kahn BB (2004) Leptin signaling in the central nervous system and the periphery. Recent Prog Horm Res 59:305–331PubMedCrossRef Bjørbaek C, Kahn BB (2004) Leptin signaling in the central nervous system and the periphery. Recent Prog Horm Res 59:305–331PubMedCrossRef
21.
Zurück zum Zitat Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM (1995) Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest 95:2409–2415PubMedCrossRef Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM (1995) Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest 95:2409–2415PubMedCrossRef
22.
Zurück zum Zitat Könner AC, Brüning JC (2012) Selective insulin and leptin resistance in metabolic disorders. Cell Metab 16:144–152PubMedCrossRef Könner AC, Brüning JC (2012) Selective insulin and leptin resistance in metabolic disorders. Cell Metab 16:144–152PubMedCrossRef
23.
Zurück zum Zitat Wauman J, Tavernier J (2011) Leptin receptor signaling: pathways to leptin resistance. Front Biosci 16:2771–2793CrossRef Wauman J, Tavernier J (2011) Leptin receptor signaling: pathways to leptin resistance. Front Biosci 16:2771–2793CrossRef
24.
Zurück zum Zitat Czeh K, Winkler G, Melczer Z, Baranyi E (2000) The role of tumour necrosis factor (TNF)-alpha resistance in obesity and insulin resistance. Diabetologia 43:525–525CrossRef Czeh K, Winkler G, Melczer Z, Baranyi E (2000) The role of tumour necrosis factor (TNF)-alpha resistance in obesity and insulin resistance. Diabetologia 43:525–525CrossRef
25.
Zurück zum Zitat Loh K, Fukushima A, Zhang X et al (2011) Elevated hypothalamic TCPTP in obesity contributes to cellular leptin resistance. Cell Metab 14:684–699PubMedCrossRef Loh K, Fukushima A, Zhang X et al (2011) Elevated hypothalamic TCPTP in obesity contributes to cellular leptin resistance. Cell Metab 14:684–699PubMedCrossRef
26.
Zurück zum Zitat Berndt J, Kovacs P, Ruschke K et al (2007) Fatty acid synthase gene expression in human adipose tissue: association with obesity and type 2 diabetes. Diabetologia 50:1472–1480PubMedCrossRef Berndt J, Kovacs P, Ruschke K et al (2007) Fatty acid synthase gene expression in human adipose tissue: association with obesity and type 2 diabetes. Diabetologia 50:1472–1480PubMedCrossRef
27.
Zurück zum Zitat Boden G (2011) Obesity, insulin resistance and free fatty acids. Curr Opin Endocrinol Diabetes Obes 18:139–143PubMedCrossRef Boden G (2011) Obesity, insulin resistance and free fatty acids. Curr Opin Endocrinol Diabetes Obes 18:139–143PubMedCrossRef
28.
Zurück zum Zitat Capurso C, Capurso A (2012) From excess adiposity to insulin resistance: the role of free fatty acids. Vascul Pharmacol 57:91–97PubMedCrossRef Capurso C, Capurso A (2012) From excess adiposity to insulin resistance: the role of free fatty acids. Vascul Pharmacol 57:91–97PubMedCrossRef
29.
Zurück zum Zitat Menendez JA, Vazquez-Martin A, Ortega FJ, Fernandez-Real JM (2009) Fatty acid synthase: association with insulin resistance, type 2 diabetes, and cancer. Clin Chem 55:425–438PubMedCrossRef Menendez JA, Vazquez-Martin A, Ortega FJ, Fernandez-Real JM (2009) Fatty acid synthase: association with insulin resistance, type 2 diabetes, and cancer. Clin Chem 55:425–438PubMedCrossRef
30.
Zurück zum Zitat Olefsky JM, Saltiel AR (2000) PPAR gamma and the treatment of insulin resistance. Trends Endocrinol Metab 11:362–368PubMedCrossRef Olefsky JM, Saltiel AR (2000) PPAR gamma and the treatment of insulin resistance. Trends Endocrinol Metab 11:362–368PubMedCrossRef
31.
Zurück zum Zitat Sugden MC, Holness MJ (2004) Potential role of peroxisome proliferator-activated receptor-alpha in the modulation of glucose-stimulated insulin secretion. Diabetes 53(Suppl 1):S71–S81PubMedCrossRef Sugden MC, Holness MJ (2004) Potential role of peroxisome proliferator-activated receptor-alpha in the modulation of glucose-stimulated insulin secretion. Diabetes 53(Suppl 1):S71–S81PubMedCrossRef
32.
Zurück zum Zitat Yamauchi T, Kadowaki T (2013) Adiponectin receptor as a key player in healthy longevity and obesity-related diseases. Cell Metab 17:185–196PubMedCrossRef Yamauchi T, Kadowaki T (2013) Adiponectin receptor as a key player in healthy longevity and obesity-related diseases. Cell Metab 17:185–196PubMedCrossRef
33.
Zurück zum Zitat Russell L, Garrett-Sinha LA (2010) Transcription factor Ets-1 in cytokine and chemokine gene regulation. Cytokine 51:217–226PubMedCrossRef Russell L, Garrett-Sinha LA (2010) Transcription factor Ets-1 in cytokine and chemokine gene regulation. Cytokine 51:217–226PubMedCrossRef
34.
Zurück zum Zitat Shu CJ, Benoist C, Mathis D (2013) The immune system’s involvement in obesity-driven type 2 diabetes. Semin Immunol 24:436–442CrossRef Shu CJ, Benoist C, Mathis D (2013) The immune system’s involvement in obesity-driven type 2 diabetes. Semin Immunol 24:436–442CrossRef
35.
Zurück zum Zitat Singh S, Barrett J, Sakata K, Tozer RG, Singh G (2002) ETS proteins and MMPs: partners in invasion and metastasis. Curr Drug Targets 3:359–367PubMedCrossRef Singh S, Barrett J, Sakata K, Tozer RG, Singh G (2002) ETS proteins and MMPs: partners in invasion and metastasis. Curr Drug Targets 3:359–367PubMedCrossRef
36.
Zurück zum Zitat Chan YC, Roy S, Huang Y, Khanna S, Sen CK (2012) The microRNA miR-199a-5p down-regulation switches on wound angiogenesis by derepressing the v-ets erythroblastosis virus E26 oncogene homolog 1-matrix metalloproteinase-1 pathway. J Biol Chem 287:41032–41043PubMedCrossRef Chan YC, Roy S, Huang Y, Khanna S, Sen CK (2012) The microRNA miR-199a-5p down-regulation switches on wound angiogenesis by derepressing the v-ets erythroblastosis virus E26 oncogene homolog 1-matrix metalloproteinase-1 pathway. J Biol Chem 287:41032–41043PubMedCrossRef
37.
Zurück zum Zitat Sung H-K, Doh K-O, Son JE et al (2013) Adipose vascular endothelial growth factor regulates metabolic homeostasis through angiogenesis. Cell Metab 17:61–72PubMedCrossRef Sung H-K, Doh K-O, Son JE et al (2013) Adipose vascular endothelial growth factor regulates metabolic homeostasis through angiogenesis. Cell Metab 17:61–72PubMedCrossRef
38.
Zurück zum Zitat Sjøttem E, Rekdal C, Svineng G et al (2007) The ePHD protein SPBP interacts with TopBP1 and together they co-operate to stimulate Ets1-mediated transcription. Nucleic Acids Res 35:6648–6662PubMedCrossRef Sjøttem E, Rekdal C, Svineng G et al (2007) The ePHD protein SPBP interacts with TopBP1 and together they co-operate to stimulate Ets1-mediated transcription. Nucleic Acids Res 35:6648–6662PubMedCrossRef
39.
Zurück zum Zitat Traurig MT, Permana PA, Nair S, Kobes S, Bogardus C, Baier LJ (2006) Differential expression of matrix metalloproteinase 3 (MMP3) in preadipocytes/stromal vascular cells from nonobese nondiabetic versus obese nondiabetic Pima Indians. Diabetes 55:3160–3165PubMedCrossRef Traurig MT, Permana PA, Nair S, Kobes S, Bogardus C, Baier LJ (2006) Differential expression of matrix metalloproteinase 3 (MMP3) in preadipocytes/stromal vascular cells from nonobese nondiabetic versus obese nondiabetic Pima Indians. Diabetes 55:3160–3165PubMedCrossRef
40.
Zurück zum Zitat Kitamura S, Miyazaki Y, Hiraoka S et al (1999) PPARgamma inhibits the expression of c-MET in human gastric cancer cells through the suppression of Ets. Biochem Biophys Res Commun 265:453–456PubMedCrossRef Kitamura S, Miyazaki Y, Hiraoka S et al (1999) PPARgamma inhibits the expression of c-MET in human gastric cancer cells through the suppression of Ets. Biochem Biophys Res Commun 265:453–456PubMedCrossRef
41.
Zurück zum Zitat Galardi S, Mercatelli N, Giorda E et al (2007) miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. J Biol Chem 282:23716–23724PubMedCrossRef Galardi S, Mercatelli N, Giorda E et al (2007) miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. J Biol Chem 282:23716–23724PubMedCrossRef
42.
Zurück zum Zitat Zhao J-J, Lin J, Yang H et al (2008) MicroRNA-221/222 negatively regulates estrogen receptor alpha and is associated with tamoxifen resistance in breast cancer. J Biol Chem 283:31079–31086PubMedCrossRef Zhao J-J, Lin J, Yang H et al (2008) MicroRNA-221/222 negatively regulates estrogen receptor alpha and is associated with tamoxifen resistance in breast cancer. J Biol Chem 283:31079–31086PubMedCrossRef
43.
Zurück zum Zitat Davis BN, Hilyard AC, Nguyen PH, Lagna G, Hata A (2009) Induction of microRNA-221 by platelet-derived growth factor signaling is critical for modulation of vascular smooth muscle phenotype. J Biol Chem 284:3728–3738PubMedCrossRef Davis BN, Hilyard AC, Nguyen PH, Lagna G, Hata A (2009) Induction of microRNA-221 by platelet-derived growth factor signaling is critical for modulation of vascular smooth muscle phenotype. J Biol Chem 284:3728–3738PubMedCrossRef
44.
Zurück zum Zitat Zhang C-Z, Zhang J-X, Zhang A-L et al (2010) MiR-221 and miR-222 target PUMA to induce cell survival in glioblastoma. Mol Cancer 9:229PubMedCrossRef Zhang C-Z, Zhang J-X, Zhang A-L et al (2010) MiR-221 and miR-222 target PUMA to induce cell survival in glioblastoma. Mol Cancer 9:229PubMedCrossRef
45.
Zurück zum Zitat Frenquelli M, Muzio M, Scielzo C et al (2010) MicroRNA and proliferation control in chronic lymphocytic leukemia: functional relationship between miR-221/222 cluster and p27. Blood 115:3949–3959PubMedCrossRef Frenquelli M, Muzio M, Scielzo C et al (2010) MicroRNA and proliferation control in chronic lymphocytic leukemia: functional relationship between miR-221/222 cluster and p27. Blood 115:3949–3959PubMedCrossRef
46.
Zurück zum Zitat Pineau P, Volinia S, McJunkin K et al (2010) miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci U S A 107:264–269PubMedCrossRef Pineau P, Volinia S, McJunkin K et al (2010) miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci U S A 107:264–269PubMedCrossRef
47.
Zurück zum Zitat Rao X, Di Leva G, Li M et al (2011) MicroRNA-221/222 confers breast cancer fulvestrant resistance by regulating multiple signaling pathways. Oncogene 30:1082–1097PubMedCrossRef Rao X, Di Leva G, Li M et al (2011) MicroRNA-221/222 confers breast cancer fulvestrant resistance by regulating multiple signaling pathways. Oncogene 30:1082–1097PubMedCrossRef
48.
Zurück zum Zitat Trojanowska M (2000) Ets factors and regulation of the extracellular matrix. Oncogene 19:6464–6471PubMedCrossRef Trojanowska M (2000) Ets factors and regulation of the extracellular matrix. Oncogene 19:6464–6471PubMedCrossRef
49.
Zurück zum Zitat Guijarro MV, Castro ME, Romero L, Moneo V, Carnero A (2007) Large scale genetic screen identifies MAP17 as protein bypassing TNF-induced growth arrest. J Cell Biochem 101:112–121PubMedCrossRef Guijarro MV, Castro ME, Romero L, Moneo V, Carnero A (2007) Large scale genetic screen identifies MAP17 as protein bypassing TNF-induced growth arrest. J Cell Biochem 101:112–121PubMedCrossRef
50.
Zurück zum Zitat Antoon JW, Lai R, Struckhoff AP et al (2012) Altered death receptor signaling promotes epithelial-to-mesenchymal transition and acquired chemoresistance. Sci Rep 2:539PubMedCrossRef Antoon JW, Lai R, Struckhoff AP et al (2012) Altered death receptor signaling promotes epithelial-to-mesenchymal transition and acquired chemoresistance. Sci Rep 2:539PubMedCrossRef
51.
Zurück zum Zitat Nakamura Y, Esnault S, Maeda T, Kelly EAB, Malter JS, Jarjour NN (2004) Ets-1 regulates TNF-alpha-induced matrix metalloproteinase-9 and tenascin expression in primary bronchial fibroblasts. J Immunol 172:1945–1952PubMed Nakamura Y, Esnault S, Maeda T, Kelly EAB, Malter JS, Jarjour NN (2004) Ets-1 regulates TNF-alpha-induced matrix metalloproteinase-9 and tenascin expression in primary bronchial fibroblasts. J Immunol 172:1945–1952PubMed
Metadaten
Titel
Human adipose microRNA-221 is upregulated in obesity and affects fat metabolism downstream of leptin and TNF-α
verfasst von
A. Meerson
M. Traurig
V. Ossowski
J. M. Fleming
M. Mullins
L. J. Baier
Publikationsdatum
01.09.2013
Verlag
Springer Berlin Heidelberg
Erschienen in
Diabetologia / Ausgabe 9/2013
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-013-2950-9

Weitere Artikel der Ausgabe 9/2013

Diabetologia 9/2013 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.