ISSN 0006-2979, Biochemistry (Moscow), 2024, Vol. 89, No. 2, pp. 313-321 © The Author(s) 2024. This article is an open access publication.
313
AgeMeta: Quantitative Gene Expression Database
of Mammalian Aging
Stanislav Tikhonov
1,2
, Mikhail Batin
3
, Vadim N. Gladyshev
4
,
Sergey E. Dmitriev
1,2
, and Alexander Tyshkovskiy
1,4,a
*
1
Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
2
Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
3
Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA 91403, USA
4
Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School,
Boston, MA 02115, USA
a
e-mail: atyshkovskii@bwh.harvard.edu
Received October 12, 2023
Revised December 12, 2023
Accepted February 13, 2024
AbstractAgeMeta is a database that provides systemic and quantitative description of mammalian aging at
the level of gene expression. It encompasses transcriptomic changes with age across various tissues of humans,
mice, and rats, based on a comprehensive meta-analysis of 122 publicly available gene expression datasets from
26 studies. AgeMeta provides an intuitive visual interface for quantification of aging-associated transcriptomics
at the level of individual genes and functional groups of genes, allowing easy comparison among various species
and tissues. Additionally, all the data in the database can be downloaded and analyzed independently. Overall, this
work contributes to the understanding of the complex network of biological processes underlying mammalian
aging and supports future advancements in this field. AgeMeta is freely available at: https://age-meta.com/.
DOI: 10.1134/S000629792402010X
Keywords: aging, ageing, gene expression, differential expression, transcriptomics, mammalian, meta-analysis,
AgeMeta, age-meta, database, GSEA
* To whom correspondence should be addressed.
DEDICATION
We dedicate this work to the cherished memory of
Vladimir P. Skulachev – our esteemed teacher, mentor,
colleague, and extraordinary scientist. His profound
influence extended beyond the confines of academia,
as he played a pivotal role in orchestrating scientific
endeavors. Academician Skulachev is renowned for
his groundbreaking contributions to the fields of cell
bioenergetics and mitochondrial function. His unwav-
ering passion has, in recent years, also encompassed
the biology of aging, yielding many remarkable dis-
coveries. Academician Skulachev’s legacy is not only
defined by his scientific achievements but also by
the enduring impression he left through his bril-
liance, wisdom, kindness, and remarkable character.
In our hearts, he will forever occupy a special place,
his memory serving as an inspiration for current and
future generations.
INTRODUCTION
Aging is the major risk factor for multiple chronic
diseases and, therefore, poses a global medical prob-
lem [1]. Understanding the molecular basis and in-
ter-species differences of the aging process is thus a
crucial step towards developing effective geroprotec-
tors and overcoming the difficulties aging brings to
our society. The systems approach to characterize ag-
ing has gained popularity in the recent years with the
advent of high-throughput sequencing and due to its
comprehensiveness, resulting in a plethora of omics
data available and a great demand for its aggregation
into databases and meta-analysis.
TIKHONOV et al.314
BIOCHEMISTRY (Moscow) Vol. 89 No. 2 2024
To date, a number of databases containing ag-
ing genomics and transcriptomics data exist, includ-
ing Aging Atlas [2], HAGR [3], Digital Ageing Atlas [4],
AGEMAP [5], and Open Genes [6]. Most of these data-
bases accommodate plenty of information in addition
to aging transcriptomics, e.g., lifespan-extending inter-
ventions, aging phenotype, and epigenetics, but when
it comes to gene expression, the information is most-
ly a collection of age-related differentially expressed
genes identified from different studies. To the best of
our knowledge, there are currently no databases that
would present meta-analysis-derived quantitative
general trends of gene expression changes with age,
including species-specific, tissue-specific, and global
trends, allowing easy comparison and visualization
for each gene at the level of individual datasets and
individual samples within each dataset. To fill this gap,
we developed the AgeMeta database that provides an
interactive visual interface to investigate these bio-
markers in particular tissues and mammalian species
as well as cellular functions enriched in the transcrip-
tomic aging signatures, both at the level of general
trends and individual datasets. To facilitate the use of
our resource, we developed a comprehensive manual
for each section of the database and made the data-
base contents downloadable.
MATERIALS AND METHODS
The full list of datasets used to construct the tran-
scriptomic signatures of mammalian aging integrated
in AgeMeta included the following study IDs from GEO
[7], ArrayExpress [8], and SRA [9] databases: GSE9103,
GSE123981, GSE3150, GSE6591, GSE74463, GSE53960,
GSE66715, GSE11291, GSE34378, GSE27625, GSE12480,
GSE36192, GSE1572, GSE28422, GSE25941, GSE53890,
GSE38718, GSE674, GSE17612, GSE21935, GSE362,
GSE132040, E-MTAB-3374, PRJNA281127, PRJNA516151.
In addition to these studies, we also used data from
GTEx [10]. The datasets contained RNA-Seq and mi-
croarray samples obtained from different tissues of
Mus musculus, Rattus norvegicus, and Homo sapiens.
The tissue content varied across species; 23 tissues
used across all three species included adipose tissue
[in certain cases labeled specifically as brown adipose
tissue (BAT), mesenteric adipose tissue (MAT), gonadal
adipose tissue (GAT) and subcutaneous adipose tissue
(SCAT)], adrenal gland, blood vessel, bone and bone
marrow, brain (in certain cases olfactory bulbs were
labeled separately as OB and cerebellum and fron-
tal cortex were labeled separately), esophagus, heart,
kidney, liver, lung, muscle, nerve, pancreas, pituitary,
prostate, salivary gland, skin, small intestine, spleen,
testis, thyroid and whole blood (in some cases white
blood cells (WBC) were labeled separately). An anno-
tation of every dataset used in this study, that includes
information about the number of samples, age range,
strain, tissue, and sex, can be found in the supplemen-
tary materials (TableS1 in the Online Resource1).
The data preprocessing pipelines for RNA-Seq and
microarray datasets were described in detail in [11].
For both types of data, all platform IDs of genes were
standardized to Entrez IDs and mapped to mouse one-
to-one orthologs (in the case of rat and human samples).
These two preprocessing steps resulted in some genes
being discarded from the data. Genes that were not de-
tected in more than 40% of the datasets used to build a
particular signature were also filtered out prior to the
corresponding meta-analysis. Consequently, if a given
gene has no entry in AgeMeta, this could have been
caused by either of the above-stated data processing
steps.
The gene expression log fold changes (logFC) uti-
lized to construct the signatures of aging were calcu-
lated as the slope coefficient of a linear model when
the sample ages included more than two unique values
and as the difference in means otherwise, using the
limmaR package [12]. Mixed-effect model was utilized
to aggregate the expression changes from all datasets
into aging signatures with the metafor R package [13].
The logFCs and their respective standard errors were
used as an input, and study ID, tissue, and species were
introduced into the model as random terms. Gene set
enrichment analysis was performed using the fgseaR
package [14] with 10,000 permutations for the individ-
ual datasets and the signatures of aging. The AgeMeta
website was created using the shiny R package [15].
Overview of the 7 transcriptomic signatures of aging
Criterion Human Mouse Rat Brain Muscle Liver Global
Number of datasets 51 52 19 25 17 11 122
Total number of genes 15,959 13,459 10,382 16,125 15,339 12,805 15,170
Number of statistically significant
age-associated genes (p adjusted<0.05)
649 184 501 1526 379 1219 7
AgeMeta: GENE EXPRESSION IN AGING 315
BIOCHEMISTRY (Moscow) Vol. 89 No. 2 2024
DATABASE CONTENT
The contents of the database are based on the
7 transcriptomic signatures of aging identified in our
recent study [11]. The transcriptomic signatures rep-
resent general trends of gene expression changes with
age and were constructed by the meta-analysis of 122
datasets from 26 studies (table). Tissue-specific signa-
tures of brain, skeletal muscle, and liver were derived
from the corresponding human, mouse, and rat data-
sets, while species-specific signatures summarize the
data across multiple different tissues of a certain spe-
cies. The global signature encompasses all of the data
from the three species and 23 tissues (see “Materials
and Methods” section).
The database consists of 5 major modules (Fig. 1).
The first one is the comparative signature interface,
which provides aggregated quantitative estimates of
the age-related expression changes (normalized logFC)
in a particular tissue and species along with the cor-
responding metrics of statistical significance (p-value
before and after the Benjamini–Hochberg adjustment
[16]) for every gene. The user can choose the main sig-
nature to work with, and sort and filter genes accord-
ing to their significance in the chosen signature. One
can easily compare the differential expression values
of chosen genes and their significance between all
7signatures of aging using the right part of the inter-
active table, where the colored arrows next to logFC val-
ues illustrate the direction of gene expression changes
and the cell hues show statistical significance (Fig. 2).
The side menu allows the user to filter genes based on
their presence and significance in all 7 signatures and
on whether they are up- or downregulated with age
according to each of the signatures.
Should one wish to inspect the origin of each
logFC value in the interactive table, one can click the
table cell with the value, which would direct them to
the mixed-effects model plot corresponding to the gene
and signature represented by the clicked cell (Fig. 3a).
The mixed-effects model was used to aggregate dif-
ferential expression data from individual datasets
into signatures (see “Materials and Methods” section).
In the plot, each point stands for a dataset, namely a
data point with a unique combination of species, tis-
sue, sex, strain, and study. The dataset specifications
are displayed for each point on mouse hover. The red
line shows the aggregated average logFC value for a
given gene presented in the comparative signature ta-
ble (this value is the output of the mixed-effects mod-
el), while the y-coordinate of each point shows the
logFC from the corresponding dataset. The color and
shape of the points can be set to discriminate between
species, tissues, studies, sexes, platforms, and types of
models used for obtaining the differential expression
values, which allows the user to tailor the plot to their
specific needs. A click of a point in this plot directs the
user to the dataset expression plot, which shows how
the expression of the selected gene changes with age
in the clicked dataset (Fig.3b). The goal of the dataset
expression plot is to display the raw data, utilized to
calculate the logFC value and its standard error for a
particular dataset.
The last two modules of the database essentially
mirror the first two, but the genes are replaced with
the functional groups of genes. Functional enrichment
for the signatures and datasets was performed using
Gene Set Enrichment Analysis [17, 18], where quanti-
tative normalized enrichment scores (NES) and Ben-
jamini–Hochberg adjusted p-values were calculated.
In the functional signature interface module, one can
compare enrichment scores and corresponding statis-
tical significances across all 7 signatures. The function-
al terms can be filtered based on their presence, statis-
tical significance, up- or downregulation according to
the signatures, as well as on the ontology these terms
came from. The utilized ontologies included GO Biolog-
ical Process [19, 20], KEGG [21-23], Reactome [24], and
Biocarta [25]. Since many functional groups of genes
are nested within each other, we introduced a feature
that allows one to simultaneously examine all parent
and child gene sets for a selected functional term, i.e.,
groups that contain the selected gene set or are sub-
sets of this gene set, respectively. Clicking a function-
al term name will reduce the functional signatures
interface table, keeping only the selected term and its
parent and child terms. The “Back to full table” button
under the table will reset the table to the default state
with all functional groups from the database. Clicking
a table cell navigates the user to the functional dataset
plot, which displays enrichment scores and statistical
significances for all individual datasets corresponding
to the clicked functional term and signature (Fig. 4).
Although aggregated functional enrichment scores of
signatures were not obtained from the scores of the
individual datasets, this plot provides a general idea
about the consistency of the age-related up- or down-
regulation of genes associated with the selected path-
way across datasets and helps to justify the values in
the interactive table. Each bar in the plot represents
one dataset, with the dataset specifications shown on
mouse hover. Green and red bars represent statistical-
ly significant up- or downregulation with age, respec-
tively, while gray bars stand for enrichment scores
that failed to cross the statistical significance threshold
set in the side menu (by default, the adjusted p-value
threshold is equal to 0.1).
To make the use of the database simple and con-
venient, we developed the “Manual” section where
each database module is explained in detail with ex-
ample tables and plots. In addition, the whole database
is available in two languages: English and Russian.
TIKHONOV et al.316
BIOCHEMISTRY (Moscow) Vol. 89 No. 2 2024
Fig. 1. The structure of the AgeMeta database. AgeMeta provides a visual interface for the 7 transcriptomic signatures of aging,
consisting of two interactive tables which display quantitative age-related transcriptomic changes for individual genes (the com-
parative signature interface) and functional groups of genes (the functional signature interface) and allow comparison between
the signatures; as well as three types of plots depicting the differential expression of a gene in a signature at the dataset level
(the mixed-effects model plot), the change of expression with age for a gene within an individual dataset (the dataset expres-
sion plot), and enrichment scores of a functional group of genes for individual datasets associated with a certain signature (the
functional dataset plot). Clicking the table cells navigates the user to the corresponding plots, while clicking the points in the
mixed-effects model plot directs the user to the dataset expression plot related to the clicked dataset. Both tables can be down-
loaded in a separate section of the database.
Fig. 2. The comparative signature interface table. The arrow colors represent the direction of gene expression changes in a given
signature (the signatures are labeled in the column names), the cell hues represent the statistical significance of those chang-
es. Adjusted p-value less than the soft threshold results in a dull hue, adjusted p-value less than the strict threshold results in
a brighter hue. Both thresholds can be set in the menu on the left-hand side of the page. All cells in the right part of the table
with the signature logFCs are clickable. Clicking them transfers the user to the corresponding mixed-effects model plot.
AgeMeta: GENE EXPRESSION IN AGING 317
BIOCHEMISTRY (Moscow) Vol. 89 No. 2 2024
Fig. 3. Examples of the mixed-effects model plot and the differential gene expression plot. a)An example of the mixed-effects
model plot. Each point represents a single dataset. Shape and color represent species and tissue, respectively. The data are mean
normalized slope/logFC ±SE. The details of each dataset are specified in the text box that appears on mouse hover. b)An exam-
ple of the dataset gene expression plot corresponding to a point clicked on the mixed-effects model plot. The slope of the regres-
sion line corresponds to the logFC or slope value for the gene in the clicked dataset (the y-axis coordinate of the clicked point),
while the width of the shaded area around the line reflects the standard error for the slope.
To allow independent analysis of our data, we made
the differential gene expression data and the function-
al enrichment data fully downloadable. They can be
accessed through the “Downloads” section.
DISCUSSION
With the use of AgeMeta, the aging-associated dif-
ferential expression patterns and their inter-species or
inter-tissue differences can be easily visualized for any
of the thousands of genes or functional groups present
in the database. The interactive tables and plots allow
users to study genes of interest at the transcriptom-
ic signature level as well as at the level of individual
datasets. Additionally, the application of these and oth-
er related transcriptomic signatures for prediction and
characterization of novel lifespan-extending and re-
juvenative interventions [11, 26] suggests that the sig-
natures of aging integrated in AgeMeta might facilitate
TIKHONOV et al.318
BIOCHEMISTRY (Moscow) Vol. 89 No. 2 2024
Fig. 4. An example of the functional enrichment dataset plot. The plot corresponds to “GOBP: Antigen processing and presen-
tation” function in the mouse signature. The bars are colored based on statistical significance: up- and downregulated func-
tions are shown in green and red, respectively, while gray bars correspond to non-significant enrichments. Dataset specification
isdisplayed in a text box on mouse hover.
the discovery and development of new potential gero-
protectors.
Several databases of aging genomics and tran-
scriptomics exist to date. Therefore, it is important to
highlight some of the differences between the most
relevant existing databases and AgeMeta. GenAge,
a database that is a part of a collection of databases
known as Human Ageing Genomic Resources, or
HAGR [3], presents effects of genomic interventions
(e.g., gene knockouts) on lifespan for model organ-
isms such as Mus musculus. The association of these
effects with age-related transcriptomic changes is
not trivial, i.e., genomic interventions that result in a
longer lifespan do not always counteract the gene ex-
pression changes that happen with age. This could be
explained by some of the age-related gene expression
changes being detrimental, while some being adap-
tive and having a positive effect on the aging pheno-
type [11]. Anexample of this phenomenon is Pparg, a
gene which is significantly upregulated with age in the
mouse signature but whose knockout significantly de-
creases the mean lifespan of mice [27].
There are other databases with data on aging
transcriptomics, e.g., Aging Atlas [2] and AGEMAP [5].
Both of them contain differential expression informa-
tion from individual studies (there is only one study
in AGEMAP), but none of them offer meta-analysis-
derived differential expression values, aggregated
across multiple studies with multiple tissues and spe-
cies, which is the key distinguishing feature of AgeMeta.
Aging Atlas also contains multi-omics data, including
protein–protein interactions, ChIP-Seq data, and single
cell transcriptomics, which were outside of the scope
of our study. Another database, Open Genes [6], con-
tains descriptions of genes with many different types
of associations with aging, including knockouts affect-
ing lifespan and age-related differential expression,
but this database also does not contain results of a
quantitative meta-analysis.
A couple of limitations of our work should be not-
ed. Firstly, the signatures of aging used in AgeMeta
were based on linear differential expression models,
which could imply that some non-linear age-related
gene expression changes were not identified as statis-
tically significant in this study. Secondly, differential
co-expression was shown to occur with age in vari-
ous mammalian species [28-30], whereas in our work
only expression changes of individual genes were ex-
amined. Expansion of AgeMeta with a meta-analysis
of non-linear differential gene expression and gene
co-expression patterns may further improve the com-
prehensiveness and the application scope of our da-
tabase as well as help enhance the understanding of
molecular mechanisms of mammalian aging by the
scientific community.
Supplementary information. The online version
contains supplementary material available at https://
doi.org/10.1134/S000629792402010X.
Contributions. A.T., S.E.D., and M.B. conceived
the study; S.T. and A.T. designed the database; S.T. per-
formed data analysis and developed the database; S.T.,
M.B., V.G., S.E.D., and A.T. interpreted the data; A.T. su-
pervised the study; S.T. and A.T. wrote the manuscript
with contributions from all other authors. All authors
read and approved the final version.
Funding. The work was supported by the Russian
Science Foundation (grant no. 23-14-00218 to S.E.D.;
data processing and creating transcriptomic signa-
tures) and Open Longevity (developing the database).
AgeMeta: GENE EXPRESSION IN AGING 319
BIOCHEMISTRY (Moscow) Vol. 89 No. 2 2024
Ethics declarations. This work does not contain
any studies involving human and animal subjects.
The authors of this work declare that they have no
conflicts of interest.
Open access. This article is licensed under a Cre-
ative Commons Attribution 4.0 International License,
which permits use, sharing, adaptation, distribution,
and reproduction in any medium or format, as long
as you give appropriate credit to the original au-
thor(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made.
Theimages or other third-party material in this article
are included in the article’s Creative Commons license,
unless indicated otherwise in a credit line to the mate-
rial. If material is not included in the article’s Creative
Commons license and your intended use is not permit-
ted by statutory regulation or exceeds the permitted
use, you will need to obtain permission directly from
the copyright holder. To view a copy of this license,
visit http://creativecommons.org/licenses/by/4.0/.
REFERENCES
1. Harman,D. (1991) The aging process: Major risk factor
for disease and death, Proc. Natl. Acad. Sci. USA, 88,
5360-5363, doi:10.1073/pnas.88.12.5360.
2. Aging Atlas Consortium (2021) Aging Atlas: a multi-om-
ics database for aging biology, Nucleic Acids Res., 49,
825-830, doi:10.1093/nar/gkaa894.
3. Tacutu,R., Thornton,D., Johnson,E., Budovsky,A., Ba-
rardo,D., Craig,T., Diana, E., Lehmann, G., Toren,D.,
Wang,J., De Magalh,P., and Fraifeld,E. (2018) Human
Ageing Genomic Resources: new and updated data-
bases, Nucleic Acids Res., 46, 1083-1090, doi:10.1093/
nar/gkx1042.
4. Craig, T., Smelick, C., Tacutu, R., Wuttke, D., Wood,
S. H., Stanley, H., Janssens, G., Savitskaya, E., Moska-
lev,A., De Magalh,P., and Arking,R. (2015) The Digital
Ageing Atlas: integrating the diversity of age-related
changes into a unified resource, Nucleic Acids Res.,
43, 873-878, doi:10.1093/nar/gku843.
5. Zahn, J. M., Poosala, S., Owen, A. B., Ingram, D. K.,
Lustig, A., Carter, A., Weeraratna, A. T., Taub, D. D.,
Gorospe, M., Mazan-mamczarz, K., Lakatta, E. G.,
Boheler, K. R., Xu, X., Mattson, M. P., Falco, G., Ko,
M.S.H., Schlessinger,D., Firman,J., Kummerfeld, S.K.,
Iii, W. H. W., Zonderman, A.B., Kim, S.K., and Beck-
er, K.G. (2007) AGEMAP: a gene expression database
for aging in mice, PLoS Genet., 3, e201, doi: 10.1371/
journal.pgen.0030201.
6. Rafikova, E., Nemirovich-Danchenko, N., Ogmen, A.,
Parfenenkova, A., Velikanova, A., Tikhonov, S., Pe-
shkin, L., Rafikov, K., Spiridonova, O., Belova,Y., Gli-
nin,T., Egorova,A., and Batin,M. (2023) Open Genes
a new comprehensive database of human genes as-
sociated with aging and longevity, Nucleic Acids Res.,
52, D950-D962, doi:10.1093/nar/gkad712.
7. Edgar,R., Domrachev,M., and Lash, A.E. (2002) Gene
expression omnibus: NCBI gene expression and hy-
bridization array data repository, Nucleic Acids Res.,
30, 207-210, doi:10.1093/nar/30.1.207.
8. Parkinson, H., Kapushesky, M., Shojatalab, M., Abey-
gunawardena,N., Coulson,R., Farne,A., Holloway,E.,
Kolesnykov,N., Lilja,P., Lukk,M., Mani,R., Rayner,T.,
Sharma, A., William, E., Sarkans, U., and Brazma, A.
(2007) ArrayExpress – a public database of microar-
ray experiments and gene expression profiles, Nucleic
Acids Res., 35, 747-750, doi:10.1093/nar/gkl995.
9. Leinonen, R., Sugawara,H., and Shumway, M. (2011)
The sequence read archive, Nucleic Acids Res., 39,
2010-2012, doi:10.1093/nar/gkq1019.
10. Lonsdale, J., Thomas, J., Salvatore, M., Phillips, R.,
Lo, E., Shad, S., Hasz, R., Walters, G., Garcia, F.,
Young,N., Foster,B., Moser,M., Karasik,E., Gillard,B.,
Ramsey, K., Sullivan, S., Bridge, J., Magazine, H., Sy-
ron,J., Fleming,J., Siminoff,L., Traino,H., Mosavel,M.,
Barker,L., Jewell,S., Rohrer,D., Maxim,D., Filkins,D.,
Harbach, P., Cortadillo, E., Berghuis, B., Turner, L.,
Hudson,E., Feenstra,K., Sobin,L., Robb,J., Branton,P.,
Korzeniewski,G., Shive,C., Tabor,D., Qi,L., Groch,K.,
Nampally,S., Buia,S., Zimmerman,A., Smith,A., Burg-
es,R., Robinson,K., Valentino,K., Bradbury,D., Cosen-
tino,M., Diaz-Mayoral,N., Kennedy,M., Engel,T., Wil-
liams,P., Erickson,K., Ardlie,K., Winckler,W., Getz,G.,
DeLuca, D., MacArthur, D., Kellis, M., Thomson, A.,
Young,T., Gelfand,E., Donovan,M., Meng,Y., Grant,G.,
Mash,D., Marcus,Y., Basile,M., Liu, J., Zhu,J., Tu,Z.,
Cox, N. J., Nicolae, D. L., Gamazon, E. R., Im, H. K.,
Konkashbaev, A., Pritchard, J., Stevens, M., Flutre, T.,
Wen,X., Dermitzakis, E.T., Lappalainen,T., Guigo,R.,
Monlong,J., Sammeth,M., Koller,D., Battle,A., Mosta-
favi, S., McCarthy, M., Rivas, M., Maller, J., Rusyn, I.,
Nobel, A., Wright, F., Shabalin, A., Feolo, M., Sharop-
ova,N., Sturcke,A., Paschal,J., Anderson, J.M., Wild-
er, E.L., Derr, L.K., Green, E.D., Struewing, J.P., Tem-
ple,G., Volpi,S., Boyer, J.T., Thomson, E.J., Guyer, M.S.,
Ng,C., Abdallah,A., Colantuoni,D., Insel, T.R., Koester,
S.E., Roger Little,A., Bender, P.K., Lehner,T., Yao,Y.,
Compton, C.C., Vaught, J.B., Sawyer,S., Lockhart, N.C.,
Demchok, J., and Moore, H. F. (2013) The Genotype-
Tissue Expression (GTEx) project, Nat. Genet., 45, 580-
585, doi:10.1038/ng.2653.
11. Tyshkovskiy, A., Ma, S., Shindyapina, A. V., Tikhon-
ov, S., Lee, S. G., Bozaykut, P., Castro, J. P., Seluan-
ov, A., Schork, N. J., Gorbunova, V., Dmitriev, S. E.,
Miller, R.A., and Gladyshev, V.N. (2023) Distinct lon-
gevity mechanisms across and within species and
their association with aging, Cell, 186, 2929-2949.e20,
doi:10.1016/j.cell.2023.05.002.
12. Ritchie, M.E., Phipson,B., Wu,D., Hu,Y., Law, C. W.,
Shi, W., and Smyth, G. K. (2015) Limma powers
TIKHONOV et al.320
BIOCHEMISTRY (Moscow) Vol. 89 No. 2 2024
differential expression analyses for RNA-sequencing
and microarray studies, Nucleic Acids Res., 43, e47,
doi:10.1093/nar/gkv007.
13. Viechtbauer,W. (2010) Conducting meta-analyses in R
with the metafor, J.Stat. Softw., 36, 1-48, doi:10.18637/
jss.v036.i03.
14. Korotkevich, G., Sukhov, V., and Sergushichev, A.
(2021) Fast gene set enrichment analysis, bioRxiv,
doi:10.1101/060012.
15. Chang, W., Cheng, J., Allaire, J., Sievert, C., Schloerke,
B., Xie, Y., Allen, J., McPherson, J., Dipert, A., and Borg-
es, B. (2022) Shiny: Web Application Framework for R..
16. Benjamini, Y., and Hochberg, Y. (1995) Controlling
the false discovery rate: a practical and powerful ap-
proach to multiple testing, J. R. Stat. Soc. Ser. B, 57,
289-300, doi:10.1111/j.2517-6161.1995.tb02031.x.
17. Subramanian,A., Tamayo, P., Mootha, V.K., Mukher-
jee,S., Ebert, B.L., Gillette, M.A., Paulovich,A., Pome-
roy, S. L., Golub, T. R., Lander, E. S., and Mesirov,
J. P. (2005) Gene set enrichment analysis: A knowl-
edge-based approach for interpreting genome-wide
expression profiles, Proc. Natl. Acad. Sci. USA, 102,
15545-15550, doi:10.1073/pnas.0506580102.
18. Mootha, V.K., Lindgren, C.M., Eriksson, K.F., Subrama-
nian,A., Sihag,S., Lehar,J., Puigserver,P., Carlsson,E.,
Ridderstråle, M., Laurila, E., Houstis, N., Daly, M. J.,
Patterson, N., Mesirov, J. P., Golub, T. R., Tamayo, P.,
Spiegelman,B., Lander, E.S., Hirschhorn, J.N., Altshul-
er,D., and Groop, L.C. (2003) PGC-1α-responsive genes
involved in oxidative phosphorylation are coordi-
nately downregulated in human diabetes, Nat. Genet.,
34, 267-273, doi:10.1038/ng1180.
19. Ashburner,M., Ball, C.A., Blake, J.A., Botstein,D., But-
ler,H., Cherry, J.M., Davis, A.P., Dolinski,K., Dwight,
S.S., Eppig, J.T., Harris, M.A., Hill, D.P., Issel-Tarver,L.,
Kasarskis,A., Lewis,S., Matese, J.C., Richardson, J.E.,
Ringwald, M., Rubin, G. M., and Sherlock, G. (2000)
Gene Ontology: tool for the unification of biology,
Nat. Genet., 25, 25-29, doi:10.1038/75556.
20. Carbon,S., Douglass,E., Good, B.M., Unni, D.R., Har-
ris, N.L., Mungall, C.J., Basu,S., Chisholm, R.L., Dod-
son, R. J., Hartline, E., Fey, P., Thomas, P. D., Albou,
L.P., Ebert,D., Kesling, M.J., Mi,H., Muruganujan,A.,
Huang, X., Mushayahama, T., LaBonte, S. A., Siegele,
D. A., Antonazzo, G., Attrill, H., Brown, N. H., Gara-
pati, P., Marygold, S. J., Trovisco, V., dos Santos, G.,
Falls,K., Tabone,C., Zhou,P., Goodman, J.L., Strelets,
V. B., Thurmond, J., Garmiri, P., Ishtiaq, R., Rodrí-
guez-López,M., Acencio, M.L., Kuiper,M., Lægreid,A.,
Logie, C., Lovering, R. C., Kramarz, B., Saverimuttu,
S.C.C., Pinheiro, S.M., Gunn,H., Su,R., Thurlow, K.E.,
Chibucos,M., Giglio,M., Nadendla,S., Munro,J., Jack-
son,R., Duesbury, M.J., Del-Toro,N., Meldal, B.H.M.,
Paneerselvam, K., Perfetto, L., Porras, P., Orchard, S.,
Shrivastava,A., Chang, H.Y., Finn, R.D., Mitchell, A.L.,
Rawlings, N. D., Richardson, L., Sangrador-Vegas, A.,
Blake, J.A., Christie, K.R., Dolan, M.E., Drabkin, H.J.,
Hill, D.P., Ni,L., Sitnikov, D.M., Harris, M.A., Oliver,
S. G., Rutherford, K., Wood, V., Hayles, J., Bähler, J.,
Bolton, E. R., de Pons, J. L., Dwinell, M. R., Hayman,
G. T., Kaldunski, M. L., Kwitek, A. E., Laulederkind,
S.J.F., Plasterer,C., Tutaj, M.A., Vedi,M., Wang, S.J.,
D’Eustachio, P., Matthews, L., Balhoff, J. P., Aleksand-
er, S. A., Alexander, M. J., Cherry, J. M., Engel, S. R.,
Gondwe,F., Karra,K., Miyasato, S.R., Nash, R.S., Simi-
son,M., Skrzypek, M.S., Weng,S., Wong, E.D., Feuer-
mann, M., Gaudet, P., Morgat,A., Bakker, E., Berardi-
ni, T.Z., Reiser,L., Subramaniam,S., Huala,E., Arighi,
C. N., Auchincloss, A., Axelsen, K., Argoud-Puy, G.,
Bateman,A., Blatter, M.C., Boutet,E., Bowler,E., Breu-
za,L., Bridge, A., Britto,R., Bye-A-Jee,H., Casas, C.C.,
Coudert, E., Denny, P., Es-Treicher, A., Famiglietti,
M.L., Georghiou, G., Gos, A. N., Gruaz-Gumowski,N.,
Hatton-Ellis, E., Hulo, C., Ignatchenko, A., Jungo, F.,
Laiho,K., Le Mercier,P., Lieberherr,D., Lock,A., Lus-
si,Y., MacDougall,A., Ma-Grane,M., Martin, M.J., Mas-
son,P., Natale, D.A., Hyka-Nouspikel,N., Orchard,S.,
Pedruzzi,I., Pourcel,L., Poux,S., Pundir,S., Rivoire,C.,
Speretta, E., Sundaram, S., Tyagi, N., Warner, K.,
Zaru,R., Wu, C.H., Diehl, A.D., Chan, J.N., Grove,C.,
Lee, R. Y. N., Muller, H. M., Raciti, D., van Auken, K.,
Sternberg, P.W., Berriman, M., Paulini,M., Howe,K.,
Gao,S., Wright,A., Stein,L., Howe, D.G., Toro,S., West-
erfield, M., Jaiswal, P., Cooper, L., and Elser, J. (2021)
The gene ontology resource: enriching a GOld mine,
Nucleic Acids Res., 49, D325-D334, doi: 10.1093/nar/
gkaa1113.
21. Kanehisa, M. (2000) KEGG: kyoto encyclopedia of
genes and genomes, Nucleic Acids Res., 28, 27-30,
doi:10.1093/nar/28.1.27.
22. Kanehisa,M. (2019) Toward understanding the origin
and evolution of cellular organisms, Protein Sci., 28,
1947-1951, doi:10.1002/pro.3715.
23. Kanehisa, M., Furumichi, M., Sato, Y., Ishiguro-Wata-
nabe, M., and Tanabe, M. (2021) KEGG: Integrating
viruses and cellular organisms, Nucleic Acids Res., 49,
D545-D551, doi:10.1093/nar/gkaa970.
24. Jassal,B., Matthews,L., Viteri,G., Gong,C., Lorente,P.,
Fabregat, A., Sidiropoulos, K., Cook, J., Gillespie, M.,
Haw,R., Loney,F., May,B., Milacic,M., Rothfels,K., Se-
villa, C., Shamovsky, V., Shorser, S., Varusai, T., Weis-
er, J., Wu, G., Stein, L., Hermjakob, H., and D’Eusta-
chio,P. (2020) The reactome pathway knowledgebase,
Nucleic Acids Res., 48, D498-D503, doi: 10.1093/nar/
gkz1031.
25. Rouillard, A. D., Gundersen, G. W., Fernandez, N. F.,
Wang, Z., Monteiro, C. D., McDermott, M. G., and
Ma’ayan, A. (2016) The harmonizome: a collec-
tion of processed datasets gathered to serve and
mine knowledge about genes and proteins, Data-
base (Oxford)
, 2016, baw100, doi: 10.1093/database/
baw100.
AgeMeta: GENE EXPRESSION IN AGING 321
BIOCHEMISTRY (Moscow) Vol. 89 No. 2 2024
26. Zhang, B., Lee, D. E., Trapp, A., Tyshkovskiy, A., Lu,
A.T., Bareja,A., Kerepesi,C., McKay, L.K., Shindyapi-
na, A.V., Dmitriev, S.E., Baht, G.S., Horvath,S., Glady-
shev, V. N., and White, J.P. (2023) Multi-omic rejuve-
nation and lifespan extension on exposure to youthful
circulation, Nat. Aging, 3, 948-964, doi:10.1038/s43587-
023-00451-9.
27. Argmann, C., Dobrin, R., Heikkinen, S., Auburtin, A.,
Pouilly,L., Cock, T.A., Koutnikova,H., Zhu,J., Schadt,
E.E., and Auwerx,J. (2009) Pparγ2 is a key driver of
longevity in the mouse, PLoS Genet., 5, e1000752,
doi:10.1371/journal.pgen.1000752.
28. Bernard,G., Teulière,J., Lopez,P., Corel,E., Lapointe,
F. J., and Bapteste, E. (2022) Aging at evolutionary
crossroads: longitudinal gene co-expression network
analyses of proximal and ultimate causes of aging
in bats, Mol. Biol. Evol., 39, msab302, doi: 10.1093/
molbev/msab302.
29. Southworth, L. K., Owen, A. B., and Kim, S.K. (2009)
Aging mice show a decreasing correlation of gene
expression within genetic modules, PLoS Genet., 5,
e1000776, doi:10.1371/journal.pgen.1000776.
30. Hu,Y., Xu,Y., Mao,L., Lei,W., Xiang,J., Gao,L., Jiang,J.,
Huang, L., Luo, O. J., Duan, J., and Chen, G. (2021)
Gene expression analysis reveals age and ethnicity
signatures between young and old adults in human
PBMC, Front. Aging, 2, 797040, doi: 10.3389/fragi.
2021.797040.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.