All Articles

The multiomics – a precision approach for aging measurement

Article
February 15, 2022
By
Agnieszka Szmitkowska, Ph.D.

Multiomics analyses data are integrated from multiple specific fields or "omes": genome, transcriptome, and proteome.

Highlights:

  • Multiomics analyses data are integrated from multiple specific fields or "omes" like genome, transcriptome, and proteome
  • Biomarkers of human aging derived from omics data could be used for personal aging speed measurement
  • Multiomics is still a costly approach in need of upgrade and standardization 

Introduction

Aging is a complex process influenced by external factors like environment, lifestyle as well as our internal biology, genes, epigenetic changes, metabolic regulation, interactions between host and microbiome, and others. Biological processes, such as the development of diseases and aging, depend on a dynamic and interactive system of molecular layers such as genetics, proteomics, or transcriptomics that, taken together, can be called multiomics. The analysis of high-throughput multiomics datasets can provide a complex and integrated profile of the multifactorial aging process in detail never available before. Multiomics strategies let scientists explore the molecular profile and regulatory status during aging and disease processes, leading to the discovery of new interventions.

The science of omics

The word omics refers to a field of biological sciences study ending with -omics: genomics, transcriptomics, proteomics, metabolomics, and others. The suffix -ome forms nouns denoting fields of a specified nature and addresses the objects of study of such fields as:

  • Genome - an organism's complete set of genetic information. Each genome contains all of the genes needed to build, grow and develop an organism
  • Transcriptome - the complete set of RNA transcripts in a specific cell type or tissue at a particular developmental stage, including messenger RNA (mRNA), transfer RNA (tRNA), ribosomal RNA (rRNA), and the non-coding RNAs (1)
  • Proteome - the sum of all the proteins in a cell, tissue, or organism.
  • Metabolome - the complete set of metabolites in the cell, tissue, organ, or organism, which are the end products of cellular processes (2)
  • Epigenome - all modifications to the genome that do not affect the DNA sequence but determine the gene expression (2)

The omics sciences identify, characterize, and quantify all biological molecules involved in the structure, function, and dynamics of a cell, tissue, or whole organism. The multiomics analysis is used in different fields of bioscience (3). It helps to understand age-related diseases such as cardiovascular disease (4), dementia (5), diabetes (6), and cancer (7), as well as aging itself. The multiomics approach provides the understanding of hidden associations and pathways which may be critical for both patients and specialists (8).

 

Biomarkers of human aging derived from omics data

Without personal aging speed measurement, successful preventive interventions for aging-related conditions are impossible (9). Aging biomarkers are molecular, cellular, or physiological parameters of the body demonstrating reproducible quantitative or qualitative changes with age. Multiomics measurement of aging is an analysis of age-related correlations among the big data obtained from the analysis of various omics. The newest way of identifying the biomarkers of human aging utilizes deep neural networks (10). The multiomics gives the possibility to evaluate complex quantitative and phenotypic aging biomarkers. This evaluation is essential due to the weak understanding of the nature of aging and the fact that distinguishing between the causes and effects of aging is difficult. Applying the whole set of omics data in aging biomarker design could create a holistic view of the aging landscape (11). Multiomics gives the possibility of growth to newly emerging companies providing easily accessible express aging clock tests that can be purchased online.

Biomarkers

Aging-associated transcriptome features

Aging is characterized by great changes in the transcriptional profile in absolutely all human tissues. It is possible to sort out six gene expression hallmarks of aging (11):

  1. genes encoding mitochondrial proteins are downregulated
  2. protein synthesis machinery genes are downregulated
  3. genes controlling immune response are deregulated
  4. the signaling cascade of growth factors has a lower expression profile
  5. the DNA damage and stress response genes are activated constantly
  6. transcriptional drift – overall expression profile is deregulated

Genomics of human aging

Genetic contribution to human variation in aging is still questioned, varying from around 15% to 30%. Genome-wide association studies (GWAS) are a powerful approach to examine the genetic architecture of aging in the genome. The increasing number of analyzed genomes of the elderly, particularly centenarians, provides insights into the genetic predisposition of exceptional longevity. Until now, only the APOE (apolipoprotein E), FOXO3 (stress-induced transcription factor forkhead box O3), and 5q33.3 (longevity locus on chromosome 5q33.3) loci are repeatedly connected with longevity across studies. The largest GWAS on human lifespan to date validated seven previously identified loci and proposed five novel genomic regions implicated in lifespan heritability, such as KCNK3 (Potassium channel subfamily K member 3) (12). However, even this study failed to thoroughly explain the heritability of lifespan and longevity. Further investigations using different populations and ethnic groups are needed (13).

Metabolome-based aging biomarkers

The whole metabolome is an informative complex biomarker, mainly when associated with other omics. It is noteworthy that metabolism can simultaneously be a driver and a marker of aging (11). Since the circulating blood collects metabolites from all organs, metabolomic profiling provides integral data of human physiology and age-related degenerative processes. In aging research, the advantages of metabolomics are enhanced sensitivity and predictability to the body's physiological state and the potential quick responsiveness changes in diet, lifestyle, or medicine intake. Fourteen circulating biomarkers were independently associated with all-cause mortality (14). The identified biomarkers are related to lipoprotein and fatty acids metabolism, glycolysis, fluid balance, and inflammation. Most of them, such as lipids, glucose, or albumin, are well-known risk factors for age-related diseases (13).


Epigenomic-based aging biomarkers

Epigenetic modifications such as DNA methylation, histone methylation, and acetylation are highly dynamic processes influenced by environmental and genetic factors. The total DNA methylation level slowly decreases with age. In contrast, cytosine methylation at specific loci containing CpG dinucleotides becomes both hyper- or hypo-methylated in different genomic locations. CpG dinucleotides are places in DNA where a nucleotide cytosine is followed by a guanine in the linear sequence (15). A considerable improvement in biological age measurement allowed to develop well-known epigenetic clocks based on a correlation between the chronological age and methylation status of selected CpG sites. Methylation clocks have been well studied and are among the most accurate chronological age predictors (13, 16). The epigenetic age may respond quickly to anti-aging interventions, offering new validating strategies to delay aging instead of conducting lengthy and expensive longitudinal trials. One of such strategy is the TRIIM (Thymus Re-generation, Immunorestoration, and Insulin Mitigation) treatment, which exhibited epigenetic rejuvenation properties. It reversed the epigenetic age by 2.5 years in the time of one-year of treatment. However, still little is known about which aging mechanisms epigenetic clocks reflect and what is the role of the changes in the epigenome with age (17)(13). 

The proteomic landscape of human aging

Proteins usually directly influence the information transduction in signaling pathways. Thus, they can tell much about the aging process more precisely (11). The number of proteins is at least two orders of magnitudes higher than the number of genes because of alternative splicing and post-translational protein modifications. More than 10 thousands proteins have been identified to date in human plasma. The proteomics study of plasma proteins using SOMAscan (a Slow Off-rate Modified Aptamer) assay discovered 13 proteins associated with chronological age and age-related phenotypes (18). Further validation using RNAseq data gave a total of 11 proteins replicated at the protein level or with consistent association with age at the gene expression level. The strongest association with age was shown for chordin-like protein 1 (CHRDL1), involved in bone morphogenic protein signaling, retinal angiogenesis, and brain plasticity. Mentioned set of 11 proteins does not include broadly accepted aging biomarkers, such as interleukin 6 (IL-6) or C-reactive protein (CRP), which is explainable by insufficient proteome coverage and specificity (13).

The Aging Multiomics Databases

The development of novel multi-omics techniques has resulted in the rapid accumulation of many aging-related datasets providing information on aging. Currently, there are several publicly available databases of aging-specific gene information, including the Human Aging Genomic Resources (HAGR)(19), AgeFactDB (20), and AGEMAP (21). They compile aging phenotypes, longevity records, aging- and longevity-related genes, and lifespan-extending factors (11). Another example is The Aging Atlas database which aims to provide a wide range of life science researchers with valuable resources that allow access to large-scale gene expression and regulation datasets created by various high-throughput omics technologies. Atlas offers user-friendly functionalities for exploring age-related changes in gene expression, as well as free raw data download services (9).

Disadvantages of a multiomic approach

Multiomics approaches are still expensive and require special equipment and qualified personnel. Moreover, the reliability of digital tools is still limited by data quality. Obvious problems occur with the collection and verification of reliable big medical data. Inaccurate data sources, problems with normalization or particular sampling features lead to the situation when algorithms trained on one data type do not fit other independent samples or make irreproducible predictions. The research platforms and bioinformatics approaches for processing extensive omics data are not unified. There is a difficult challenge of big data compatibility obtained by different techniques (8, 11).

Conclusions

Aging is a complex process occurring at all levels of the organization of biological systems. That is why every new anti-aging intervention implemented in the clinical practice needs a multidimensional systemic approach to measuring the patient's rate of aging and biological age. Original multiomics studies have begun to appear regularly and become a rich database for new aging biomarkers (11). The multiomics approach gives the most comprehensive and complex insight into age-related diseases or aging and can be personalized. Unfortunately, it still has many disadvantages and limitations that require further investigation of available multiomics datasets and new algorithms testing. 

References

1.            Dong Z, Chen Y. Transcriptomics: advances and approaches. Sci China Life Sci. 2013;56(10):960-7.

2.            Vailati-Riboni M, Palombo V, Loor JJ. What Are Omics Sciences? In: Ametaj BN, editor. Periparturient Diseases of Dairy Cows: A Systems Biology Approach. Cham: Springer International Publishing; 2017. p. 1-7.

3.            Aon MA, Bernier M, Mitchell SJ, Di Germanio C, Mattison JA, Ehrlich MR, et al. Untangling determinants of enhanced health and lifespan through a multi-omics approach in mice. Cell metabolism. 2020;32(1):100-16. e4.

4.            Leon-Mimila P, Wang J, Huertas-Vazquez A. Relevance of multi-omics studies in cardiovascular diseases. Frontiers in cardiovascular medicine. 2019;6:91.

5.            Currais A, Goldberg J, Farrokhi C, Chang M, Prior M, Dargusch R, et al. A comprehensive multiomics approach toward understanding the relationship between aging and dementia. Aging (Albany NY). 2015;7(11):937-55.

6.            Faulkner A, Dang Z, Avolio E, Thomas AC, Batstone T, Lloyd GR, et al. Multi-omics analysis of diabetic heart disease in the db/db model reveals potential targets for treatment by a longevity-associated gene. Cells. 2020;9(5):1283.

7.            Dayan IE, Arga KY, Ulgen KO. Multiomics Approach to Novel Therapeutic Targets for Cancer and Aging-Related Diseases: Role of Sld7 in Yeast Aging Network. OMICS. 2017;21(2):100-13.

8.            Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome Biol. 2017;18(1):83.

9.            Aging Atlas C. Aging Atlas: a multi-omics database for aging biology. Nucleic acids research. 2021;49(D1):D825-D30.

10.          Putin E, Mamoshina P, Aliper A, Korzinkin M, Moskalev A, Kolosov A, et al. Deep biomarkers of human aging: Application of deep neural networks to biomarker development. Aging. 2016;8(5):1021-33.

11.          Solovev I, Shaposhnikov M, Moskalev A. Multi-omics approaches to human biological age estimation. Mechanisms of Ageing and Development. 2020;185:111192.

12.          Timmers PR, Mounier N, Lall K, Fischer K, Ning Z, Feng X, et al. Genomics of 1 million parent lifespans implicates novel pathways and common diseases and distinguishes survival chances. Elife. 2019;8.

13.          Kudryashova KS, Burka K, Kulaga AY, Vorobyeva NS, Kennedy BK. Aging Biomarkers: From Functional Tests to Multi‐Omics Approaches. Proteomics. 2020;20(5-6):1900408.

14.          Deelen J, Kettunen J, Fischer K, van der Spek A, Trompet S, Kastenmüller G, et al. A metabolic profile of all-cause mortality risk identified in an observational study of 44,168 individuals. Nature Communications. 2019;10(1):3346.

15.          Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, et al. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 2010;20(4):440-6.

16.          Horvath S, Raj K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nature Reviews Genetics. 2018;19(6):371-84.

17.          Fahy GM, Brooke RT, Watson JP, Good Z, Vasanawala SS, Maecker H, et al. Reversal of epigenetic aging and immunosenescent trends in humans. Aging Cell. 2019;18(6):e13028.

18.          Lehallier B, Gate D, Schaum N, Nanasi T, Lee SE, Yousef H, et al. Undulating changes in human plasma proteome profiles across the lifespan. Nature medicine. 2019;25(12):1843-50.

19.          De Magalhaes JP, Budovsky A, Lehmann G, Costa J, Li Y, Fraifeld V, et al. The Human Ageing Genomic Resources: online databases and tools for biogerontologists. Aging cell. 2009;8(1):65-72.

20.          Hühne R, Thalheim T, Sühnel J. AgeFactDB—the JenAge Ageing Factor Database—towards data integration in ageing research. Nucleic acids research. 2014;42(D1):D892-D6.

21.          Zahn JM, Poosala S, Owen AB, Ingram DK, Lustig A, Carter A, et al. AGEMAP: a gene expression database for aging in mice. PLoS genetics. 2007;3(11):e201.


Highlights:

  • Multiomics analyses data are integrated from multiple specific fields or "omes" like genome, transcriptome, and proteome
  • Biomarkers of human aging derived from omics data could be used for personal aging speed measurement
  • Multiomics is still a costly approach in need of upgrade and standardization 

Introduction

Aging is a complex process influenced by external factors like environment, lifestyle as well as our internal biology, genes, epigenetic changes, metabolic regulation, interactions between host and microbiome, and others. Biological processes, such as the development of diseases and aging, depend on a dynamic and interactive system of molecular layers such as genetics, proteomics, or transcriptomics that, taken together, can be called multiomics. The analysis of high-throughput multiomics datasets can provide a complex and integrated profile of the multifactorial aging process in detail never available before. Multiomics strategies let scientists explore the molecular profile and regulatory status during aging and disease processes, leading to the discovery of new interventions.

The science of omics

The word omics refers to a field of biological sciences study ending with -omics: genomics, transcriptomics, proteomics, metabolomics, and others. The suffix -ome forms nouns denoting fields of a specified nature and addresses the objects of study of such fields as:

  • Genome - an organism's complete set of genetic information. Each genome contains all of the genes needed to build, grow and develop an organism
  • Transcriptome - the complete set of RNA transcripts in a specific cell type or tissue at a particular developmental stage, including messenger RNA (mRNA), transfer RNA (tRNA), ribosomal RNA (rRNA), and the non-coding RNAs (1)
  • Proteome - the sum of all the proteins in a cell, tissue, or organism.
  • Metabolome - the complete set of metabolites in the cell, tissue, organ, or organism, which are the end products of cellular processes (2)
  • Epigenome - all modifications to the genome that do not affect the DNA sequence but determine the gene expression (2)

The omics sciences identify, characterize, and quantify all biological molecules involved in the structure, function, and dynamics of a cell, tissue, or whole organism. The multiomics analysis is used in different fields of bioscience (3). It helps to understand age-related diseases such as cardiovascular disease (4), dementia (5), diabetes (6), and cancer (7), as well as aging itself. The multiomics approach provides the understanding of hidden associations and pathways which may be critical for both patients and specialists (8).

 

Biomarkers of human aging derived from omics data

Without personal aging speed measurement, successful preventive interventions for aging-related conditions are impossible (9). Aging biomarkers are molecular, cellular, or physiological parameters of the body demonstrating reproducible quantitative or qualitative changes with age. Multiomics measurement of aging is an analysis of age-related correlations among the big data obtained from the analysis of various omics. The newest way of identifying the biomarkers of human aging utilizes deep neural networks (10). The multiomics gives the possibility to evaluate complex quantitative and phenotypic aging biomarkers. This evaluation is essential due to the weak understanding of the nature of aging and the fact that distinguishing between the causes and effects of aging is difficult. Applying the whole set of omics data in aging biomarker design could create a holistic view of the aging landscape (11). Multiomics gives the possibility of growth to newly emerging companies providing easily accessible express aging clock tests that can be purchased online.

Biomarkers

Aging-associated transcriptome features

Aging is characterized by great changes in the transcriptional profile in absolutely all human tissues. It is possible to sort out six gene expression hallmarks of aging (11):

  1. genes encoding mitochondrial proteins are downregulated
  2. protein synthesis machinery genes are downregulated
  3. genes controlling immune response are deregulated
  4. the signaling cascade of growth factors has a lower expression profile
  5. the DNA damage and stress response genes are activated constantly
  6. transcriptional drift – overall expression profile is deregulated

Genomics of human aging

Genetic contribution to human variation in aging is still questioned, varying from around 15% to 30%. Genome-wide association studies (GWAS) are a powerful approach to examine the genetic architecture of aging in the genome. The increasing number of analyzed genomes of the elderly, particularly centenarians, provides insights into the genetic predisposition of exceptional longevity. Until now, only the APOE (apolipoprotein E), FOXO3 (stress-induced transcription factor forkhead box O3), and 5q33.3 (longevity locus on chromosome 5q33.3) loci are repeatedly connected with longevity across studies. The largest GWAS on human lifespan to date validated seven previously identified loci and proposed five novel genomic regions implicated in lifespan heritability, such as KCNK3 (Potassium channel subfamily K member 3) (12). However, even this study failed to thoroughly explain the heritability of lifespan and longevity. Further investigations using different populations and ethnic groups are needed (13).

Metabolome-based aging biomarkers

The whole metabolome is an informative complex biomarker, mainly when associated with other omics. It is noteworthy that metabolism can simultaneously be a driver and a marker of aging (11). Since the circulating blood collects metabolites from all organs, metabolomic profiling provides integral data of human physiology and age-related degenerative processes. In aging research, the advantages of metabolomics are enhanced sensitivity and predictability to the body's physiological state and the potential quick responsiveness changes in diet, lifestyle, or medicine intake. Fourteen circulating biomarkers were independently associated with all-cause mortality (14). The identified biomarkers are related to lipoprotein and fatty acids metabolism, glycolysis, fluid balance, and inflammation. Most of them, such as lipids, glucose, or albumin, are well-known risk factors for age-related diseases (13).


Epigenomic-based aging biomarkers

Epigenetic modifications such as DNA methylation, histone methylation, and acetylation are highly dynamic processes influenced by environmental and genetic factors. The total DNA methylation level slowly decreases with age. In contrast, cytosine methylation at specific loci containing CpG dinucleotides becomes both hyper- or hypo-methylated in different genomic locations. CpG dinucleotides are places in DNA where a nucleotide cytosine is followed by a guanine in the linear sequence (15). A considerable improvement in biological age measurement allowed to develop well-known epigenetic clocks based on a correlation between the chronological age and methylation status of selected CpG sites. Methylation clocks have been well studied and are among the most accurate chronological age predictors (13, 16). The epigenetic age may respond quickly to anti-aging interventions, offering new validating strategies to delay aging instead of conducting lengthy and expensive longitudinal trials. One of such strategy is the TRIIM (Thymus Re-generation, Immunorestoration, and Insulin Mitigation) treatment, which exhibited epigenetic rejuvenation properties. It reversed the epigenetic age by 2.5 years in the time of one-year of treatment. However, still little is known about which aging mechanisms epigenetic clocks reflect and what is the role of the changes in the epigenome with age (17)(13). 

The proteomic landscape of human aging

Proteins usually directly influence the information transduction in signaling pathways. Thus, they can tell much about the aging process more precisely (11). The number of proteins is at least two orders of magnitudes higher than the number of genes because of alternative splicing and post-translational protein modifications. More than 10 thousands proteins have been identified to date in human plasma. The proteomics study of plasma proteins using SOMAscan (a Slow Off-rate Modified Aptamer) assay discovered 13 proteins associated with chronological age and age-related phenotypes (18). Further validation using RNAseq data gave a total of 11 proteins replicated at the protein level or with consistent association with age at the gene expression level. The strongest association with age was shown for chordin-like protein 1 (CHRDL1), involved in bone morphogenic protein signaling, retinal angiogenesis, and brain plasticity. Mentioned set of 11 proteins does not include broadly accepted aging biomarkers, such as interleukin 6 (IL-6) or C-reactive protein (CRP), which is explainable by insufficient proteome coverage and specificity (13).

The Aging Multiomics Databases

The development of novel multi-omics techniques has resulted in the rapid accumulation of many aging-related datasets providing information on aging. Currently, there are several publicly available databases of aging-specific gene information, including the Human Aging Genomic Resources (HAGR)(19), AgeFactDB (20), and AGEMAP (21). They compile aging phenotypes, longevity records, aging- and longevity-related genes, and lifespan-extending factors (11). Another example is The Aging Atlas database which aims to provide a wide range of life science researchers with valuable resources that allow access to large-scale gene expression and regulation datasets created by various high-throughput omics technologies. Atlas offers user-friendly functionalities for exploring age-related changes in gene expression, as well as free raw data download services (9).

Disadvantages of a multiomic approach

Multiomics approaches are still expensive and require special equipment and qualified personnel. Moreover, the reliability of digital tools is still limited by data quality. Obvious problems occur with the collection and verification of reliable big medical data. Inaccurate data sources, problems with normalization or particular sampling features lead to the situation when algorithms trained on one data type do not fit other independent samples or make irreproducible predictions. The research platforms and bioinformatics approaches for processing extensive omics data are not unified. There is a difficult challenge of big data compatibility obtained by different techniques (8, 11).

Conclusions

Aging is a complex process occurring at all levels of the organization of biological systems. That is why every new anti-aging intervention implemented in the clinical practice needs a multidimensional systemic approach to measuring the patient's rate of aging and biological age. Original multiomics studies have begun to appear regularly and become a rich database for new aging biomarkers (11). The multiomics approach gives the most comprehensive and complex insight into age-related diseases or aging and can be personalized. Unfortunately, it still has many disadvantages and limitations that require further investigation of available multiomics datasets and new algorithms testing. 

References

1.            Dong Z, Chen Y. Transcriptomics: advances and approaches. Sci China Life Sci. 2013;56(10):960-7.

2.            Vailati-Riboni M, Palombo V, Loor JJ. What Are Omics Sciences? In: Ametaj BN, editor. Periparturient Diseases of Dairy Cows: A Systems Biology Approach. Cham: Springer International Publishing; 2017. p. 1-7.

3.            Aon MA, Bernier M, Mitchell SJ, Di Germanio C, Mattison JA, Ehrlich MR, et al. Untangling determinants of enhanced health and lifespan through a multi-omics approach in mice. Cell metabolism. 2020;32(1):100-16. e4.

4.            Leon-Mimila P, Wang J, Huertas-Vazquez A. Relevance of multi-omics studies in cardiovascular diseases. Frontiers in cardiovascular medicine. 2019;6:91.

5.            Currais A, Goldberg J, Farrokhi C, Chang M, Prior M, Dargusch R, et al. A comprehensive multiomics approach toward understanding the relationship between aging and dementia. Aging (Albany NY). 2015;7(11):937-55.

6.            Faulkner A, Dang Z, Avolio E, Thomas AC, Batstone T, Lloyd GR, et al. Multi-omics analysis of diabetic heart disease in the db/db model reveals potential targets for treatment by a longevity-associated gene. Cells. 2020;9(5):1283.

7.            Dayan IE, Arga KY, Ulgen KO. Multiomics Approach to Novel Therapeutic Targets for Cancer and Aging-Related Diseases: Role of Sld7 in Yeast Aging Network. OMICS. 2017;21(2):100-13.

8.            Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome Biol. 2017;18(1):83.

9.            Aging Atlas C. Aging Atlas: a multi-omics database for aging biology. Nucleic acids research. 2021;49(D1):D825-D30.

10.          Putin E, Mamoshina P, Aliper A, Korzinkin M, Moskalev A, Kolosov A, et al. Deep biomarkers of human aging: Application of deep neural networks to biomarker development. Aging. 2016;8(5):1021-33.

11.          Solovev I, Shaposhnikov M, Moskalev A. Multi-omics approaches to human biological age estimation. Mechanisms of Ageing and Development. 2020;185:111192.

12.          Timmers PR, Mounier N, Lall K, Fischer K, Ning Z, Feng X, et al. Genomics of 1 million parent lifespans implicates novel pathways and common diseases and distinguishes survival chances. Elife. 2019;8.

13.          Kudryashova KS, Burka K, Kulaga AY, Vorobyeva NS, Kennedy BK. Aging Biomarkers: From Functional Tests to Multi‐Omics Approaches. Proteomics. 2020;20(5-6):1900408.

14.          Deelen J, Kettunen J, Fischer K, van der Spek A, Trompet S, Kastenmüller G, et al. A metabolic profile of all-cause mortality risk identified in an observational study of 44,168 individuals. Nature Communications. 2019;10(1):3346.

15.          Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, et al. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 2010;20(4):440-6.

16.          Horvath S, Raj K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nature Reviews Genetics. 2018;19(6):371-84.

17.          Fahy GM, Brooke RT, Watson JP, Good Z, Vasanawala SS, Maecker H, et al. Reversal of epigenetic aging and immunosenescent trends in humans. Aging Cell. 2019;18(6):e13028.

18.          Lehallier B, Gate D, Schaum N, Nanasi T, Lee SE, Yousef H, et al. Undulating changes in human plasma proteome profiles across the lifespan. Nature medicine. 2019;25(12):1843-50.

19.          De Magalhaes JP, Budovsky A, Lehmann G, Costa J, Li Y, Fraifeld V, et al. The Human Ageing Genomic Resources: online databases and tools for biogerontologists. Aging cell. 2009;8(1):65-72.

20.          Hühne R, Thalheim T, Sühnel J. AgeFactDB—the JenAge Ageing Factor Database—towards data integration in ageing research. Nucleic acids research. 2014;42(D1):D892-D6.

21.          Zahn JM, Poosala S, Owen AB, Ingram DK, Lustig A, Carter A, et al. AGEMAP: a gene expression database for aging in mice. PLoS genetics. 2007;3(11):e201.


Article reviewed by
Dr. Ana Baroni MD. Ph.D.
SCIENTIFIC & MEDICAL ADVISOR
Quality Garant
Close

Dr. Ana Baroni MD. Ph.D.

Scientific & Medical Advisor
Quality Garant

Ana has over 20 years of consultancy experience in longevity, regenerative and precision medicine. She has a multifaceted understanding of genomics, molecular biology, clinical biochemistry, nutrition, aging markers, hormones and physical training. This background allows her to bridge the gap between longevity basic sciences and evidence-based real interventions, putting them into the clinic, to enhance the healthy aging of people. She is co-founder of Origen.life, and Longevityzone. Board member at Breath of Health, BioOx and American Board of Clinical Nutrition. She is Director of International Medical Education of the American College of Integrative Medicine, Professor in IL3 Master of Longevity at Barcelona University and Professor of Nutrigenomics in Nutrition Grade in UNIR University.

DISCOVER
HealthyLongevity.guide
4.6 / 5
Professional science-based education
250+ Articles, video lectures, webinars
Community of 1000+ verified professionals
Sign Up

Read the latest articles

News
HealthyLongevity.guide

Longevity Conferences 2023

January 19, 2023

Curated list of Longevity Conferences, where you can explore the latest research and developments in the field of aging and longevity.

Ehab Naim, MBA.
News
Body
Diagnostics
Medicine

Vascular endothelial senescence: Exploring current and future diagnostic and therapeutic potential

January 18, 2023

Researchers examined endothelial dysfunction, and identified its causes and effects. They evaluated the use of senotherapeutics in fighting age-related ailments.

Ehab Naim, MBA.
News
Nutrition

A Controlled Trial: Eating Almonds May Help in Reducing Obesity

January 4, 2023

Almond consumption might affect appetite and reduce obesity. Researchers analyzed the blood of obese people after eating almonds to find out more.

Reem Abedi
News
Technology
Lifestyle

Physical activity trackers: Shaping behavior to promote healthy aging among older adults

December 29, 2022

Wearing physical activity tracking devices can promote healthy aging, improve population's quality of life, and reduce the risk of non-communicable diseases.

Ehab Naim, MBA.
News
Body
Longevity

Gut microbiota taxa analysis could offer a better definition of a healthy microbiome

December 26, 2022

One of the drivers behind the age-related decline is the gut microbiome. In their study, Ghosh et al. analyzed over 21,000 microbiome profiles from seven databases across five continents.

Ehab Naim, MBA.
News
Nutrition

Flavonols consumption from fruit and vegetables delays cognitive decline

December 23, 2022

Flavonols are a type of flavonoids that slow down the decline in episodic and semantic memory, perceptual speed, and working memory. A study anaylzed what exactly is their impact on cognitive health.

Agnieszka Szmitkowska, Ph.D.
News
Disease

Saffron Combined with Aerobic Exercise to address Type 2 Diabetes Mellitus

December 21, 2022

Rajabi et al. conducted extensive research on type-2 diabetes patients to investigate the effect of aerobic training and Saffron supplementation for two months.

Reem Abedi
Article
Body
Lifestyle

Which Sport is Best for Longevity?

December 20, 2022

Researchers evaluated which sport is best for longevity. Key components of highly beneficial sports are including a social aspect, engaging both arms and legs, or including whole-body movements.

Jiří Kaloč
News
Aging
Lifestyle
Prevention

Simple lifestyle modifications could reduce the risk of dementia

December 16, 2022

American Heart Association defined ideal values and levels for seven modifiable factors that directly affect cardiovascular health. They include physical activity, smoking, or fasting plasma glucose.

Ehab Naim, MBA.
Video
Lifestyle
Longevity

NATURE EXPOSURE & LONGEVITY (Webinar with Sarah Nielson follow-up + recording)

December 15, 2022

Sarah Nielsen explained the impact of nature exposure on heart rate and blood pressure; how it affects cortisol, inflammation, or anti-cancer proteins; and what you can recommend to your clients.

Reem Abedi
News
No Tag Added

Lower birth weight vs. cardiovascular disease in adulthood

December 15, 2022

A recent article published in the Heart journal demonstrates a connection between lower birth weight, the incidence of myocardial infarction, and adverse left ventricular remodeling.

Agnieszka Szmitkowska, Ph.D.
Article
Lifestyle
Prevention
Aging
Longevity
Nutrition

Key Blue Zones patterns could help with physician burnout

December 6, 2022

There are five areas on Earth where people live significantly longer and disease-free into their late years. What makes them so special? People who live there follow nine simple rules.

Agnieszka Szmitkowska, Ph.D.
News
Medicine
Prevention

Daylight saving time (DST) and mortality patterns in Europe

December 5, 2022

Researchers examined whether daylight saving time affects European mortality patterns. They compared the daily death rates (DDR) for 2 months prior to and after each DST transition.

Reem Abedi
News
Disease

Prostaglandin E2 potentially increases susceptibility to influenza A infection in the elderly

November 30, 2022

A new study tested whether age-related elevation in Prostaglandin E2 is a driver that impairs host defense against influenza.

Ehab Naim, MBA.
Article
Lifestyle
Prevention

Future healthy longevity starts at conception

November 29, 2022

The habits we develop as children significantly impact lifespan and healthspan in adulthood. Dietary choices, exercise, or for example daily screen time can lead to lasting changes in the organism.

Agnieszka Szmitkowska, Ph.D.
Article
No Tag Added

Every move counts: Non-exercise physical activity for cardiovascular health and longevity

December 13, 2022

Increasing movement and reducing sedentary time lead to significant reductions in the occurrence of many diseases. It is important to encourage people to increase their non-exercise physical activity.

Reem Abedi
No items found.