Integrating Machine Learning With Multi-Omics Technologies in Geroscience Towards Personalized Medicine
Integrating Machine Learning With Multi-Omics Technologies in Geroscience Towards Personalized Medicine
Personalized
Medicine
Review
Integrating Machine Learning with Multi-Omics Technologies in
Geroscience: Towards Personalized Medicine
Nikolaos Theodorakis 1,2 , Georgios Feretzakis 3 , Lazaros Tzelves 4 , Evgenia Paxinou 3 , Christos Hitas 1 ,
Georgia Vamvakou 1 , Vassilios S. Verykios 3, * and Maria Nikolaou 1
1 Department of Cardiology & 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str.,
15127 Melissia, Greece; [email protected] (N.T.); [email protected] (C.H.);
[email protected] (G.V.); [email protected] (M.N.)
2 School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
3 School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece;
[email protected] (G.F.); [email protected] (E.P.)
4 2nd Department of Urology, Sismanoglio General Hospital, Sismanogliou 37, National and Kapodistrian
University of Athens, 15126 Athens, Greece; [email protected]
* Correspondence: [email protected]; Tel.: +30-2610367698
1.4. Objectives
This review aims to provide a comprehensive overview of how integrating multi-
omics data with ML techniques can enhance our understanding of the hallmarks of aging.
We will discuss the role of different omics technologies in studying each hallmark of
aging. Furthermore, we will explore how ML models can be applied to predict and
analyze age-related changes. Additionally, we will highlight case studies and applications
that demonstrate the potential of this integrative approach. Moreover, we will report
the challenges and limitations in the field. Finally, we will suggest future directions for
advancing aging research and developing personalized strategies to extend health span
and lifespan.
J. Pers. Med. 2024, 14, 931 3 of 27
2.1.2. Machine Learning Models to Predict Genomic Instability and Its Effects
ML models are particularly useful in predicting the effects of genomic instability by
analyzing large-scale genomic datasets. Supervised learning ML algorithms, like support
vector machines (SVMs) and neural networks, can be trained on labeled genomic data
to classify mutations as benign or pathogenic, which is critical for understanding which
mutations contribute to aging and age-related diseases. Unsupervised learning techniques
like clustering can identify novel mutational signatures that may not be evident through
traditional analysis. These signatures can be associated with aging, leading to insights into
the genomic changes that accumulate over time [13].
In a specific example, Xu et al. developed a deep learning model to predict genomic
instability from histopathology slides. When applied to 1010 patients with breast cancer,
the model accurately classified chromosomal instability status with with 81.2% sensitivity
and 68.7% specificity in the test set [14].
2.4.2. Omics Data to Study Proteostasis and Machine Learning Models to Predict
Proteostasis-Related Disorders
ML models, especially deep learning approaches, can analyze large-scale proteomic
data to identify patterns of protein misfolding, aggregation, and degradation that are
characteristic of aging cells. Supervised learning can predict the likelihood of proteostasis-
related disorders, such as neurodegenerative diseases, by analyzing protein expression
profiles and identifying key proteins that lose stability with age [22,23].
In a specific example, Eshari et al. developed an logistic regression model to predict
protein aggregation propensity from sequence data. Their model outperformed existing
methods in predicting aggregation-prone regions in proteins. This approach could be
valuable for understanding protein misfolding in age-related neurodegenerative diseases
and for designing therapeutic interventions [24].
2.6.2. Integrative Models to Study the Regulation and Deregulation of These Pathways
Supervised ML algorithms can predict the activation state of nutrient sensing pathways
(like mTOR, AMPK, and sirtuins) based on gene expression and proteomic data, aiding
in the understanding of how these pathways contribute to aging. Unsupervised learning
techniques can identify novel regulatory networks within nutrient sensing pathways,
providing deeper insights into how these pathways are deregulated in aging [29].
In a specific example, Drewe et al. used machine learning models to identify AMPK
activators, which play a crucial role in regulating cellular metabolism and are beneficial in
treating diseases like diabetes and cancer. Various algorithms, including random forest and
deep neural networks, were tested and showed high accuracy in distinguishing activators
from controls. The models are suitable for screening potential AMPK activators, including
natural compounds, and can guide further in vitro testing to identify promising candidates
for drug development [30].
2.8.2. Using Omics Data and Machine Learning to Identify Senescent Cells and
Develop Interventions
Supervised ML models can classify cells based on their senescence status by analyzing
the expression of cell cycle inhibitors and SASP factors, allowing for the identification of
senescent cells within tissues. Furthermore, ML can also model the composition and dynam-
ics of the SASP, providing insights into how these factors contribute to tissue dysfunction
and aging [35].
In a specific example, Tuttle et al. conducted a systematic review and meta-analysis
of cellular senescence markers across various human tissues. While not directly applying
ML, their comprehensive analysis provides a foundation for future ML studies on cellular
senescence. They synthesized data from multiple studies to identify consistent markers of
senescence across different tissues and age groups. This work is crucial for developing ML
models to predict cellular senescence and its impact on aging [36].
2.9.2. Integrative Approaches to Study Stem Cell Biology and Predictive Models
ML models can predict the likelihood of stem cell exhaustion by analyzing gene
expression and protein interaction networks, helping to identify key factors that lead to the
decline in stem cell function with age. These models can also help in planning interventions
by predicting the effects of potential treatments on stem cell function, guiding strategies to
rejuvenate aged stem cells [38].
In a specific example, Barardo et al. developed an ML model to predict lifespan-
extending compounds, which could potentially address stem cell exhaustion. They used
a random forest classifier trained on various molecular and chemical features to predict
compounds that might extend lifespan. While not directly focused on stem cells, many
lifespan-extending compounds work by preserving stem cell function. This approach
demonstrates how ML can be used to identify potential interventions for age-related stem
cell exhaustion [39].
2.10.2. Omics Data to Study Intercellular Communication and Machine Learning Models to
Predict Alterations
ML can classify cells based on their signaling activity by integrating transcriptomic
and proteomic data, helping to identify disruptions in cell communication that contribute
to aging. By revealing how altered intercellular communication interacts with processes
like inflammation, ML models can guide the development of therapies aimed at restoring
normal cell signaling [41].
In a specific example, Wang et al. developed iTALK, an R package that uses ML algo-
rithms to analyze single-cell RNA-seq data and predict cell–cell communication networks.
While not specifically focused on aging, this tool has significant potential for studying how
intercellular communication changes with age. iTALK can identify ligand-receptor pairs
and visualize communication networks, providing insights into how cellular interactions
are altered in aging tissues [42].
2.12. Dysbiosis
2.12.1. Age-Related Changes in the Microbiome
Dysbiosis refers to changes in the composition and function of the microbiome. The
microbiome, which consists of trillions of microorganisms residing in the gut and other
tissues, plays a critical role in regulating metabolism, immunity, and overall health. Alter-
ations in the microbiome can influence various aspects of health and contribute to aging
and age-related diseases. Studies have shown that aged individuals exhibit a decline in
microbial diversity and an increase in the abundance of pathogenic bacteria. Experimental
evidence from model organisms has demonstrated that the restoration of a healthy micro-
biome, through the administration of probiotics or fecal microbiota transplantation, can
improve metabolic health and extend lifespan, suggesting that targeting dysbiosis could be
a viable strategy to combat aging [46].
J. Pers. Med. 2024, 14, 931 9 of 27
2.12.2. Integrative Approaches to Study the Microbiome and Machine Learning Models to
Predict Dysbiosis
ML can classify microbiome samples based on their age-related profiles, using metage-
nomic and metabolomic data to identify signatures of dysbiosis that contribute to aging.
By integrating omics data, ML models can reveal how dysbiosis interacts with host pro-
cesses, such as metabolism and immune function, guiding the development of microbiome-
targeted interventions [46].
In a specific example, Wilmanski et al. (2021) used ML to analyze microbiome data and
predict biological age. They developed a random forest model trained on gut microbiome
composition data from a large cohort of individuals. The model identified specific microbial
features associated with healthy aging and longevity. The study demonstrated how ML
can be used to understand the complex relationship between the gut microbiome and
aging, providing insights into potential interventions to promote healthy aging through
microbiome modulation [47].
A summary of the aging hallmarks, their molecular mechanisms, and potential inter-
ventions is presented in Table 1.
Table 1. Summary of aging hallmarks, their molecular mechanisms, and potential interventions.
Changes in DNA methylation, histone • HDAC inhibitors (e.g., suberoylanilide hydroxamic acid).
Epigenetic Alterations [2,18,19,52,53]
modifications, and chromatin remodeling. • DNMT inhibitors (e.g., 5-azacytidine).
Permanent cell cycle arrest due to DNA • Senolytics (e.g., dasatinib, quercetin).
Cellular Senescence [2,34–36,60,61]
damage, oxidative stress, etc. • SASP inhibitors (e.g., ruxolitinib).
Reduction in the number and function of • Wnt pathway modulators (e.g., R-spondin1).
Stem Cell Exhaustion [2,37,38,62]
stem cells. • Young blood factors (e.g., GDF-11, TIMP-2).
Abbreviations. AMPK (AMP-activated protein kinase); DNA (deoxyribonucleic acid); DNMT (DNA methyltrans-
ferase); GDF-11 (growth differentiation factor-11); HDAC (histone deacetylase); IGF-1 (insulin-like growth factor 1);
IL-10 (interleukin-10); mTOR (mechanistic target of rapamycin); NAD+ (nicotinamide adenine dinucleotide);
NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells); PARP (poly(ADP-ribose) polymerase);
PGC-1α (peroxisome proliferator-activated receptor gamma coactivator 1-alpha); ROS (reactive oxygen species);
SASP (senescence-associated secretory phenotype); TA-65 (telomerase activator-65); TIMP-2 (tissue inhibitor of
metalloproteinases-2); Wnt (wingless/integrated).
J. Pers. Med. 2024, 14, 931 10 of 27
Figure
Figure 1. Workflow
1. Workflow ofofmulti-omics
multi-omics data
dataintegration withwith
integration machine learning
machine in aging research.
learning in aging research.
A summary of ML techniques and their applications in multi-omics aging research is
A summary
presented of ML
in Table 2. techniques and their applications in multi-omics aging research
presented in Table 2.
J. Pers. Med. 2024, 14, 931 12 of 27
Table 2. Summary of machine learning techniques and their applications in multi-omics aging research.
Models that capture relationships and • Modeling interactions between genes, proteins,
Graph Neural Networks interactions between entities in a and metabolites.
graph structure. • Integrating multi-omics data for comprehensive analysis.
Convolutional Neural Deep learning models are particularly effective • Integrating and analyzing imaging data.
Networks for analyzing spatial and visual data. • Detecting patterns in omics data.
3.4. Practical Guidance on Selecting Machine Learning Methods for Multi-Omics Research
in Geroscience
Data from multi-omics technologies are often characterized by diversity and com-
plexity, hindering analysis and drawing conclusions. When selecting ML methods for
multi-omics integration in geroscience, it is crucial to consider the specific characteristics of
the data and the research objectives. As a practical guide, we can apply the following ML
methods for each of the following basic indications [20,77–80]:
• Genomic, epigenomic, and proteomic data analysis.
- Random forests: ideal for telomere length analysis.
- Support vector machines (SVMs): Suitable for distinguishing between benign and
pathogenic mutations in genomic instability studies. Also ideal for identification
of proteins with high aggregation potential.
- Gradient boosting machines (GBMs): effective in identifying key epigenetic modi-
fications that contribute to aging.
- CNNs: best for analyzing proteomic data, particularly when dealing with struc-
tural data like protein imaging or spatial transcriptomics, where the spatial rela-
tionships between features are important.
- Recurrent neural networks (RNNs) or long short-term memory (LSTM): Ideal for
analyzing genomic sequences where the order of nucleotides (sequential data) is
crucial. These models are particularly effective in understanding mutations that
affect protein structure and function.
J. Pers. Med. 2024, 14, 931 13 of 27
ple, genomic data can identify genetic predispositions to CVD, while transcriptomic and
proteomic data can reveal changes in gene and protein expression associated with disease
states. Metabolomic profiling can provide information on metabolic shifts that contribute
to CVD. ML models have been used to integrate these datasets, leading to the development
of predictive models for CVD risk and progression. A recent study has utilized ML to
investigate the presence of multiple factors in the risk of aortic stenosis. The findings
indicated that significant features present in aortic stenosis patients included older age,
arterial hypertension, aortic regurgitation, ascending aortic dilatation, and bicuspid aortic
valve. These insights suggest that hypertension and other factors play a crucial role in the
hemodynamic and anatomical progression of AS, highlighting the implications of ML for a
comprehensive risk assessment and early intervention strategy [52].
mTOR inhibitors. As mentioned earlier, rapamycin and its analogs inhibit mTOR
signaling, which can mimic the effects of caloric restriction and promote longevity. Pre-
clinical studies have demonstrated that mTOR inhibitors can extend lifespan and improve
metabolic health in various animal models. ML can identify individuals with overactive
mTOR signaling through integrated omics data analysis, suggesting those who may benefit
from such interventions [30].
Sirtuin activators. Resveratrol and other sirtuin activators enhance the activity of
sirtuins, a family of NAD+-dependent deacetylases involved in metabolic regulation and
stress resistance. These compounds have been shown to improve mitochondrial function,
enhance stress resistance, and extend lifespan in animal models. ML can analyze sirtuin
activity from proteomic and metabolomic data to identify candidates who would benefit
from sirtuin activators [62].
predict the benefits of young blood factors by analyzing circulating biomarkers and stem cell
function data, and identifying individuals who might benefit from such interventions [67].
both deep learning and ML to improve image analysis accuracy to 86%, surpassing tradi-
tional methods. By integrating CNN for robust feature extraction and ELM for efficient
classification, this model exemplifies the potential of hybrid ML systems in not only en-
hancing diagnostic accuracy but also in facilitating early disease detection, which is critical
in treatment planning and outcome improvement [90].
Additionally, AI is increasingly playing a transformative role in cardiology, enabling
enhanced diagnostic and predictive capabilities. ML algorithms, particularly neural net-
works, have been applied successfully in interpreting complex cardiovascular imaging
data, leading to improved diagnostic accuracy in conditions such as coronary artery dis-
ease, hypertrophic cardiomyopathy, and atrial fibrillation. These AI-driven tools assist
clinicians by providing more precise and rapid interpretations of echocardiograms and
electrocardiograms, potentially leading to better patient outcomes by enabling timely and
accurate treatment decisions [91]. Specifically, a recent study has explored the application
of ML in predicting atrial fibrillation in patients with embolic strokes of undetermined
sources (ESUS), showcasing another promising dimension of AI in cardiovascular health
diagnostics. A study utilized ML models, including SVMs, multilayer perceptron (MLP),
XGBoost, and random forest, to analyze clinical and echocardiographic data of 157 ESUS
patients. The SVM model exhibited the highest efficacy, with an area under the curve (AUC)
of approximately 0.736, demonstrating a robust capability to predict AF occurrences [92].
Another study focused on predicting myocardial ischemia by integrating exploratory data
analysis and ML models, achieving accuracies above 80% [93].
Furthermore, recent advancements in ML have significantly enhanced prostate cancer
diagnostics, particularly through the use of radiomics and AI in molecular imaging. By ap-
plying data-characterization algorithms to positron emission tomography scans, radiomics
helps to accurately distinguish between pathological and physiological tracer uptakes.
ML models, including CNNs, can analyze these radiomics features to predict Gleason
scores, influencing treatment decisions and enhancing personalized patient management
in oncology [94].
Such innovative approaches highlight the transformative impact of advanced ML
techniques in medical diagnosis, offering new pathways for the effective analysis of complex
biological datasets.
Additionally, techniques like cross-validation and bootstrapping can help assess model
robustness and reliability [96].
6. Future Directions
6.1. Advances in Omics Technologies
Emerging omics technologies, such as single-cell omics and spatial transcriptomics,
have the potential to provide unprecedented insights into the aging process. These tech-
nologies can capture molecular changes at higher resolution and across different tissues,
offering new opportunities for aging research.
J. Pers. Med. 2024, 14, 931 21 of 27
6.2.2. Explainable AI
Explainable AI techniques aim to provide interpretable and transparent ML models.
These methods can help researchers understand the decision-making process of ML models,
ensuring that the results are biologically meaningful and trustworthy. Explainable AI can
enhance the interpretability of multi-omics data integration and guide the development
of personalized interventions [76]. This is particularly important in precision medicine,
where the ability to interpret and trust AI models can significantly impact patient outcomes.
Explainable AI not only supports the integration of AI with digital health data but also
helps in improving the accuracy and reliability of predictive models, ensuring that the
results are actionable and meaningful for clinical practice [103].
Futuredirections
Figure2.2.Future
Figure directions
in in
thethe integration
integration of machine
of machine learning
learning withwith multi-omics
multi-omics technologies.
technologies.
6.4.Collaborative
6.4. CollaborativeEfforts
Efforts
in in Data
Data Sharing
Sharing andand Open
Open Science
Science
Collaborativeefforts
Collaborative efforts
in in data
data sharing
sharing andand open
open science
science are are essential
essential for advancing
for advancing
aging research. Sharing data and resources can facilitate the replication and
aging research. Sharing data and resources can facilitate the replication and validation validation
of of
findings, improve the robustness of results, and accelerate the translation of research into into
findings, improve the robustness of results, and accelerate the translation of research
clinicalapplications.
clinical applications.
7. Conclusions
The integration of multi-omics data with ML offers a powerful approach to delve into
a better understanding of the molecular hallmarks of aging. Advances in these technologies
can have a significant impact on aging research. This approach has the potential to identify
critical biomarkers and therapeutic targets, paving the way for personalized interventions
aimed at extending both health span and lifespan. The future of aging research lies in
the seamless integration of these technologies, supported by collaborative efforts in data
sharing and open science. Such advancements will be essential in overcoming current
challenges and translating research findings into clinical practice. Ultimately, by leveraging
the power of multi-omics data and ML, we can move closer to a future where healthy aging
is achievable for all.
Author Contributions: Conceptualization, N.T. and V.S.V.; Methodology, G.F.; Validation, L.T.;
Writing—Original Draft Preparation, N.T., G.F., L.T. and E.P.; Writing—Review and Editing, C.H.,
G.V., V.S.V. and M.N.; Visualization, N.T.; Supervision, V.S.V. and M.N.; Project Administration, M.N.
All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: No new data were created or analyzed in this study. Data sharing is
not applicable to this article.
Conflicts of Interest: The authors declare no conflicts of interest.
References
1. Li, Z.; Zhang, Z.; Ren, Y.; Wang, Y.; Fang, J.; Yue, H.; Ma, S.; Guan, F. Aging and Age-Related Diseases: From Mechanisms to
Therapeutic Strategies. Biogerontology 2021, 22, 165–187. [CrossRef]
2. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of Aging: An Expanding Universe. Cell 2023, 186,
243–278. [CrossRef]
3. Jay, F.B. An Overview of Deep Generative Models in Functional and Evolutionary Genomics. Annu. Rev. Biomed. Data Sci. 2023, 6,
173–189.
4. Evans, M.J.; Smith, R.L. Epigenetic Mechanisms in Aging. Trends Cell Biol. 2021, 31, 347–358.
5. Chambers, D.C.; Carew, A.M.; Lukowski, S.W.; Powell, J.E. Transcriptomics and Single-Cell RNA-Sequencing. Respirology 2019,
24, 29–36. [CrossRef] [PubMed]
6. Moaddel, R.; Ubaida-Mohien, C.; Tanaka, T.; Lyashkov, A.; Basisty, N.; Schilling, B.; Semba, R.D.; Franceschi, C.; Gorospe, M.;
Ferrucci, L. Proteomics in Aging Research: A Roadmap to Clinical, Translational Research. Aging Cell 2021, 20, e13325. [CrossRef]
7. Fang, W.; Chen, S.; Jin, X.; Liu, S.; Cao, X.; Liu, B. Metabolomics in Aging Research: Aging Markers from Organs. Front. Cell Dev.
Biol. 2023, 11, 1071–1078. [CrossRef] [PubMed]
8. Picard, M.; Scott-Boyer, M.-P.; Bodein, A.; Périn, O.; Droit, A. Integration Strategies of Multi-Omics Data for Machine Learning
Analysis. Comput. Struct. Biotechnol. J. 2021, 19, 3735–3746. [CrossRef] [PubMed]
9. Odden, M.C.; Melzer, D. Machine Learning in Aging Research. J. Gerontol. A Biol. Sci. Med. Sci. 2019, 74, 1901–1902. [CrossRef]
10. Nelson, B.C.; Dizdaroglu, M. Implications of DNA Damage and DNA Repair on Human Diseases. Mutagenesis 2020, 35, 1–3.
[CrossRef]
11. Lima, T.; Li, T.Y.; Mottis, A.; Auwerx, J. Pleiotropic Effects of Mitochondria in Aging. Nat. Aging 2022, 2, 199–213. [CrossRef]
[PubMed]
12. Nikfarjam, S.; Singh, K.K. DNA Damage Response Signaling: A Common Link Between Cancer and Cardiovascular Diseases.
Cancer Med. 2023, 12, 4380–4404. [CrossRef]
13. López-Gil, L.; Pascual-Ahuir, A.; Proft, M. Genomic Instability and Epigenetic Changes during Aging. Int. J. Mol. Sci. 2023, 24,
14279. [CrossRef]
14. Xu, Z.; Verma, A.; Naveed, U.; Bakhoum, S.F.; Khosravi, P.; Elemento, O. Deep learning predicts chromosomal instability from
histopathology images. iScience 2021, 24, 102394. [CrossRef] [PubMed]
15. Armanios, M. The Role of Telomeres in Human Disease. Annu. Rev. Genom. Hum. Genet. 2022, 23, 363–381. [CrossRef] [PubMed]
16. Sohn, E.J.; Goralsky, J.A.; Shay, J.W.; Min, J. The Molecular Mechanisms and Therapeutic Prospects of Alternative Lengthening of
Telomeres (ALT). Cancers 2023, 15, 1945. [CrossRef] [PubMed]
J. Pers. Med. 2024, 14, 931 24 of 27
17. Zhang, H.; Kong, W.; Xie, Y.; Zhao, X.; Luo, D.; Chen, S.; Pan, Z. Telomere-related genes as potential biomarkers to predict
endometriosis and immune response: Development of a machine learning-based risk model. Front. Med. 2023, 10, 1132676.
[CrossRef]
18. Zhang, J.; Wang, S.; Liu, B. New Insights into the Genetics and Epigenetics of Aging Plasticity. Genes 2023, 14, 329. [CrossRef]
19. Zhou, J.; Chen, Q.; Braun, P.R.; Mandell, K.A.P.; Jaffe, A.E.; Tan, H.Y.; Hyde, T.M.; Kleinman, J.E.; Potash, J.B.; Shinozaki, G.; et al.
Deep Learning Predicts DNA Methylation Regulatory Variants in the Human Brain and Elucidates the Genetics of Psychiatric
Disorders. Proc. Natl. Acad. Sci. USA 2022, 119, e2206069119. [CrossRef]
20. Higgins-Chen, A.T.; Thrush, K.L.; Wang, Y.; Minteer, C. A Computational Solution for Bolstering Reliability of Epigenetic Clocks:
Implications for Clinical Trials and Longitudinal Tracking. Nat. Aging 2022, 2, 644–661. [CrossRef]
21. Klaips, C.L.; Jayaraj, G.G.; Hartl, F.U. Pathways of Cellular Proteostasis in Aging and Disease. J. Cell Biol. 2018, 217, 51–63.
[CrossRef]
22. Chen, X.Q.; Shen, T.; Fang, S.J.; Sun, X.M.; Li, G.Y.; Li, Y.F. Protein Homeostasis in Aging and Cancer. Front. Cell Dev. Biol. 2023,
11, 1143532. [CrossRef]
23. Thompson, M.A.; De-Souza, E.A. A Year at the Forefront of Proteostasis and Aging. Biol. Open 2023, 12, bio059750. [CrossRef]
24. Eshari, F.; Momeni, F.; Faraj Nezhadi, A.; Shemehsavar, S.; Habibi-Rezaei, M. Prediction of protein aggregation propensity
employing SqFt-based logistic regression model. Int. J. Biol. Macromol. 2023, 249, 126036. [CrossRef] [PubMed]
25. Lu, G.; Wang, Y.; Shi, Y.; Zhang, Z.; Huang, C.; He, W.; Wang, C.; Shen, H.-M. Autophagy in Health and Disease: From Molecular
Mechanisms to Therapeutic Target. MedComm 2022, 3, e150. [CrossRef] [PubMed]
26. Gómez-Virgilio, L.; Silva-Lucero, M.-D.-C.; Flores-Morelos, D.-S.; Gallardo-Nieto, J.; Lopez-Toledo, G.; Abarca-Fernandez, A.-M.;
Zacapala-Gómez, A.-E.; Luna-Muñoz, J.; Montiel-Sosa, F.; Soto-Rojas, L.O.; et al. Autophagy: A Key Regulator of Homeostasis
and Disease: An Overview of Molecular Mechanisms and Modulators. Cells 2022, 11, 2262. [CrossRef] [PubMed]
27. Dong, G.; Gao, H.; Chen, Y.; Yang, H. Machine learning and bioinformatics analysis to identify autophagy-related biomarkers in
peripheral blood for rheumatoid arthritis. Front. Genet. 2023, 14, 1238407. [CrossRef]
28. Johnson, S.C. Nutrient Sensing, Signaling and Ageing: The Role of IGF-1 and mTOR in Ageing and Age-Related Disease. Subcell
Biochem. 2018, 90, 49–97.
29. Fu, W.; Wu, G. Targeting mTOR for Anti-Aging and Anti-Cancer Therapy. Molecules 2023, 28, 3157. [CrossRef]
30. Drewe, J.; Küsters, E.; Hammann, F.; Kreuter, M.; Boss, P.; Schöning, V. Modeling Structure–Activity Relationship of AMPK
Activation. Molecules 2021, 26, 6508. [CrossRef]
31. Videla, L.A.; Marimán, A.; Ramos, B.; José Silva, M.; Del Campo, A. Standpoints in Mitochondrial Dysfunction: Underlying
Mechanisms in Search of Therapeutic Strategies. Mitochondrion 2022, 63, 9–22. [CrossRef] [PubMed]
32. Amorim, J.A.; Coppotelli, G.; Rolo, A.P.; Palmeira, C.M.; Ross, J.M.; Sinclair, D.A. Mitochondrial and Metabolic Dysfunction in
Ageing and Age-Related Diseases. Nat. Rev. Endocrinol. 2022, 18, 243–258. [CrossRef] [PubMed]
33. Qin, H.; Abulaiti, A.; Maimaiti, A.; Abulaiti, Z.; Fan, G.; Aili, Y.; Ji, W.; Wang, Z.; Wang, Y. Integrated machine learning survival
framework develops a prognostic model based on inter-crosstalk definition of mitochondrial function and cell death patterns in a
large multicenter cohort for lower-grade glioma. J. Transl. Med. 2023, 21, 588. [CrossRef] [PubMed]
34. Kuehnemann, C.; Wiley, C.D. Senescent Cells at the Crossroads of Aging, Disease, and Tissue Homeostasis. Aging Cell 2024, 23,
e13988. [CrossRef] [PubMed]
35. Cuollo, L.; Antonangeli, F.; Santoni, A.; Soriani, A. The Senescence-Associated Secretory Phenotype (SASP) in the Challenging
Future of Cancer Therapy and Age-Related Diseases. Biology 2020, 9, 485. [CrossRef] [PubMed]
36. Tuttle, C.S.L.; Waaijer, M.E.C.; Slee-Valentijn, M.S.; Stijnen, T.; Westendorp, R.; Maier, A.B. Cellular Senescence and Chronological
Age in Various Human Tissues: A Systematic Review and Meta-Analysis. Aging Cell 2021, 20, e13083. [CrossRef]
37. Mi, L.; Hu, J.; Li, N.; Gao, J.; Huo, R.; Peng, X.; Zhang, N.; Liu, Y.; Zhao, H.; Liu, R.; et al. The Mechanism of Stem Cell Aging.
Stem Cell Rev. Rep. 2022, 18, 1281–1293. [CrossRef]
38. Weng, Z.; Wang, Y.; Ouchi, T.; Liu, H.; Qiao, X.; Wu, C.; Zhao, Z.; Li, L.; Li, B. Mesenchymal Stem/Stromal Cell Senescence:
Hallmarks, Mechanisms, and Combating Strategies. Stem Cells Transl. Med. 2022, 11, 356–371. [CrossRef]
39. Barardo, D.G.; Newby, D.; Thornton, D.; Ghafourian, T.; de Magalhães, J.P.; Freitas, A.A. Machine Learning for Predicting
Lifespan-Extending Chemical Compounds. Aging 2022, 14, 2163–2189. [CrossRef]
40. Liu, Z.; Liang, Q.; Ren, Y.; Guo, C.; Ge, X.; Wang, L.; Cheng, Q.; Luo, P.; Zhang, Y.; Han, X. Immunosenescence: Molecular
Mechanisms and Diseases. Signal Transduct. Target. Ther. 2023, 8, 200. [CrossRef]
41. Baechle, J.J.; Chen, N.; Makhijani, P.; Winer, S.; Furman, D.; Winer, D.A. Chronic Inflammation and the Hallmarks of Aging. Mol.
Metab. 2023, 74, 101755. [CrossRef] [PubMed]
42. Wang, Y.; Wang, R.; Zhang, S.; Song, S. iTALK: An R Package to Characterize and Illustrate Intercellular Communication. bioRxiv
2021, 507871. [CrossRef]
43. Dugan, B.; Conway, J.; Duggal, N.A. Inflammaging as a Target for Healthy Ageing. Age Ageing 2023, 52, afac328. [CrossRef]
44. Libby, P. Inflammatory Mechanisms: The Molecular Basis of Inflammation and Disease. Nature 2007, 420, 868–874. [CrossRef]
[PubMed]
45. Bobrov, E.; Georgievskaya, A.; Kiselev, K.; Sevastopolsky, A.; Zhavoronkov, A.; Gurov, S.; Rudakov, K.; Del Pilar Bonilla Tobar, M.;
Jaspers, S.; Clemann, S. PhotoAgeClock: Deep Learning Algorithms for Development of Non-Invasive Visual Biomarkers of
Aging. Aging 2022, 14, 1500–1519. [CrossRef] [PubMed]
J. Pers. Med. 2024, 14, 931 25 of 27
46. Salazar, J.; Durán, P.; Díaz, M.P.; Chacín, M.; Santeliz, R.; Mengual, E.; Gutiérrez, E.; León, X.; Díaz, A.; Bernal, M.; et al. Exploring
the Relationship Between the Gut Microbiota and Ageing: A Possible Age Modulator. Int. J. Environ. Res. Public Health 2023, 20,
5845. [CrossRef]
47. Wilmanski, T.; Diener, C.; Rappaport, N.; Patwardhan, S.; Wiedrick, J.; Lapidus, J.; Earls, J.C.; Zimmer, A.; Glusman, G.; Robinson,
M.; et al. Gut Microbiome Pattern Reflects Healthy Ageing and Predicts Survival in Humans. Nat. Metab. 2021, 3, 274–286.
[CrossRef]
48. Shi, Y.; Zhang, H.; Huang, S.; Yin, L.; Wang, F.; Luo, P.; Huang, H. Epigenetic Regulation in Cardiovascular Disease: Mechanisms
and Advances in Clinical Trials. Signal Transduct. Target. Ther. 2022, 7, 200. [CrossRef]
49. Bhole, R.P.; Chikhale, R.V.; Rathi, K.M. Current Biomarkers and Treatment Strategies in Alzheimer Disease: An Overview and
Future Perspectives. IBRO Neurosci. Rep. 2023, 16, 8–42. [CrossRef]
50. Veneziani, I.; Marra, A.; Formica, C.; Grimaldi, A.; Marino, S.; Quartarone, A.; Maresca, G. Applications of Artificial Intelligence
in the Neuropsychological Assessment of Dementia: A Systematic Review. J. Pers. Med. 2024, 14, 113. [CrossRef]
51. Formica, C.; Bonanno, L.; Giambò, F.M.; Maresca, G.; Latella, D.; Marra, A.; Cucinotta, F.; Bonanno, C.; Lombardo, M.; Tomarchio,
O.; et al. Paving the Way for Predicting the Progression of Cognitive Decline: The Potential Role of Machine Learning Algorithms
in the Clinical Management of Neurodegenerative Disorders. J. Pers. Med. 2023, 13, 1386. [CrossRef] [PubMed]
52. Irtyuga, O.; Babakekhyan, M.; Kostareva, A.; Uspensky, V.; Gordeev, M.; Faggian, G.; Malashicheva, A.; Metsker, O.; Shlyakhto,
E.; Kopanitsa, G. Analysis of Prevalence and Clinical Features of Aortic Stenosis in Patients with and Without Bicuspid Aortic
Valve Using Machine Learning Methods. J. Pers. Med. 2023, 13, 1588. [CrossRef]
53. Paluch-Shimon, S.; Cardoso, F. PARP Inhibitors Coming of Age. Nat. Rev. Clin. Oncol. 2021, 18, 69–70. [CrossRef] [PubMed]
54. Sharma, A.; Chabloz, S.; Lapides, R.A.; Roider, E.; Ewald, C.Y. Potential Synergistic Supplementation of NAD+ Promoting
Compounds as a Strategy for Increasing Healthspan. Nutrients 2023, 15, 445. [CrossRef]
55. Derevyanko, A.; Skowronska, A.; Skowronski, M.T.; Kordowitzki, P. The Interplay Between Telomeres, Mitochondria, and
Chronic Stress Exposure in the Aging Egg. Cells 2022, 11, 2612. [CrossRef] [PubMed]
56. Cao, Q.; Tian, Y.; Deng, Z.; Yang, F.; Chen, E. Epigenetic Alteration in Colorectal Cancer: Potential Diagnostic and Prognostic
Implications. Int. J. Mol. Sci. 2024, 25, 3358. [CrossRef] [PubMed]
57. Wang, K.; Liu, H.; Hu, Q.; Wang, L.; Liu, J.; Zheng, Z.; Zhang, W.; Ren, J.; Zhu, F.; Liu, G.-H. Epigenetic Regulation of Aging:
Implications for Interventions of Aging and Diseases. Signal Transduct. Target. Ther. 2022, 7, 374. [CrossRef]
58. Türker, F.; Cook, E.K.; Margolis, S.S. The Proteasome and Its Role in the Nervous System. Cell Chem. Biol. 2021, 28, 903–917.
[CrossRef]
59. Kaushik, S.; Tasset, I.; Arias, E.; Pampliega, O.; Wong, E.; Martinez-Vicente, M.; Cuervo, A.M. Autophagy and the Hallmarks of
Aging. Ageing Res. Rev. 2021, 72, 101468. [CrossRef]
60. Sharp, Z.D.; Strong, R. Rapamycin, the Only Drug That Has Been Consistently Demonstrated to Increase Mammalian Longevity:
An Update. Exp. Gerontol. 2023, 176, 112166. [CrossRef]
61. Zhu, X.; Shen, W.; Liu, Z.; Sheng, S.; Xiong, W.; He, R.; Zhang, X.; Ma, L.; Ju, Z. Effect of Metformin on Cardiac Metabolism and
Longevity in Aged Female Mice. Front. Cell Dev. Biol. 2021, 8, 626011. [CrossRef] [PubMed]
62. Iside, C.; Scafuro, M.; Nebbioso, A.; Altucci, L. SIRT1 Activation by Natural Phytochemicals: An Overview. Front. Pharmacol.
2020, 11, 1225. [CrossRef] [PubMed]
63. Norat, P.; Soldozy, S.; Sokolowski, J.D.; Gorick, C.M.; Kumar, J.S.; Chae, Y.; Yağmurlu, K.; Prada, F.; Walker, M.; Levitt, M.R.; et al.
Mitochondrial Dysfunction in Neurological Disorders: Exploring Mitochondrial Transplantation. NPJ Regen. Med. 2020, 5, 22.
[CrossRef]
64. Lu, Y.; Fujioka, H.; Wang, W.; Zhu, X. Bezafibrate Confers Neuroprotection in the 5xFAD Mouse Model of Alzheimer’s Disease.
Biochim. Biophys. Acta Mol. Basis Dis. 2023, 1869, 166841. [CrossRef] [PubMed]
65. Kirkland, J.L.; Tchkonia, T. Senolytic Drugs: From Discovery to Translation. J. Intern. Med. 2020, 288, 518–536. [CrossRef]
[PubMed]
66. Xu, M.; Tchkonia, T.; Kirkland, J.L.; Nakahara, T. Senolytics Improve Physical Function and Increase Lifespan in Old Age. Nat.
Med. 2018, 21, 857–862. [CrossRef]
67. O’Loghlen, A. The Potential of Aging Rejuvenation. Cell Cycle 2022, 21, 111–116. [CrossRef]
68. Jiang, T.; Gradus, J.L.; Rosellini, A.J. Supervised Machine Learning: A Brief Primer. Behav. Ther. 2020, 51, 675–687. [CrossRef]
69. Eckhardt, C.M.; Madjarova, S.J.; Williams, R.J.; Ollivier, M.; Karlsson, J.; Pareek, A.; Nwachukwu, B.U. Unsupervised Machine
Learning Methods and Emerging Applications in Healthcare. Knee Surg. Sports Traumatol. Arthrosc. 2023, 31, 376–381. [CrossRef]
70. Al-Hamadani, M.N.A.; Fadhel, M.A.; Alzubaidi, L.; Balazs, H. Reinforcement Learning Algorithms and Applications in Healthcare
and Robotics: A Comprehensive and Systematic Review. Sensors 2024, 24, 2461. [CrossRef]
71. LeCun, Y.; Bengio, Y.; Hinton, G. Deep Learning. Nature 2015, 521, 436–444. [CrossRef] [PubMed]
72. Subramanian, I.; Verma, S.; Kumar, S.; Jere, A.; Anamika, K. Multi-Omics Data Integration, Interpretation, and Its Application.
Bioinform. Biol. Insights 2020, 14, 1177932219899051. [CrossRef] [PubMed]
73. Athieniti, E.; Spyrou, G.M. A Guide to Multi-Omics Data Collection and Integration for Translational Medicine. Comput. Struct.
Biotechnol. J. 2022, 20, 134–149. [CrossRef]
74. Zitnik, M.; Agrawal, M.; Leskovec, J. Modeling Polypharmacy Side Effects with Graph Convolutional Networks. Bioinformatics
2018, 34, i457–i466. [CrossRef] [PubMed]
J. Pers. Med. 2024, 14, 931 26 of 27
75. Theodoris, C.V.; Xiao, L.; Chopra, A.; Chaffin, M.D.; Al Sayed, Z.R.; Hill, M.C.; Mantineo, H.; Brydon, E.M.; Zeng, Z.; Liu, X.S.;
et al. Transfer Learning Enables Predictions in Network Biology. Nature 2023, 618, 616–624. [CrossRef] [PubMed]
76. Wu, W.-T.; Li, Y.-J.; Feng, A.-Z.; Li, L.; Huang, T.; Xu, A.-D.; Lyu, J. Data Mining in Clinical Big Data: The Frequently Used
Databases, Steps, and Methodological Models. Mil. Med. Res. 2021, 8, 44. [CrossRef]
77. Ketkar, Y.; Gawade, S. A Decision Support System for Selecting the Most Suitable Machine Learning in Healthcare Using User
Parameters and Requirements. Healthc. Anal. 2022, 2, 100117. [CrossRef]
78. Huang, S.; Cai, N.; Pacheco, P.P.; Narrandes, S.; Wang, Y.; Xu, W. Applications of Support Vector Machine (SVM) Learning in
Cancer Genomics. Cancer Genom. Proteom. 2018, 15, 41–51.
79. Quist, J.; Taylor, L.; Staaf, J.; Grigoriadis, A. Random Forest Modelling of High-Dimensional Mixed-Type Data for Breast Cancer
Classification. Cancers 2021, 13, 991. [CrossRef]
80. Navarro, S.; Ventura, S. Computational Methods to Predict Protein Aggregation. Curr. Opin. Struct. Biol. 2022, 73, 102343.
[CrossRef]
81. Whittemore, K.; Derevyanko, A.; Martinez, P.; Serrano, R.; Pumarola, M.; Bosch, F.; Blasco, M.A. Telomerase Gene Therapy
Ameliorates the Effects of Neurodegeneration Associated with Short Telomeres in Mice. Aging 2019, 11, 2916–2948. [CrossRef]
[PubMed]
82. Squarzoni, S.; Schena, E.; Sabatelli, P.; Mattioli, E.; Capanni, C.; Cenni, V.; D’Apice, M.R.; Andrenacci, D.; Sarli, G.; Pellegrino, V.;
et al. Interleukin-6 Neutralization Ameliorates Symptoms in Prematurely Aged Mice. Aging Cell 2021, 20, e13285. [CrossRef]
[PubMed]
83. Liu, J.; Wang, Y.J. Rejuvenating the Immune System: Insights for Anti-Neurodegeneration Strategies. Neurosci. Bull. 2022, 38,
107–109. [CrossRef] [PubMed] [PubMed Central]
84. Zhang, L.; Zhao, J.; Mu, X.; McGowan, S.J.; Angelini, L.; O’Kelly, R.D.; Yousefzadeh, M.J.; Sakamoto, A.; Aversa, Z.; LeBrasseur,
N.K.; et al. Novel small molecule inhibition of IKK/NF-κB activation reduces markers of senescence and improves healthspan in
mouse models of aging. Aging Cell 2021, 20, e13486. [CrossRef] [PubMed]
85. Dagdeviren, S.; Jung, D.Y.; Friedline, R.H.; Noh, H.L.; Kim, J.H.; Patel, P.R.; Tsitsilianos, N.; Inashima, K.; Tran, D.A.; Hu, X.; et al.
IL-10 prevents aging-associated inflammation and insulin resistance in skeletal muscle. FASEB J. 2017, 31, 701–710. [CrossRef]
[PubMed]
86. Parker, A.; Romano, S.; Ansorge, R.; Aboelnour, A.; Le Gall, G.; Savva, G.M.; Pontifex, M.G.; Telatin, A.; Baker, D.; Jones, E.; et al.
Fecal microbiota transfer between young and aged mice reverses hallmarks of the aging gut, eye, and brain. Microbiome 2022, 10,
68. [CrossRef]
87. Shapiro, S.C. Biomarkers in Rheumatoid Arthritis. Cureus 2021, 13, e15063. [CrossRef]
88. Hsu, L.-C.; Li, C.-J.; Lin, L.-T.; Pan, L.-F.; Wen, Z.-H.; Sheu, J.J.-C.; Tsui, K.-H. Multi-Omics Reveals the Role of Osteopon-
tin/Secreted Phosphoprotein 1 in Regulating Ovarian Aging. J. Pers. Med. 2024, 14, 78. [CrossRef]
89. Carrillo-Perez, F.; Morales, J.C.; Castillo-Secilla, D.; Gevaert, O.; Rojas, I.; Herrera, L.J. Machine-Learning-Based Late Fusion on
Multi-Omics and Multi-Scale Data for Non-Small-Cell Lung Cancer Diagnosis. J. Pers. Med. 2022, 12, 601. [CrossRef]
90. Sureshkumar, V.; Prasad, R.S.N.; Balasubramaniam, S.; Jagannathan, D.; Daniel, J.; Dhanasekaran, S. Breast Cancer Detection and
Analytics Using Hybrid CNN and Extreme Learning Machine. J. Pers. Med. 2024, 14, 792. [CrossRef]
91. Patrascanu, O.S.; Tutunaru, D.; Musat, C.L.; Dragostin, O.M.; Fulga, A.; Nechita, L.; Ciubara, A.B.; Piraianu, A.I.; Stamate, E.;
Poalelungi, D.G.; et al. Future Horizons: The Potential Role of Artificial Intelligence in Cardiology. J. Pers. Med. 2024, 14, 656.
[CrossRef] [PubMed]
92. Ming, C.; Lee, G.J.W.; Teo, Y.H.; Teo, Y.N.; Toh, E.M.S.; Li, T.Y.W.; Guo, C.Y.; Ding, J.; Zhou, X.; Teoh, H.L.; et al. Machine Learning
Modeling to Predict Atrial Fibrillation Detection in Embolic Stroke of Undetermined Source Patients. J. Pers. Med. 2024, 14, 534.
[CrossRef]
93. Elvas, L.B.; Nunes, M.; Ferreira, J.C.; Dias, M.S.; Rosário, L.B. AI-Driven Decision Support for Early Detection of Cardiac Events:
Unveiling Patterns and Predicting Myocardial Ischemia. J. Pers. Med. 2023, 13, 1421. [CrossRef] [PubMed]
94. Tapper, W.; Carneiro, G.; Mikropoulos, C.; Thomas, S.A.; Evans, P.M.; Boussios, S. The Application of Radiomics and AI to
Molecular Imaging for Prostate Cancer. J. Pers. Med. 2024, 14, 287. [CrossRef] [PubMed]
95. Martínez-García, M.; Hernández-Lemus, E. Data Integration Challenges for Machine Learning in Precision Medicine. Front. Med.
2022, 8, 784455. [CrossRef]
96. Bernardi, F.A.; Alves, D.; Crepaldi, N.; Yamada, D.B.; Lima, V.C.; Rijo, R. Data Quality in Health Research: Integrative Literature
Review. J. Med. Internet Res. 2023, 25, e41446. [CrossRef]
97. Vikas, H.; Kini, A.; Sharma, N.; Gowda, N.R.; Gupta, A. How informed is the informed consent? J. Fam. Med. Prim. Care 2021, 10,
2299–2303. [CrossRef]
98. Yadav, N.; Pandey, S.; Gupta, A.; Dudani, P.; Gupta, S.; Rangarajan, K. Data Privacy in Healthcare: In the Era of Artificial
Intelligence. Indian Dermatol. Online J. 2023, 14, 788–792. [CrossRef]
99. Wiltshire, D.; Alvanides, S. Ensuring the Ethical Use of Big Data: Lessons from Secure Data Access. Heliyon 2022, 8, e08981.
[CrossRef]
100. Washington, V.; Franklin, J.B.; Huang, E.S.; Mega, J.L.; Abernethy, A.P. Diversity, Equity, and Inclusion in Clinical Research: A
Path Toward Precision Health for Everyone. Clin. Pharmacol. Ther. 2023, 113, 575–584. [CrossRef]
J. Pers. Med. 2024, 14, 931 27 of 27
101. Stein, C.M.; Weiskirchen, R.; Damm, F.; Strzelecka, P.M. Single-Cell Omics: Overview, Analysis, and Application in Biomedical
Science. J. Cell. Biochem. 2021, 122, 1571–1578. [CrossRef]
102. Bressan, D.; Battistoni, G.; Hannon, G.J. The Dawn of Spatial Omics. Science 2023, 381, eabq4964. [CrossRef] [PubMed]
103. Allen, B. The Promise of Explainable AI in Digital Health for Precision Medicine: A Systematic Review. J. Pers. Med. 2024, 14, 277.
[CrossRef] [PubMed]
104. Devriendt, T.; Borry, P.; Shabani, M. Factors That Influence Data Sharing Through Data Sharing Platforms: A Qualitative Study
on the Views and Experiences of Cohort Holders and Platform Developers. PLoS ONE 2021, 16, e0254202. [CrossRef] [PubMed]
105. Isaacowitz, D.M.; Lind, M. Open Science Is for Aging Research, Too. Innov. Aging 2019, 3, igz028. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.