ArticlePDF Available

A Systems Biology Approach Identifies Inflammatory Abnormalities Between Mouse Strains Prior to Development of Metabolic Disease

Authors:

Abstract and Figures

Type 2 diabetes and obesity are increasingly affecting human populations around the world. Our goal was to identify early molecular signatures predicting genetic risk to these metabolic diseases using two strains of mice that differ greatly in disease susceptibility. We integrated metabolic characterization, gene expression, protein-protein interaction networks, RT-PCR, and flow cytometry analyses of adipose, skeletal muscle, and liver tissue of diabetes-prone C57BL/6NTac (B6) mice and diabetes-resistant 129S6/SvEvTac (129) mice at 6 weeks and 6 months of age. At 6 weeks of age, B6 mice were metabolically indistinguishable from 129 mice, however, adipose tissue showed a consistent gene expression signature that differentiated between the strains. In particular, immune system gene networks and inflammatory biomarkers were upregulated in adipose tissue of B6 mice, despite a low normal fat mass. This was accompanied by increased T-cell and macrophage infiltration. The expression of the same networks and biomarkers, particularly those related to T-cells, further increased in adipose tissue of B6 mice, but only minimally in 129 mice, in response to weight gain promoted by age or high-fat diet, further exacerbating the differences between strains. Insulin resistance in mice with differential susceptibility to diabetes and metabolic syndrome is preceded by differences in the inflammatory response of adipose tissue. This phenomenon may serve as an early indicator of disease and contribute to disease susceptibility and progression.
Content may be subject to copyright.
A Systems Biology Approach Identifies Inflammatory
Abnormalities Between Mouse Strains Prior to
Development of Metabolic Disease
Marcelo A. Mori,
1
Manway Liu,
2
Olivier Bezy,
1
Katrine Almind,
1
Hagit Shapiro,
1
Simon Kasif,
2
and
C. Ronald Kahn
1
OBJECTIVE—Type 2 diabetes and obesity are increasingly
affecting human populations around the world. Our goal was to
identify early molecular signatures predicting genetic risk to
these metabolic diseases using two strains of mice that differ
greatly in disease susceptibility.
RESEARCH DESIGN AND METHODS—We integrated meta-
bolic characterization, gene expression, protein-protein interac-
tion networks, RT-PCR, and flow cytometry analyses of adipose,
skeletal muscle, and liver tissue of diabetes-prone C57BL/6NTac
(B6) mice and diabetes-resistant 129S6/SvEvTac (129) mice at 6
weeks and 6 months of age.
RESULTS—At 6 weeks of age, B6 mice were metabolically
indistinguishable from 129 mice, however, adipose tissue showed
a consistent gene expression signature that differentiated be-
tween the strains. In particular, immune system gene networks
and inflammatory biomarkers were upregulated in adipose tissue
of B6 mice, despite a low normal fat mass. This was accompanied
by increased T-cell and macrophage infiltration. The expression
of the same networks and biomarkers, particularly those related
to T-cells, further increased in adipose tissue of B6 mice, but only
minimally in 129 mice, in response to weight gain promoted by
age or high-fat diet, further exacerbating the differences between
strains.
CONCLUSIONS—Insulin resistance in mice with differential
susceptibility to diabetes and metabolic syndrome is preceded by
differences in the inflammatory response of adipose tissue. This
phenomenon may serve as an early indicator of disease and
contribute to disease susceptibility and progression. Diabetes
59:2960–2971, 2010
Type 2 diabetes and obesity are major causes of
mortality and morbidity worldwide (1). Accord-
ing to World Health Organization estimates,
more than 1 billion adults are overweight and
more than 200 million individuals have type 2 diabetes.
The etiologies of obesity and diabetes are complex and
created by interactions between environmental factors
(i.e., high caloric intake and reduced energy expenditure)
(1,2) and genetic background (3–5).
Inbred mouse strains are useful models for studying the
role of the environment and genes in differential suscep-
tibility to diabetes and obesity (6 –14). In response to
high-fat diet, aging, or genetic challenge, C57BL/6NTac
(B6) mice become severely insulin resistant, hyperinsu-
linemic, and diabetic, whereas 129S6/SvEvTac (129) mice
are resistant to these challenges (7,8). Using intercross
and F2 mice, our group and others have previously shown
that the difference in disease susceptibility between the
strains is inherited in a dominant fashion and linked to
quantitative traits on at least three different chromosomes
(8,15).
In the present study, we compared gene expression
profiles of B6 and 129 mice in different tissues, ages, and
diets. For each comparison, we identified the differentially
expressed network of genes between the strains using a
novel variant of gene network enrichment analysis
(GNEA) (16), an algorithm that integrates gene expression
data together with protein-protein interaction networks.
We subsequently intersected the results of the compari-
sons between B6 and 129 mice to identify genes and
pathways that were significant to each or all conditions,
i.e., different tissues, ages, and diets. Of the numerous,
differentially expressed subnetworks in the adipose tissue
of B6 versus 129 mice; the most significant related to the
immune system. Importantly, these differences were iden-
tified even at 6 weeks, an age when the mouse strains were
indistinguishable by fat mass or metabolic phenotyping.
RT-PCR and flow cytometry confirmed higher expression
of major inflammatory markers and higher infiltration of
macrophages and T-cells in adipose tissue of 6-week-old
B6 versus 129 mice. Weight gain associated with aging or
high-fat diet further increased inflammation in adipose
tissue of B6 but not in 129, mice. Taken together, these
results demonstrate that measurable differences in the
inflammatory milieu of the adipose tissue precede measur-
able differences in insulin sensitivity in response to weight
gain and contribute to the differences in diabetes risk
between B6 and 129 mice.
RESEARCH DESIGN AND METHODS
C57BL/6NTac and 129S6/SvEvTac male mice were obtained from Taconic
(Germantown, NY). Mice were maintained on a 12-h light-dark cycle with ad
libitum access to tap water (reverse osmosis purified) and a chow diet
containing 21% calories from fat, 23% from protein, and 55% from carbohy-
drates (Mouse Diet 9F; PharmaServ, Framingham, MA). For the feeding
studies, 6-week-old mice were submitted to low-fat diet (Rodent NIH-31M
Auto: 14% calories from fat, 25% from protein, and 61% from carbohydrates
[Taconic]) or high-fat diet (TD.93075: 55% calories from fat, 21% calories from
protein, and 24% calories from carbohydrates [Harlan Teklad, Madison, WI])
for 18 weeks before they were killed. In addition to fat content, these diets
From the
1
Section on Integrative Physiology and Metabolism, Joslin Diabetes
Center, Harvard Medical School, Boston, Massachusetts; and the
2
Depart-
ment of Biomedical Engineering, Boston University, Boston, Massachusetts.
Corresponding author: C. Ronald Kahn, c.ronald.kahn@joslin.harvard.edu.
Received 16 March 2010 and accepted 4 August 2010. Published ahead of
print at http://diabetes.diabetesjournals.org on 16 August 2010. DOI:
10.2337/db10-0367.
M.A.M. and M.L. contributed equally to this work.
© 2010 by the American Diabetes Association. Readers may use this article as
long as the work is properly cited, the use is educational and not for profit,
and the work is not altered. See http://creativecommons.org/licenses/by
-nc-nd/3.0/ for details.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked “advertisement” in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
ORIGINAL ARTICLE
2960 DIABETES, VOL. 59, NOVEMBER 2010 diabetes.diabetesjournals.org
differ in other components that could also influence the results of our analysis.
For example, our chow diet contained fat from animal sources, whereas both
low-fat and high-fat diets contained vegetable fat. These differences may have
contributed to the fact that a few metabolic parameters displayed in Table 1,
such as insulin levels, did not necessarily track with the percentage of fat in
the diet. However, in this study and in others, these diets have been primarily
used as independent paradigms to investigate the impact of the environment
on weight gain of animal models, showing metabolic effects that closely
resembled different characteristics of human obesity (8). Caloric-restricted
animals were obtained from the National Institute on Aging. Caloric restric-
tion was initiated at 14 weeks of age with a 10% decrease in calories, increased
to 25% restriction at 15 weeks, and to 40% restriction at 16 weeks, which was
maintained until the age of 6 months when the mice were killed. Protocols for
animal use were reviewed and approved by the Animal Care Committee of the
Joslin Diabetes Center and were in accordance with the National Institutes of
Health guidelines.
Gene expression analysis. RNA pooled from tissues of two or three mice
was used for microarray analysis. Two to five Affymetrix murine chips
U74Av.2 (Santa Clara, CA) were used per mouse strain, for each comparison
of B6 versus 129 mice. Quantitative RT-PCR was performed using a SYBR
Green-based ABI Prism 7900 Sequence Detection System (Applied Biosys-
tems, Foster City, CA). For details, refer to the supplementary METHODS in the
online appendix available at http://diabetes.diabetesjournals.org/cgi/
content/full/db10-0367/DC1.
Gene network enrichment analysis. A novel version of Gene Network
Enrichment Analysis (16) was applied to each comparison of B6 versus 129
mice to identify gene networks (subnetworks of genes with edges connecting
physically interacting protein products) with differential activation between
these mice, and to test such networks for statistically significant, over-
represented pathways and processes. Briefly, the algorithm was composed of
the following steps (see supplementary METHODS for details).
First, for each gene, a differential expression significance value was
calculated in B6 versus 129 mice, converted to zscores, and used to annotate
the corresponding proteins in a protein-protein interaction network curated
from literature (17) (Fig. 1A).
Subsequently, a computationally intensive, stochastic optimization algo-
rithm (18) was employed to identify a subnetwork of genes with a high mean
zscore (reflecting high significance). This process was repeated many times
to yield a multitude of subnetworks (Fig. 1B). The hypothesis was that a
consensus among the stochastically detected subnetworks represented a
prediction of the true, differentially active gene network in B6 versus 129 mice.
Thus, each gene was associated with a Pvalue based on the observed number
of subnetworks in which it appeared, reflecting its chance probability of
belonging to the true network (Fig. 1C).
In the next step of the algorithm, each biological process and pathway from
the Gene Ontology and Molecular Signatures Database were tested for
over-representation of genes with low Pvalues, using standard statistical
methods (Fig. 1D).
Finally, for each statistically significant pathway or biological process, the
algorithm produced a gene network consisting of pathway member genes and
all additional genes with significant, adjusted Pvalues (false discovery rate
0.25) that shared an edge with a pathway member gene in the protein-
protein interaction network (Fig. 1E), providing an intuitively appealing
visualization aid that helped in interpreting the enrichment results.
Flow cytometry. Erythrocyte-free stromavascular cells were isolated from
epididymal fat pads after collagenase digestion as previously described (19).
These cells were incubated with a mix of antibodies against different surface
markers and analyzed using the LSRII flow cytometer (BD Biosciences, San
Jose, CA). See supplementary METHODS for detailed protocol.
Statistics. Statistical analyses for GNEA are described in supplementary
METHODS. All other experiments were compared using ANOVA. Results are
expressed as mean SEM. Pvalues 0.05 were considered significant.
RESULTS
Gene network signatures associated with the predis-
position to metabolic diseases in B6 versus 129 mice.
At 6 months of age, on a chow diet, B6 mice were
overweight, hyperglycemic, and exhibited higher plasma
leptin and insulin levels than 129 mice of the same age
(Table 1, supplementary Fig. S1) (8). B6 mice showed
clear signs of insulin resistance when compared with 129
mice, even on low-fat diet (LFD), and this difference was
further exacerbated when the mice were placed on a
high-fat diet (Table 1) (8). At 6 weeks of age, however, no
phenotypic differences or serum markers indicative of
diabetes and obesity could be detected between the two
strains (Table 1) (8). Indeed, 6-week-old 129 mice have the
same weight as B6 mice and actually have significantly
more fat mass, as indicated by higher fat pad weight (Table
1). By studying B6 versus 129 mice at different ages and on
different diets, one may therefore elucidate the back-
ground inherited phenomena predisposing to obesity-ac-
companied insulin resistance.
To identify gene networks that might provide early
biomarkers of adult metabolic disease, we initially per-
formed microarray analyses on RNA from adipose tissue,
liver, and skeletal muscle of B6 and 129 mice at 6 weeks
and 6 months of age on a standard chow diet. We then
applied GNEA to detect networks that were differentially
active in a predisease stage and to estimate how they
changed over time in disease-prone versus diabetes-resis-
tant strains (Fig. 1). Since disease susceptibility is an
inherited phenomenon in these animals, we hypothesized
that the predisposing hereditable biological processes
would be those showing differential activity in young mice
and maintained or further increased with age.
Among the three different tissues in which B6 versus
129 mice were compared, only adipose tissue exhibited
significantly enriched biological processes (Qvalue
0.33) at 6 weeks of age by GNEA, with 12 enriched
gene sets, and this increased to 25 enriched gene sets at
6 months of age. Of these, only two sets (“immune
system process” and “regulation of cytokine secretion”)
were found enriched at both ages (supplementary Table
1), and both of these related to inflammatory pathways.
Furthermore, the “immune system process” was the
TABLE 1
Metabolic parameters in B6 vs. 129 mice at different ages and dietary conditions
Chow diet (21% fat) Low-fat diet (14% fat) High-fat diet (55% fat)
6 weeks old 6 months old
B6 129 B6 129 B6 129 B6 129
Body weight (g) 21.9 0.6 23.5 0.5 45.5 1.0 30.4 1.2 41.5 1.4 30.9 1.2 46.4 1.1 39.2 1.5
Epididymal fat
mass (g) 0.09 0.01 0.21 0.02 1.67 0.14 2.14 0.30 1.62 0.17 0.58 0.11 3.39 0.33 3.92 0.14
Blood glucose–
random fed
(mg/dl) 155.5 3.1 129.3 2.9 168.2 12.3 106.6 3.5 148.5 7.4 102.5 3.4 153.3 6.7 105.5 3.7
Insulin (ng/ml) 2.26 0.48 3.12 0.58 92.14 25.71 3.96 2.04 24.78 12.51 1.29 0.32 32.73 12.46 3.58 0.41
Leptin (ng/ml) 7.14 0.70 9.00 0.90 76.18 3.82 16.67 4.5 72.50 8.06 4.97 0.77 82.38 7.41 46.81 6.02
Values are expressed as mean SE of 8 animals per group.
M.A. MORI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 59, NOVEMBER 2010 2961
most significantly enriched gene set at 6 months of age
(Qvalue 0.008; supplementary Table 1). In contrast, in
the liver, 37 gene sets (Qvalue 0.33) were differen-
tially regulated in B6 versus 129 at 6 months of age, but
none was significantly different in the 6-week-old ani-
mals. In skeletal muscle, no significant difference was
observed at either age (data not shown).
In addition to GNEA, the adipose tissue dataset was
analyzed using gene set enrichment analysis (GSEA) (20)
and the standard hypergeometric test (21). GSEA also
Diet
(% lipid)
Tissue
Age
(weeks)
(months)
129
B6 vs.
55%
Liver Adipose Skeletal Muscle
14% 55%
21% 14% 55%
21% 14% 21%
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 mos
6 wks
6 wks
6 wks
6 wks
6 wks
6 wks
Map individual p-values onto
a network of genes.
Find differentially active
subnetworks
G1 G2 G3 G4 G5 G6 G7 G8 G9 G10 G11 G12
C1
C1
C1
C1
C2
C2
C2
C2
P
Identify biological processes
enriched in genes with high
MAN-scores and visualize in
the network context.
G1G2GN
G3
...
Order genes by MAN-scores
Differential gene
expression (B6 vs. 129)
Network of genes labeled
with their p-values
Differentially active
subnetworks of genes
Computational Analysis
Experimental Setup
CD45
IKBKA
CD75
MCP1
SDF1
Calculate the Membership
in Differentially Active
SubNetwork score (MAN-score)
per gene.
...
MAN-score
Network view of a biological process
A
B
CD
E
FIG. 1. Overview of gene network enrichment analysis. Individual comparisons of B6 and 129 mice were performed in different tissue types
(adipose, skeletal muscle, and liver), ages (6 weeks and 6 months), and diets (high fat, low fat, and standard chow). A: For each comparison, a
differential expression significance value was determined for each gene, converted to a zscore, and mapped onto a network of protein-protein
interactions. The colors in the network represent the different ranges of zscores. B: A stochastic search algorithm (18) was then applied to search
for subnetworks with high mean zscore (high significance). Each application of the algorithm identifies one subnetwork; hence, to detect a
multitude of subnetworks, the algorithm was run many times. The consensus among these subnetworks represented approximations of the true,
differentially active network in B6 vs. 129 mice. C: A membership in differentially active subnetwork (MAN) score was determined for each gene.
MAN scores are Pvalues and represent the probability of genes to belong to the true, differentially active network. This score was calculated as
the random chance probability that the gene is detected in the observed number of subnetworks computed over permutations of the data. D:
Biological processes and pathways were tested for enrichment in genes with high MAN scores. Statistically significant pathways and processes
were predicted to be differentially active in B6 vs. 129 mice. E: For each significant process and pathway, a network visualization was generated
to aid interpretation.
SYSTEMS BIOLOGY AND SUSCEPTIBILITY TO DIABETES
2962 DIABETES, VOL. 59, NOVEMBER 2010 diabetes.diabetesjournals.org
identified differences in inflammation at 6 weeks. In par-
ticular, it found a significant difference in the “humoral
immune response” biological process (supplementary Ta-
ble 2). On the other hand, GSEA failed to identify any gene
set that was differentially expressed at both ages (supple-
mentary Table 3). By comparison, the standard hypergeo-
metric test (21) failed to identify either a clear immune
signature at 6 weeks or any enriched gene sets at 6 months
(supplementary Tables 2 and 3).
Gene networks associated with the immune system
process are differentially altered in the adipose tis-
sue of B6 versus 129 mice. To determine how genes
related to the immune response were differentially ex-
pressed in the fat tissue of young B6 and 129 mice and
changed in expression as the mice aged, we identified the
network associated with the “immune system process”
gene set (supplementary Fig. S2) and plotted the histo-
gram of differential expression for its genes in B6 versus
129 mice at 6 weeks and 6 months (Fig. 2). A few
individual genes, such as apolipoprotein A (APOA1 and
APOA2) and mannose-binding lectin (MBL2) were more
strongly downregulated in B6 versus 129 mice at 6 months
and less so at 6 weeks. Nonetheless, much of the immune
system process network was upregulated at 6 months
compared with 6 weeks (note the right-shift in the histo-
gram for 6 months vs. 6 weeks in Fig. 2). Lymphoid and
myeloid markers, such as CD45, CD3, and chemokine (C-C
motif) receptor 5 (CCR5), monocyte chemotactic protein-1
(MCP1), as well as proinflammatory genes such as chemo-
kine (C-C motif) ligand 5 (CCL5) and interleukin-6 (IL6),
were more highly expressed in B6 compared with 129 mice
at 6 weeks, and this increased further at 6 months of age
(supplementary Fig. S2, left panel,arrows). Moreover, as
shown by the histogram (Fig. 2), many of the immune
system process genes already showed twofold or greater
differences in expression in B6 versus 129 mice at 6 weeks,
an age when no significant metabolic differences were
observed between strains.
Genes that have protein products that physically inter-
act tend to correlate in expression (22) and are likely
related by function or membership in the same signaling
pathways. We consequently examined the subnetworks of
genes that interact at the protein level with inflammatory
markers identified in the strain comparisons (Fig. 3). For
the T-cell marker Thy1, minor changes in the expression of
its interacting genes were observed in the 6-week-old B6
versus 129 mice comparisons (Fig. 3A,left panel). How-
ever, by 6 months of age, more than 90% of the nodes in the
network were differentially active between the two strains
(large green triangles in Fig. 3A,right panel). Hence, genes
interacting with Thy1 were dramatically more upregulated
with age-associated weight gain in adipose tissue of B6
mice than in 129 mice, suggesting that B6 mice have more
T-cells recruited to adipose tissue in response to increas-
0
10
20
30
40
50
60
70
-6.88 -5.86 -4.85 -3.83 -2.82 -1.80 -0.79 0.23 1.24 2.26 3.27 4.29 5.31
6 weeks
6 months
Fold-change (log
2
) in B6 vs. 129 mice
# of genes
APOA1 APOA2 MBL2
CCR5, CD64
FIG. 2. Histogram of differential expression between B6 and 129 mice at 6 weeks and 6 months of age for genes in the immune system process
network. The x-axis values are the upper bounds of the histogram bins and correspond to the log
2
fold-change difference in B6 vs. 129 mice. Few
network genes are unchanged (absolute log
2
fold change <1) in B6 vs. 129 mice at 6 months. The majority are strongly upregulated (log
2
fold
change >1), although a few are strongly downregulated (log
2
fold change <1). A network view of the same biological process demonstrating
the fold changes of individual genes is provided in supplementary Fig. S2.
M.A. MORI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 59, NOVEMBER 2010 2963
CD11B CD11B
CD18
CD18
CD61
CD61
CD51
CD51
CD87
CD87
CD23
CD23
CD152
CD152
CD49E
CD49E
CCBP1
CCBP1
MCP1 MCP1
A
CD75 CD75
B
C
Fold Change 6 mos > 6 wks
Fold Change 6 mos < 6 wks
1
< 0.5
> 2
Fold Change 6 wks
1
< 0.5
> 2
Fold Change 6 mos
FIG. 3. Gene networks associated with inflammatory markers in adipose tissue of B6 and 129 mice at 6 weeks (left) and 6 months (right) of age. Gene
networks were generated by mapping genes that were significantly overrepresented (Qvalue <0.25) among the gene network enrichment analysis
results at 6 months intersected with those at 6 weeks onto protein-protein interaction networks involving at least one interactor to each inflammatory
marker: (A) Thy1, (B) CD45, and (C) MCP1. The CD45 network was further restricted only to genes showing a greater than twofold magnitude
difference in B6 vs. 129 mice between 6 weeks and 6 months. This was done to aid visualization since the unrestricted network consisted of 416
interactions and 141 genes. Colors range from bright red to green, corresponding to twofold less and twofold greater differences in expression between
B6 and 129 at each age, respectively. Nodes point upward if the fold-change difference between B6 and 129 is greater at 6 months than at 6 weeks, and
downward otherwise. The node size corresponds to the magnitude of that fold-change difference between the two ages. The inflammatory network
around each biomarker is drawn to scale between the two ages. Networks around different biomarkers are not to scale with one another.
SYSTEMS BIOLOGY AND SUSCEPTIBILITY TO DIABETES
2964 DIABETES, VOL. 59, NOVEMBER 2010 diabetes.diabetesjournals.org
ing adiposity than 129 mice. The same pattern was ob-
served in subnetworks generated for CD45, a leukocyte
marker (Fig. 3B); CD11c, a dendritic cell marker (supple-
mentary Fig. S3A); CD11b, a macrophage marker (supple-
mentary Fig. S3B); MCP1, a monocyte chemokine (Fig.
3C); and tumor necrosis factor (TNF), a proinflamma-
tory cytokine (supplementary Fig. S3C). Together these
data suggest that B6 mice show a proinflammatory ten-
dency in fat at 6 weeks of age not observed in 129 mice,
and that this differentially increases with weight gain
promoted by aging, affecting the pool of inflammatory
cells.
Changes in expression of inflammatory markers and
cytokines/chemokines in adipose tissue of B6 versus
129 mice. To validate the observations obtained using
network analysis, we measured markers of inflammatory
cells and cytokine/chemokine genes in the adipose tissue
of B6 and 129 mice using quantitative RT-PCR (qPCR)
(Fig. 4). In agreement with the microarray data, at 6 weeks
of age, there were higher levels of macrophage/myeloid
cell markers (CD68, CD11b, and CD18), lymphocyte mark-
ers (CD3, CD72, and CD80), T-cell attractants/activators
CCL5/RANTES, stroma cell-derived factor-1(SDF1),
and interferon-(IFN), among others, in B6 versus 129
mice (Fig. 4Aand B). For most of these genes, the
differences between strains were maintained or further
exacerbated at 6 months of age. MCP1, a monocyte
attractant chemokine, despite being similarly expressed in
young B6 and 129 mice, exhibited a 4.4-fold increase in B6
mice between 6 weeks and 6 months of age, whereas in
129 mice, there was a 78% reduction with age (Fig. 4B). For
a few cytokines, namely TNF, IL6, and macrophage
migration inhibitory factor (MIF), no differences were
observed between the strains at any age (Fig. 4B).
We also validated by RT-PCR the expression of selected
genes identified as differentially present in the differen-
tially active subnetworks identified by GNEA. Two medi-
ators of lipopolysaccharide (LPS) signaling, i.e., the
lymphocyte antigen 86 (Ly86) and LPS-binding protein
(LBP), were highly ranked by GNEA and were substan-
tially higher in B6 compared with 129 mice at both ages (Fig.
4C).
To determine if the higher expression of inflammation
markers in B6 versus 129 mice was restricted to the
adipose tissue or present in other tissues, we assessed the
expression of two macrophage markers (CD68 and F4/80),
two T-cell markers (CD3 and Thy1), and two chemokines
(MCP1 and SDF1) in the liver, skeletal muscle, and
spleen of B6 and 129 mice at both ages using qPCR. The
pattern of expression in liver was qualitatively similar to
that in adipose tissue, but with smaller fold differences
(supplementary Fig. S4A). In contrast, no differences were
observed in the skeletal muscle or spleen in the expression
for most of these genes (supplementary Fig. S4Band C).
The two exceptions were MCP1, which was increased in
all tissues in the 6-month-old B6 mice when compared
with 129 mice, and CD3, which increased in both B6 and
129 mice with age in skeletal muscle only (supplementary
Fig. S4).
An altered repertoire of inflammatory cells in the
adipose tissue of B6 versus 129 mice. To determine if
there was a cellular shift in the adipose tissue contributing
A
mRNA expression
(Fold Change)
mRNA expression
(Fold Change)
mRNA expression
(Fold Change)
0
0.5
1
1.5
2
2.5
3
3.5
CD45 CCR5 CD68 F4/80 CD11b CD11c CD18 CD3 Thy1 CD72 CD80
B6 6wk
129 6wk
B6 6 mo
129 6 mo
B6 6wk
129 6wk
B6 6 mo
129 6 mo
B6 6wk
129 6wk
B6 6 mo
129 6 mo
**
#
*
*** **
*
*
#
**
****
#
**
###
*
*
*
**
#
#
B
0
1
2
3
4
5
6
TNF IL6 IFNγMIF MCP1 CCL5 SDF
**
##
***
##
##
**
***
0
0.2
0.4
0.6
0.8
1
1.2
1.4
LBP Ly86
*** ***
***
*
Myeloid markers Lymphoid markers
Leukocyte markers
Proinflammatory
cytokines
Chemokines
C
α
FIG. 4. Expression of inflammatory markers in
adipose tissue of 6-week-old (6 weeks) or
6-month-old (6 months) B6 and 129 mice. A:
Leukocyte markers, including selective myeloid
cell markers (e.g., macrophages and dendritic
cells) and lymphoid markers (e.g., lymphocytes).
B: Cytokines and chemokines implicated with
inflammatory responses. C: Mediators of LPS
signaling. mRNA expression was assessed by
qPCR. All values are normalized by TATA-bind-
ing protein (TBP) and expressed as fold change
of the 6-week-old B6 average value. Results rep-
resent mean SEM of 5–7 animals. *P<0.05;
**P<0.01; ***P<0.001 vs. 129. #P<0.05;
##P<0.01; ###P<0.001 vs. 6 weeks.
M.A. MORI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 59, NOVEMBER 2010 2965
to our computational and qPCR results, we analyzed the
inflammatory cell repertoire in the fat tissue of B6 versus
129 mice using flow cytometry (Fig. 5). This revealed a
significant twofold increase in the number of nonerythro-
cyte stromavascular fraction cells per gram of epididymal
fat in B6 compared with 129 mice at both ages (Fig. 5A). At
6 weeks of age, 75% of this difference was explained by a
higher number of leukocytes in B6 fat versus 129 fat.
Although the number of leukocytes per gram of adipose
tissue decreased in both strains with age as adipose mass
increased, at both time points the number of leukocytes
was about 2.5-fold higher in the stromavascular fraction
from B6 fat than in 129 fat (Fig. 5A). Likewise, the number
of macrophages per gram of fat decreased with age
because of growing fat mass, but was higher in the B6
versus 129 mice at both ages (Fig. 5A). These data corre-
lated with histologic evidence of crown-like structures,
i.e., collections of macrophages surrounding dead or dying
adipocytes, in the adipose tissue of 6-month-old B6 mice
(supplementary Fig. S1).
Somewhat surprisingly, the density of CD11c
-myeloid
cells, which have been previously associated with insulin
resistance states in mice (23), was decreased in B6 versus
129 mice at 6 weeks and 6 months of age by 65 and 37%,
respectively (Fig. 5A). In contrast, the T-cell population
was dramatically increased by 7.1-fold in B6 mice at 6
weeks of age (Fig. 5A). Furthermore, the density of T-cells
increased by 25-fold in B6 mice with age-associated weight
gain, but showed only a threefold increase in 129 mice
(Fig. 5A). Thus, at 6 months of age, B6 mice had 55-fold
more total T-cells and 60-fold more CD4
T-helper cells
in the adipose tissue compared with the 129 mice (Fig.
5A). B-cells were also present in the adipose tissue of
these mice; however, they were not considered in the
analysis because of their modest numbers in comparison
with other leukocytes (1%).
A similar pattern of lymphocyte/monocyte infiltration
was observed in the 6-week-old mice when the total
number of cells in the epididymal fat pads was calculated,
i.e., cells per gram total grams of fat (Fig. 5B). When
expressed as total cells, however, it was apparent that the
number of each of the myeloid and lymphoid cell popula-
tion increased with age in both strains. Importantly, the
differences between strains were maintained in both
younger and older mice (Fig. 5B). These results confirm
the gene network and expression analysis and demon-
strate that most of these differences are consistent with
differences in the lymphoid/myeloid population in the
adipose tissue of B6 versus 129 mice.
Effects of high-fat diet on the inflammatory status of
the adipose tissue of B6 versus 129 mice. To investi-
gate how differences in the genetic background could
impact the effects of diet-induced obesity on the inflam-
matory cell repertoire of the adipose tissue, cohorts of B6
and 129 mice were placed on a high-fat diet (HFD) for 18
weeks and compared with mice on LFD. On both LFD and
HFD, the mRNA expression of the macrophage marker
CD68 was higher by twofold or greater in B6 compared
with 129 mice (Fig. 6A). Similarly, MCP1 expression was
higher in B6 than in 129 on LFD, and this was maintained
with HFD (Fig. 6A). The macrophage marker F4/80 and
SDF1both trended higher in B6 than in 129 mice on LFD,
but neither reached statistical significance (Fig. 6A). Inter-
estingly, the HFD-induced increases in the expression of
the T-cell markers CD3 and Thy1 were completely abro-
gated in the 129 mice (Fig. 6A). Caloric restriction, an
intervention that drastically reduces fat mass, helps pre-
vents age-associated diseases, and prolongs life span (24),
decreased the expression of CD3 and Thy1 in the adipose
tissue of B6 mice by 41 and 51%, respectively, without
affecting the expression of macrophage markers (supple-
mentary Fig. S5). These results demonstrate a positive
correlation between adiposity and the expression of T-
cells in adipose tissue of B6 mice, a phenomenon that is
significantly impaired in 129 mice.
Accordingly, analysis of B6 versus 129 mice under LFD
and HFD using network analysis revealed a substantial
number of genes in the “immune system process” gene set
*** *** **
***
###
###
###
###
##
## ###
###
### ###
##
##
*
*
###
##
##
### ### ###
***
***
**
**
**
** ****
###
A
B6 6wk
1800
Cells (x10
3
) / g of fat
Cells (x10
5
)
1600
1400
1200
1200
800
600
400
200
0
Nonerythrocytes
stromavascular
fraction cells
Leukocytes
Macrophages
CD11c
+
-Myeloid cells
T-cells
T-helper cells
Nonerythrocytes
stromavascular
fraction cells
Leukocytes
Macrophages
CD11c
+
-Myeloid cells
T-cells
T-helper cells
0
5
10
15
20
25
129 6wk
B6 6 mo
129 6 mo
B6 6wk
129 6wk
B6 6 mo
129 6 mo
B
FIG. 5. Immune cell repertoire in adipose tissue of 6-week-old (6 weeks) or 6-month-old (6 months) B6 and 129 mice. Cells in the stromavascular
fraction were labeled with fluorescence-conjugated antibodies against different myeloid cell and lymphoid markers and counted using flow
cytometry. Nonerythrocytes stromavascular cells, PI-/Ter-119-; leukocytes, PI-/Ter-119-/CD45; macrophages, PI-/Ter-119-/CD45/CD11b/F4/
80/CD11c-; CD11c
-Myeloid cells, PI-/Ter-119-/CD45/CD11b/F4/80/CD11c; T-cells, PI-/Ter-119-/CD45/CD3; T-helper cells, PI-/Ter-119-/
CD45/CD3/CD4.A: Represents number of cells per gram of fat tissue and (B) the total number of cells in the epididymal fat depot. All values
are mean SEM of 4 8 animals. *P<0.05; **P<0.01; ***P<0.001 vs. 129 mice. #P<0.05; ##P<0.01; ###P<0.001 vs. 6 weeks.
SYSTEMS BIOLOGY AND SUSCEPTIBILITY TO DIABETES
2966 DIABETES, VOL. 59, NOVEMBER 2010 diabetes.diabetesjournals.org
with higher expression in B6 compared with 129 mice, as
evidenced by the large, bright green nodes in Fig. 6B. The
majority of these genes had greater differences between
B6 and 129 mice under HFD than under LFD conditions, as
shown by the high number of upward triangles in Fig. 6.
In particular, T-cell marker CD3 and the subnetwork
around T-helper marker CD4 were among the most up-
regulated genes with HFD in B6 versus 129 mice (arrows
A
0
0.5
1
1.5
2
2.5
F4/80 M CP1 CD3 T h y 1 SDF1
CD68
mRNA ex
(Fold Change)
#
***
#
***
*
**
**
#
**
*
0
0.5
1
1.5
2
2.5 B6 LFD
129 LFD
B6 HFD
129 HFD
B6 LFD
129 LFD
B6 HFD
129 HFD
F4/80 M CP1 CD3 T h y 1CD68
mRNA expression
(Fold Change)
#
***
#
***
*
**
**
#
**
*
#
1
< 0.5
> 2
Fold Change
Fold Change HFD > LFD
Fold Change HFD < LFD
B
IKBKA
CD18
MCP1
SDF1
IKBKA
CD18
MCP1
SDF1
FIG. 6. Expression of inflammatory markers in adipose tissue of 6-month-old B6 and 129 mice fed with LFD or HFD. A: Expression of mRNA
was assessed by qPCR. All values are normalized by TBP and expressed as fold change of the average value of the LFD B6 mice. Results
represent mean SEM of 4 6 animals. *P<0.05; **P<0.01; ***P<0.001 vs. 129 mice. #P<0.05 vs. LFD. B: The network view of the
immune system process differences between B6 and 129 mouse strains on a LFD (left) and HFD (right). The gene network was generated
by mapping genes that were significantly overrepresented in B6 vs. 129 mice (Qvalue <0.25) among the GNEA results on HFD intersected
with those on LFD onto protein-protein interaction networks involving genes annotated with the immune system process gene set. Genes
in red are more than twofold higher in 129 mice compared with B6 mice; genes in green are more than twofold higher in B6 mice compared
with 129 mice. Genes are denoted by a downward triangle if the fold change of the HFD animals is less than those on the LFD, and by an
upward triangle for the converse; the size of the triangle denotes the magnitude of the difference. Arrows represent genes with specific
interest commented on in the main text.
M.A. MORI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 59, NOVEMBER 2010 2967
in Fig. 6B). Conversely, anti-inflammatory cytokines, such
as interleukin-4 (IL-4) and interleukin-10 (IL-10) were
reduced after HFD in the B6 mice, but not in 129 mice
(arrows in Fig. 6B).
DISCUSSION
Although a relationship between obesity and inflammation
in fat has been previously observed, in most cases these
inflammatory changes have been viewed as being second-
ary to obesity. Our data, comparing diabetes-prone B6
mouse and diabetes-resistant 129 mouse at 6 weeks of age,
show alterations in the inflammatory process in adipose
tissue even before differences in metabolic parameters can
be detected. Thus, B6 mice exhibit increased expression of
the T-cell chemokines SDF1and CCL5/RANTES and an
increased number of T-cells in the fat tissue. These differ-
ences are associated with higher IFNand CD80 levels in
the B6 mice— both molecules are known to participate in
T-cell function and activation (25) (summarized in the
model shown in Fig. 7).
Recent work has shown that T-cells infiltrate into the
visceral adipose tissue of obese animals and humans with
type 2 diabetes, and this is followed by recruitment of
macrophages and development of insulin resistance (26
29). Based on these findings, a model for the role of T-cells
in the pathogenesis of obesity and insulin resistance has
been proposed in which increases in SDF1and CCL5/
RANTES levels in adipose tissue occur in response to an
obesogenic environment and promote infiltration with T
lymphocytes (29). IFNderived from these T-cells then
promotes MCP1 secretion by preadipocytes (and possibly
other cell types), resulting in recruitment of macrophages
that further contribute to insulin resistance by production
of proinflammatory cytokines (29).
Our study provides evidence to support a major impact
of the genetic background of different mouse strains in the
migration of T-cells to the adipose tissue, both in the basal
state and in response to weight gain. In B6 mice, the
number of T-cells in the adipose tissue correlates posi-
tively with the increase in adipose mass as a consequence
of aging or HFD. By contrast, this response is practically
absent in 129 mice despite a significant increase in adipos-
ity in response to age or HFD. Thus, similar to mice with
ablation of T-cells (27,28), 129 mice develop only mild
insulin resistance in response to obesity. To what extent
aging and the composition of the diet impact the infiltra-
tion of T-cells into adipose tissue, in addition to the effects
of weight gain, remain to be determined. It is clear though
that insulin resistance in response to increased adiposity
differ substantially between mouse strains, and this phe-
nomenon is correlated with the migration of T-cell to the
adipose tissue. Thus, inflammation in the adipose tissue
does not always correlate with weight gain and is strongly
dependent on the genetic background of the host. Similar
differences dependent on genetic background are likely to
occur in humans and contribute to differences in obesity-
induced diabetes risk in different ethnic groups or even
different individuals, allowing for some of the “fat-fit”
phenotype.
Among other implications, these observations clearly
impact the choice of models for metabolic studies, in
particular when knockout mice are used. Knockout mice
are often derived from the embryonic stem cells of 129
mice, and therefore, in many cases, studies are performed
on mice on a pure 129 background or mixed 129/B6
background. In these cases, backcrossing to B6 mice for
several generations usually potentiates the inflammatory
and metabolic response of the model to environmental
factors. However, it is not clear which of these back-
grounds (B6, 129, or mixed) most closely mimic the human
condition.
Other recent reports support our findings that metabolic
disease traits can be associated with alterations of inflam-
matory gene expression networks in the adipose tissue
and liver (30,31). However, to our knowledge, no study has
previously demonstrated a measurable difference in in-
flammation preceding any measurable phenotypic differ-
ence associated with metabolic diseases. The novelty of
our study is that, by comparing young B6 and 129 animals
using a sensitive computational approach, we can focus on
factors that can potentially predispose to disease in a
prospective manner and avoid findings that are mainly
secondary to obesity or metabolic differences. Our inves-
tigation also examined differences between B6 and 129
mice in multiple organs, allowing us to conclude that
inflammation in adipose tissue, and to a lesser extent in
liver, but not in skeletal muscle or spleen, is associated
with the predisposition to insulin resistance. In addition,
our computational analysis was able to set forth hypothe-
ses that led to subsequent biologic validation experiments
which provided further insight into the components of the
immune system that may contribute to metabolic diseases
(i.e., T-cell recruitment).
In addition to inflammation, other phenomena have
been shown to participate in the multifactorial pathophys-
iology of insulin resistance. Alterations in insulin receptor
levels and insulin signaling through IRS-proteins (32),
induction of the unfolded protein response (33) and oxi-
dative stress pathways (34), and changes in lipid (35) and
amino acid metabolism (36) can all promote insulin resis-
tance and contribute to the final phenotype. Many of these
pathways act by producing post-translational modifica-
tions of signaling proteins, such as phosphorylation or
alterations in compartmentalization, which would not be
detected as changes at the gene expression level. In this
regard it is worth noting that in addition to differences in
inflammation, GNEA analysis was able to identify other
gene sets differentially expressed between the mouse
strains in adipose tissue at 6 weeks of age (supplementary
Table S2), including networks related to signal transduc-
tion, protein secretion pathways, and glucose catabolism.
Many of these pathways can interact with inflammatory
pathways, and this crosstalk could represent an entry
point to the manifestation of metabolic diseases. Thus,
although changes in the immune response are definitely
one of the factors that precedes and predicts the tendency
of B6 mice to have greater insulin resistance than 129, it is
unlikely that inflammation is the only predisposing risk
factor associated with diabetes between these two mouse
strains.
In summary, it has been proposed that type 2 diabetes
and obesity are diseases associated with an immune
system that cannot cope appropriately with environmental
threats (37–39). Based on this hypothesis, anti-inflamma-
tory drugs, such as salicylates (40,41) and interleukin-1
blockers (42), have been used to improve glycemia in
individuals with type 2 diabetes. In this study, we demon-
strate that pre-existing differences in the inflammatory
milieu in metabolically active tissues may represent an
important component of the genetic background as a risk
factor to metabolic diseases. Thus, inflammation cannot
SYSTEMS BIOLOGY AND SUSCEPTIBILITY TO DIABETES
2968 DIABETES, VOL. 59, NOVEMBER 2010 diabetes.diabetesjournals.org
be viewed as only a mechanism that links susceptibility
factors such as overfeeding, underactivity, aging, and
stress to metabolic diseases—it may also link these pa-
thologies to heritability. Our study indicates that inflam-
mation is an important early variable in the metabolic
response to environmental challenges and suggests sev-
eral potential targets for intervention, including LBP, Ly86,
SDF1, CCL5/RANTES, and MCP1. This provides new
Age/HFD
129
mouse
129
mouse
B6
mouse
B6
mouse
SDF1α
SDF1α
CCL5
CCL5
IFN-γ
IFN-γ
MCP1
MCP1
Monocyte
T-cell
Low
Low
Adipocyte
Macrophage
Low
Blood vessel
SVF cell
SDF1α
SDF1α
CCL5
CCL5
IFN-γ
IFN-γ
MCP1
MCP1
Monocyte
T-cell
Low
Low
Adipocyte
Macrophage
Low
Blood vessel
SVF cell
SDF1α
SDF1α
CCL5
CCL5
IFN-γ
IFN-γ
MCP1
MCP1
Monocyte
T-cell
High
High
Adipocyte
Obesity
Obesity
Obesity
Cytokines
Chemokines
Macrophage
High
Blood vessel
Insulin
Resistance
Insulin
Insulin
Resistance
Resistance
SVF cell
SDF1α
SDF1α
CCL5
CCL5
IFN-γ
IFN-γ
MCP1
MCP1
Monocyte
T-cell
High
High
Adipocyte
Obesity
Obesity
Obesity
Cytokines
Chemokines
Macrophage
High
Blood vessel
Insulin
Resistance
Insulin
Insulin
Resistance
Resistance
SVF cell
129
mouse
129
mouse
B6
mouse
B6
mouse
SDF1α
SDF1α
CCL5
CCL5
IFN-γ
IFN-γ
MCP1
MCP1
Monocyte
T-cell
Low
Low
Adipocyte
Macrophage
Low
Blood vessel
SVF cell
SDF1α
SDF1α
CCL5
CCL5
IFN-γ
IFN-γ
MCP1
MCP1
Monocyte
T-cell
Low
Low
Adipocyte
Macrophage
Low
Blood vessel
SVF cell
SDF1α
SDF1α
CCL5
CCL5
IFN-γ
IFN-γ
MCP1
MCP1
Monocyte
T-cell
High
High
Adipocyte
Obesity
Obesity
Obesity
Cytokines
Chemokines
Macrophage
High
Blood vessel
Insulin
Resistance
Insulin
Insulin
Resistance
Resistance
SVF cell
SDF1α
SDF1α
CCL5
CCL5
IFN-γ
IFN-γ
MCP1
MCP1
Monocyte
T-cell
High
High
Adipocyte
Obesity
Obesity
Obesity
Cytokines
Chemokines
Macrophage
High
Blood vessel
Insulin
Resistance
Insulin
Insulin
Resistance
Resistance
SVF cell
FIG. 7. Schematic model illustrating the potential causes and consequences related to the different repertoire of immune cells in adipose tissue
of B6 and 129 mice. Solid arrows, secretion; dashed arrows, migration; dotted arrows, migration/differentiation.
M.A. MORI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 59, NOVEMBER 2010 2969
strategies for reducing the epidemic of type 2 diabetes and
metabolic diseases in spite of increasing obesity by attack-
ing the variable genetic risk.
ACKNOWLEDGMENTS
M.A.M., O.B., K.A., and C.R.K. were supported by grants
from the National Institutes of Health: DK-082659, DK-
033201, and DK-060837 (Diabetes Genome Anatomy
Project), as well as the Joslin Diabetes and Endocrinology
Research Center cores (DK-036836) and the Mary K.
Iacocca Professorship. The work of M.L. and S.K. was
supported by grants from the National Human Genome
Research Institute (R01 HG003367-01A1) and the National
Institutes of Health (U54 LM008748) National Center for
Biomedical Computing.
No potential conflicts of interest relevant to this article
were reported.
M.A.M. and M.L. researched data, contributed to discus-
sion, and wrote the manuscript. O.B. researched data and
contributed to discussion. K.A. planned the study and
researched data. H.S. researched data. S.K. researched
data, contributed to discussion, and reviewed/edited the
manuscript. C.R.K. planned the study, researched data,
contributed to discussion, and reviewed/edited the
manuscript.
The authors thank J. LaVecchio, G. Buruzula, and the
flow cytometry core at the Joslin Diabetes Center for help
with cytometry; J. Schroeder and the genomics core at the
Joslin Diabetes Center for help with microarray process-
ing; M. Rourk, Joslin Diabetes Center, for his expertise in
animal care; and S.E. Shoelson, MD, PhD, Joslin Diabetes
Center, for his helpful advice and discussion.
REFERENCES
1. Hossain P, Kawar B, El Nahas M. Obesity and diabetes in the developing
world—a growing challenge. N Engl J Med 2007;356:213–215
2. Lazar MA. How obesity causes diabetes: not a tall tale. Science 2005;307:
373–375
3. Doria A, Patti ME, Kahn CR. The emerging genetic architecture of type 2
diabetes. Cell Metab 2008;8:186 –200
4. Rankinen T, Zuberi A, Chagnon YC, Weisnagel SJ, Argyropoulos G, Walts
B, Perusse L, Bouchard C. The human obesity gene map: the 2005 update.
Obesity (Silver Spring) 2006;14:529 644
5. Walley AJ, Asher JE, Froguel P. The genetic contribution to non-syndromic
human obesity. Nat Rev Genet 2009;10:431– 442
6. Almind K, Kulkarni RN, Lannon SM, Kahn CR. Identification of interactive
loci linked to insulin and leptin in mice with genetic insulin resistance.
Diabetes 2003;52:1535–1543
7. Kulkarni RN, Almind K, Goren HJ, Winnay JN, Ueki K, Okada T, Kahn CR.
Impact of genetic background on development of hyperinsulinemia and
diabetes in insulin receptor/insulin receptor substrate-1 double heterozy-
gous mice. Diabetes 2003;52:1528 –1534
8. Almind K, Kahn CR. Genetic determinants of energy expenditure and
insulin resistance in diet-induced obesity in mice. Diabetes 2004;53:3274
3285
9. Haluzik M, Colombo C, Gavrilova O, Chua S, Wolf N, Chen M, Stannard B,
Dietz KR, Le RD, Reitman ML. Genetic background (C57BL/6J versus
FVB/N) strongly influences the severity of diabetes and insulin resistance
in ob/ob mice. Endocrinology 2004;145:3258 –3264
10. Bachmanov AA, Reed DR, Tordoff MG, Price RA, Beauchamp GK. Nutrient
preference and diet-induced adiposity in C57BL/6ByJ and 129P3/J mice.
Physiol Behav 2001;72:603– 613
11. Rossmeisl M, Rim JS, Koza RA, Kozak LP. Variation in type 2 diabetes-
related traits in mouse strains susceptible to diet-induced obesity. Diabe-
tes 2003;52:1958 –1966
12. West DB, York B. Dietary fat, genetic predisposition, and obesity: lessons
from animal models. Am J Clin Nutr 1998;67:505S–512S
13. Leiter EH, Lee CH. Mouse models and the genetics of diabetes: is there
evidence for genetic overlap between type 1 and type 2 diabetes? Diabetes
2005;54(Suppl. 2):S151–S158
14. Ferrara CT, Wang P, Neto EC, Stevens RD, Bain JR, Wenner BR, Ilkayeva
OR, Keller MP, Blasiole DA, Kendziorski C, Yandell BS, Newgard CB, Attie
AD. Genetic networks of liver metabolism revealed by integration of
metabolic and transcriptional profiling. PLoS Genet 2008;4:e1000034
15. Schmidt C, Gonzaludo NP, Strunk S, Dahm S, Schuchhardt J, Kleinjung F,
Wuschke S, Joost HG, Al-Hasani H. A meta-analysis of QTL for diabetes-
related traits in rodents. Physiol Genomics 2008;34:42–53
16. Liu M, Liberzon A, Kong SW, Lai WR, Park PJ, Kohane IS, Kasif S.
Network-based analysis of affected biological processes in type 2 diabetes
models. PLoS Genet 2007;3:e96
17. Keshava Prasad TS, Goel R, Kandasamy K, Keerthikumar S, Kumar S,
Mathivanan S, Telikicherla D, Raju R, Shafreen B, Venugopal A, Balakrish-
nan L, Marimuthu A, Banerjee S, Somanathan DS, Sebastian A, Rani S, Ray
S, Harrys Kishore CJ, Kanth S, Ahmed M, Kashyap MK, Mohmood R,
Ramachandra YL, Krishna V, Rahiman BA, Mohan S, Ranganathan P,
Ramabadran S, Chaerkady R, Pandey A. Human Protein Reference Data-
base–2009 update. Nucleic Acid Res 2009;37:D767–D772
18. Ideker T, Ozier O, Schwikowski B, Siegel AF. Discovering regulatory and
signalling circuits in molecular interaction networks. Bioinformatics 2002;
18(Suppl. 1):S233–S240
19. Gesta S, Bluher M, Yamamoto Y, Norris AW, Berndt J, Kralisch S, Boucher
J, Lewis C, Kahn CR. Evidence for a role of developmental genes in the
origin of obesity and body fat distribution. Proc Natl Acad SciUSA
2006;103:6676 – 6681
20. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette
MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP. Gene set
enrichment analysis: a knowledge-based approach for interpreting ge-
nome-wide expression profiles. Proc Natl Acad SciUSA2005;102:15545–
15550
21. Fisher RA. On the interpretation of -square from contingency tables, and
the calculation of P. J Royal Statistical Society 1922;85:87–94
22. Ge H, Liu Z, Church GM, Vidal M. Correlation between transcriptome and
interactome mapping data from Saccharomyces cerevisiae. Nat Genet
2001;29:482– 486
23. Nguyen MT, Favelyukis S, Nguyen AK, Reichart D, Scott PA, Jenn A,
Liu-Bryan R, Glass CK, Neels JG, Olefsky JM. A subpopulation of macro-
phages infiltrates hypertrophic adipose tissue and is activated by free fatty
acids via toll-like receptors 2 and 4 and JNK-dependent pathways. J Biol
Chem 2007;282:35279 –35292
24. Fontana L, Partridge L, Longo VD. Extending healthy life span–from yeast
to humans. Science 2010;328:321–326
25. Freedman AS, Freeman GJ, Rhynhart K, Nadler LM. Selective induction of
B7/BB-1 on interferon-stimulated monocytes: a potential mechanism for
amplification of T cell activation through the CD28 pathway. Cell Immunol
1991;137:429 – 437
26. Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, Lee J,
Goldfine AB, Benoist C, Shoelson S, Mathis D. Lean, but not obese, fat is
enriched for a unique population of regulatory T cells that affect metabolic
parameters. Nat Med 2009;15:930 –939
27. Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J, Dorfman R, Wang
Y, Zielenski J, Mastronardi F, Maezawa Y, Drucker DJ, Engleman E, Winer
D, Dosch HM. Normalization of obesity-associated insulin resistance
through immunotherapy. Nat Med 2009;15:921–929
28. Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, Otsu
M, Hara K, Ueki K, Sugiura S, Yoshimura K, Kadowaki T, Nagai R. CD8
effector T cells contribute to macrophage recruitment and adipose tissue
inflammation in obesity. Nat Med 2009;15:914 –920
29. Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz M, Wabitsch M,
Fischer-Posovszky P, Barth TF, Dragun D, Skurk T, Hauner H, Bluher M,
Unger T, Wolf AM, Knippschild U, Hombach V, Marx N. T-lymphocyte
infiltration in visceral adipose tissue: a primary event in adipose tissue
inflammation and the development of obesity-mediated insulin resistance.
Arterioscler Thromb Vasc Biol 2008;28:1304 –1310
30. Emilsson V, Thorleifsson G, Zhang B, Leonardson AS, Zink F, Zhu J,
Carlson S, Helgason A, Walters GB, Gunnarsdottir S, Mouy M, Steinthors-
dottir V, Eiriksdottir GH, Bjornsdottir G, Reynisdottir I, Gudbjartsson D,
Helgadottir A, Jonasdottir A, Jonasdottir A, Styrkarsdottir U, Gretarsdottir
S, Magnusson KP, Stefansson H, Fossdal R, Kristjansson K, Gislason HG,
Stefansson T, Leifsson BG, Thorsteinsdottir U, Lamb JR, Gulcher JR,
Reitman ML, Kong A, Schadt EE, Stefansson K. Genetics of gene expres-
sion and its effect on disease. Nature 2008;452:423–428
31. Chen Y, Zhu J, Lum PY, Yang X, Pinto S, Macneil DJ, Zhang C, Lamb J,
Edwards S, Sieberts SK, Leonardson A, Castellini LW, Wang S, Champy
MF, Zhang B, Emilsson V, Doss S, Ghazalpour A, Horvath S, Drake TA,
Lusis AJ, Schadt EE. Variations in DNA elucidate molecular networks that
cause disease. Nature 2008;452:429 435
32. Bruning JC, Winnay J, Bonner-Weir S, Taylor SI, Accili D, Kahn CR.
SYSTEMS BIOLOGY AND SUSCEPTIBILITY TO DIABETES
2970 DIABETES, VOL. 59, NOVEMBER 2010 diabetes.diabetesjournals.org
Development of a novel polygenic model of NIDDM in mice heterozygous
for IR and IRS-1 null alleles. Cell 1997;88:561–572
33. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, Tuncman
G, Gorgun C, Glimcher LH, Hotamisligil GS. Endoplasmic reticulum
stress links obesity, insulin action, and type 2 diabetes. Science
2004;306:457– 461
34. Houstis N, Rosen ED, Lander ES. Reactive oxygen species have a causal
role in multiple forms of insulin resistance. Nature 2006;440:944 –948
35. Boden G. Role of fatty acids in the pathogenesis of insulin resistance and
NIDDM. Diabetes 1997;46:3–10
36. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, Haqq
AM, Shah SH, Arlotto M, Slentz CA, Rochon J, Gallup D, Ilkayeva O,
Wenner BR, Yancy WS Jr, Eisenson H, Musante G, Surwit RS, Millington
DS, Butler MD, Svetkey LP. A branched-chain amino acid-related meta-
bolic signature that differentiates obese and lean humans and contributes
to insulin resistance. Cell Metab 2009;9:311–326
37. Fernandez-Real JM, Pickup JC. Innate immunity, insulin resistance and
type 2 diabetes. Trends Endocrinol Metab 2008;19:10 –16
38. Hotamisligil GS, Erbay E. Nutrient sensing and inflammation in metabolic
diseases. Nat Rev Immunol 2008;8:923–934
39. Schenk S, Saberi M, Olefsky JM. Insulin sensitivity: modulation by nutri-
ents and inflammation. J Clin Invest 2008;118:2992–3002
40. Goldfine AB, Silver R, Aldhahi W, Cai D, Tatro E, Lee J, Shoelson SE. Use
of salsalate to target inflammation in the treatment of insulin resistance
and type 2 diabetes. Clin Transl Sci 2008;1:36 43
41. Hundal RS, Petersen KF, Mayerson AB, Randhawa PS, Inzucchi S,
Shoelson SE, Shulman GI. Mechanism by which high-dose aspirin
improves glucose metabolism in type 2 diabetes. J Clin Invest 2002;109:
1321–1326
42. Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B,
Mandrup-Poulsen T, Donath MY. Interleukin-1-receptor antagonist in type
2 diabetes mellitus. N Engl J Med 2007;356:1517–1526
M.A. MORI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 59, NOVEMBER 2010 2971

Supplementary resource (1)

... Indeed, earlier spinal cord inflammation is concomitant with a higher defective protein catabolic response, mitochondrial dysfunction, and axonal function impairment in the CNS of fast than slow-progressing mice. In addition, 129Sv mice have a higher basal metabolic rate and lower circulating insulin levels than the C57Bl6 mice [63], which may account for increased hypermetabolism, one of the hallmarks associated with ALS pathogenesis [64]. Here, we pinpointed that genetic background also affects the extent of activation of presynaptic and postsynaptic compensatory mechanisms to impinge on muscle atrophy and contribute to the variability of the disease course. ...
Article
Full-text available
Following publication of the original article [1], the authors reported that Fig. 7 needed to be amended. The correct Fig. 7 has been provided in this Correction. The original article [1] has been corrected.
... Indeed, earlier spinal cord inflammation is concomitant with a higher defective protein catabolic response, mitochondrial dysfunction, and axonal function impairment in the CNS of fast than slow-progressing mice. In addition, 129Sv mice have a higher basal metabolic rate and lower circulating insulin levels than the C57Bl6 mice [63], which may account for increased hypermetabolism, one of the hallmarks associated with ALS pathogenesis [64]. Here, we pinpointed that genetic background also affects the extent of activation of presynaptic and postsynaptic compensatory mechanisms to impinge on muscle atrophy and contribute to the variability of the disease course. ...
Article
Full-text available
Background Amyotrophic lateral sclerosis (ALS) is a heterogeneous disease in terms of onset and progression rate. This may account for therapeutic clinical trial failure. Transgenic SOD1G93A mice on C57 or 129Sv background have a slow and fast disease progression rate, mimicking the variability observed in patients. Based on evidence inferring the active influence of skeletal muscle on ALS pathogenesis, we explored whether dysregulation in hindlimb skeletal muscle reflects the phenotypic difference between the two mouse models. Methods Ex vivo immunohistochemical, biochemical, and biomolecular methodologies, together with in vivo electrophysiology and in vitro approaches on primary cells, were used to afford a comparative and longitudinal analysis of gastrocnemius medialis between fast- and slow-progressing ALS mice. Results We reported that slow-progressing mice counteracted muscle denervation atrophy by increasing acetylcholine receptor clustering, enhancing evoked currents, and preserving compound muscle action potential. This matched with prompt and sustained myogenesis, likely triggered by an early inflammatory response switching the infiltrated macrophages towards a M2 pro-regenerative phenotype. Conversely, upon denervation, fast-progressing mice failed to promptly activate a compensatory muscle response, exhibiting a rapidly progressive deterioration of muscle force. Conclusions Our findings further pinpoint the pivotal role of skeletal muscle in ALS, providing new insights into underestimated disease mechanisms occurring at the periphery and providing useful (diagnostic, prognostic, and mechanistic) information to facilitate the translation of cost-effective therapeutic strategies from the laboratory to the clinic.
... All mice were maintained under a 12/12 h light/dark cycle, 60 ± 5% humidity, and 23 ± 5°C temperature. The C57BL/6 mice strain has been shown to be a suitable strain mimicking the human metabolism observed in obesity (Lang et al., 2019;Mori et al., 2010). ...
Article
Full-text available
Aerobic exercise is an effective intervention in preventing obesity and is also an important factor associated with thermogenesis. There is an increasing interest in the factors and mechanisms induced by aerobic exercise that can influence the metabolism and thermogenic activity in an individual. Recent studies suggest that exercise induced circulating factors (known as ‘exerkines’), which are able to modulate activation of brown adipose tissue (BAT) and browning of white adipose tissue. However, the underlying molecular mechanisms associated with the effect of exercise‐induced peripheral factors on BAT activation remain poorly understood. Furthermore, the role of exercise training in BAT activation is still debatable. Hence, the purpose of our study is to assess whether exercise training affects the expression of uncoupled protein 1 (UCP1) in brown adipocytes via release of different blood factors. Four weeks of exercise training significantly decreased the body weight gain and fat mass gain. Furthermore, trained mice exhibit higher levels of energy expenditure and UCP1 expression than untrained mice. Surprisingly, treatment with serum from exercise‐trained mice increased the expression of UCP1 in differentiated brown adipocytes. To gain a better understanding of these mechanisms, we analysed the conditioned media obtained after treating the C2C12 myotubes with an AMP‐activated protein kinase (AMPK) activator (AICAR; 5‐aminoimidazole‐4‐carboxamide ribonucleotide), which leads to an increased expression of UCP1 when added to brown adipocytes. Our observations suggest the possibility of aerobic exercise‐induced BAT activation via activation of AMPK in skeletal muscles. Key points Exercise promotes thermogenesis by activating uncoupling protein 1 (UCP1), which leads to a decrease in the body weight gain and body fat content. However, little is known about the role of exerkines in modulating UCP1 expression and subsequent brown adipose tissue (BAT) activation. Four weeks of voluntary wheel‐running exercise reduces body weight and fat content. Exercise induces the increase in AMP‐activated protein kinase (AMPK) and slow‐type muscle fibre marker genes in skeletal muscles and promotes UCP1 expression in white and brown adipose tissues. Incubation of brown adipocytes with serum isolated from exercise‐trained mice significantly increased their UCP1 gene and protein levels; moreover, conditioned media of AMPK‐activator‐treated C2C12 myotubes induces increased UCP1 expression in brown adipocytes. These results show that aerobic exercise‐induced skeletal muscle AMPK has a significant effect on UCP1 expression in BAT.
... Accordingly, KO mice generated using 129 derived ES cells are routinely backcrossed onto a B6 background. Importantly, the 129 and B6 strains vary widely in their biology and in the pathology of multiple systems and organs, including aging, 2,3 behavior, 4-6 bone, 7 brain, [8][9][10] cancer, 11 cardiovascular biology, 12-14 cellular death, 15 coagulation, 16 immunology, [17][18][19] infection, [20][21][22] inflammation, 23,24 kidney, [25][26][27] liver, 28,29 metabolism, 30,31 neurology, 32,33 nutrition, 2 placenta, 34 reproductive biology, 35 skin, 36,37 transfusion, 30 and vascular biology. 38 Thus, if residual 129 genetic elements remain after backcrossing onto a B6 background, and one compares a KO strain to WT B6, then one risks mistaking a phenotypic difference between 129 and B6 mice for a result caused by the KO. ...
Article
Background Genetically modified mice are used widely to explore mechanisms in most biomedical fields—including transfusion. Concluding that a gene modification is responsible for a phenotypic change assumes no other differences between the gene-modified and wild-type mice besides the targetted gene. Study Design and Methods To test the hypothesis that the N-terminus of Band3, which regulates metabolism, affects RBC storage biology, RBCs from mice with a modified N-terminus of Band3 were stored under simulated blood bank conditions. All strains of mice were generated with the same initial embryonic stem cells from 129 mice and each strain was backcrossed with C57BL/6 (B6) mice. Both 24-h recoveries post-transfusion and metabolomics were determined for stored RBCs. Genetic profiles of mice were assessed by a high-resolution SNP array. Results RBCs from mice with a mutated Band3 N-terminus had increased lipid oxidation and worse 24-h recoveries, “demonstrating” that Band3 regulates oxidative injury during RBC storage. However, SNP analysis demonstrated variable inheritance of 129 genetic elements between strains. Controlled interbreeding experiments demonstrated that the changes in lipid oxidation and some of the decreased 24-hr recovery were caused by inheritance of a region of chromosome 1 of 129 origin, and not due to the modification of Band 3. SNP genotyping of a panel of commonly used commercially available KO mice showed considerable 129 contamination, despite wild-type B6 mice being listed as the correct control. Discussion Thousands of articles published each year use gene-modified mice, yet genetic background issues are rarely considered. Assessment of such issues are not, but should become, routine norms of murine experimentation.
... In this study, we examined the effect of chronic administration of an SGLT2 inhibitor (ipragliflozin) on energy metabolism in HFD-fed 129S6/Sv mice. 129S6/Sv mice are resistant to the stimulus of HFD, showing the induction of beige fat with higher browning propensity and less susceptibility to diabetes compared with C57BL/6J mice [13,14]. We chose this model to investigate the effect of ipragliflozin, focusing on adipose tissue browning, irrespective of the glucose-lowering mechanism. ...
Article
Full-text available
Background: Sodium-glucose co-transporter 2 (SGLT2) inhibitors are a new class of antidiabetic drugs that exhibit multiple extraglycemic effects. However, there are conflicting results regarding the effects of SGLT2 inhibition on energy expenditure and thermogenesis. Therefore, we investigated the effect of ipragliflozin (a selective SGLT2 inhibitor) on energy metabolism. Methods: Six-week-old male 129S6/Sv mice with a high propensity for adipose tissue browning were randomly assigned to three groups: normal chow control, 60% high-fat diet (HFD)-fed control, and 60% HFD-fed ipragliflozin-treated groups. The administration of diet and medication was continued for 16 weeks. Results: The HFD-fed mice became obese and developed hepatic steatosis and adipose tissue hypertrophy, but their random glucose levels were within the normal ranges; these features are similar to the metabolic features of a prediabetic condition. Ipragliflozin treatment markedly attenuated HFD-induced hepatic steatosis and reduced the size of hypertrophied adipocytes to that of smaller adipocytes. In the ipragliflozin treatment group, uncoupling protein 1 (Ucp1) and other thermogenesis-related genes were significantly upregulated in the visceral and subcutaneous adipose tissue, and fatty acid oxidation was increased in the brown adipose tissue. These effects were associated with a significant reduction in the insulin-to-glucagon ratio and the activation of the AMP-activated protein kinase (AMPK)/sirtuin 1 (SIRT1) pathway in the liver and adipose tissue. Conclusion: SGLT2 inhibition by ipragliflozin showed beneficial metabolic effects in 129S6/Sv mice with HFD-induced obesity that mimics prediabetic conditions. Our data suggest that SGLT2 inhibitors, through their upregulation of energy expenditure, may have therapeutic potential in prediabetic obesity.
... In addition, a range of antecedent diseases have been associated with a delayed ALS onset age but a shorter disease duration (Hollinger et al., 2016). In this context, it is relevant that in wild type mice, the C57 genetic background renders the mice more prone to become obese, insulin resistant, and glucose intolerant and develop diabetes (Almind and Kahn, 2004;Mori et al., 2010). Interestingly, in SOD1 G93A mice, the C57 strain exhibits slower disease progression and lower weight loss compared to the 129S (Nardo et al., 2016). ...
Article
Full-text available
The rate of disease progression in amyotrophic lateral sclerosis (ALS) is highly variable, even between patients with the same genetic mutations. Metabolic alterations may affect disease course variability in ALS patients, but challenges in identifying the preclinical and early phases of the disease limit our understanding of molecular mechanisms underlying differences in the rate of disease progression. We examined effects of SOD1G93A on thoracic and lumbar spinal cord metabolites in two mouse ALS models with different rates of disease progression: the transgenic SOD1G93A-C57BL/6JOlaHsd (C57-G93A, slow progression) and transgenic SOD1G93A-129SvHsd (129S-G93A, fast progression) strains. Samples from three timepoints (presymptomatic, disease onset, and late stage disease) were analyzed using Gas Chromatography-Mass Spectrometry metabolomics. Tissue metabolome differences in the lumbar spinal cord were driven primarily by mouse genetic background, although larger responses were observed in metabolic trajectories after the onset of symptoms. The significantly affected lumbar spinal cord metabolites were involved in energy and lipid metabolism. In the thoracic spinal cord, metabolic differences related to genetic background, background-SOD1 genotype interactions, and longitudinal SOD1G93A effects. The largest responses in thoracic spinal cord metabolic trajectories related to SOD1G93A effects before onset of visible symptoms. More metabolites were significantly affected in the thoracic segment, which were involved in energy homeostasis, neurotransmitter synthesis and utilization, and the oxidative stress response. We find evidence that initial metabolic alterations in SOD1G93A mice confer disadvantages for maintaining neuronal viability under ALS-related stressors, with slow-progressing C57-G93A mice potentially having more favorable spinal cord bioenergetic profiles than 129S-G93A. These genetic background-associated metabolic differences together with the different early metabolic responses underscore the need to better characterize the impact of germline genetic variation on cellular responses to ALS gene mutations both before and after the onset of symptoms in order to understand their impact on disease development.
... Low-grade inflammation in adipose tissue and liver is a common feature of obesity and metabolic syndrome (Franceschi et al., 2018;Hotamisligil, 2017;Mori et al., 2010;Odegaard and Chawla, 2013). This is characterized by a polarization of adipose tissue macrophages toward the M1-like phenotype and increased pro-inflammatory cytokine production (Castoldi et al., 2015;Lackey and Olefsky, 2016;Shapouri-Moghaddam et al., 2018). ...
Article
miRNAs can be found in serum and other body fluids and serve as biomarkers for disease. More importantly, secreted miRNAs, especially those in extracellular vesicles (EVs) such as exosomes, may mediate paracrine and endocrine communication between different tissues and thus modulate gene expression and the function of distal cells. When impaired, these processes can lead to tissue dysfunction, aging, and disease. Adipose tissue is an especially important contributor to the pool of circulating exosomal miRNAs. As a result, alterations in adipose tissue mass or function, which occur in many metabolic conditions, can lead to changes in circulating miRNAs, which then function systemically. Here we review the findings that led to these conclusions and discuss how this sets the stage for new lines of investigation in which extracellular miRNAs are recognized as important mediators of intercellular communication and potential candidates for therapy of disease.
Article
Full-text available
We examined effects of exposing female and male mice for 33 weeks to 45% or 60% high fat diet (HFD). Males fed with either diet were more vulnerable than females, displaying higher and faster increase in body weight and more elevated cholesterol and liver enzymes levels. Higher glucose metabolism was revealed by PET in the olfactory bulbs of both sexes. However, males also displayed altered anterior cortex and cerebellum metabolism, accompanied by a more prominent brain inflammation relative to females. Although both sexes displayed reduced transcripts of neuronal and synaptic genes in anterior cortex, only males had decreased protein levels of AMPA and NMDA receptors. Oppositely, to anterior cortex, cerebellum of HFD-exposed mice displayed hypometabolism and transcriptional up-regulation of neuronal and synaptic genes. These results indicate that male brain is more susceptible to metabolic changes induced by HFD and that the anterior cortex versus cerebellum display inverse susceptibility to HFD. Analysis of male and female mice exposed to a high fat diet reveals that the male mouse brain is more susceptible to resultant metabolic changes.
Article
Full-text available
The mesenchymal stromal cells (MSCs) residing within the stromal component of visceral adipose tissue appear to be greatly affected by obesity, with impairment of their functions and presence of senescence. To gain further insight into these phenomena, we analyzed the changes in total proteome content and secretome of mouse MSCs after a high-fat diet (HFD) treatment compared to a normal diet (ND). In healthy conditions, MSCs are endowed with functions mainly devoted to vesicle trafficking. These cells have an immunoregulatory role, affecting leukocyte activation and migration, acute inflammation phase response, chemokine signaling, and platelet activities. They also present a robust response to stress. We identified four signaling pathways (TGF-β, VEGFR2, HMGB1, and Leptin) that appear to govern the cells’ functions. In the obese mice, MSCs showed a change in their functions. The immunoregulation shifted toward pro-inflammatory tasks with the activation of interleukin-1 pathway and of Granzyme A signaling. Moreover, the methionine degradation pathway and the processing of capped intronless pre-mRNAs may be related to the inflammation process. The signaling pathways we identified in ND MSCs were replaced by MET, WNT, and FGFR2 signal transduction, which may play a role in promoting inflammation, cancer, and aging.
Article
Obesity has emerged as a leading cause of death in the last few decades, mainly due to associated cardiovascular diseases. Obesity, inflammation, and insulin resistance are strongly interlinked. Lisofylline (LSF), an anti-inflammatory agent, demonstrated protection against type 1 diabetes, as well as reduced obesity-induced insulin resistance and adipose tissue inflammation. However, its role in mitigating cardiac inflammation associated with obesity is not well studied. Mice were divided into 4 groups; the first group was fed regular chow diet, the second was fed regular chow diet and treated with LSF, the third was fed high fat diet (HFD), and the fourth was fed HFD and treated with LSF. Cardiac inflammation was interrogated via expression levels of TNF α, interleukins 6 and 10, phosphorylated STAT4 and lipoxygenases 12 and 12/15. Apoptosis and expression of the survival gene, AMPK, were also evaluated. We observed that LSF alleviated obesity-induced cardiac injury indirectly by improving both pancreatic β-cell function and insulin sensitivity, as well as, directly via upregulation of cardiac AMPK expression and downregulation of cardiac inflammation and apoptosis. LSF may represent an effective therapy targeting obesity-induced metabolic and cardiovascular complications.
Article
Full-text available
Common human diseases result from the interplay of many genes and environmental factors. Therefore, a more integrative biology approach is needed to unravel the complexity and causes of such diseases. To elucidate the complexity of common human diseases such as obesity, we have analysed the expression of 23,720 transcripts in large population-based blood and adipose tissue cohorts comprehensively assessed for various phenotypes, including traits related to clinical obesity. In contrast to the blood expression profiles, we observed a marked correlation between gene expression in adipose tissue and obesity-related traits. Genome-wide linkage and association mapping revealed a highly significant genetic component to gene expression traits, including a strong genetic effect of proximal (cis) signals, with 50% of the cis signals overlapping between the two tissues profiled. Here we demonstrate an extensive transcriptional network constructed from the human adipose data that exhibits significant overlap with similar network modules constructed from mouse adipose data. A core network module in humans and mice was identified that is enriched for genes involved in the inflammatory and immune response and has been found to be causally associated to obesity-related traits.
Article
Full-text available
Inflammation is increasingly regarded as a key process underlying metabolic diseases in obese individuals. In particular, obese adipose tissue shows features characteristic of active local inflammation. At present, however, little is known about the sequence of events that comprises the inflammatory cascade or the mechanism by which inflammation develops. We found that large numbers of CD8(+) effector T cells infiltrated obese epididymal adipose tissue in mice fed a high-fat diet, whereas the numbers of CD4(+) helper and regulatory T cells were diminished. The infiltration by CD8(+) T cells preceded the accumulation of macrophages, and immunological and genetic depletion of CD8(+) T cells lowered macrophage infiltration and adipose tissue inflammation and ameliorated systemic insulin resistance. Conversely, adoptive transfer of CD8(+) T cells to CD8-deficient mice aggravated adipose inflammation. Coculture and other in vitro experiments revealed a vicious cycle of interactions between CD8(+) T cells, macrophages and adipose tissue. Our findings suggest that obese adipose tissue activates CD8(+) T cells, which, in turn, promote the recruitment and activation of macrophages in this tissue. These results support the notion that CD8(+) T cells have an essential role in the initiation and propagation of adipose inflammation.
Article
Full-text available
Obesity and its associated metabolic syndromes represent a growing global challenge, yet mechanistic understanding of this pathology and current therapeutics are unsatisfactory. We discovered that CD4(+) T lymphocytes, resident in visceral adipose tissue (VAT), control insulin resistance in mice with diet-induced obesity (DIO). Analyses of human tissue suggest that a similar process may also occur in humans. DIO VAT-associated T cells show severely biased T cell receptor V(alpha) repertoires, suggesting antigen-specific expansion. CD4(+) T lymphocyte control of glucose homeostasis is compromised in DIO progression, when VAT accumulates pathogenic interferon-gamma (IFN-gamma)-secreting T helper type 1 (T(H)1) cells, overwhelming static numbers of T(H)2 (CD4(+)GATA-binding protein-3 (GATA-3)(+)) and regulatory forkhead box P3 (Foxp3)(+) T cells. CD4(+) (but not CD8(+)) T cell transfer into lymphocyte-free Rag1-null DIO mice reversed weight gain and insulin resistance, predominantly through T(H)2 cells. In obese WT and ob/ob (leptin-deficient) mice, brief treatment with CD3-specific antibody or its F(ab')(2) fragment, reduces the predominance of T(H)1 cells over Foxp3(+) cells, reversing insulin resistance for months, despite continuation of a high-fat diet. Our data suggest that the progression of obesity-associated metabolic abnormalities is under the pathophysiological control of CD4(+) T cells. The eventual failure of this control, with expanding adiposity and pathogenic VAT T cells, can successfully be reversed by immunotherapy.
Article
Full-text available
Obesity is accompanied by chronic, low-grade inflammation of adipose tissue, which promotes insulin resistance and type-2 diabetes. These findings raise the question of how fat inflammation can escape the powerful armamentarium of cells and molecules normally responsible for guarding against a runaway immune response. CD4(+) Foxp3(+) T regulatory (T(reg)) cells with a unique phenotype were highly enriched in the abdominal fat of normal mice, but their numbers were strikingly and specifically reduced at this site in insulin-resistant models of obesity. Loss-of-function and gain-of-function experiments revealed that these T(reg) cells influenced the inflammatory state of adipose tissue and, thus, insulin resistance. Cytokines differentially synthesized by fat-resident regulatory and conventional T cells directly affected the synthesis of inflammatory mediators and glucose uptake by cultured adipocytes. These observations suggest that harnessing the anti-inflammatory properties of T(reg) cells to inhibit elements of the metabolic syndrome may have therapeutic potential.
Article
Results At 13 weeks, in the anakinra group, the glycated hemoglobin level was 0.46 per- centage point lower than in the placebo group (P=0.03); C-peptide secretion was enhanced (P=0.05), and there were reductions in the ratio of proinsulin to insulin (P = 0.005) and in levels of interleukin-6 (P<0.001) and C-reactive protein (P = 0.002). Insulin resistance, insulin-regulated gene expression in skeletal muscle, serum adipokine levels, and the body-mass index were similar in the two study groups. Symptomatic hypoglycemia was not observed, and there were no apparent drug- related serious adverse events.
Article
When the food intake of organisms such as yeast and rodents is reduced (dietary restriction), they live longer than organisms fed a normal diet. A similar effect is seen when the activity of nutrient-sensing pathways is reduced by mutations or chemical inhibitors. In rodents, both dietary restriction and decreased nutrient-sensing pathway activity can lower the incidence of age-related loss of function and disease, including tumors and neurodegeneration. Dietary restriction also increases life span and protects against diabetes, cancer, and cardiovascular disease in rhesus monkeys, and in humans it causes changes that protect against these age-related pathologies. Tumors and diabetes are also uncommon in humans with mutations in the growth hormone receptor, and natural genetic variants in nutrient-sensing pathways are associated with increased human life span. Dietary restriction and reduced activity of nutrient-sensing pathways may thus slow aging by similar mechanisms, which have been conserved during evolution. We discuss these findings and their potential application to prevention of age-related disease and promotion of healthy aging in humans, and the challenge of possible negative side effects.