Invest Clin 66(4): 390- 407, 2025 https://doi.org/10.54817/IC.v66n4a04
Corresponding author: Xinwei Su. Department of Obstetrics and Gynecology, The Fifth Affiliated Hospital of Sun
Yat-sen University, 52 Meihua East Road, Zhuhai, Guangdong 519000, China. Tel: +86 0756 2528453.
Email: XXinnwei_ss@outlook.com
Comprehensive analysis of autophagy-
and glycolysis-related differentially
expressed genes involved in chronic
inflammation in obese patients.
Simin Yang1, Rexidanmu Hudabai1 and Xinwei Su2
1Department of Anesthesiology, The Fifth Affiliated Hospital of Sun Yat-sen University,
Zhuhai, Guangdong, China.
2Department of Obstetrics and Gynecology, The Fifth Affiliated Hospital of Sun Yat-sen
University, Zhuhai, Guangdong, China.
Keywords: Obesity; Autophagy; Glycolysis; Inflammation.
Abstract. The interaction between glycolysis and autophagy contributes
to reprogramming chronic inflammation in obesity, but the knowledge about
this interaction remains limited. Publicly available data were used to analyze
autophagy- and glycolysis-related differentially expressed genes (A&GRDEGs)
in two datasets comparing patients with obesity and normal-weight patients.
A total of 5 A&GRDEGs were obtained through screening, namely recombi-
nant eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1),
transforming growth factor beta 1 (TGFB1), fatty acid synthase (FASN), alpha-
synuclein (SNCA), and C-X-C chemokine receptor 4 (CXCR4). Levels of autoph-
agy and glycolysis exhibit substantial predictive value for obesity development
and mechanistically contribute to disease pathogenesis through immunometa-
bolic dysregulation.
Multi-Omics map of obesity inflammation 391
Vol. 66(4): 390 - 407, 2025
Análisis exhaustivo de los genes expresados diferencialmente,
relacionados con la autofagia y la glucólisis, que intervienen
en la inflamación crónica en pacientes obesos.
Invest Clin 2025; 66 (4): 390 – 407
Palabras clave: Obesidad; Autofagia; Glucólisis; Inflamación.
Resumen. La interacción entre la glucólisis y la autofagia contribuye a la
reprogramación de la inflamación crónica en la obesidad, pero el conocimiento
sobre esta interacción es limitado. Se utilizaron datos públicamente disponi-
bles para analizar los genes expresados diferencialmente relacionados con la
autofagia y la glucólisis (A&GRDEG) entre pacientes con obesidad y con peso
normal en dos conjuntos de datos. Se obtuvo un total de 5 A&GRDEG mediante
cribado, a saber: la proteína recombinante de unión al factor de iniciación de la
traducción eucariótica 4E 1 (EIF4EBP1), el factor de crecimiento transforman-
te beta 1 (TGFB1), la sintasa de ácidos grasos (FASN), la alfa-sinucleína (SNCA)
y el receptor de quimiocina C-X-C 4 (CXCR4). Los niveles de autofagia y de glu-
cólisis mostraron un valor predictivo elevado para el desarrollo de la obesidad
y contribuyen mecánicamente a la patogénesis de la enfermedad mediante la
desregulación inmunometabólica.
Received: 03-08-2025 Accepted: 21-09-2025
INTRODUCTION
According to projections, the global
population of people living with obesity, a
metabolic syndrome, will reach one billion
by 2030, affecting one in five women and one
in seven men 1 Obesity is characterized by
excess weight (body mass index, BMI ≥30
kg/m2), accompanied by chronic low-grade
inflammation, oxidative stress, insulin resis-
tance, hypertension, dyslipidemia, and other
abnormalities. It could also increase the sus-
ceptibility of individuals to chronic diseases,
such as type 2 diabetes mellitus (T2DM),
non-alcoholic fatty liver disease (NAFLD),
and specific malignancies 2. Furthermore,
adipocytes serve as central metabolic and
inflammatory regulators through their en-
docrine capacity, secreting both pro- and an-
ti-inflammatory mediators, including leptin
(pro-inflammatory) and adiponectin (anti-
inflammatory), which systemically influence
energy homeostasis and immune responses1.
Leptin and adiponectin exert pro-inflam-
matory and anti-inflammatory functions,
respectively, and mutually regulate carcino-
genesis 2.
Adequate lipid storage prevents ectopic
lipid accumulation in non-specific organs
(e.g. muscle, liver, and heart) and toxic lipid
accumulation (e.g., lipotoxicity) and is also
associated with stable metabolic function 3.
Lipid droplet accumulation in the liver and
other organs has been linked with obesity-
associated autophagic dysfunction 4. The
seminal recognition of autophagy through
the 2016 Nobel Prize in Physiology or Medi-
cine underscores its pivotal role as a funda-
mental cellular clearance mechanism for
superfluous or deleterious constituents5.
Chronic inflammation in individuals with
obesity may suppress autophagy. Moreover,
392 Yang et al.
Investigación Clínica 66(4): 2025
obesity-associated autophagic dysfunction
may cause protein and organelle degrada-
tion, cellular dysfunction, and cell death 6.
Autophagy is presumed to be inactive during
obesity owing to the chronic upregulation of
mammalian target of rapamycin complex 1
(mTORC1) 7. Obesity could contribute to an
elevated risk of cancer through autophagic
impairment; thus, treatment methods in-
volving the enhancement of autophagy may
serve as a practical approach against obesi-
ty-associated cancers 8.
Metabolism is widely known as a core
process underpinning all biological phenom-
ena, supplying energy and building blocks
for macromolecules 9. Restricting glycolysis
has been shown to impede cytokine produc-
tion but not cellular proliferation, suggest-
ing that glycolysis plays a critical role in
regulating inflammation 10. However, infor-
mation on the exact mechanism by which
the interaction between glycolysis and au-
tophagy contributes to reprogramming the
progression of chronic inflammation in obe-
sity remains limited.
Individuals with obesity exhibit adipo-
cyte hypoxia, which could lead to the elevat-
ed expression of hypoxia-inducible factors,
activation of adipocytes, production and re-
lease of free fatty acids and pro-inflammato-
ry mediators, induction of circulating mono-
cyte recruitment, and aggregation of adipose
tissue macrophages 11. Studies have revealed
that individuals with obesity have a higher
macrophage count in white adipose tissues
than individuals with normal BMI, showing
increases ranging from 10% to >50% of the
total cell count 12. Metabolically activated
macrophages show a higher rate of glycoly-
sis and produce more lactic acid in the fatty
tissues of individuals with obesity than in the
tissues of normal-weight individuals 13. Alter-
ations in immune cell infiltration dynamics
and their secretion of pro-inflammatory cy-
tokines critically contribute to the sustained
low-grade inflammation observed in obesity.
Consequently, targeting immune cell re-
cruitment and activation has emerged as a
key therapeutic strategy for obesity-associat-
ed chronic inflammation 14.
In this study, screening was first per-
formed to identify genes frequently involved
in autophagy and glycolysis. Next, we lever-
aged publicly available datasets to compare
the expression profiles of autophagy- and gly-
colysis-related genes between obese and nor-
mal-weight individuals. Additionally, we as-
sessed immune cell infiltration patterns and
examined their correlations with the differen-
tially expressed genes. These findings could
help elucidate the key mechanisms underly-
ing the pathogenesis of chronic inflammation
in patients with obesity and provide potential
targets for prevention and treatment.
MATERIALS AND METHODS
Dataset download and processing
We obtained two obesity-related gene
expression datasets (GSE134913 15 and
GSE59034 16) from the Gene Expression
Omnibus (GEO) database (https://www.
ncbi.nlm.nih.gov/geo/). The GSE134913
dataset included samples. We included 14
pre-surgery samples from obese patients
(Obese group) who underwent metabolic
surgery and six controls (Control group),
forming a subset of 20 samples.
The GSE134913 dataset is a subset of
participants from the GSE135066 cohort.
It comes from the clinical study “Dynamic
Changes in Muscle Insulin Sensitivity after
Metabolic Surgery” by Gancheva et al. 15.
The original cohort included six control sub-
jects and 16 obese individuals who had met-
abolic surgery, with longitudinal gene ex-
pression profiling done at three timepoints:
before surgery, two weeks after surgery, and
52 weeks after surgery. For this analysis, we
included all six control subjects and 14 of
the 16 obese participants who had surgery,
based on data completeness criteria.
Although aggregate baseline character-
istics of the entire cohort are provided in the
original publication, detailed demographic
and clinical metadata for the specific genet-
Multi-Omics map of obesity inflammation 393
Vol. 66(4): 390 - 407, 2025
ic sequencing subset were unavailable. As a
result, participant-level clinical information
could not be included in our analyses.
The GSE59034 dataset contained 48
samples. We selected 16 pre-surgery obese
samples and 16 controls, yielding a total of
32 samples. This dataset served as the pri-
mary cohort for all subsequent analyses. De-
tailed information about the datasets is pro-
vided in Table S1.
Autophagy-related genes (ARGs) and
glycolysis-related genes (GRGs) were sys-
tematically identified through comprehen-
sive searches of the GeneCards database
(https://www.genecards.org), followed by
literature validation using PubMed (https://
pubmed.ncbi.nlm.nih.gov/) with “Autopha-
gy17-19 and “Glycolysis” 20-22 as the primary
search terms, respectively. The intersection
of ARGs and GRGs defined the autophagy-
and glycolysis-related genes (A&GRGs) used
in this study (Fig. 1).
Differentially expressed genes analysis
Based on the original study designs,
samples were stratified into control and
obese groups. Differentially expressed genes
(DEGs) were identified using the limma
package (v3.58.1) in R (v4.2.2), with signifi-
cance defined as |logFC| > 0.5 and p<0.05.
Results were visualized through volcano
plots (ggplot2, v3.4.4).
To obtain the autophagy- and glycol-
ysis-related differentially expressed genes
(A&GRDEGs), we first intersected all signifi-
cant DEGs with our curated list of A&GRGs,
and a Venn diagram was plotted to provide
the A&GRDEGs. Expression patterns of
these A&GRDEGs were subsequently dis-
played as clustered heatmaps (pheatmap
package, v1.0.12) in R, using z-score nor-
malized expression values.
Fig. 1. Technology roadmap.
ARGs, Autophagy Related Ge-
nes. GRGs, Glycolysis Related
Genes. A&GRGs, Autophagy
& Glycolysis Related Genes.
A&GRDEGs, Autophagy &
Glycolysis Related Differen-
tially Expressed Genes. DEGs,
Differentially expressed genes.
GSEA, Gene Set Enrichment
Analysis. GO, Gene Ontology.
KEGG, Kyoto Encyclopedia of
Genes and Genomes. ssGSEA,
single-sample gene-set enrich-
ment Analysis. ROC, receiver
operating characteristic cur-
ve. PPI, Protein-protein inte-
raction network. TF, Transcrip-
tion factors.
394 Yang et al.
Investigación Clínica 66(4): 2025
Gene ontology and Kyoto Encyclopedia of
Genes and Genomes pathway enrichment
analyses
Functional enrichment analysis was
performed using the clusterProfiler pack-
age (v4.10.0) in R. Gene Ontology (GO) and
Kyoto Encyclopedia of Genes and Genomes
(KEGG) pathway analyses were conducted
on the differentially expressed genes. Terms
with p<0.05 and false discovery rate (FDR;
q) <0.25 were considered statistically sig-
nificant.
Gene Set Enrichment Analysis
Gene Set Enrichment Analysis (GSEA)
was conducted on the GSE59034 dataset us-
ing the clusterProfiler package. Genes were
ranked by log fold-change and analyzed
against the ‘c2.all.v2022.1.Hs.symbols.gmt’
gene set (All Canonical Pathways, n=3,050)
from MSigDB, with Homo sapiens as the ref-
erence species. Significance thresholds were
set at p<0.05 and q <0.25.
Differential expression and receiver
operating characteristic analyses
of A&GRDEGs
The expression patterns of A&GRDEGs
were compared between the obese and con-
trol groups across both datasets. The diag-
nostic potential was evaluated using receiver
operating characteristic (ROC) analysis per-
formed with the pROC package (v1.18.5).
Area under the curve (AUC) values were de-
termined to measure the predictive ability.
Analysis of immune infiltration
Immune cell infiltration levels were
quantified using single-sample Gene Set En-
richment Analysis (ssGSEA) implemented in
the GSVA package (v1.50.0). Correlations
among differentially abundant immune cell
populations (obese vs. control), and between
immune cells and A&GRDEG expression,
were analyzed in the GSE59034 dataset. Re-
sults were visualized as correlation dot plots
(ggplot2).
Statistical analysis
All statistical analyses were conducted
using R software (R Foundation for Statis-
tical Computing, Vienna, Austria). Con-
tinuous variables were compared with Stu-
dent’s t-test (for normally distributed data)
or Wilcoxon rank-sum test (for non-normal
data). Multi-group comparisons employed
the Kruskal-Wallis test. Categorical variables
were analyzed with χ² tests or Fisher’s exact
tests, depending on the situation. Correla-
tions were assessed with Spearman’s rank
correlation. A two-tailed α level of 0.05 was
deemed statistically significant unless other-
wise noted.
RESULTS
Identification of A&GRDEGs
Differential expression analysis re-
vealed 628 significant DEGs (|logFC| >0.5
and p<0.05) in the GSE134913 dataset,
comprising 316 upregulated and 312 down-
regulated genes (Fig. 2A). The GSE59034
dataset showed more pronounced differen-
tial expression with 904 DEGs under the
same thresholds (653 upregulated, 251
downregulated; Fig. 2B). Volcano plots vi-
sually represent these expression patterns,
highlighting the asymmetric distribution
of upregulated versus downregulated genes
between datasets.
To identify the A&GRDEGs, we inter-
sected the significant DEGs (|logFC| > 0.5
and p<0.05) from both datasets with our
curated A&GRG list, revealing five overlap-
ping genes (Fig. 2C): eukaryotic transla-
tion initiation factor 4E binding protein 1
(EIF4EBP1), transforming growth factor
beta 1 (TGFB1), fatty acid synthase (FASN),
alpha-synuclein (SNCA), and C-X-C chemo-
kine receptor type 4 (CXCR4) (Table 1).
We subsequently analyzed and visualized
the differential expression patterns of these
A&GRDEGs between sample groups in the
GSE134913 dataset (Fig. 2D) and in the
GSE59034 dataset (Fig. 2E).
Multi-Omics map of obesity inflammation 395
Vol. 66(4): 390 - 407, 2025
Construction and analysis
of the prediction model
We systematically investigated the func-
tional associations of the five A&GRDEGs
with obesity through comprehensive GO and
KEGG enrichment analyses (Table 2). GO
analysis identified significant enrichment in
biological processes (BP) such as positive reg-
ulation of glial cell differentiation, receptor
metabolic process, and gliogenesis; cellular
Fig. 2. Differentially expressed genes (DEGs) analysis.
(A) Volcano plot of differential analysis results between the obese and control groups in the GSE134913
dataset. (B) Volcano plot of differential analysis results between the obese and control groups in the
GSE59034 dataset. (C) Venn diagram of DEGs in the GSE134913 and GSE59034 datasets and A&GRGs.
(D) Differential expression heatmap of DEGs in the GSE134913 dataset. (E) Differential expression
heatmap of DEGs in the GSE59034 dataset. Green indicates the control group, and orange indicates
the obese group. In the heatmaps, blue represents low expression, and red represents high expression.
Table 1. A&GRDEGs in GSE59034 and GSE134913.
Gene logFC AveExpr t p p adjust B group
GSE59034
EIF4EBP -0.75697 6.168652 -7.58047 8.52E-09 3.51E-06 10.16121 down
TGFB1 0.571166 6.797319 5.933876 1.06E-06 4.33E-05 5.525466 up
FASN -0.64127 10.46345 -4.02795 0.0003 0.002172 0.123576 down
SNCA 0.596172 7.741707 2.504538 0.017237 0.049432 -3.63114 up
CXCR4 0.512738 6.194027 2.197896 0.034888 0.085782 -4.25343 up
GSE134913
EIF4EBP -0.79216 9.560483 -4.21636 0.00039 0.901994 -3.5489 down
SNCA 1.325622 9.526431 2.616228 0.016162 0.999958 -4.10701 up
CXCR4 0.785232 7.648631 2.278605 0.033299 0.999958 -4.22823 up
TGFB1 1.137448 6.567356 2.192964 0.039755 0.999958 -4.25833 up
FASN -0.57511 7.727896 -2.11711 0.046404 0.999958 -4.28468 down
396 Yang et al.
Investigación Clínica 66(4): 2025
Table 2. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG)
enrichment analysis results.
Ontology ID Description Gene
Ratio
Bg
Ratio
p p adjust q geneID Count
BP GO:0032103 positive regulation
of response to
external stimulus
3/5 442/
18800
0.00012461 0.02430796 0.00669754 TGFB1/
SNCA/
CXCR4
3
BP GO:0045687 positive regulation
of glial cell
differentiation
2/5 42/
18800
4.8517E-05 0.02430796 0.00669754 TGFB1/
CXCR4
2
BP GO:0043112 receptor metabolic
process
2/5 61/
18800
0.00010291 0.02430796 0.00669754 TGFB1/
SNCA
2
BP GO:0014015 positive regulation
of gliogenesis
2/5 64/
18800
0.00011333 0.02430796 0.00669754 TGFB1/
CXCR4
2
BP GO:2000379 positive regulation
of reactive oxygen
species metabolic
process
2/5 71/
18800
0.0001396 0.02430796 0.00669754 TGFB1/
SNCA
2
CC GO:0031091 platelet alpha
granule
2/5 91/
19594
0.0002114 0.00718767 0.00400552 TGFB1/
SNCA
2
CC GO:0031092 platelet alpha
granule membrane
1/5 17/
19594
0.00433098 0.07362671 0.04103037 SNCA 1
CC GO:0031093 platelet alpha
granule lumen
1/5 67/
19594
0.01698227 0.08811426 0.04910392 TGFB1 1
CC GO:0016234 inclusion body 1/5 74/
19594
0.01874314 0.08811426 0.04910392 SNCA 1
CC GO:0005902 microvillus 1/5 90/
19594
0.0227585 0.08811426 0.04910392 TGFB1 1
MF GO:0003779 actin binding 2/5 439/
18410
0.00540893 0.04141684 0.01278836 SNCA/
CXCR4
2
MF GO:0008190 eukaryotic
initiation factor 4E
binding
1/5 10/
18410
0.00271326 0.04141684 0.01278836 EIF4EBP1 1
MF GO:0034713 Type I transforming
growth factor beta
receptor binding
1/5 10/
18410
0.00271326 0.04141684 0.01278836 TGFB1 1
MF GO:0004312 fatty acid synthase
activity
1/5 13/
18410
0.00352609 0.04141684 0.01278836 FASN 1
MF GO:0043027 cysteine-type
endopeptidase
inhibitor activity
involved in the
apoptotic process
1/5 22/
18410
0.0059614 0.04141684 0.01278836 SNCA 1
KEGG hsa04672 Intestinal immune
network for IgA
production
2/5 49/
8164
0.00034888 0.02163052 0.01652586 TGFB1/
CXCR4
2
KEGG hsa04152 AMPK signaling
pathway
2/5 121/
8164
0.00211594 0.05414617 0.04136804 FASN/
EIF4EBP1
2
Multi-Omics map of obesity inflammation 397
Vol. 66(4): 390 - 407, 2025
components (CC) such as platelet alpha gran-
ule and its subcompartments (membrane, lu-
men); molecular functions (MF) such as fatty
acid synthase activity, TGF-β receptor type I
binding, and eukaryotic initiation factor 4E
binding. KEGG pathway analysis revealed in-
volvement in the intestinal immune network
for IgA production, the insulin signaling path-
way, and the AMP-activated protein kinase
(AMPK) signaling pathway. Results were vi-
sualized as bar charts (Fig. 3A) and bubble
plots (Fig. 3B), and functional networks were
constructed for BP (Fig. 3C), MF (Fig. 3D),
CC (Fig. 3E), and KEGG pathways (Fig. 3F).
Results of Gene Set Enrichment Analysis
(GSEA)
GSEA of the GSE59034 dataset re-
vealed significant associations between
global gene expression patterns and key bio-
logical processes (Fig. 4A), including inter-
leukin-10 signaling (Fig. 4B), neutrophil de-
granulation (Fig. 4C), Leishmania infection
responses (Fig. 4D), and proinflammatory/
profibrotic mediator networks (Fig. 4E).
These findings, detailed in Table S2, demon-
strate broad enrichment in immune-meta-
bolic pathways, highlighting their potential
role in obesity-related pathophysiology.
Differential expression and ROC analyses
of A&GRDEGs
Wilcoxon rank-sum tests revealed sig-
nificant differential expression (p<0.05)
of three A&GRDEGs (EIF4EBP1, TGFB1,
SNCA) between obese and control groups
in the GSE134913 dataset (Fig. 5A). ROC
analysis demonstrated strong diagnostic
potential for EIF4EBP1 (AUC = 0.921, Fig.
5D) and moderate predictive accuracy for
FASN (AUC = 0.750), SNCA (AUC = 0.833),
TGFB1 (AUC = 0.798), and CXCR4 (AUC =
0.762) (Fig. 5C-E), with AUC values indicat-
ing their utility as potential biomarkers for
obesity classification.
Consistent analysis of the GSE59034
dataset revealed significant differential ex-
pression (p<0.05) for all five A&GRDEGs
between obese and control groups (Fig. 5B).
ROC analysis demonstrated excellent diag-
nostic performance for EIF4EBP1 (AUC =
0.980) and TGFB1 (AUC = 0.910), moderate
accuracy for FASN (AUC = 0.828), and lim-
ited predictive value for SNCA (AUC = 0.699)
and CXCR4 (AUC = 0.691) (Fig. 5F-H).
Analysis of immune cell infiltration
Comparative analysis of immune cell
infiltration using Wilcoxon rank-sum tests
revealed significant abundance differences
(p<0.05) for 26 of 28 immune cell types
between obese and control groups in the
GSE59034 dataset (Fig. 6A). Correlation
analysis of these differentially abundant im-
mune populations identified a particularly
strong positive association (r = 0.98) be-
tween activated dendritic cells and myeloid-
derived suppressor cells (MDSCs) (Fig. 6B).
Ontology ID Description Gene
Ratio
Bg
Ratio
p p adjust q geneID Count
KEGG hsa04910 Insulin signaling
pathway
2/5 137/
8164
0.00270445 0.05414617 0.04136804 FASN/
EIF4EBP1
2
KEGG hsa04218 Cellular senescence 2/5 156/
8164
0.0034933 0.05414617 0.04136804 TGFB1/
EIF4EBP1
2
KEGG hsa05163 Human
cytomegalovirus
infection
2/5 225/
8164
0.00715783 0.08875712 0.06781104 CXCR4/
EIF4EBP1
2
BP: biological process; CC: cellular component; MF: molecular function.
Table 2. CONTINUATION
398 Yang et al.
Investigación Clínica 66(4): 2025
Fig. 3. GO and KEGG analyses.
(A) Bar charts of GO and KEGG analysis results for A&GRDEGs. The vertical axis shows GO and KEGG
terms. (B) Bubble charts of GO and KEGG analysis results for A&GRDEGs. The vertical axis shows GO
and KEGG terms. (C–E) Network diagrams of GO enrichment analysis for A&GRDEGs (C: BP, D: CC,
E: MF). (F) Network diagrams of KEGG enrichment analysis for A&GRDEGs. In the network diagrams
(C–F), pink dots represent specific pathways and blue dots represent specific genes. In the bubble
charts, the bubble size represents the number of genes, and the bubble color represents the p-value,
with redder hues indicating smaller values and bluer hues indicating larger values. The screening cri-
teria for GO and KEGG analyses were p<0.05 and q <0.25.
Multi-Omics map of obesity inflammation 399
Vol. 66(4): 390 - 407, 2025
Correlation analysis between the 26 dif-
ferentially abundant immune cell types and
A&GRDEG expression levels in GSE59034
revealed a strong positive association be-
tween TGFB1 expression and MDSC infiltra-
tion (r = 0.94), and a significant negative
correlation between EIF4EBP1 levels and
Th1 cell abundance (r = -0.87) (Fig. 6C).
DISCUSSION
The continuous rise of the obesity rate
requires multifaceted and effective preven-
tion and treatment. Inflammatory programs
are activated during the early stages of adi-
pose tissue expansion and during chronic
obesity, thus perpetuating a proinflamma-
tory phenotype in the immune system. This
metabolic dysregulation may elevate the risk
of developing severe comorbidities, includ-
ing insulin resistance, T2DM, cardiovascular
disease, NAFLD, certain malignancies, and
neurodegenerative disorders 23. Inhibition of
glycolysis has been shown to suppress mac-
roautophagy and chaperone-mediated au-
tophagy, leading to increased lipid accumu-
lation 24. The mechanisms of both autophagy
and glycolysis in obesity-associated diseases
are currently hot topics in research. Further
research on the regulatory mechanisms of
autophagy and the effects of glycolysis lev-
els on obesity and chronic inflammation will
help formulate more effective treatment
strategies for obesity control.
By phosphorylating the EIF4EBP1,
mTOR could dissociate from eIF4E and pro-
mote the initiation of translation 25. There-
fore, EIF4EBP1 is believed to be closely re-
lated to a wide range of diseases through its
regulation of autophagy and glycolysis26,27.
Fig. 4. GSEA of the GSE59034 dataset.
(A) Ridgeline plots of the four main biological functions in GSEA for the GSE59034 dataset. (B–E)
Genes in the GSE59034 dataset were significantly enriched in interleukin 10 signaling (B), neutrophil
degranulation (C), leishmania infection (D) and overview of proinflammatory and profibrotic mediators
(E). The screening criteria for GSEA were p<0.05 and q <0.25. NES, normalized enrichment score.
400 Yang et al.
Investigación Clínica 66(4): 2025
Experimental studies demonstrate that
mTOR target proteins EIF4EBP1 and EIF-
4EBP2 play critical roles in metabolic regula-
tion. Genetic ablation of both EIF4EBP1 and
EIF4EBP2 exacerbates diet-induced obesity
in murine models 28. Conversely, enhanced
EIF4EBP1 activity in skeletal muscle confers
metabolic protection, mitigating age- and
diet-induced insulin resistance while main-
taining energy expenditure. This protec-
tive effect is associated with reduced white
adipose tissue accumulation and preserved
brown adipose tissue mass 29. The above re-
sults are consistent with the findings of this
Fig. 5. Differential expression and ROC analyses of A&GRDEGs.
(A) Group comparison plot of A&GRDEGs between the obese and control groups in the GSE134913
dataset. (B) Group comparison plot of A&GRDEGs between the obese and control groups in the
GSE59034 dataset. (C–E) ROC curves of A&GRDEGs: FASN and SNCA (C), EIF4EBP1 and TGFB1
(D), and CXCR4 (E) between different groups (obese or control) in the GSE134913 dataset. (F-H)
ROC curves of A&GRDEGs: FASN and SNCA (F), EIF4EBP1 and TGFB1 (G), and CXCR4 (H) between
different groups (obese or control) in the GSE59034 dataset. *p<0.05; ***p<0.001.
Sensitivity (TPR)
Sensitivity (TPR)
Sensitivity (TPR)
Sensitivity (TPR)
Sensitivity (TPR)
Sensitivity (TPR)
1-Specicity (FPR)
1-Specicity (FPR)
1-Specicity (FPR)
1-Specicity (FPR)
1-Specicity (FPR)
1-Specicity (FPR)
Multi-Omics map of obesity inflammation 401
Vol. 66(4): 390 - 407, 2025
study, EIF4EBP1 is closely related to the reg-
ulatory mechanisms of obesity and chronic
inflammation. However, the mechanisms
by which EIF4EBP1 regulates obesity and
chronic inflammation through autophagy
and glycolysis require further study.
Patients with hyperlipidemia exhibit
elevated levels of TGF-β1, which could sup-
press the function of natural killer (NK)
cells, whereas restoring NK cell function
could improve the prognosis of patients with
metabolic syndrome 30. Studies have shown
that TGF-β1 could promote autophagy via
the Smad and non-Smad pathways, driving
hepatic fibrosis in NAFLD, and the progres-
sion of cardiac and renal fibrosis after ioniz-
ing radiation 31, 32. TGF-β1 induces metabolic
reprogramming in target cells, shifting ener-
gy production from mitochondrial oxidative
phosphorylation to glycolytic metabolism
– a phenomenon consistent with the War-
burg effect observed in many pathological
states33. Systemic blocking of TGF-β1 signal-
ing regulates glucose tolerance and energy
homeostasis, protecting mice from obesity,
diabetes, and fatty liver disease 34. TGF-β1
directly modulates PBX-regulating protein-
1expression in both adipose-derived stem
cells and mature adipocytes, thereby regu-
lating adipogenic differentiation and insulin
Fig. 6. ssGSEA of the GSE59034 dataset.
(A) Group comparison box plots of immune cells under the obese and control groups in the GSE59034
dataset. (B) Heatmap of correlations among the 26 immune cell types with significant differences
in the GSE59034 dataset. (C) Correlation dot plot between the expression levels of A&GRDEGs and
the abundance of 26 infiltrating immune cell types. Red indicates positive correlation and blue
indicates negative correlation, with the shade of color indicating the strength of correlation. ns
p≥0.05; * p<0.05; ** p<0.01; *** p<0.001.
402 Yang et al.
Investigación Clínica 66(4): 2025
sensitivity 35. Many studies have shown a re-
lationship between TGF-β1 and obesity and
chronic inflammation, consistent with our
study. However, there is no research on the
mechanism of TGF-β1 inhibiting obesity and
chronic inflammation by regulating autoph-
agy and glycolysis levels, which is worthy of
further exploration.
FASN is a key enzyme in hepatic de
novo lipogenesis, while its upregulation is
associated with insulin resistance 36. Com-
pared with normal-weight patients, FASN
expression is significantly downregulated in
patients with obesity 37. Studies have dem-
onstrated that the effect of hepatic FASN
deficiency on NAFLD and diabetes is depen-
dent on the etiology of obesity 36. In mice
with diet-induced obesity, adipose-specific
FASN knockout aggravated high-fat diet-
induced metabolic disturbances, exacerbat-
ing both hyperglycemia and hepatic dysfunc-
tion. These effects may be mediated through
impaired hepatic glucose uptake secondary
to glycogen accumulation and suppressed
glycolytic flux 36. The elevated FASN expres-
sion observed in obesity may confer a prolif-
erative advantage to malignant cells through
enhanced lipogenesis, potentially promoting
tumorigenesis in obese microenvironments
38. However, research has shown that renal
cancer patients with obesity or overweight
have a longer median overall survival owing
to low FASN expression 37. Therefore, consid-
ering the presence of obesity or overweight
is important in future interventional treat-
ments targeting the FASN pathway.
The high expression levels of the SNCA
gene are closely associated with earlier on-
set, more rapid progression, and more severe
manifestation of Parkinson’s disease 39. The
SNCA gene encodes α-synuclein. Mitochon-
drial accumulation of α-synuclein aggre-
gates triggers apoptotic pathways through
multiple mechanisms, including mitochon-
drial permeability transition pore open-
ing, calcium efflux, cytochrome C release,
and subsequent mitochondrial swelling 40.
Importantly, high-fat diet-induced obesity
was shown to accelerate the onset of mo-
tor deficits in human α-synuclein-expressing
transgenic mice, correlating with premature
α-synucleinopathy development and astrogli-
osis 41. These findings suggest that diet-in-
duced metabolic dysfunction may represent
a significant environmental risk factor for
α-synuclein pathology progression. Hence,
molecular and cellular mechanisms under-
lying the interactions between obesity and
SNCA in cognitive disorders await further
elucidation.
CXCR4, the exclusive receptor for che-
mokine (C-X-C motif) ligand 12, orchestrates
neutrophil metabolic reprogramming toward
glycolysis and lactate production, while driv-
ing neutrophil accumulation in both circu-
lation and psoriatic skin lesions 42. Beyond
its inflammatory functions, metabolic stress
induces fat mass and obesity-related protein
(FTO)-dependent N6-methyadenosine (m6A)
mRNA demethylation, which upregulates
CXCR4 expression via autophagic pathways.
This mechanism critically contributes to mel-
anoma pathogenesis and confers resistance
to PD-1 checkpoint inhibition 43. Moreover,
inhibition of CXCR4 led to the sensitization
of osteosarcoma to doxorubicin via the induc-
tion of autophagic cell death 44. In summary,
CXCR4 is closely related to inflammation, au-
tophagy, and glycolysis, and hence its regu-
latory role in patients with obesity warrants
further exploration.
Functional enrichment analysis revealed
that the five A&GRDEGs were significantly
associated with five key biological processes:
insulin signaling, AMPK-mediated metabolic
regulation, intestinal immune network for
IgA production, cellular senescence path-
ways, and human cytomegalovirus (CMV)
infection response. Among these, biological
processes such as insulin resistance, AMPK
signaling pathway, intestinal IgA immunity,
and cellular senescence are thought to be
closely related to chronic low-grade inflam-
mation in patients with obesity 45-48. GSEA
of the GSE59034 dataset demonstrated sig-
nificant enrichment of pro-inflammatory
Multi-Omics map of obesity inflammation 403
Vol. 66(4): 390 - 407, 2025
and pro-fibrotic pathways in obesity. Nota-
bly, three metabolic regulators (EIF4EBP1,
FASN, and TGFB1) exhibited consistent
diagnostic accuracy across both datasets
(Fig. 5). These findings implicate autopha-
gy-glycolysis crosstalk in obesity pathogen-
esis, potentially through the amplification
of inflammatory and fibrotic cascades. How-
ever, whether these five A&GRDEGs could
be applied clinically for the prevention and
treatment of obesity needs to be confirmed
through further investigations.
Among the five A&GRDEGs, TGFB1,
SNCA, and CXCR4 expression positively cor-
related with immune cell recruitment, while
FASN and EIF4EBP1 showed significant
inverse associations with immune infiltra-
tion levels. This dichotomous relationship
suggests differential roles in immunometa-
bolic regulation within obese adipose tissue.
TGFB1 showed the strongest positive corre-
lation with MDSCs, while EIF4EBP1 showed
the strongest negative correlation with Th1
cells. Th1 cells, playing a facilitatory role in
the pathogenesis of autoimmune diseases
and tissue inflammatory responses, is regu-
lated by cytokines IFN49. Furthermore, an
elevated Th1 cell count was linked with ele-
vated adipose tissue inflammation and insu-
lin resistance, with its cell count increasing
alongside increasing obesity 50. In vitro evi-
dence demonstrates that IFN-γ exacerbates
adipose tissue inflammation in obesity, while
IFN-γ knockout mice show improved meta-
bolic profiles, including enhanced insulin
sensitivity and reduced inflammatory mark-
ers in high-fat diet models 51. However, the
mechanistic relationship between EIF4EBP1
expression and Th1 cell infiltration in obese
adipose tissue remains unexplored. This
knowledge gap highlights the need for fo-
cused investigations into EIF4EBP1-mediat-
ed immunometabolic regulation in chronic
inflammation associated with obesity.
This study also has some limitations.
First, the results from the two datasets in-
cluded were not completely consistent, high-
lighting the need for experimental validation
and prospective clinical studies, which rep-
resent an important direction for our future
research. Second, although screening was
performed on obesity-related A&GRDEGs,
the specific mechanisms of action involved
were not clarified, which warrants further in-
vestigation.
In conclusion, levels of autophagy and
glycolysis exhibit substantial predictive value
for obesity development and mechanistically
contribute to disease pathogenesis through
immunometabolic dysregulation. These re-
sults provide a foundational framework for
understanding obesity-associated chronic in-
flammation, offering new molecular targets
for both basic research and potential thera-
peutic development.
Acknowledgements
The authors gratefully acknowledge the
data provided by patients and researchers
participating in GEO.
Funding
This research received no specific grant
from any funding agency in the public, com-
mercial, or not-for-profit sectors.
Conflict of interests
All authors declare that they have no
conflict of interest.
Ethics approval and consent
to participate
Not applicable, because GEO belongs
to public databases, the patients involved in
the database have obtained ethical approval,
users can download relevant data for free
for research and publish relevant articles,
and our study is based on open-source data,
and the The Fifth Affiliated Hospital of Sun
Yat-sen University does not require research
using publicly available data to be submit-
ted for review to their ethics committee, so
there are no ethical issues and other con-
flicts of interest.
404 Yang et al.
Investigación Clínica 66(4): 2025
Consent for publication
Not applicable.
Availability of data and materials
The original contributions presented in
the study are included in the article/Supple-
mentary material; further inquiries can be
directed to the corresponding author.
ORCID Numbers
Simin Yang (SY):
0009-0008-4837-9124
Rexidanmu Hudabai (RH):
0009-0005-8911-5916
Xinwei Su (XS):
0009-0001-4289-7018
Authors’ contributions
SY designed the research, collected the
data, performed the statistical analysis, and
drafted the manuscript. RH collected the
data and performed the statistical analysis.
XS supervised the statistical analysis, re-
viewed, and edited the manuscript. All au-
thors have approved the final manuscript.
REFERENCES
1. Ghaben AL, Scherer PE. Adipogenesis
and metabolic health. Nat Rev Mol Cell
Biol. 2019;20(4):242-258. https://doi.
org/10.1038/s41580-018-0093-z.
2. Kim DS, Scherer PE. Obesity, Diabetes,
and Increased Cancer Progression. Diabe-
tes Metab J. 2021;45(6):799-812. https://
doi.org/10.4093/dmj.2021.0077.
3. Calle EE, Kaaks R. Overweight, obesi-
ty and cancer: epidemiological eviden-
ce and proposed mechanisms. Nat Rev
Cancer. 2004;4(8):579-591. https://doi.
org/10.1038/nrc1408.
4. Namkoong S, Cho CS, Semple I, Lee
JH. Autophagy Dysregulation and
Obesity-Associated Pathologies. Mol
Cells. 2018;41(1):3-10. https://doi.
org/10.14348/molcells.2018.2213.
5. Tooze SA, Dikic I. Autophagy Captures
the Nobel Prize. Cell. 2016;167(6):1433-
1435. https://doi.org/10.1016/j.cell.2016.
11.023.
6. Zhang Y, Sowers JR, Ren J. Targeting
autophagy in obesity: from pathophysio-
logy to management. Nat Rev Endocri-
nol. 2018;14(6):356-376. https://doi.
org/10.1038/s41574-018-0009-1.
7. Dann SG, Selvaraj A, Thomas G. mTOR
Complex1-S6K1 signaling: at the crossro-
ads of obesity, diabetes and cancer. Trends
Mol Med. 2007;13(6):252-259. https://
doi.org/10.1016/j.molmed.2007.04.002.
8. Behrooz AB, Cordani M, Fiore A, Donade-
lli M, Gordon JW, Klionsky DJ, et al. The
obesity-autophagy-cancer axis: Mechanistic
insights and therapeutic perspectives. Se-
min Cancer Biol. 2024;99:24-44. https://
doi.org/10.1016/j.semcancer.2024.01.003.
9. Soto-Heredero G, Gomez de Las He-
ras MM, Gabande-Rodriguez E, Oller
J, Mittelbrunn M. Glycolysis - a key pla-
yer in the inflammatory response. FEBS
J. 2020;287(16):3350-3369. https://doi.
org/10.1111/febs.15327.
10. Chang CH, Curtis JD, Maggi LB, Jr.,
Faubert B, Villarino AV, O’Sullivan D, et
al. Posttranscriptional control of T cell
effector function by aerobic glycolysis.
Cell. 2013;153(6):1239-1251. https://doi.
org/10.1016/j.cell.2013.05.016.
11. Jiao P, Chen Q, Shah S, Du J, Tao B,
Tzameli I, et al. Obesity-related upregu-
lation of monocyte chemotactic factors in
adipocytes: involvement of nuclear factor-
kappaB and c-Jun NH2-terminal kinase
pathways. Diabetes. 2009;58(1):104-115.
https://doi.org/10.2337/db07-1344.
12. O’Rourke RW, White AE, Metcalf MD, Oli-
vas AS, Mitra P, Larison WG, et al. Hypoxia-
induced inflammatory cytokine secretion
in human adipose tissue stromovascular
cells. Diabetologia. 2011;54(6):1480-1490.
https://doi.org/10.1007/s00125-011-2103-y.
13. Boutens L, Hooiveld GJ, Dhingra S,
Cramer RA, Netea MG, Stienstra R.
Unique metabolic activation of adipose
Multi-Omics map of obesity inflammation 405
Vol. 66(4): 390 - 407, 2025
tissue macrophages in obesity promo-
tes inflammatory responses. Diabetolo-
gia. 2018;61(4):942-953. https://doi.
org/10.1007/s00125-017-4526-6.
14. Jacks RD, Lumeng CN. Macrophage and
T cell networks in adipose tissue. Nat Rev
Endocrinol. 2024;20(1):50-61. https://
doi.org/10.1038/s41574-023-00908-2.
15. Gancheva S, Ouni M, Jelenik T, Ko-
liaki C, Szendroedi J, Toledo FGS, et
al. Dynamic changes of muscle insulin
sensitivity after metabolic surgery. Nat
Commun. 2019;10(1):4179. https://doi.
org/10.1038/s41467-019-12081-0.
16. Petrus P, Mejhert N, Corrales P, Lecoutre
S, Li Q, Maldonado E, et al. Transforming
Growth Factor-β3 Regulates Adipocyte Num-
ber in Subcutaneous White Adipose Tissue.
Cell Rep. 2018;25(3):551-560.e5. https://
doi.org/10.1016/j.celrep.2018.09.069.
17. Sun S, Shen Y, Wang J, Li J, Cao J, Zhang
J. Identification and Validation of Autopha-
gy-Related Genes in Chronic Obstructive
Pulmonary Disease. Int J Chron Obstruct
Pulmon Dis. 2021;16:67-78. https://doi.
org/10.2147/copd.S288428.
18. Fu D, Zhang B, Wu S, Zhang Y, Xie J, Ning
W, et al. Prognosis and Characterization
of Immune Microenvironment in Acute
Myeloid Leukemia Through Identification
of an Autophagy-Related Signature. Front
Immunol. 2021;12:695865. https://doi.
org/10.3389/fimmu.2021.695865.
19. Wang N, Wei L, Liu D, Zhang Q, Xia X,
Ding L, et al. Identification and Vali-
dation of Autophagy-Related Genes in
Diabetic Retinopathy. Front Endocrinol
(Lausanne). 2022;13:867600. https://doi.
org/10.3389/fendo.2022.867600.
20. Zhang D, Zheng Y, Yang S, Li Y, Wang
M, Yao J, et al. Identification of a Novel
Glycolysis-Related Gene Signature for
Predicting Breast Cancer Survival. Front
Oncol. 2020;10:596087. https://doi.
org/10.3389/fonc.2020.596087.
21. Zheng J, Guo J, Zhu L, Zhou Y, Tong J.
Comprehensive analyses of glycolysis-rela-
ted lncRNAs for ovarian cancer patients. J
Ovarian Res. 2021;14(1):124. https://doi.
org/10.1186/s13048-021-00881-2.
22. Bi J, Bi F, Pan X, Yang Q. Establishment of
a novel glycolysis-related prognostic gene
signature for ovarian cancer and its rela-
tionships with immune infiltration of the
tumor microenvironment. J Transl Med.
2021;19(1):382. https://doi.org/10.1186/
s12967-021-03057-0.
23. Saltiel AR, Olefsky JM. Inflammatory me-
chanisms linking obesity and metabolic
disease. J Clin Invest. 2017;127(1):1-4.
https://doi.org/10.1172/JCI92035.
24. Zhang T, Linghu KG, Tan J, Wang M,
Chen D, Shen Y, et al. TIGAR exacer-
bates obesity by triggering LRRK2-me-
diated defects in macroautophagy and
chaperone-mediated autophagy in adi-
pocytes. Autophagy. 2024;20(8):1741-
1761. https://doi.org/10.1080/1554862
7.2024.2338576.
25. Bjornsti MA, Houghton PJ. Lost in
translation: dysregulation of cap-de-
pendent translation and cancer. Cancer
Cell. 2004;5(6):519-523. https://doi.
org/10.1016/j.ccr.2004.05.027.
26. Chen C, Chen W, Nong Z, Nie Y, Chen X,
Pan X, et al. Hyperbaric oxygen allevia-
ted cognitive impairments in mice indu-
ced by repeated cerebral ischemia-reper-
fusion injury via inhibition of autophagy.
Life Sci. 2020;241:117170. https://doi.
org/10.1016/j.lfs.2019.117170.
27. Selvarajah B, Azuelos I, Plate M, Gui-
llotin D, Forty EJ, Contento G, et al.
mTORC1 amplifies the ATF4-depen-
dent de novo serine-glycine pathway to
supply glycine during TGF-beta(1)-in-
duced collagen biosynthesis. Sci Signal.
2019;12(582):eaav3048. https://doi.
org/10.1126/scisignal.aav3048.
28. Le Bacquer O, Petroulakis E, Paglialun-
ga S, Poulin F, Richard D, Cianflone K,
et al. Elevated sensitivity to diet-induced
obesity and insulin resistance in mice
lacking 4E-BP1 and 4E-BP2. J Clin In-
vest. 2007;117(2):387-396. https://doi.
org/10.1172/JCI29528.
29. Tsai S, Sitzmann JM, Dastidar SG, Ro-
driguez AA, Vu SL, McDonald CE, et
al. Muscle-specific 4E-BP1 signaling ac-
tivation improves metabolic parameters
406 Yang et al.
Investigación Clínica 66(4): 2025
during aging and obesity. J Clin Invest.
2015;125(8):2952-2964. https://doi.org/
10.1172/JCI77361.
30. Hu X, Jia X, Xu C, Wei Y, Wang Z, Liu G,
et al. Downregulation of NK cell activities
in Apolipoprotein C-III-induced hyper-
lipidemia resulting from lipid-induced
metabolic reprogramming and crosstalk
with lipid-laden dendritic cells. Meta-
bolism. 2021;120:154800. https://doi.
org/10.1016/j.metabol.2021.154800.
31. Siapoush S, Rezaei R, Alavifard H, Ha-
tami B, Zali MR, Vosough M, et al.
Therapeutic implications of targeting
autophagy and TGF-beta crosstalk for
the treatment of liver fibrosis. Life Sci.
2023;329:121894. https://doi.org/10.101
6/j.lfs.2023.121894.
32. Ahamed J, Laurence J. Role of Platelet-
Derived Transforming Growth Factor-beta1
and Reactive Oxygen Species in Radiation-
Induced Organ Fibrosis. Antioxid Redox
Signal. 2017;27(13):977-988. https://doi.
org/10.1089/ars.2017.7064.
33. Hewitson TD, Smith ER. A Metabolic
Reprogramming of Glycolysis and Glu-
tamine Metabolism Is a Requisite for Re-
nal Fibrogenesis-Why and How? Front
Physiol. 2021;12:645857. https://doi.
org/10.3389/fphys.2021.645857.
34. Yadav H, Quijano C, Kamaraju AK, Ga-
vrilova O, Malek R, Chen W, et al. Pro-
tection from obesity and diabetes by bloc-
kade of TGF-beta/Smad3 signaling. Cell
Metab. 2011;14(1):67-79. https://doi.
org/10.1016/j.cmet.2011.04.013.
35. Bruno A, Di Sano C, Simon HU, Chanez P,
Patti AM, Di Vincenzo S, et al. Leptin and
TGF-beta1 Downregulate PREP1 Expression
in Human Adipose-Derived Mesenchymal
Stem Cells and Mature Adipocytes. Front
Cell Dev Biol. 2021;9:700481. https://doi.
org/10.3389/fcell.2021.700481.
36. Matsukawa T, Yagi T, Uchida T, Sakai M,
Mitsushima M, Naganuma T, et al. Hepa-
tic FASN deficiency differentially affects
nonalcoholic fatty liver disease and dia-
betes in mouse obesity models. JCI In-
sight. 2023;8(17):e161282. https://doi.
org/10.1172/jci.insight.161282.
37. Albiges L, Hakimi AA, Xie W, McKay RR,
Simantov R, Lin X, et al. Body Mass In-
dex and Metastatic Renal Cell Carcinoma:
Clinical and Biological Correlations. J Clin
Oncol. 2016;34(30):3655-3663. https://
doi.org/10.1200/JCO.2016.66.7311.
38. Khandekar MJ, Cohen P, Spiegelman
BM. Molecular mechanisms of cancer
development in obesity. Nat Rev Cancer.
2011;11(12):886-895. https://doi.org/10.
1038/nrc3174.
39. Ross OA, Braithwaite AT, Skipper LM,
Kachergus J, Hulihan MM, Middleton FA,
et al. Genomic investigation of alpha-sy-
nuclein multiplication and parkinsonism.
Ann Neurol. 2008;63(6):743-750. https://
doi.org/10.1002/ana.21380.
40. Malpartida AB, Williamson M, Na-
rendra DP, Wade-Martins R, Ryan
BJ. Mitochondrial Dysfunction and
Mitophagy in Parkinson’s Disease: From Me-
chanism to Therapy. Trends Biochem Sci.
2021;46(4):329-343. https://doi.org/10.10
16/j.tibs.2020.11.007.
41. Rotermund C, Truckenmuller FM, Schell
H, Kahle PJ. Diet-induced obesity accele-
rates the onset of terminal phenotypes in
alpha-synuclein transgenic mice. J Neuro-
chem. 2014;131(6):848-858. https://doi.
org/10.1111/jnc.12813.
42. Chen J, Bai Y, Xue K, Li Z, Zhu Z, Li
Q, et al. CREB1-driven CXCR4(hi) neu-
trophils promote skin inflammation in
mouse models and human patients. Nat
Commun. 2023;14(1):5894. https://doi.
org/10.1038/s41467-023-41484-3.
43. Yang S, Wei J, Cui YH, Park G, Shah P,
Deng Y, et al. m(6)A mRNA demethylase
FTO regulates melanoma tumorigenicity
and response to anti-PD-1 blockade. Nat
Commun. 2019;10(1):2782. https://doi.
org/10.1038/s41467-019-10669-0.
44. Liao YX, Lv JY, Zhou ZF, Xu TY, Yang D, Gao
QM, et al. CXCR4 blockade sensitizes osteo-
sarcoma to doxorubicin by inducing autopha-
gic cell death via PI3KAktmTOR pathway
inhibition. Int J Oncol. 2021;59(1):49.
https://doi.org/10.3892/ijo.2021.5229.
45. Esser N, Legrand-Poels S, Piette J,
Scheen AJ, Paquot N. Inflammation as a
Multi-Omics map of obesity inflammation 407
Vol. 66(4): 390 - 407, 2025
link between obesity, metabolic syndro-
me and type 2 diabetes. Diabetes Res Clin
Pract. 2014;105(2):141-150. https://doi.
org/10.1016/j.diabres.2014.04.006.
46. Foretz M, Guigas B, Viollet B. Understan-
ding the glucoregulatory mechanisms of met-
formin in type 2 diabetes mellitus. Nat Rev
Endocrinol. 2019;15(10):569-589. https://
doi.org/10.1038/s41574-019-0242-2.
47. Huus KE, Petersen C, Finlay BB. Diversity
and dynamism of IgA-microbiota interac-
tions. Nat Rev Immunol. 2021;21(8):514-
525. https://doi.org/10.1038/s41577-021-
00506-1.
48. Ferrucci L, Fabbri E. Inflammageing:
chronic inflammation in ageing, cardio-
vascular disease, and frailty. Nat Rev Car-
diol. 2018;15(9):505-522. https://doi.
org/10.1038/s41569-018-0064-2.
49. Huh JY, Park YJ, Ham M, Kim JB.
Crosstalk between adipocytes and immune
cells in adipose tissue inflammation and
metabolic dysregulation in obesity. Mol
Cells. 2014;37(5):365-371. https://doi.
org/10.14348/molcells.2014.0074.
50. Schipper HS, Prakken B, Kalkhoven E,
Boes M. Adipose tissue-resident immune
cells: key players in immunometabolism.
Trends Endocrinol Metab. 2012;23(8):407-
415. https://doi.org/10.1016/j.tem.2012.
05.011.
51. Rocha VZ, Folco EJ, Sukhova G, Shimizu
K, Gotsman I, Vernon AH, et al. Interfe-
ron-gamma, a Th1 cytokine, regulates fat
inflammation: a role for adaptive immuni-
ty in obesity. Circ Res. 2008;103(5):467-
476. https://doi.org/10.1161/CIRCRESA-
HA.108.177105.