Epigenetic Signatures of Aging: A Comprehensive Study of Biomarker Discovery

Abstract

Background: Aging is a complex biological process that is associated with a decline in physiological functions and an increased risk of age-related diseases. Despite advances in molecular biology and genetics, the underlying mechanisms of aging remain largely unknown. Study: The identification of biomarkers of aging would provide a powerful tool for monitoring the effects of aging and for developing interventions to improve healthspan. Aging is associated with alterations in genetics, epigenetic marks, telomere shortening, cell senescence, and changes in the expression of genes involved in metabolism, inflammation, and DNA damage repair. Epigenetic changes, including modifications to DNA methylation and histone acetylation patterns, play a critical role in the aging process. As we age, these changes can lead to altered gene expression and contribute to the development of age-related diseases such as cancer, Alzheimer’s disease (AD) and cardiovascular disease (CVD). Conclusion: The discovery of aging biomarkers that are sensitive to these epigenetic changes has the potential to revolutionize our understanding of the aging process and inform the development of interventions to improve healthspan and extend lifespan.

Share and Cite:

Lee, M. , Kwon, H. , Kim, Y. , Min, N. , Lee, S. and Lee, I. (2023) Epigenetic Signatures of Aging: A Comprehensive Study of Biomarker Discovery. Advances in Aging Research, 12, 11-38. doi: 10.4236/aar.2023.122002.

1. Introduction

Aging is the gradual accumulation of cellular and molecular changes over time that lead to the decline of physiological functions and an increased risk of age-related diseases [1]. Recent aging research has focused on understanding the underlying mechanisms of aging and developing interventions that can extend a healthy lifespan [2] [3]. Aging biological studies are a field of research focused on understanding the underlying mechanisms of aging and developing interventions to improve health and extend lifespan [4] [5]. This interdisciplinary field encompasses genetics, molecular biology, biochemistry, and physiology, and involves the study of changes in cells, tissues, and organisms over time [6]. The primary aim of aging biological studies is to uncover the molecular and cellular processes that contribute to aging and age-related diseases, such as cardiovascular disease [7], cancer [8], and neurodegeneration [9]. Researchers use a variety of approaches, including genetic screens [10], epigenetic changes [11], transcriptomics [12], proteomics [13], and functional assays [14] to study changes in gene expression and regulation, protein levels, and cellular behavior that occur with age. Another key aspect of aging biological studies is the development of interventions to delay the onset of aging and age-related diseases [2] [15]. These interventions may include the use of drugs that target specific molecular pathways, dietary and lifestyle changes, and the application of regenerative medicine approaches, such as stem cell therapies [16]. Overall, the goal of aging biological studies is to improve the health and quality of life of individuals as they age and to extend the human lifespan. The findings of these studies have the potential to have a significant impact on public health and the global economy by reducing the burden of age-related diseases and improving the health of aging populations.

2. Aging Biological Studies

Aging biological studies are a field of research focused on understanding the underlying mechanisms of aging and developing interventions to improve health and extend lifespan [17]. Aging is a biological process characterized by a gradual decline in physiological function and increased vulnerability to disease and death overtime [18]. The exact mechanisms underlying aging are not fully understood, but it is thought to result from a combination of genetic lifestyle and environmental factors. Studies have investigated the role of biological process, lifestyle factors, and environmental exposures in aging and age-related diseases [19] [20]. Various studies have focused on understanding the molecular basis of biological processes in aging, providing important insights into the underlying mechanisms of aging. These studies have led to the identification of potential targets for interventions that reduce the risk of certain age-related diseases and can potentially extend healthy lifespan [21] [22]. Some of the most promising areas of aging research in biological process are genetics, epigenetics, senolytics, telomere, metabolism, immune system, and neurodegeneration.

Recently there has been a growing interest in the study of DNA epigenetic changes, which is a process that modifies the activity of genes without changing the underlying DNA sequence. Epigenetic changes, including DNA methylation and histone modifications, play an important role in aging [23] [24]. These changes can affect gene expression and cellular function, leading to the decline of physiological functions and an increased risk of age-related diseases (Figure 1). The field of epigenetics has seen significant technological advances in recent years, allowing for more comprehensive and in-depth studies of the changes in gene expression histone modification and DNA methylation patterns that occur with aging and contribute to disease development. Some of the key technological advances in this field of next-generation sequencing (NGS) [25] and microarray technologies [26] that allow for high-throughput and cost-effective analysis of large-scale epigenetic data include bisulfite sequencing [27], methylation-specific restriction enzyme sequencing (MSRE-seq) [28], chromatin immunoprecipitation sequencing (ChIP-seq) [29], RNA sequencing (RNA-seq) [30], assay for transposase-accessible chromatin sequencing (ATAC-seq) [31] and methylation microarrays (Figure 2). These technologies enable the identification of changes in epigenetic marks, such as DNA methylation and histone modifications that are associated with aging and age-related disease. These technologies, combined with new computational and data analysis tools, have allowed for major advances in our understanding of the role of epigenetics in aging and disease, and the discovery of novel biomarkers [32] [33].

Figure 1. The illustration of epigenetic changes contributing in aging and age-related diseases. The epigenetic changes are caused by any of the three factors i.e. they could be inherited, caused by environment, or lifestyle.

Figure 2. Different molecular technologies to understand the mechanisms of aging and its contribution to different diseases.

These biomarkers can be used for early detection, diagnosis and prognosis of age-related disease, and measure biological aging as an indicator of health and disease status. The discovery of these biomarkers also allows the development of therapeutic targets for a new drug and a novel therapy to slow down aging or reduce the risk of certain diseases that are associated with aging process.

3. Biological Cause of Aging

There are several theories about the underlying mechanisms of aging, including genetics, telomere shortening, cell senescence, epigenetic changes, inflammation, oxidative stress and mitochondrial dysfunctions. Aging is regulated by genetic pathways that govern cellular metabolism and stress resistance. Some of the key genetic pathways thought to play a role in aging include SIRT1 [34], TERT genes [35], and the insulin/IGF-1 signaling pathway [36], the mTOR pathway [37], and the FOXO pathway [38]. Telomeres are the protective caps on the ends of our chromosomes that shorten as we age, which can lead to cellular aging and an increased risk of age-related diseases [39]. Cell senescence is a state of a cell that has ceased to divide and is no longer able to replicate. In aging research, cellular senescence is considered an important contributor to the aging process [40]. Epigenetic changes, such as DNA methylation and histone modifications, can influence gene expression and contribute to aging [41]. Other areas of biological aging research are also in Oxidative stress [42], Inflammation [43] and Mitochondrial dysfunctions [44] : Oxidative stress is caused by the accumulation of cellular damage from free radicals, which are unstable molecules that can damage cells and lead to oxidative stress [45]. Chronic low-grade inflammation has been linked to aging and age-related diseases, including cardiovascular disease, diabetes, and certain cancers [46]. The mitochondria are the cellular powerhouses, and can become damaged over time, leading to decreased energy production and an increased risk of age-related diseases [44] (Figure 3). Despite the many challenges associated with aging research, scientists continue to work towards a better understanding of the underlying biological causes of aging and the development of interventions to improve health and extend lifespan.

4. Genetics in Aging

One area of research in molecular biology is the study of the genetic basis of aging. Studies have identified a number of genes that play a role in aging and age-related diseases [47]. Many genes have been identified that are associated with aging and age-related diseases. One of the most well-known genes associated

Figure 3. Different factors that contribute to the underlying aging process.

with aging is the SIRT1 gene, which encodes for a protein called sirtuin1. This protein is involved in regulating cellular metabolism, DNA repair, and cell survival [48]. Studies have shown that increasing SIRT1 activity can extend lifespan in animals and protect against age-related diseases [49]. Another gene associated with aging is the FOXO3 gene. This gene encodes for a protein called Forkhead box O3, which regulates the expression of other genes involved in stress response, insulin signaling, and DNA repair [50]. Studies have shown that individuals with a particular variant of the FOXO3 gene have a longer lifespan and a reduced risk of age-related diseases [51]. Another gene that has been associated with aging is the telomerase reverse transcriptase (TERT) gene. TERT is a protein that is responsible for telomerase activity, and it is known that telomerase activity is associated with aging, as telomeres shorten with age. Studies have shown that mutations in the TERT gene can lead to telomere shortening and an increased risk of age-related diseases [52]. There are many other genes that have been associated with aging, including the p53 gene, which is involved in DNA repair and cell survival, and the mTOR gene, which regulates cell growth and metabolism [53]. It’s worth noting that while these genes have been associated with aging, they don’t determine the aging process solely.

5. Epigenetics in Aging

The study of epigenetic changes in aging is an important area of research that has the potential to provide new insights into the underlying mechanisms of aging and to develop interventions that can delay or prevent age-related diseases [54]. Epigenetics is a field of study that examines how changes in gene expression and activity are regulated, without altering the underlying DNA sequence. In aging research, epigenetic changes are increasingly recognized as key contributors to the aging process [55]. Age-related changes in epigenetic marks, such as DNA methylation, and histone modification have been implicated in the regulation of gene expression patterns that are associated with aging, such as changes in stem cell function, oxidative stress, inflammation, and DNA damage [56]. Epigenetic changes can also affect the regulation of telomere length, a hallmark of aging, and contribute to age-related diseases such as cancer and neurodegenerative diseases.

6. DNA Methylation in Aging

DNA methylation patterns change with age and are thought to play a role in aging and age-related diseases [57]. DNA methylation is a type of epigenetic modification that occurs throughout the genome, and it plays an important role in regulating gene expression. Methylation involves the addition of methyl groups to the DNA molecule, and it can affect the accessibility of the genetic material to the cell’s machinery responsible for transcribing DNA into RNA [58] [59]. Research has shown that changes in DNA methylation patterns occur as a part of the aging process. With aging, there are global reductions in DNA methylation levels, as well as changes in the methylation patterns of specific genes [60] [61]. Some studies have suggested that these changes in methylation patterns may contribute to the development of age-related diseases such as cancer, cardiovascular disease, and neurodegenerative disease [62]. In addition, certain genes have been identified that are particularly vulnerable to age-associated changes in methylation. For example, the promoter regions of certain tumor suppressor genes, such as p16INK4a, have been found to be highly methylated in aging cells, leading to a reduction in the expression of these genes and an increased risk of cancer [63] [64]. One area of research is the study of epigenetic clock, which is a set of DNA methylation markers that can be used to predict chronological age. Studies have found that these markers are highly correlated with age, and that they change in a consistent way as people age [65] [66]. Epigenetic clock based on DNA methylation markers has been found to be associated with age-related diseases [67]. Another area of research is the study of how environmental and lifestyle factors affect DNA methylation patterns. Studies have shown that factors such as diet, exercise, stress, and exposure to pollutants can affect DNA methylation patterns and contribute to aging and age-related diseases [68]. Additionally, research is ongoing on the use of interventions that can slow or reverse DNA methylation changes associated with aging [69]. Some studies have shown that certain drugs, such as methylation inhibitors, can slow or reverse these changes, potentially leading to new therapies for aging and age-related diseases [70].

7. Histone Acetylation in Aging

Histone acetylation is an epigenetic mechanism that regulates gene expression and plays an important role in aging [71]. Histones are the proteins around which DNA is wrapped to form chromosomes, and acetylation of histones is associated with increased gene expression, whereas deacetylation of histones is associated with decreased gene expression [72] [73]. In aging, changes in histone acetylation patterns have been observed, with decreased histone acetylation levels in some regions of the genome and increased histone acetylation levels in others. These changes are thought to contribute to aging-associated changes in gene expression, leading to the development of age-related diseases such as Alzheimer’s disease, cardiovascular disease, cancer and others [74] [75]. To target histone acetylation in aging, drugs known as histone deacetylase inhibitors (HDAC inhibitors) have been developed [76] [77]. These drugs increase histone acetylation levels and are being investigated as potential therapies for a range of age-related diseases, including Alzheimer’s disease and cancer [78] [79].

8. Telomeres Shortening in Aging

Telomeres shorten as cells divide and age, and this shortening is thought to play a role in the aging of cells and the development of age-related diseases [80]. Research on telomerase in aging has focused on understanding how telomerase activity affects the aging process and identifying ways to modulate telomerase activity to extend healthy lifespan [81]. Telomerase is an enzyme that adds repetitive DNA sequences called telomeres to the ends of chromosomes [82]. Studies have shown that cells with high levels of telomerase activity, known as telomerase-positive cells, have a greater capacity for proliferation and are less likely to undergo senescence or apoptosis [83]. One area of research is on telomerase activation as a potential anti-aging therapy. Studies have shown that activating telomerase in certain cells can increase their proliferation and delay the onset of senescence. Some studies in animals showed that activating telomerase in certain tissues can extend lifespan and improve health [84] [85]. Another area of research is on telomerase inhibition as a cancer therapy [86]. Cancer cells have been found to have high levels of telomerase activity, allowing them to divide and grow indefinitely. Inhibiting telomerase activity in cancer cells can induce cell death and may be a promising strategy for cancer treatment [86] [87]. One area of research is the study of telomere length in cfDNA [88]. Studies have shown that telomere length in cfDNA decreases with age and that shorter telomeres in cfDNA are associated with an increased risk of age-related diseases [89]. Overall, telomerase research is providing new insights into the underlying mechanisms of aging and is helping to identify potential targets for interventions that can extend healthy lifespan.

9. Cell Senescence in Aging

Another area of research is the study of senescence, a state where cells stop dividing and enter a phase of permanent growth arrest [90]. Senescent cells release a variety of factors that promote inflammation, oxidative stress, and other changes in the surrounding tissues. This can lead to further damage to cells, tissues, and organs, contributing to the progression of age-related diseases [91]. Research in senescence has led to the development of drugs called senolytics, which are designed to target and eliminate senescent cells, thus slowing down aging. Senolytic drugs are a class of drugs that are designed to target and eliminate senescent cells, which are cells that have stopped dividing and can contribute to aging and age-related diseases [92] [93]. These drugs work by inducing apoptosis, or programmed cell death, in senescent cells. Senescent cells are thought to contribute to aging by secreting pro-inflammatory factors and other molecules that can damage surrounding cells and tissues [94]. By eliminating senescent cells, senolytic drugs can reduce inflammation and improve tissue function.

10. Antiaging Medication

There are currently no FDA-approved medications specifically marketed as “anti-aging” drugs. However, certain medications have been shown to have potential anti-aging effects such as caloric restriction mimetics [95], growth hormone receptor antagonists [96], sirtuin activators [97], and mTOR inhibitors [98]. These drugs target processes and pathways associated with aging, such as oxidative stress, inflammation, and cellular senescence. Caloric restriction mimetics are compounds or interventions that mimic the effects of caloric restriction, which is a reduction in caloric intake without malnutrition [95]. Caloric restriction has been shown to extend lifespan and improve health in many species, including yeast, worms, flies, rodents, and primates [99] [100]. However, caloric restriction is not a practical or feasible approach for most people, so researchers are looking for ways to achieve similar benefits through other means. Caloric restriction mimetics have been developed to target various biological pathways involved in aging, such as insulin/IGF-1 signaling, sirtuins, and AMP-activated protein kinase (AMPK). Some examples of caloric restriction mimetics include rapamycin, resveratrol, metformin, and spermidine [100]. Growth hormone receptor (GHR) antagonists are drugs that target the growth hormone (GH) receptor and block its signaling. They have been studied for their potential anti-aging effects as GH signaling has been linked to the aging process and age-related diseases [96]. Some studies have shown that blocking GH signaling with GHR antagonists can extend lifespan and improve healthspan in animal models [101] [102]. Sirtuin activators are a class of compounds that are thought to promote longevity and delay aging by activating sirtuins, a family of proteins with histone deacetylase activity that regulate various cellular processes. Sirtuins have been shown to play a role in cellular stress resistance, DNA repair, and the regulation of metabolic pathways [97]. Some sirtuin activators have been developed as potential anti-aging drugs, although their efficacy and safety have yet to be fully established. The most well-known sirtuin activator is resveratrol, a naturally occurring compound found in red wine [103] [104]. mTOR inhibitors are drugs that inhibit the mechanistic target of rapamycin (mTOR) signaling pathway, which is involved in regulation of cell growth, proliferation, metabolism, and aging. In preclinical and early clinical studies, mTOR inhibitors have shown the potential to extend lifespan and delay age-related diseases.

11. Drug Development in Antiaging

While aging is an inevitable process, there are several interventions that have been proposed to slow down the aging process and extend healthy lifespan. These include telomerase activation, senolytic drugs, DNA repair and epigenetic regulators. Studies have shown that activating telomerase in certain cells can increase their proliferation and delay the onset of senescence, potentially extending healthy lifespan [84] [85]. Telomerase activation is being researched as a potential target for anti-aging and anti-cancer therapies. In certain types of cancer cells, telomerase is activated, which leads to the cells becoming immortal and contributing to the development of tumors. Thus, inhibiting telomerase activity has been a target for cancer therapy. However, in normal aging, telomere shortening is a hallmark and contributes to cellular senescence and a decline in tissue functions [105]. Hence, reactivating telomerase to increase telomere length has been suggested as a way to potentially reverse some aspects of aging. Several compounds are being researched for their potential to activate telomerase, including astragalosides [106], huperzine A [107], cycloastragenol [108], and TA-65 [109]. Senolytic drugs target senescent cells, which are thought to contribute to aging and age-related diseases. Navitoclax is an inhibitor of the anti-apoptotic Bcl-2 protein. It has been shown to selectively eliminate senescent cells in preclinical studies [110]. Dasatinib and quercetin (D+Q) is a small molecule inhibitor of the Src family of tyrosine kinases, and quercetin is a flavonoid. These drugs have been found to have senolytic effects in preclinical studies [111]. ABT-263 is a BH3 mimetic, a small molecule that mimics the activity of pro-apoptotic BH3 proteins. It has been shown to selectively eliminate senescent cells in preclinical studies [112]. FOXO4-DRI is a synthetic peptide that targets the FOXO4 protein. It has been shown to selectively eliminate senescent cells in preclinical studies [113]. As DNA repair mechanisms become less effective with age, some researchers are investigating ways to enhance these mechanisms and slow down the aging process. Some examples of DNA repair in aging intervention are NAD+ precursors. NAD+ is a coenzyme that plays a key role in energy metabolism and DNA repair. As NAD+ levels decrease with age, some researchers are investigating ways to increase NAD+ levels through supplementation with NAD+ precursors [114]. Epigenetic modification such as DNA methylation and Histone modification is an important biological process associated with aging- and age-related disease development. Drugs that target methylation are a class of drugs that specifically target the process of methylation in the DNA, which can affect the function of the gene to which it is attached [115]. These drugs can be used to modulate methylation patterns in the DNA and potentially restore normal gene expression. These enzymes include DNA methyltransferases (DNMTs) [116] and histone deacetylases (HDACs) [117]. By inhibiting these enzymes, these drugs can modulate methylation patterns and potentially restore normal gene expression.

12. Technology Advance for Epigenetic Study

The field of epigenetics has seen significant technological advances in recent years, allowing for more comprehensive and in-depth studies of the changes in gene expression, chromatin modification and DNA methylation patterns that occur with aging and contribute to disease development. Some of the key technological advances in this field are included in Table 1.

Next-generation sequencing (NGS) technologies use parallel processing to sequence many small fragments of DNA simultaneously, producing millions or billions of sequences in a single run. NGS technologies allow for high-throughput and cost-effective analysis of large-scale epigenetic data [118]. ChIP-seq (Chromatin Immunoprecipitation Sequencing) is a method used to study the interaction of DNA with histone proteins and other chromatin-associated proteins, and provides insight into epigenetic regulation of gene expression [119]. It combines

Table 1. Different molecular technologies to study DNA methylation, gene expression, and chromatin modifications occur during aging and age-related diseases.

chromatin immunoprecipitation (ChIP), which selectively captures protein-DNA interactions, with NGS technology. In ChIP-seq, the chromatin is fragmented and cross-linked to the associated proteins. An antibody specific to a target protein, such as a histone modification, is then used to pull down the protein-DNA complexes. The resulting DNA is then sequenced and the data is analyzed to determine the locations of the target protein along the genome. ChIP-seq is widely used to study the regulation of gene expression by histone modifications, such as acetylation, methylation, and phosphorylation [120]. RNA sequencing (RNA-seq) allows for the quantification of transcriptome-wide changes in gene expression levels, providing a comprehensive view of the effects of epigenetic changes on gene expression [121]. Assay for transposase-accessible chromatin sequencing (ATAC-seq) is a method for mapping open chromatin regions, providing insight into changes in chromatin accessibility with aging and disease [122]. Bisulfite sequencing works by converting unmethylated cytosine bases into uracil, while methylated cytosine bases remain unchanged. The treated DNA is then sequenced using NGS technology, and the resulting data is analyzed to identify the methylation status of individual cytosine residues. Bisulfite sequencing is a method for quantifying DNA methylation levels at a single-base resolution, providing a detailed view of the changes in DNA methylation patterns that occur with aging and disease [123]. Methylation Specific Restriction Enzyme Sequencing (MSREs) are a class of enzymes that specifically recognize and cut methylated DNA sequences. These enzymes can be used to analyze methylation patterns in the DNA by cutting only the methylated regions of the DNA and leaving the unmethylated regions intact. By cutting only the methylated regions of the specific DNA sequence, these enzymes can be used to analyze the methylation status of specific genes or regions of the genome [124]. Methylation arrays such as Illumina Infinium array enable quantitative interrogation of selected methylation sites across the genome, offering high-throughput capabilities that minimize the cost per sample. Histone modification assay uses antibodies specific to histone modifications to quantify the levels of these modifications at specific sites along the genome [125]. Chromatin conformation capture (3C) method is used to study the spatial organization of the genome and to identify interactions between different regions of the genome [126]. These technologies, combined with new computational and data analysis tools, have allowed for major advances in our understanding of the role of epigenetics in aging and disease.

13. Geriatric Disease

Age-related diseases or geriatric diseases are conditions that primarily affect older adults and are caused by the natural aging process. These diseases often occur because of accumulated damage to cells and tissues over time and are characterized by a progressive decline in function and an increased risk of disability and death. The most common aging-related diseases include cancer, cardiovascular and neurodegenerative disease [128]. Aging also involves diseases such as diabetes, osteoarthritis, osteoporosis, and many other aging-related diseases including cataract, glaucoma, macular degeneration, sarcopenia, and frailty [129] [130]. It also involves some cosmetic changes such as skin wrinkling and sagging, and hair loss and whitening (Figure 4). Preventing and managing these diseases is critical to maintaining a healthy and independent older age. These conditions are not inevitable and can be influenced by a combination of genetic, epigenetic, environmental, and lifestyle factors. Further research is needed to fully understand the underlying mechanisms of these diseases and to develop effective interventions for their prevention and treatment.

14. Alzheimer’s Disease in Aging

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that is characterized by memory loss, cognitive decline, and changes in behavior. It is the most common cause of dementia in older adults and is typically diagnosed in individuals over the age of 65. Aging is the major risk factor for Alzheimer’s disease, and as the population ages, the number of individuals affected by AD is expected to increase [131]. While the exact cause of AD is not fully understood, several factors have been identified that may contribute to the development of the disease. These include genetics, epigenetics, lifestyle, environment, inflammation, oxidative stress, and metabolic factors [132]. In terms of genetics, several genetic mutations and variants have been identified that significantly increase the risk of developing Alzheimer’s disease. The most well-known of these is the apolipoprotein E (APOE) gene. This protein combines with fats (lipids) in the body to form molecules called lipoproteins. Lipoproteins are responsible for packaging cholesterol and other fats and carrying them through the bloodstream. The APOE ε4 allele is the strongest genetic risk factor for late-onset Alzheimer’s disease [133]. Other genetic mutations that increase the risk of Alzheimer’s disease

Figure 4. Different diseases associated with aging due to environmental changes, lifestyle, and inherited epigenetic changes.

include those in genes involved in the formation and clearance of beta-amyloid plaques, tau protein aggregation, and inflammation. Several rare mutations linked to familial AD (FAD) on the Aβ precursor protein (APP), Presenilin-1 (PS1), Presenilin-2 (PS2), Adamalysin10, and other genetic risk factors [134] [135]. By studying the genetic causes of Alzheimer’s disease, researchers hope to identify new therapeutic targets and develop new drugs to treat the disease. Epigenetic factors play a role in the development of Alzheimer’s disease. Changes in DNA methylation, histone modification and microRNA expression have been implicated in the pathology of Alzheimer’s disease and can influence gene expression, synaptic function and neuro-inflammation. Several genes have been identified to be involved in DNA methylation changes in Alzheimer’s disease, including APOE, BIN1, CLU, PICALM, and CR1 [136]. Other genes such as SORL1, ABCA7, and MS4A have also been found to be associated with DNA methylation changes and Alzheimer’s risk. In addition to the genetic and epigenetic, environmental factors such as head trauma, high blood pressure, and exposure to toxins may increase the risk of AD. Lifestyle factors for example poor diet, lack of physical activity, and smoking have been linked to an increased risk of AD [137]. Chronic inflammation has been implicated as a contributing factor in the development and progression of Alzheimer’s disease (AD). Studies have shown that elevated levels of pro-inflammatory cytokines and immune cells in the brain can contribute to the oxidative stress, neural injury, and neurodegeneration that are hallmarks of AD. The connection between inflammation and AD may also be influenced by epigenetic changes, such as DNA methylation, that affect the expression of genes involved in immune and oxidative stress response [138]. Metabolic factors such as diabetes, obesity, and high cholesterol have been linked to an increased risk of AD. Additionally, alterations in energy metabolism, oxidative stress, and insulin resistance have been observed in the brains of patients with AD. Research on AD in aging model is ongoing, and new findings are emerging as the field is advancing. This can include not only drug development but also lifestyle changes, early detection and intervention, and the identification of new targets for therapy.

15. Cancer in Aging

Cancer is a disease characterized by the uncontrolled growth and spread of abnormal cells, and it is a leading cause of death worldwide [139]. The risk of developing cancer increases with aging. This is due to several factors including the accumulation of mutations in cells over time, exposure to environmental toxins, changes in hormonal balance, and a decline in the function of the immune system. In fact, the majority of cancer cases are diagnosed in individuals over the age of 65 [140]. Aging is considered as the major risk factor for cancer, mainly because the incidence of cancer increases with age. The underlying mechanisms of cancer in aging are complex and not fully understood, but several factors have been identified that may contribute to the development of cancer. Accumulation of genetic mutations (somatic) can increase as people age. This is because as cells divide and replicate over time, they accumulate genetic changes (mutations) that can lead to the development of cancer. Some of the most well-known somatic genetic mutations in cancer include TP53, KRAS, and APC [141]. These mutations can activate oncogenes and inactivate tumor suppressor genes, leading to the development of cancer as people get aged. Epigenetic pattern of certain genes is changed with age. Some tumor suppressor genes or oncogenes can become either silenced or activated when the genes are hypermethylated or hypomethylated during the development and progression of cancer [142] [143] [144]. Examples of such genes include p16INK4a in colon cancer, BRCA1 in breast cancer, and RASSF1A in lung cancer [145] [146]. Understanding the genetic mutations and epigenetic changes that accumulate in aging and how they contribute to the development of cancer is an important area of research that will help inform the development of new treatments and prevention strategies. Additionally, exposure to environmental factors such as radiation, chemicals and toxins, and certain viruses and bacteria have been linked to an increased risk of cancer. Lifestyle factors such as smoking, poor diet, and lack of physical activity have been linked to an increased risk of cancer. Chronic inflammation has been associated with the development of cancer. Oxidative stress, which is an imbalance between the production of reactive oxygen species and the body’s ability to neutralize them, has been linked to the development of cancer. Metabolic factors such as obesity, diabetes, and high cholesterol have been linked to an increased risk of cancer that is associated with aging.

16. Cardiovascular Disease in Aging

Cardiovascular disease (CVD) is a broad term that refers to a group of disorders that affect the heart and blood vessels, including coronary artery disease, heart failure, and stroke. As people age, they have an increased risk of developing CVD, and it is a leading cause of death worldwide. Aging is considered as a major risk factor for CVD, mainly because the incidence of CVD increases with age. The underlying mechanisms of CVD in aging are complex and not fully understood, but both genetic and non-genetic factors have been identified that may contribute to the development of CVD. Certain genetic mutations and variations have been linked to an increased risk of CVD, including mutations in genes that regulate blood pressure, cholesterol, and clotting. Some common genetic risk factors for CVD include mutations in genes involved in lipid metabolism, such as the LDL receptor and PCSK9 genes, as well as genetic variations in genes involved in inflammation and blood clotting [147]. Other genetic factors that have been associated with an increased risk of CVD include those involved in hypertension, such as the AGT gene, and those involved in the regulation of glucose and insulin metabolism, such as the PPARG gene [148]. Several genes have been associated with epigenetic changes in cardiovascular disease (CVD). For example, studies have shown that changes in DNA methylation patterns in genes involved in inflammation and lipid metabolism can increase the risk of CVD. Additionally, epigenetic changes in genes related to blood pressure regulation and blood vessel growth have also been linked to the development of CVD. Some of these genes include APOA1, APOE, FGF5, and LEP. Epigenetic changes in these genes can affect their expression levels, which in turn may contribute to the development and progression of CVD [149]. Environmental factors such as exposure to pollution, radiation and toxins have been linked to an increased risk of CVD Lifestyle factors such as smoking, poor diet, lack of physical activity, and high alcohol consumption have been linked to an increased risk of CVD. Chronic inflammation has been associated with the development of CVD. Chronic inflammation can cause damage to the arteries, leading to plaque buildup and ultimately, cardiovascular events such as heart attacks and strokes. Additionally, chronic inflammation can contribute to the development of conditions like atherosclerosis, which is a major risk factor for CVD. Studies have shown that elevated levels of inflammatory markers such as C-reactive protein are associated with an increased risk of CVD, and that reducing levels of these markers can reduce the risk of cardiovascular events. Metabolic factors such as obesity, diabetes, and high cholesterol have been linked to an increased risk of CVD. High blood pressure or hypertension is one of the most important risk factors for CVD.

17. Discussion

This article provides a comprehensive overview of the current state of epigenetic research related to aging. The objective of the study is to identify the epigenetic changes that occur during aging and evaluate their potential as biomarkers of aging. Multiple studies have shown that aging is associated with various epigenetic changes, including changes in DNA methylation, histone modification, and chromatin accessibility. The study of “Epigenetic Signatures of Aging” sheds light on the significance of these epigenetic changes in aging and their potential use as biomarkers. The study emphasizes the importance of epigenetic changes in aging and suggests that they can be utilized to enhance our understanding of age-related diseases and develop new diagnostic tests and interventions to improve health in older adults.

Although epigenetic studies of aging have the promise of unlocking new ways to intervene in the aging process and its associated diseases, there are several limitations to aging research. Aging is a complex process that is influenced by a variety of factors, including genetics, environmental factors, lifestyle, and other biological processes. This complexity makes it difficult to isolate the specific mechanisms and factors that contribute to aging. Furthermore, there is no single, agreed-upon measure of aging, making it difficult to accurately track and compare changes in aging over time. Although aging can be studied in model organisms such as yeast, worms, flies, and mice, these models have limitations and do not always accurately reflect the complex aging processes that occur in humans. Additionally, studying aging in humans is logistically challenging due to the long lifespan and a large number of variables that can influence aging. Longitudinal studies, which follow the same individuals over time, are necessary to understand the dynamics of epigenetic changes during aging and their impact on health and disease. However, such studies can be challenging and time-consuming to carry out, and many existing aging studies are cross-sectional in nature, limiting their ability to reveal longitudinal changes. Some potential interventions for aging, such as life extension strategies, raise ethical concerns about their impact on society and the consequences of a rapidly aging population. Despite these limitations, there have been significant advances in our understanding of aging in recent years, and researchers are continuing to work towards finding ways to delay and prevent age-related diseases.

18. Conclusion

In conclusion, biological studies of aging play a crucial role in understanding the underlying mechanisms of aging and aging-related diseases. From genetics to epigenetics, telomere shortening, senescence cells and various research areas are exploring the different factors that contribute to aging and the potential for developing interventions that can slow or reverse its effects. The discovery of aging biomarkers and the exploration of the role of these marks in aging have provided valuable insights into the biological processes of aging and its relationship with age-related diseases such as cancer, Alzheimer’s disease (AD) and cardiovascular disease (CVD). Epigenetic studies in aging have shown significant progress in uncovering the mechanisms of aging and the potential for developing new therapies to delay aging-related diseases. Epigenetic modifications, such as DNA methylation and histone modification, play a critical role in regulating gene expression and cellular processes that contribute to aging. Epigenetic changes can be influenced by environmental factors such as diet, lifestyle, and exposure to toxins, as well as by genetic factors. By exploring the interplay between genetics and epigenetics in aging, researchers are gaining insight into the complex processes underlying aging and developing new strategies for preventing and treating aging-related diseases. Despite advances in the field, there is still much work to be done to fully understand the role of epigenetics in aging and to translate this knowledge into effective interventions.

Funding

We would like to express our sincere gratitude to Eone Diagnomics Genome Center for their generous support towards the publication of the article.

Competing Interests

We declare that there are no conflicts of interest related to the contents presented in this article. However, we acknowledge that there may be differing interpretations or opinions or potential limitations in our article, and we welcome further discussion and scrutiny of our work.

References

  1. 1. Jaul, E. and Barron, J. (2017) Age-Related Diseases and Clinical and Public Health Implications for the 85 Years Old and Over Population. Frontiers in Public Health, 5, 335. https://doi.org/10.3389/fpubh.2017.00335

  2. 2. Li, Z., et al. (2021) Aging and Age-Related Diseases: From Mechanisms to Therapeutic Strategies. Biogerontology, 22, 165-187. https://doi.org/10.1007/s10522-021-09910-5

  3. 3. Liu, J.-K. (2022) Antiaging Agents: Safe Interventions to Slow Aging and Healthy Life Span Extension. Natural Products and Bioprospecting, 12, 18. https://doi.org/10.1007/s13659-022-00339-y

  4. 4. Bravo-San Pedro, J.M. and Senovilla, L. (2013) Immunostimulatory Activity of Lifespan-Extending Agents. Aging, 5, 793-801. https://doi.org/10.18632/aging.100619

  5. 5. Longo, V.D., Antebi, A., Bartke, A., Barzilai, N., Brown-Borg, H.M., Caruso, C., Curiel, T.J., Cabo, R., Franceschi, C., Gems, D., Ingram, D.K., Johnson, T.E., Kennedy, B.K., Kenyon, C., Klein, S., Kopchick, J.J., Lepperdinger, G., Madeo, F., Mirisola, M.G. and Mitchell, J.R. (2015) Interventions to Slow Aging in Humans: Are We Ready? Aging Cell, 14, 497-510. https://doi.org/10.1111/acel.12338

  6. 6. Kirkwood, T.B.L. (2005) Understanding the Odd Science of Aging. Cell, 120, 437-447. https://doi.org/10.1016/j.cell.2005.01.027

  7. 7. Yan, M., Sun, S., Xu, K., Huang, X., Dou, L., Pang, J., Tang, W., Shen, T. and Li, J. (2021) Cardiac Aging: From Basic Research to Therapeutics. Oxidative Medicine and Cellular Longevity, 2021, Article ID: 9570325. https://doi.org/10.1155/2021/9570325

  8. 8. Berben, L., Floris, G., Wildiers, H. and Hatse, S. (2021) Cancer and Aging: Two Tightly Interconnected Biological Processes. Cancers, 13, 1400. https://doi.org/10.3390/cancers13061400

  9. 9. Wyss-Coray, T. (2016) Ageing, Neurodegeneration and Brain Rejuvenation. Nature, 539, 180-186. https://doi.org/10.1038/nature20411

  10. 10. Waltz, M., Cadigan, R.J., Prince, A.E.R., Skinner, D. and Henderson, G.E. (2018) Age and Perceived Risks and Benefits of Preventive Genomic Screening. Genetics in Medicine, 20, 1038-1044. https://doi.org/10.1038/gim.2017.206

  11. 11. Martin-Herranz, D.E., Aref-Eshghi, E., Bonder, M.J., Stubbs, T.M., Choufani, S., Weksberg, R., Stegle, O., Sadikovic, B., Reik, W. and Thornton, J.M. (2019) Screening for Genes That Accelerate the Epigenetic Aging Clock in Humans Reveals a Role for the H3K36 Methyltransferase NSD1. Genome Biology, 20, 146. https://doi.org/10.1186/s13059-019-1753-9

  12. 12. Janssens, G.E., Lin, X.-X., Millan-Ariño, L., Kavšek, A., Sen, I., Seinstra, R.I., Stroustrup, N., Nollen, E.A.A. and Riedel, C.G. (2019) Transcriptomics-Based Screening Identifies Pharmacological Inhibition of Hsp90 as a Means to Defer Aging. Cell Reports, 27, 467-480.e6. https://doi.org/10.1016/j.celrep.2019.03.044

  13. 13. Moaddel, R., Ubaida-Mohien, C., Tanaka, T., Lyashkov, A., Basisty, N., Schilling, B., Semba, R.D., Franceschi, C., Gorospe, M. and Ferrucci, L. (2021) Proteomics in Aging Research: A Roadmap to Clinical, Translational Research. Aging Cell, 20, e13325. https://doi.org/10.1111/acel.13325

  14. 14. Whiting, C.C., Siebert, J., Newman, A.M., Du, H., Alizadeh, A.A., Goronzy, J., Weyand, C.M., Krishnan, E., Fathman, C.G. and Maecker, H.T. (2015) Large-Scale and Comprehensive Immune Profiling and Functional Analysis of Normal Human Aging. PLOS ONE, 10, e0133627. https://doi.org/10.1371/journal.pone.0133627

  15. 15. Nguyen, H., Zarriello, S., Coats, A., Nelson, C., Kingsbury, C., Gorsky, A., Rajani, M., Neal, E.G. and Borlongan, C.V. (2019) Stem Cell Therapy for Neurological Disorders: A Focus on Aging. Neurobiology of Disease, 126, 85-104. https://doi.org/10.1016/j.nbd.2018.09.011

  16. 16. Oh, J., Lee, Y.D. and Wagers, A.J. (2014) Stem Cell Aging: Mechanisms, Regulators and Therapeutic Opportunities. Nature Medicine, 20, 870-880. https://doi.org/10.1038/nm.3651

  17. 17. Hamczyk, M.R., et al. (2022) Biological versus Chronological Aging: JACC Focus Seminar. Journal of the American College of Cardiology, 75, 919-930.

  18. 18. López-Otín, C., Blasco, M.A., Partridge, L., Serrano, M. and Kroemer, G. (2013) The Hallmarks of Aging. Cell, 153, 1194-1217. https://doi.org/10.1016/j.cell.2013.05.039

  19. 19. Askarova, S., Umbayev, B., Masoud, A.-R., Kaiyrlykyzy, A., Safarova, Y., Tsoy, A., Olzhayev, F. and Kushugulova, A. (2020) The Links between the Gut Microbiome, Aging, Modern Lifestyle and Alzheimer’s Disease. Frontiers in Cellular and Infection Microbiology, 10, 104. https://doi.org/10.3389/fcimb.2020.00104

  20. 20. Parrado, C., Mercado-Saenz, S., Perez-Davo, A., Gilaberte, Y., Gonzalez, S. and Juarranz, A. (2019) Environmental Stressors on Skin Aging. Mechanistic Insights. Frontiers in Pharmacology, 10, 759. https://doi.org/10.3389/fphar.2019.00759

  21. 21. Yu, M., Zhang, H., Wang, B., Zhang, Y., Zheng, X., Shao, B., Zhuge, Q. and Jin, K. (2021) Key Signaling Pathways in Aging and Potential Interventions for Healthy Aging. Cells, 10, 660. https://doi.org/10.3390/cells10030660

  22. 22. Bird, A. (2002) DNA Methylation Patterns and Epigenetic Memory. Genes & Development, 16, 6-21. https://doi.org/10.1101/gad.947102

  23. 23. Unnikrishnan, A., Freeman, W.M., Jackson, J., Wren, J.D., Porter, H. and Richardson, A. (2019) The Role of DNA Methylation in Epigenetics of Aging. Pharmacology & Therapeutics, 195, 172-185. https://doi.org/10.1016/j.pharmthera.2018.11.001

  24. 24. Ciccarone, F., Tagliatesta, S., Caiafa, P. and Zampieri, M. (2018) DNA Methylation Dynamics in Aging: How Far Are We from Understanding the Mechanisms? Mechanisms of Ageing and Development, 174, 3-17. https://doi.org/10.1016/j.mad.2017.12.002

  25. 25. Behzadi, P. and Ranjbar, R. (2018) DNA Microarray Technology and Bioinformatic Web Services. Acta Microbiologica et Immunologica Hungarica, 66, 19-30. https://doi.org/10.1556/030.65.2018.028

  26. 26. Dang, K., Zhang, W., Jiang, S., Lin, X. and Qian, A. (2020) Application of Lectin Microarrays for Biomarker Discovery. ChemistryOpen, 9, 285-300. https://doi.org/10.1002/open.201900326

  27. 27. Gong, T., Borgard, H., Zhang, Z., Chen, S., Gao, Z. and Deng, Y. (2022) Analysis and Performance Assessment of the Whole Genome Bisulfite Sequencing Data Workflow: Currently Available Tools and a Practical Guide to Advance DNA Methylation Studies. Small Methods, 6, Article ID: 2101251. https://doi.org/10.1002/smtd.202101251

  28. 28. Yamazaki, J., Matsumoto, Y., Jelinek, J., Ishizaki, T., Maeda, S., Watanabe, K., Ishihara, G., Yamagishi, J. and Takiguchi, M. (2021) DNA Methylation Landscape of 16 Canine Somatic Tissues by Methylation-Sensitive Restriction Enzyme-Based Next Generation Sequencing. Scientific Reports, 11, Article No. 10005. https://doi.org/10.1038/s41598-021-89279-0

  29. 29. Pei, J., van den Dungen, N.A.M., Asselbergs, F.W., Mokry, M. and Harakalova, M. (2022) Chromatin Immunoprecipitation Sequencing (ChIP-seq) Protocol for Small Amounts of Frozen Biobanked Cardiac Tissue. Methods in Molecular Biology, 2458, 97-111. https://doi.org/10.1007/978-1-0716-2140-0_6

  30. 30. Kukurba, K.R. and Montgomery, S.B. (2015) RNA Sequencing and Analysis. Cold Spring Harbor Protocols, 2015, 951-969. https://doi.org/10.1101/pdb.top084970

  31. 31. Grandi, F.C., Modi, H., Kampman, L. and Corces, M.R. (2022) Chromatin Accessibility Profiling by ATAC-seq. Nature Protocols, 17, 1518-1552. https://doi.org/10.1038/s41596-022-00692-9

  32. 32. Saul, D. and Kosinsky, R.L. (2021) Epigenetics of Aging and Aging-Associated Diseases. International Journal of Molecular Sciences, 22, 401. https://doi.org/10.3390/ijms22010401

  33. 33. Chen, C., Zhou, M., Ge, Y. and Wang, X. (2020) SIRT1 and Aging Related Signaling Pathways. Mechanisms of Ageing and Development, 187, Article ID: 111215. https://doi.org/10.1016/j.mad.2020.111215

  34. 34. Yan, J., Luo, A., Sun, R., Tang, X., Zhao, Y., Zhang, J., Zhou, B., Zheng, H., Yu, H. and Li, S. (2020) Resveratrol Mitigates Hippocampal Tau Acetylation and Cognitive Deficit by Activation SIRT1 in Aged Rats Following Anesthesia and Surgery. Oxidative Medicine and Cellular Longevity, 2020, Article ID: 4635163. https://doi.org/10.1155/2020/4635163

  35. 35. Salminen, A., Kaarniranta, K. and Kauppinen, A. (2021) Insulin/IGF-1 Signaling Promotes Immunosuppression via the STAT3 Pathway: Impact on the Aging Process and Age-Related Diseases. Inflammation Research, 70, 1043-1061. https://doi.org/10.1007/s00011-021-01498-3

  36. 36. Papadopoli, D., Boulay, K., Kazak, L., Pollak, M., Mallette, F., Topisirovic, I. and Hulea, L. (2019) mTOR as a Central Regulator of Lifespan and Aging. F1000Research, 8, 998. https://doi.org/10.12688/f1000research.17196.1

  37. 37. Du, S. and Zheng, H. (2021) Role of FoxO Transcription Factors in Aging and Age-Related Metabolic and Neurodegenerative Diseases. Cell & Bioscience, 11, 188. https://doi.org/10.1186/s13578-021-00700-7

  38. 38. Chakravarti, D., LaBella, K.A. and DePinho, R.A. (2021) Telomeres: History, Health, and Hallmarks of Aging. Cell, 184, 306-322. https://doi.org/10.1016/j.cell.2020.12.028

  39. 39. Di Micco, R., Krizhanovsky, V., Baker, D. and d’Adda di Fagagna, F. (2020) Cellular Senescence in Ageing: From Mechanisms to Therapeutic Opportunities. Nature Reviews Molecular Cell Biology, 22, 75-95. https://doi.org/10.1038/s41580-020-00314-w

  40. 40. Kane, A.E. and Sinclair, D.A. (2019) Epigenetic Changes during Aging and Their Reprogramming Potential. Critical Reviews in Biochemistry and Molecular Biology, 54, 61-83.

  41. 41. Hajam, Y.A., Rani, R., Ganie, S.Y., Sheikh, T.A., Javaid, D., Qadri, S.S., Pramodh, S., Alsulimani, A., Alkhanani, M.F., Harakeh, S., Hussain, A., Haque, S. and Reshi, M.S. (2022) Oxidative Stress in Human Pathology and Aging: Molecular Mechanisms and Perspectives. Cells, 11, 552. https://doi.org/10.3390/cells11030552

  42. 42. Wu, M., et al. (2021) Potential Implications of Polyphenols on Aging Considering Oxidative Stress, Inflammation, Autophagy, and Gut Microbiota. Critical Reviews in Food Science and Nutrition, 61, 2175-2193.

  43. 43. Haas, R.H. (2019) Mitochondrial Dysfunction in Aging and Diseases of Aging. Biology, 8, 48. https://doi.org/10.3390/biology8020048

  44. 44. Sies, H. (2020) Oxidative Stress: Concept and Some Practical Aspects. Antioxidants, 9, 852. https://doi.org/10.3390/antiox9090852

  45. 45. Furman, D., Campisi, J., Verdin, E., Carrera-Bastos, P., Targ, S., Franceschi, C., Ferrucci, L., Gilroy, D.W., Fasano, A., Miller, G.W., Miller, A.H., Mantovani, A., Weyand, C.M., Barzilai, N., Goronzy, J.J., Rando, T.A., Effros, R.B., Lucia, A., Kleinstreuer, N. and Slavich, G.M. (2019) Chronic Inflammation in the Etiology of Disease across the Life Span. Nature Medicine, 25, 1822-1832. https://doi.org/10.1038/s41591-019-0675-0

  46. 46. Wheeler, H.E. and Kim, S.K. (2021) Genetics and Genomics of Human Ageing. Philosophical Transactions of the Royal Society B: Biological Sciences, 366, 43-50.

  47. 47. Wang, J., Zhang, S., Wang, Y., Chen, L. and Zhang, X.-S. (2009) Disease-Aging Network Reveals Significant Roles of Aging Genes in Connecting Genetic Diseases. PLoS Computational Biology, 5, e1000521. https://doi.org/10.1371/journal.pcbi.1000521

  48. 48. Yao, Y., Liu, L., Guo, G., Zeng, Y. and Ji, J.S. (2021) Interaction of Sirtuin 1 (SIRT1) Candidate Longevity Gene and Particulate Matter (PM2.5) on All-Cause Mortality: A Longitudinal Cohort Study in China. Environmental Health, 20, 25. https://doi.org/10.1186/s12940-021-00718-x

  49. 49. Morris, B.J., Willcox, D.C., Donlon, T.A. and Willcox, B.J. (2015) FOXO3: A Major Gene for Human Longevity—A Mini-Review. Gerontology, 61, 515-525. https://doi.org/10.1159/000375235

  50. 50. Sanese, Forte, G., Disciglio, V., Grossi, V. and Simone, C. (2019) FOXO3 on the Road to Longevity: Lessons from SNPs and Chromatin Hubs. Computational and Structural Biotechnology Journal, 17, 737-745. https://doi.org/10.1016/j.csbj.2019.06.011

  51. 51. Zhao, Y. and Liu, Y.-S. (2021) Longevity Factor FOXO3: A Key Regulator in Aging-Related Vascular Diseases. Frontiers in Cardiovascular Medicine, 8, Article ID: 778674. https://doi.org/10.3389/fcvm.2021.778674

  52. 52. Colebatch, A.J., Dobrovic, A. and Cooper, W.A. (2019) TERT Gene: Its Function and Dysregulation in Cancer. Journal of Clinical Pathology, 72, 281-284. https://doi.org/10.1136/jclinpath-2018-205653

  53. 53. Feng, Z., Hu, W., Teresky, A.K., Hernando, E., Cordon-Cardo, C. and Levine, A.J. (2007) Declining p53 Function in the Aging Process: A Possible Mechanism for the Increased Tumor Incidence in Older Populations. Proceedings of the National Academy of Sciences, 104, 16633-16638. https://doi.org/10.1073/pnas.0708043104

  54. 54. Wang, K., Liu, H., Hu, Q., Wang, L., Liu, J., Zheng, Z., Zhang, W., Ren, J., Zhu, F. and Liu, G.-H. (2022) Epigenetic Regulation of Aging: Implications for Interventions of Aging and Diseases. Signal Transduction and Targeted Therapy, 7, Article No. 374. https://doi.org/10.1038/s41392-022-01211-8

  55. 55. Li, A., Koch, Z. and Ideker, T. (2022) Epigenetic Aging: Biological Age Prediction and Informing a Mechanistic Theory of Aging. Journal of Internal Medicine, 292, 733-744. https://doi.org/10.1111/joim.13533

  56. 56. Zhang, W., Qu, J., Liu, G.-H. and Belmonte, J.C.I. (2020) The Ageing Epigenome and Its Rejuvenation. Nature Reviews Molecular Cell Biology, 21, 137-150. https://doi.org/10.1038/s41580-019-0204-5

  57. 57. Xiao, F.-H., Wang, H.-T. and Kong, Q.-P. (2019) Dynamic DNA Methylation during Aging: A “Prophet” of Age-Related Outcomes. Frontiers in Genetics, 10, Article No. 107. https://doi.org/10.3389/fgene.2019.00107

  58. 58. Moore, L.D., Le, T. and Fan, G. (2012) DNA Methylation and Its Basic Function. Neuropsychopharmacology, 38, 23-38. https://doi.org/10.1038/npp.2012.112

  59. 59. Jin, B.L., Li, Y.J. and Robertson, K.D. (2011) DNA Methylation: Superior or Subordinate in the Epigenetic Hierarchy? Genes & Cancer, 2, 607-617.

  60. 60. Madrigano, J., et al. (2023) Aging and Epigenetics: Longitudinal Changes in Gene-Specific DNA Methylation. Epigenetics, 7, 63-70.

  61. 61. Franzago, M., Pilenzi, L., Di Rado, S., Vitacolonna, E. and Stuppia, L. (2022) The Epigenetic Aging, Obesity, and Lifestyle. Frontiers in Cell and Developmental Biology, 10, Article ID: 985274. https://doi.org/10.3389/fcell.2022.985274

  62. 62. Li, X., Hui, A.-M., Sun, L., Hasegawa, K., Torzilli, G., Minagawa, M., Takayama, T. and Makuuchi, M. (2004) p16INK4A Hypermethylation Is Associated with Hepatitis Virus Infection, Age, and Gender in Hepatocellular Carcinoma. Clinical Cancer Research, 10, 7484-7489. https://doi.org/10.1158/1078-0432.CCR-04-1715

  63. 63. Boltze, C., Zack, S., Quednow, C., Bettge, S., Roessner, A. and Schneider-Stock, R. (2003) Hypermethylation of the CDKN2/p16INK4A Promotor in Thyroid Carcinogenesis. Pathology—Research and Practice, 199, 399-404. https://doi.org/10.1078/0344-0338-00436

  64. 64. Duan, R., Fu, Q., Sun, Y. and Li, Q. (2022) Epigenetic Clock: A Promising Biomarker and Practical Tool in Aging. Ageing Research Reviews, 81, Article ID: 101743. https://doi.org/10.1016/j.arr.2022.101743

  65. 65. Oblak, L., van der Zaag, J., Higgins-Chen, A.T., Levine, M.E. and Boks, M.P. (2021) A Systematic Review of Biological, Social and Environmental Factors Associated with Epigenetic Clock Acceleration. Ageing Research Reviews, 69, Article ID: 101348. https://doi.org/10.1016/j.arr.2021.101348

  66. 66. Horvath, S. and Raj, K. (2018) DNA Methylation-Based Biomarkers and the Epigenetic Clock Theory of Ageing. Nature Reviews Genetics, 19, 371-384. https://doi.org/10.1038/s41576-018-0004-3

  67. 67. Ryan, J., Wrigglesworth, J., Loong, J., Fransquet, D. and Woods, R.L. (2019) A Systematic Review and Meta-Analysis of Environmental, Lifestyle, and Health Factors Associated with DNA Methylation Age. The Journals of Gerontology: Series A, 75, 481-494. https://doi.org/10.1093/gerona/glz099

  68. 68. Partridge, L., Fuentealba, M. and Kennedy, B.K. (2020) The Quest to Slow Ageing through Drug Discovery. Nature Reviews Drug Discovery, 19, 513-532. https://doi.org/10.1038/s41573-020-0067-7

  69. 69. Kane, A.E. and Sinclair, D.A. (2019) Epigenetic Changes during Aging and Their Reprogramming Potential. Critical Reviews in Biochemistry and Molecular Biology, 54, 61-83.

  70. 70. Reale, A., Tagliatesta, S., Zardo, G. and Zampieri, M. (2022) Counteracting Aged DNA Methylation States to Combat Ageing and Age-Related Diseases. Mechanisms of Ageing and Development, 206, Article ID: 111695. https://doi.org/10.1016/j.mad.2022.111695

  71. 71. Fang, Z., Wang, X., Sun, X., Hu, W. and Miao, Q.R. (2021) The Role of Histone Protein Acetylation in Regulating Endothelial Function. Frontiers in Cell and Developmental Biology, 9, Article ID: 672447. https://doi.org/10.3389/fcell.2021.672447

  72. 72. Eberharter, A. and Becker, B. (2002) Histone Acetylation: A Switch between Repressive and Permissive Chromatin. EMBO Reports, 3, 224-229. https://doi.org/10.1093/embo-reports/kvf053

  73. 73. Yi, S.-J. and Kim, K. (2020) New Insights into the Role of Histone Changes in Aging. International Journal of Molecular Sciences, 21, 8241. https://doi.org/10.3390/ijms21218241

  74. 74. Stilling, R.M. and Fischer, A. (2011) The Role of Histone Acetylation in Age-Associated Memory Impairment and Alzheimer’s Disease. Neurobiology of Learning and Memory, 96, 19-26. https://doi.org/10.1016/j.nlm.2011.04.002

  75. 75. Yu, R., Cao, X., Sun, L., Zhu, J., Wasko, B.M., Liu, W., Crutcher, E., Liu, H., Jo, M.C., Qin, L., Kaeberlein, M., Han, Z. and Dang, W. (2021) Inactivating Histone Deacetylase HDA Promotes Longevity by Mobilizing Trehalose Metabolism. Nature Communications, 12, Article No. 1981. https://doi.org/10.1038/s41467-021-22257-2

  76. 76. Pasyukova, E.G. and Vaiserman, A.M. (2017) HDAC Inhibitors: A New Promising Drug Class in Anti-Aging Research. Mechanisms of Ageing and Development, 166, 6-15. https://doi.org/10.1016/j.mad.2017.08.008

  77. 77. Shukla, S. and Tekwani, B.L. (2020) Histone Deacetylases Inhibitors in Neurodegenerative Diseases, Neuroprotection and Neuronal Differentiation. Frontiers in Pharmacology, 11, Article No. 537. https://doi.org/10.3389/fphar.2020.00537

  78. 78. Eckschlager, T., Plch, J., Stiborova, M. and Hrabeta, J. (2017) Histone Deacetylase Inhibitors as Anticancer Drugs. International Journal of Molecular Sciences, 18, 1414. https://doi.org/10.3390/ijms18071414

  79. 79. Jiang, H., Ju, Z. and Rudolph, K.L. (2007) Telomere Shortening and Ageing. Zeitschrift für Gerontologie und Geriatrie, 40, 314-324. https://doi.org/10.1007/s00391-007-0480-0

  80. 80. Vaiserman, A. and Krasnienkov, D. (2021) Telomere Length as a Marker of Biological Age: State-of-the-Art, Open Issues, and Future Perspectives. Frontiers in Genetics, 11, Article ID: 630186. https://doi.org/10.3389/fgene.2020.630186

  81. 81. Zvereva, M.I., Shcherbakova, D.M. and Dontsova, O.A. (2010) Telomerase: Structure, Functions, and Activity Regulation. Biochemistry (Moscow), 75, 1563-1583. https://doi.org/10.1134/S0006297910130055

  82. 82. Roake, C.M. and Artandi, S.E. (2020) Regulation of Human Telomerase in Homeostasis and Disease. Nature Reviews Molecular Cell Biology, 21, 384-397. https://doi.org/10.1038/s41580-020-0234-z

  83. 83. Logeswaran, D. and Chen, J.J.-L. (2019) Effects of Telomerase Activation. In: Gu, D. and Dupre, M.E., Eds., Encyclopedia of Gerontology and Population Aging, Springer, Berlin, 1-8. https://doi.org/10.1007/978-3-319-69892-2_42-1

  84. 84. de Jesus, B.B. and Blasco, M.A. (2012) Potential of Telomerase Activation in Extending Health Span and Longevity. Current Opinion in Cell Biology, 24, 739-743. https://doi.org/10.1016/j.ceb.2012.09.004

  85. 85. Guterres, A.N. and Villanueva, J. (2020) Targeting Telomerase for Cancer Therapy. Oncogene, 39, 5811-5824. https://doi.org/10.1038/s41388-020-01405-w

  86. 86. Jafri, M.A., Ansari, S.A., Alqahtani, M.H. and Shay, J.W. (2016) Roles of Telomeres and Telomerase in Cancer, and Advances in Telomerase-Targeted Therapies. Genome Medicine, 8, Article No. 69. https://doi.org/10.1186/s13073-016-0324-x

  87. 87. Robinson, N.J. and Schiemann, W.P. (2022) Telomerase in Cancer: Function, Regulation, and Clinical Translation. Cancers, 14, 808. https://doi.org/10.3390/cancers14030808

  88. 88. Shi, Y., Zhang, Y., Zhang, L., Ma, J.-L., Zhou, T., Li, Z.-X., Liu, W.-D., Li, W.-Q., Deng, D.-J., You, W.-C. and Pan, K.-F. (2019) Telomere Length of Circulating Cell-Free DNA and Gastric Cancer in a Chinese Population at High-Risk. Frontiers in Oncology, 9, Article No. 1434. https://doi.org/10.3389/fonc.2019.01434

  89. 89. Wu, X. and Tanaka, H. (2015) Aberrant Reduction of Telomere Repetitive Sequences in Plasma Cell-Free DNA for Early Breast Cancer Detection. Oncotarget, 6, 29795-29807. https://doi.org/10.18632/oncotarget.5083

  90. 90. van Deursen, J.M. (2014) The Role of Senescent Cells in Ageing. Nature, 509, 439-446. https://doi.org/10.1038/nature13193

  91. 91. McHugh, D. and Gil, J. (2017) Senescence and Aging: Causes, Consequences, and Therapeutic Avenues. Journal of Cell Biology, 217, 65-77. https://doi.org/10.1083/jcb.201708092

  92. 92. Chaib, S., Tchkonia, T. and Kirkland, J.L. (2022) Cellular Senescence and Senolytics: The Path to the Clinic. Nature Medicine, 28, 1556-1568. https://doi.org/10.1038/s41591-022-01923-y

  93. 93. Novais, E.J., Tran, V.A., Johnston, S.N., Darris, K.R., Roupas, A.J., Sessions, G.A., Shapiro, I.M., Diekman, B.O. and Risbud, M.V. (2021) Long-Term Treatment with Senolytic Drugs Dasatinib and Quercetin Ameliorates Age-Dependent Intervertebral Disc Degeneration in Mice. Nature Communications, 12, Article No. 5213. https://doi.org/10.1038/s41467-021-25453-2

  94. 94. Mylonas, A. and O’Loghlen, A. (2022) Cellular Senescence and Ageing: Mechanisms and Interventions. Frontiers in Aging, 3, Article ID: 866718. https://doi.org/10.3389/fragi.2022.866718

  95. 95. Hofer, S.J., Davinelli, S., Bergmann, M., Scapagnini, G. and Madeo, F. (2021) Caloric Restriction Mimetics in Nutrition and Clinical Trials. Frontiers in Nutrition, 8, Article ID: 717343. https://doi.org/10.3389/fnut.2021.717343

  96. 96. Schally, A.V., Zhang, X., Cai, R., Hare, J.M., Granata, R. and Bartoli, M. (2019) Actions and Potential Therapeutic Applications of Growth Hormone-Releasing Hormone Agonists. Endocrinology, 160, 1600-1612. https://doi.org/10.1210/en.2019-00111

  97. 97. Zemel, M.B. (2021) Modulation of Energy Sensing by Leucine Synergy with Natural Sirtuin Activators: Effects on Health Span. Journal of Medicinal Food, 23, 1129-1135.

  98. 98. Dumas, S.N. and Lamming, D.W. (2019) Next Generation Strategies for Geroprotection via mTORC1 Inhibition. The Journals of Gerontology: Series A, 75, 14-23. https://doi.org/10.1093/gerona/glz056

  99. 99. Balasubramanian, P., Howell, R. and Anderson, R.M. (2017) Aging and Caloric Restriction Research: A Biological Perspective with Translational Potential. EBioMedicine, 21, 37-44. https://doi.org/10.1016/j.ebiom.2017.06.015

  100. 100. Redmancorresponding, L.M. and Ravussin, E. (2021) Caloric Restriction in Humans: Impact on Physiological, Psychological, and Behavioral Outcomes. Antioxidants & Redox Signaling, 14, 275-287.

  101. 101. Duran-Ortiz, S., List, E.O., Basu, R. and Kopchick, J.J. (2021) Extending Lifespan by Modulating the Growth Hormone/Insulin-Like Growth Factor-1 Axis: Coming of Age. Pituitary, 24, 438-456. https://doi.org/10.1007/s11102-020-01117-0

  102. 102. Salehi, B., Mishra, A., Nigam, M., Sener, B., Kilic, M., Sharifi-Rad, M., Fokou, P., Martins, N. and Sharifi-Rad, J. (2018) Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines, 6, 91. https://doi.org/10.3390/biomedicines6030091

  103. 103. Zhang, L.-X., Li, C.-X., Kakar, M.U., Khan, M.S., Wu, F., Amir, R.M., Dai, D.-F., Naveed, M., Li, Q.-Y., Saeed, M., Shen, J.-Q., Rajput, S.A. and Li, J.-H. (2021) Resveratrol (RV): A Pharmacological Review and Call for Further Research. Biomedicine & Pharmacotherapy, 143, Article ID: 112164. https://doi.org/10.1016/j.biopha.2021.112164

  104. 104. Wallerath, T., Deckert, G., Ternes, T., Anderson, H., Li, H., Witte, K. and Förstermann, U. (2002) Resveratrol, a Polyphenolic Phytoalexin Present in Red Wine, Enhances Expression and Activity of Endothelial Nitric Oxide Synthase. Circulation, 106, 1652-1658. https://doi.org/10.1161/01.CIR.0000029925.18593.5C

  105. 105. Low, K.C. and Tergaonkar, V. (2013) Telomerase: Central Regulator of All of the Hallmarks of Cancer. Trends in Biochemical Sciences, 38, 426-434. https://doi.org/10.1016/j.tibs.2013.07.001

  106. 106. Zhou, L., Li, M., Chai, Z., Zhang, J., Cao, K., Deng, L., Liu, Y., Jiao, C., Zou, G.-M., Wu, J. and Han, F. (2022) Anticancer Effects and Mechanisms of Astragaloside IV (Review). Oncology Reports, 49, Article No. 5. https://doi.org/10.3892/or.2022.8442

  107. 107. Tsai, S.-J. (2019) Huperzine-A, a Versatile Herb, for the Treatment of Alzheimer’s Disease. Journal of the Chinese Medical Association, 82, 750-751. https://doi.org/10.1097/JCMA.0000000000000151

  108. 108. Yu, Y., Zhou, L., Yang, Y. and Liu, Y. (2018) Cycloastragenol: An Exciting Novel Candidate for Age Associated Diseases. Experimental and Therapeutic Medicine, 16, 2175-2182. https://doi.org/10.3892/etm.2018.6501

  109. 109. Alshinnawy, A.S., El-Sayed, W.M., Sayed, A.A., Salem, A.M. and Taha, A.M. (2021) Telomerase Activator-65 and Pomegranate Peel Improved the Health Status of the Liver in Aged Rats; Multi-Targets Involved. Iranian Journal of Basic Medical Sciences, 24, 842-850.

  110. 110. Sharma, A.K., Roberts, R.L., Benson, R.D., Pierce, J.L., Yu, K., Hamrick, M.W. and McGee-Lawrence, M.E. (2020) The Senolytic Drug Navitoclax (ABT-263) Causes Trabecular Bone Loss and Impaired Osteoprogenitor Function in Aged Mice. Frontiers in Cell and Developmental Biology, 8, 354. https://doi.org/10.3389/fcell.2020.00354

  111. 111. Krzystyniak, A., Wesierska, M., Petrazzo, G., Gadecka, A., Dudkowska, M., Bielak-Zmijewska, A., Mosieniak, G., Figiel, I., Wlodarczyk, J. and Sikora, E. (2022) Combination of Dasatinib and Quercetin Improves Cognitive Abilities in Aged Male Wistar Rats, Alleviates Inflammation and Changes Hippocampal Synaptic Plasticity and Histone H3 Methylation Profile. Aging, 14, 572-595. https://doi.org/10.18632/aging.203835

  112. 112. Chang, J., Wang, Y., Shao, L., Laberge, R.-M., Demaria, M., Campisi, J., Janakiraman, K., Sharpless, N.E., Ding, S., Feng, W., Luo, Y., Wang, X., Aykin-Burns, N., Krager, K., Ponnappan, U., Hauer-Jensen, M., Meng, A. and Zhou, D. (2015) Clearance of Senescent Cells by ABT263 Rejuvenates Aged Hematopoietic Stem Cells in Mice. Nature Medicine, 22, 78-83. https://doi.org/10.1038/nm.4010

  113. 113. Zhang, C., Xie, Y., Chen, H., Lv, L., Yao, J., Zhang, M., Xia, K., Feng, X., Li, Y., Liang, X., Sun, X., Deng, C. and Liu, G. (2020) FOXO4-DRI Alleviates Age-Related Testosterone Secretion Insufficiency by Targeting Senescent Leydig Cells in Aged Mice. Aging, 12, 1272-1284. https://doi.org/10.18632/aging.102682

  114. 114. Reiten, O.K., Wilvang, M.A., Mitchell, S.J., Hu, Z. and Fang, E.F. (2021) Preclinical and Clinical Evidence of NAD+ Precursors in Health, Disease, and Ageing. Mechanisms of Ageing and Development, 199, Article ID: 111567. https://doi.org/10.1016/j.mad.2021.111567

  115. 115. Cheng, Y., et al. (2019) Targeting Epigenetic Regulators for Cancer Therapy: Mechanisms and Advances in Clinical Trials. Signal Transduction and Targeted Therapy, 4, 1-39. https://doi.org/10.1038/s41392-019-0095-0

  116. 116. Giri, A.K. and Aittokallio, T. (2019) DNMT Inhibitors Increase Methylation in the Cancer Genome. Frontiers in Pharmacology, 10, Article No. 385. https://www.frontiersin.org/articles/10.3389/fphar.2019.00385 https://doi.org/10.3389/fphar.2019.00385

  117. 117. Xu, W.S., Parmigiani, R.B. and Marks, A. (2007) Histone Deacetylase Inhibitors: Molecular Mechanisms of Action. Oncogene, 26, 5541-5552. https://doi.org/10.1038/sj.onc.1210620

  118. 118. Reuter, J.A., Spacek, D.V. and Snyder, M.P. (2015) High-Throughput Sequencing Technologies. Molecular Cell, 58, 586-597. https://doi.org/10.1016/j.molcel.2015.05.004

  119. 119. Wu, D.-Y., Bittencourt, D., Stallcup, M.R. and Siegmund, K.D. (2015) Identifying Differential Transcription Factor Binding in ChIP-seq. Frontiers in Genetics, 6, Article No. 169. https://doi.org/10.3389/fgene.2015.00169

  120. 120. Muhammad, I.I., Kong, S.L., Akmar Abdullah, S.N. and Munusamy, U. (2019) RNA-seq and ChIP-seq as Complementary Approaches for Comprehension of Plant Transcriptional Regulatory Mechanism. International Journal of Molecular Sciences, 21, 167. https://doi.org/10.3390/ijms21010167

  121. 121. Hong, M., Tao, S., Zhang, L., Diao, L.-T., Huang, X., Huang, S., Xie, S.-J., Xiao, Z.-D. and Zhang, H. (2020) RNA Sequencing: New Technologies and Applications in Cancer Research. Journal of Hematology & Oncology, 13, Article No. 166. https://doi.org/10.1186/s13045-020-01005-x

  122. 122. Yan, F., Powell, D.R., Curtis, D.J. and Wong, N.C. (2020) From Reads to Insight: A Hitchhiker’s Guide to ATAC-seq Data Analysis. Genome Biology, 21, Article No. 22. https://doi.org/10.1186/s13059-020-1929-3

  123. 123. Zhou, L., Ng, H.K., Drautz-Moses, D.I., Schuster, S.C., Beck, S., Kim, C., Chambers, J.C. and Loh, M. (2019) Systematic Evaluation of Library Preparation Methods and Sequencing Platforms for High-Throughput Whole Genome Bisulfite Sequencing. Scientific Reports, 9, Article No. 10383. https://doi.org/10.1038/s41598-019-46875-5

  124. 124. Bonora, G., Rubbi, L., Morselli, M., Ma, F., Chronis, C., Plath, K. and Pellegrini, M. (2019) DNA Methylation Estimation Using Methylation-Sensitive Restriction Enzyme Bisulfite Sequencing (MREBS). PLOS ONE, 14, e0214368. https://doi.org/10.1371/journal.pone.0214368

  125. 125. Ma, F., Jiang, S. and Zhang, C.-Y. (2019) Recent Advances in Histone Modification and Histone Modifying Enzyme Assays. Expert Review of Molecular Diagnostics, 19, 27-36.

  126. 126. Lafontaine, D.L., Yang, L., Dekker, J. and Gibcus, J.H. (2021) Hi-C 3.0: Improved Protocol for Genome-Wide Chromosome Conformation Capture. Current Protocols, 1, e198. https://doi.org/10.1002/cpz1.198

  127. 127. Arneson, A., Haghani, A., Thompson, M.J., Pellegrini, M., Kwon, S.B., Vu, H., Maciejewski, E., Yao, M., Li, C.Z., Lu, A.T., Morselli, M., Rubbi, L., Barnes, B., Hansen, K.D., Zhou, W., Breeze, C.E., Ernst, J. and Horvath, S. (2022) A Mammalian Methylation Array for Profiling Methylation Levels at Conserved Sequences. Nature Communications, 13, Article No. 783. https://doi.org/10.1038/s41467-022-28355-z

  128. 128. Liu, Y., Weng, W., Gao, R. and Liu, Y. (2019) New Insights for Cellular and Molecular Mechanisms of Aging and Aging-Related Diseases: Herbal Medicine as Potential Therapeutic Approach. Oxidative Medicine and Cellular Longevity, 2019, Article ID: 4598167. https://doi.org/10.1155/2019/4598167

  129. 129. Nanayakkara, N., Curtis, A.J., Heritier, S., Gadowski, A.M., Pavkov, M.E., Kenealy, T., Owens, D.R., Thomas, R.L., Song, S., Wong, J., Chan, J.C.-N., Luk, A.O.-Y., Penno, G., Ji, L., Mohan, V., Amutha, A., Romero-Aroca, P., Gasevic, D., Magliano, D.J. and Teede, H.J. (2020) Impact of Age at Type 2 Diabetes Mellitus Diagnosis on Mortality and Vascular Complications: Systematic Review and Meta-Analyses. Diabetologia, 64, 275-287. https://doi.org/10.1007/s00125-020-05319-w

  130. 130. Driban, J.B., Harkey, M.S., Liu, S.-H., Salzler, M. and McAlindon, T.E. (2020) Osteoarthritis and Aging: Young Adults with Osteoarthritis. Current Epidemiology Reports, 7, 9-15. https://doi.org/10.1007/s40471-020-00224-7

  131. 131. Breijyeh, Z. and Karaman, R. (2020) Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules, 25, 5789. https://doi.org/10.3390/molecules25245789

  132. 132. Sengoku, R. (2019) Aging and Alzheimer’s Disease Pathology. Neuropathology, 40, 22-29. https://doi.org/10.1111/neup.12626

  133. 133. Saddiki, H., Fayosse, A., Cognat, E., Sabia, S., Engelborghs, S., Wallon, D., Alexopoulos, P., Blennow, K., Zetterberg, H., Parnetti, L., Zerr, I., Hermann, P., Gabelle, A., Boada, M., Orellana, A., de Rojas, I., Lilamand, M., Bjerke, M., Van Broeckhoven, C. and Farotti, L. (2020) Age and the Association between Apolipoprotein E Genotype and Alzheimer Disease: A Cerebrospinal Fluid Biomarker-Based Case-Control Study. PLOS Medicine, 17, e1003289. https://doi.org/10.1371/journal.pmed.1003289

  134. 134. Han, J., Park, H., Maharana, C., Gwon, A-Ryeong., Park, J., Baek, S.H., Bae, H.-G., Cho, Y., Kim, H.K., Sul, J.H., Lee, J., Kim, E., Kim, J., Cho, Y., Park, S., Palomera, L.F., Arumugam, T.V., Mattson, M.P. and Jo, D.-G. (2021) Alzheimer’s Disease-Causing Presenilin-1 Mutations Have Deleterious Effects on Mitochondrial Function. Theranostics, 11, 8855-8873. https://doi.org/10.7150/thno.59776

  135. 135. Galla, L., Redolfi, N., Pozzan, T., Pizzo, P. and Greotti, E. (2020) Intracellular Calcium Dysregulation by the Alzheimer’s Disease-Linked Protein Presenilin 2. International Journal of Molecular Sciences, 21, 770. https://doi.org/10.3390/ijms21030770

  136. 136. Mitsumori, R., Sakaguchi, K., Shigemizu, D., Mori, T., Akiyama, S., Ozaki, K., Niida, S. and Shimoda, N. (2020) Lower DNA Methylation Levels in CpG Island Shores of CR1, CLU, and PICALM in the Blood of Japanese Alzheimer’s Disease Patients. PLOS ONE, 15, e0239196. https://doi.org/10.1371/journal.pone.0239196

  137. 137. Fratiglioni, L., Marseglia, A. and Dekhtyar, S. (2020) Ageing without Dementia: Can Stimulating Psychosocial and Lifestyle Experiences Make a Difference? The Lancet Neurology, 19, 533-543. https://doi.org/10.1016/S1474-4422(20)30039-9

  138. 138. McGrattan, A.M., McGuinness, B., McKinley, M.C., Kee, F., Passmore, P., Woodside, J.V. and McEvoy, C.T. (2019) Diet and Inflammation in Cognitive Ageing and Alzheimer’s Disease. Current Nutrition Reports, 8, 53-65. https://doi.org/10.1007/s13668-019-0271-4

  139. 139. Bray, F., Laversanne, M., Weiderpass, E. and Soerjomataram, I. (2021) The Ever-Increasing Importance of Cancer as a Leading Cause of Premature Death Worldwide. Cancer, 127, 3029-3030. https://doi.org/10.1002/cncr.33587

  140. 140. Laconi, E., Marongiu, F. and DeGregori, J. (2020) Cancer as a Disease of Old Age: Changing Mutational and Microenvironmental Landscapes. British Journal of Cancer, 122, 943-952. https://doi.org/10.1038/s41416-019-0721-1

  141. 141. Marbun, V.M.G., Erlina, L. and Lalisang, T.J.M. (2022) Genomic Landscape of Pathogenic Mutation of APC, KRAS, TP53, PIK3CA, and MLH1 in Indonesian Colorectal Cancer. PLOS ONE, 17, e0267090. https://doi.org/10.1371/journal.pone.0267090

  142. 142. Madakashira, B.P. and Sadler, K.C. (2017) DNA Methylation, Nuclear Organization, and Cancer. Frontiers in Genetics, 8, Article No. 76.https://www.frontiersin.org/articles/10.3389/fgene.2017.00076 https://doi.org/10.3389/fgene.2017.00076

  143. 143. Pfeifer, G.P. (2018) Defining Driver DNA Methylation Changes in Human Cancer. International Journal of Molecular Sciences, 19, 1166. https://doi.org/10.3390/ijms19041166

  144. 144. Tang, Q., Cheng, J., et al. (2016) Blood-Based DNA Methylation as Biomarker for Breast Cancer: A Systematic Review. Clinical Epigenetics, 8, 115. https://doi.org/10.1186/s13148-016-0282-6

  145. 145. Nassar, F.J., et al. (2021) Methylated Circulating Tumor DNA as a Biomarker for Colorectal Cancer Diagnosis, Prognosis, and Prediction. Clinical Epigenetics, 13, 111. https://doi.org/10.1186/s13148-021-01095-5

  146. 146. Wang, Y., et al. (2007) Identification of Epigenetic Aberrant Promoter Methylation of RASSF1A in Serum DNA and Its Clinicopathological Significance in Lung Cancer. Lung Cancer, 56, 289-294. https://doi.org/10.1016/j.lungcan.2006.12.007

  147. 147. Krittanawong, C., Khawaja, M., Rosenson, R.S., Amos, C.I., Nambi, V., Lavie, C.J. and Virani, S.S. (2022) Association of PCSK9 Variants with the Risk of Atherosclerotic Cardiovascular Disease and Variable Responses to PCSK9 Inhibitor Therapy. Current Problems in Cardiology, 47, Article ID: 101043. https://doi.org/10.1016/j.cpcardiol.2021.101043

  148. 148. Wagner, N. and Wagner, K.-D. (2023) Pharmacological Utility of PPAR Modulation for Angiogenesis in Cardiovascular Disease. International Journal of Molecular Sciences, 24, 2345. https://doi.org/10.3390/ijms24032345

  149. 149. Balcerzyk-Matić, A., Nowak, T., Mizia-Stec, K., Iwanicka, J., Iwanicki, T., Bańka, P., Jarosz, A., Filipecki, A., Żak, I., Krauze, J. and Niemiec, P. (2022) Polymorphic Variants of AGT, ABCA1, and CYBA Genes Influence the Survival of Patients with Coronary Artery Disease: A Prospective Cohort Study. Genes, 13, 2148. https://doi.org/10.3390/genes13112148

Conflicts of Interest

We declare that there are no conflicts of interest related to the contents presented in this article. However, we acknowledge that there may be differing interpretations or opinions or potential limitations in our article, and we welcome further discussion and scrutiny of our work.

References

[1] Jaul, E. and Barron, J. (2017) Age-Related Diseases and Clinical and Public Health Implications for the 85 Years Old and Over Population. Frontiers in Public Health, 5, 335.
https://doi.org/10.3389/fpubh.2017.00335
[2] Li, Z., et al. (2021) Aging and Age-Related Diseases: From Mechanisms to Therapeutic Strategies. Biogerontology, 22, 165-187.
https://doi.org/10.1007/s10522-021-09910-5
[3] Liu, J.-K. (2022) Antiaging Agents: Safe Interventions to Slow Aging and Healthy Life Span Extension. Natural Products and Bioprospecting, 12, 18.
https://doi.org/10.1007/s13659-022-00339-y
[4] Bravo-San Pedro, J.M. and Senovilla, L. (2013) Immunostimulatory Activity of Lifespan-Extending Agents. Aging, 5, 793-801.
https://doi.org/10.18632/aging.100619
[5] Longo, V.D., Antebi, A., Bartke, A., Barzilai, N., Brown-Borg, H.M., Caruso, C., Curiel, T.J., Cabo, R., Franceschi, C., Gems, D., Ingram, D.K., Johnson, T.E., Kennedy, B.K., Kenyon, C., Klein, S., Kopchick, J.J., Lepperdinger, G., Madeo, F., Mirisola, M.G. and Mitchell, J.R. (2015) Interventions to Slow Aging in Humans: Are We Ready? Aging Cell, 14, 497-510.
https://doi.org/10.1111/acel.12338
[6] Kirkwood, T.B.L. (2005) Understanding the Odd Science of Aging. Cell, 120, 437-447.
https://doi.org/10.1016/j.cell.2005.01.027
[7] Yan, M., Sun, S., Xu, K., Huang, X., Dou, L., Pang, J., Tang, W., Shen, T. and Li, J. (2021) Cardiac Aging: From Basic Research to Therapeutics. Oxidative Medicine and Cellular Longevity, 2021, Article ID: 9570325.
https://doi.org/10.1155/2021/9570325
[8] Berben, L., Floris, G., Wildiers, H. and Hatse, S. (2021) Cancer and Aging: Two Tightly Interconnected Biological Processes. Cancers, 13, 1400.
https://doi.org/10.3390/cancers13061400
[9] Wyss-Coray, T. (2016) Ageing, Neurodegeneration and Brain Rejuvenation. Nature, 539, 180-186.
https://doi.org/10.1038/nature20411
[10] Waltz, M., Cadigan, R.J., Prince, A.E.R., Skinner, D. and Henderson, G.E. (2018) Age and Perceived Risks and Benefits of Preventive Genomic Screening. Genetics in Medicine, 20, 1038-1044.
https://doi.org/10.1038/gim.2017.206
[11] Martin-Herranz, D.E., Aref-Eshghi, E., Bonder, M.J., Stubbs, T.M., Choufani, S., Weksberg, R., Stegle, O., Sadikovic, B., Reik, W. and Thornton, J.M. (2019) Screening for Genes That Accelerate the Epigenetic Aging Clock in Humans Reveals a Role for the H3K36 Methyltransferase NSD1. Genome Biology, 20, 146.
https://doi.org/10.1186/s13059-019-1753-9
[12] Janssens, G.E., Lin, X.-X., Millan-Ariño, L., Kavšek, A., Sen, I., Seinstra, R.I., Stroustrup, N., Nollen, E.A.A. and Riedel, C.G. (2019) Transcriptomics-Based Screening Identifies Pharmacological Inhibition of Hsp90 as a Means to Defer Aging. Cell Reports, 27, 467-480.e6.
https://doi.org/10.1016/j.celrep.2019.03.044
[13] Moaddel, R., Ubaida-Mohien, C., Tanaka, T., Lyashkov, A., Basisty, N., Schilling, B., Semba, R.D., Franceschi, C., Gorospe, M. and Ferrucci, L. (2021) Proteomics in Aging Research: A Roadmap to Clinical, Translational Research. Aging Cell, 20, e13325.
https://doi.org/10.1111/acel.13325
[14] Whiting, C.C., Siebert, J., Newman, A.M., Du, H., Alizadeh, A.A., Goronzy, J., Weyand, C.M., Krishnan, E., Fathman, C.G. and Maecker, H.T. (2015) Large-Scale and Comprehensive Immune Profiling and Functional Analysis of Normal Human Aging. PLOS ONE, 10, e0133627.
https://doi.org/10.1371/journal.pone.0133627
[15] Nguyen, H., Zarriello, S., Coats, A., Nelson, C., Kingsbury, C., Gorsky, A., Rajani, M., Neal, E.G. and Borlongan, C.V. (2019) Stem Cell Therapy for Neurological Disorders: A Focus on Aging. Neurobiology of Disease, 126, 85-104.
https://doi.org/10.1016/j.nbd.2018.09.011
[16] Oh, J., Lee, Y.D. and Wagers, A.J. (2014) Stem Cell Aging: Mechanisms, Regulators and Therapeutic Opportunities. Nature Medicine, 20, 870-880.
https://doi.org/10.1038/nm.3651
[17] Hamczyk, M.R., et al. (2022) Biological versus Chronological Aging: JACC Focus Seminar. Journal of the American College of Cardiology, 75, 919-930.
[18] López-Otín, C., Blasco, M.A., Partridge, L., Serrano, M. and Kroemer, G. (2013) The Hallmarks of Aging. Cell, 153, 1194-1217.
https://doi.org/10.1016/j.cell.2013.05.039
[19] Askarova, S., Umbayev, B., Masoud, A.-R., Kaiyrlykyzy, A., Safarova, Y., Tsoy, A., Olzhayev, F. and Kushugulova, A. (2020) The Links between the Gut Microbiome, Aging, Modern Lifestyle and Alzheimer’s Disease. Frontiers in Cellular and Infection Microbiology, 10, 104.
https://doi.org/10.3389/fcimb.2020.00104
[20] Parrado, C., Mercado-Saenz, S., Perez-Davo, A., Gilaberte, Y., Gonzalez, S. and Juarranz, A. (2019) Environmental Stressors on Skin Aging. Mechanistic Insights. Frontiers in Pharmacology, 10, 759.
https://doi.org/10.3389/fphar.2019.00759
[21] Yu, M., Zhang, H., Wang, B., Zhang, Y., Zheng, X., Shao, B., Zhuge, Q. and Jin, K. (2021) Key Signaling Pathways in Aging and Potential Interventions for Healthy Aging. Cells, 10, 660.
https://doi.org/10.3390/cells10030660
[22] Bird, A. (2002) DNA Methylation Patterns and Epigenetic Memory. Genes & Development, 16, 6-21.
https://doi.org/10.1101/gad.947102
[23] Unnikrishnan, A., Freeman, W.M., Jackson, J., Wren, J.D., Porter, H. and Richardson, A. (2019) The Role of DNA Methylation in Epigenetics of Aging. Pharmacology & Therapeutics, 195, 172-185.
https://doi.org/10.1016/j.pharmthera.2018.11.001
[24] Ciccarone, F., Tagliatesta, S., Caiafa, P. and Zampieri, M. (2018) DNA Methylation Dynamics in Aging: How Far Are We from Understanding the Mechanisms? Mechanisms of Ageing and Development, 174, 3-17.
https://doi.org/10.1016/j.mad.2017.12.002
[25] Behzadi, P. and Ranjbar, R. (2018) DNA Microarray Technology and Bioinformatic Web Services. Acta Microbiologica et Immunologica Hungarica, 66, 19-30.
https://doi.org/10.1556/030.65.2018.028
[26] Dang, K., Zhang, W., Jiang, S., Lin, X. and Qian, A. (2020) Application of Lectin Microarrays for Biomarker Discovery. ChemistryOpen, 9, 285-300.
https://doi.org/10.1002/open.201900326
[27] Gong, T., Borgard, H., Zhang, Z., Chen, S., Gao, Z. and Deng, Y. (2022) Analysis and Performance Assessment of the Whole Genome Bisulfite Sequencing Data Workflow: Currently Available Tools and a Practical Guide to Advance DNA Methylation Studies. Small Methods, 6, Article ID: 2101251.
https://doi.org/10.1002/smtd.202101251
[28] Yamazaki, J., Matsumoto, Y., Jelinek, J., Ishizaki, T., Maeda, S., Watanabe, K., Ishihara, G., Yamagishi, J. and Takiguchi, M. (2021) DNA Methylation Landscape of 16 Canine Somatic Tissues by Methylation-Sensitive Restriction Enzyme-Based Next Generation Sequencing. Scientific Reports, 11, Article No. 10005.
https://doi.org/10.1038/s41598-021-89279-0
[29] Pei, J., van den Dungen, N.A.M., Asselbergs, F.W., Mokry, M. and Harakalova, M. (2022) Chromatin Immunoprecipitation Sequencing (ChIP-seq) Protocol for Small Amounts of Frozen Biobanked Cardiac Tissue. Methods in Molecular Biology, 2458, 97-111.
https://doi.org/10.1007/978-1-0716-2140-0_6
[30] Kukurba, K.R. and Montgomery, S.B. (2015) RNA Sequencing and Analysis. Cold Spring Harbor Protocols, 2015, 951-969.
https://doi.org/10.1101/pdb.top084970
[31] Grandi, F.C., Modi, H., Kampman, L. and Corces, M.R. (2022) Chromatin Accessibility Profiling by ATAC-seq. Nature Protocols, 17, 1518-1552.
https://doi.org/10.1038/s41596-022-00692-9
[32] Saul, D. and Kosinsky, R.L. (2021) Epigenetics of Aging and Aging-Associated Diseases. International Journal of Molecular Sciences, 22, 401.
https://doi.org/10.3390/ijms22010401
[33] Chen, C., Zhou, M., Ge, Y. and Wang, X. (2020) SIRT1 and Aging Related Signaling Pathways. Mechanisms of Ageing and Development, 187, Article ID: 111215.
https://doi.org/10.1016/j.mad.2020.111215
[34] Yan, J., Luo, A., Sun, R., Tang, X., Zhao, Y., Zhang, J., Zhou, B., Zheng, H., Yu, H. and Li, S. (2020) Resveratrol Mitigates Hippocampal Tau Acetylation and Cognitive Deficit by Activation SIRT1 in Aged Rats Following Anesthesia and Surgery. Oxidative Medicine and Cellular Longevity, 2020, Article ID: 4635163.
https://doi.org/10.1155/2020/4635163
[35] Salminen, A., Kaarniranta, K. and Kauppinen, A. (2021) Insulin/IGF-1 Signaling Promotes Immunosuppression via the STAT3 Pathway: Impact on the Aging Process and Age-Related Diseases. Inflammation Research, 70, 1043-1061.
https://doi.org/10.1007/s00011-021-01498-3
[36] Papadopoli, D., Boulay, K., Kazak, L., Pollak, M., Mallette, F., Topisirovic, I. and Hulea, L. (2019) mTOR as a Central Regulator of Lifespan and Aging. F1000Research, 8, 998.
https://doi.org/10.12688/f1000research.17196.1
[37] Du, S. and Zheng, H. (2021) Role of FoxO Transcription Factors in Aging and Age-Related Metabolic and Neurodegenerative Diseases. Cell & Bioscience, 11, 188.
https://doi.org/10.1186/s13578-021-00700-7
[38] Chakravarti, D., LaBella, K.A. and DePinho, R.A. (2021) Telomeres: History, Health, and Hallmarks of Aging. Cell, 184, 306-322.
https://doi.org/10.1016/j.cell.2020.12.028
[39] Di Micco, R., Krizhanovsky, V., Baker, D. and d’Adda di Fagagna, F. (2020) Cellular Senescence in Ageing: From Mechanisms to Therapeutic Opportunities. Nature Reviews Molecular Cell Biology, 22, 75-95.
https://doi.org/10.1038/s41580-020-00314-w
[40] Kane, A.E. and Sinclair, D.A. (2019) Epigenetic Changes during Aging and Their Reprogramming Potential. Critical Reviews in Biochemistry and Molecular Biology, 54, 61-83.
[41] Hajam, Y.A., Rani, R., Ganie, S.Y., Sheikh, T.A., Javaid, D., Qadri, S.S., Pramodh, S., Alsulimani, A., Alkhanani, M.F., Harakeh, S., Hussain, A., Haque, S. and Reshi, M.S. (2022) Oxidative Stress in Human Pathology and Aging: Molecular Mechanisms and Perspectives. Cells, 11, 552.
https://doi.org/10.3390/cells11030552
[42] Wu, M., et al. (2021) Potential Implications of Polyphenols on Aging Considering Oxidative Stress, Inflammation, Autophagy, and Gut Microbiota. Critical Reviews in Food Science and Nutrition, 61, 2175-2193.
[43] Haas, R.H. (2019) Mitochondrial Dysfunction in Aging and Diseases of Aging. Biology, 8, 48.
https://doi.org/10.3390/biology8020048
[44] Sies, H. (2020) Oxidative Stress: Concept and Some Practical Aspects. Antioxidants, 9, 852.
https://doi.org/10.3390/antiox9090852
[45] Furman, D., Campisi, J., Verdin, E., Carrera-Bastos, P., Targ, S., Franceschi, C., Ferrucci, L., Gilroy, D.W., Fasano, A., Miller, G.W., Miller, A.H., Mantovani, A., Weyand, C.M., Barzilai, N., Goronzy, J.J., Rando, T.A., Effros, R.B., Lucia, A., Kleinstreuer, N. and Slavich, G.M. (2019) Chronic Inflammation in the Etiology of Disease across the Life Span. Nature Medicine, 25, 1822-1832.
https://doi.org/10.1038/s41591-019-0675-0
[46] Wheeler, H.E. and Kim, S.K. (2021) Genetics and Genomics of Human Ageing. Philosophical Transactions of the Royal Society B: Biological Sciences, 366, 43-50.
[47] Wang, J., Zhang, S., Wang, Y., Chen, L. and Zhang, X.-S. (2009) Disease-Aging Network Reveals Significant Roles of Aging Genes in Connecting Genetic Diseases. PLoS Computational Biology, 5, e1000521.
https://doi.org/10.1371/journal.pcbi.1000521
[48] Yao, Y., Liu, L., Guo, G., Zeng, Y. and Ji, J.S. (2021) Interaction of Sirtuin 1 (SIRT1) Candidate Longevity Gene and Particulate Matter (PM2.5) on All-Cause Mortality: A Longitudinal Cohort Study in China. Environmental Health, 20, 25.
https://doi.org/10.1186/s12940-021-00718-x
[49] Morris, B.J., Willcox, D.C., Donlon, T.A. and Willcox, B.J. (2015) FOXO3: A Major Gene for Human Longevity—A Mini-Review. Gerontology, 61, 515-525.
https://doi.org/10.1159/000375235
[50] Sanese, Forte, G., Disciglio, V., Grossi, V. and Simone, C. (2019) FOXO3 on the Road to Longevity: Lessons from SNPs and Chromatin Hubs. Computational and Structural Biotechnology Journal, 17, 737-745.
https://doi.org/10.1016/j.csbj.2019.06.011
[51] Zhao, Y. and Liu, Y.-S. (2021) Longevity Factor FOXO3: A Key Regulator in Aging-Related Vascular Diseases. Frontiers in Cardiovascular Medicine, 8, Article ID: 778674.
https://doi.org/10.3389/fcvm.2021.778674
[52] Colebatch, A.J., Dobrovic, A. and Cooper, W.A. (2019) TERT Gene: Its Function and Dysregulation in Cancer. Journal of Clinical Pathology, 72, 281-284.
https://doi.org/10.1136/jclinpath-2018-205653
[53] Feng, Z., Hu, W., Teresky, A.K., Hernando, E., Cordon-Cardo, C. and Levine, A.J. (2007) Declining p53 Function in the Aging Process: A Possible Mechanism for the Increased Tumor Incidence in Older Populations. Proceedings of the National Academy of Sciences, 104, 16633-16638.
https://doi.org/10.1073/pnas.0708043104
[54] Wang, K., Liu, H., Hu, Q., Wang, L., Liu, J., Zheng, Z., Zhang, W., Ren, J., Zhu, F. and Liu, G.-H. (2022) Epigenetic Regulation of Aging: Implications for Interventions of Aging and Diseases. Signal Transduction and Targeted Therapy, 7, Article No. 374.
https://doi.org/10.1038/s41392-022-01211-8
[55] Li, A., Koch, Z. and Ideker, T. (2022) Epigenetic Aging: Biological Age Prediction and Informing a Mechanistic Theory of Aging. Journal of Internal Medicine, 292, 733-744.
https://doi.org/10.1111/joim.13533
[56] Zhang, W., Qu, J., Liu, G.-H. and Belmonte, J.C.I. (2020) The Ageing Epigenome and Its Rejuvenation. Nature Reviews Molecular Cell Biology, 21, 137-150.
https://doi.org/10.1038/s41580-019-0204-5
[57] Xiao, F.-H., Wang, H.-T. and Kong, Q.-P. (2019) Dynamic DNA Methylation during Aging: A “Prophet” of Age-Related Outcomes. Frontiers in Genetics, 10, Article No. 107.
https://doi.org/10.3389/fgene.2019.00107
[58] Moore, L.D., Le, T. and Fan, G. (2012) DNA Methylation and Its Basic Function. Neuropsychopharmacology, 38, 23-38.
https://doi.org/10.1038/npp.2012.112
[59] Jin, B.L., Li, Y.J. and Robertson, K.D. (2011) DNA Methylation: Superior or Subordinate in the Epigenetic Hierarchy? Genes & Cancer, 2, 607-617.
[60] Madrigano, J., et al. (2023) Aging and Epigenetics: Longitudinal Changes in Gene-Specific DNA Methylation. Epigenetics, 7, 63-70.
[61] Franzago, M., Pilenzi, L., Di Rado, S., Vitacolonna, E. and Stuppia, L. (2022) The Epigenetic Aging, Obesity, and Lifestyle. Frontiers in Cell and Developmental Biology, 10, Article ID: 985274.
https://doi.org/10.3389/fcell.2022.985274
[62] Li, X., Hui, A.-M., Sun, L., Hasegawa, K., Torzilli, G., Minagawa, M., Takayama, T. and Makuuchi, M. (2004) p16INK4A Hypermethylation Is Associated with Hepatitis Virus Infection, Age, and Gender in Hepatocellular Carcinoma. Clinical Cancer Research, 10, 7484-7489.
https://doi.org/10.1158/1078-0432.CCR-04-1715
[63] Boltze, C., Zack, S., Quednow, C., Bettge, S., Roessner, A. and Schneider-Stock, R. (2003) Hypermethylation of the CDKN2/p16INK4A Promotor in Thyroid Carcinogenesis. Pathology—Research and Practice, 199, 399-404.
https://doi.org/10.1078/0344-0338-00436
[64] Duan, R., Fu, Q., Sun, Y. and Li, Q. (2022) Epigenetic Clock: A Promising Biomarker and Practical Tool in Aging. Ageing Research Reviews, 81, Article ID: 101743.
https://doi.org/10.1016/j.arr.2022.101743
[65] Oblak, L., van der Zaag, J., Higgins-Chen, A.T., Levine, M.E. and Boks, M.P. (2021) A Systematic Review of Biological, Social and Environmental Factors Associated with Epigenetic Clock Acceleration. Ageing Research Reviews, 69, Article ID: 101348.
https://doi.org/10.1016/j.arr.2021.101348
[66] Horvath, S. and Raj, K. (2018) DNA Methylation-Based Biomarkers and the Epigenetic Clock Theory of Ageing. Nature Reviews Genetics, 19, 371-384.
https://doi.org/10.1038/s41576-018-0004-3
[67] Ryan, J., Wrigglesworth, J., Loong, J., Fransquet, D. and Woods, R.L. (2019) A Systematic Review and Meta-Analysis of Environmental, Lifestyle, and Health Factors Associated with DNA Methylation Age. The Journals of Gerontology: Series A, 75, 481-494.
https://doi.org/10.1093/gerona/glz099
[68] Partridge, L., Fuentealba, M. and Kennedy, B.K. (2020) The Quest to Slow Ageing through Drug Discovery. Nature Reviews Drug Discovery, 19, 513-532.
https://doi.org/10.1038/s41573-020-0067-7
[69] Kane, A.E. and Sinclair, D.A. (2019) Epigenetic Changes during Aging and Their Reprogramming Potential. Critical Reviews in Biochemistry and Molecular Biology, 54, 61-83.
[70] Reale, A., Tagliatesta, S., Zardo, G. and Zampieri, M. (2022) Counteracting Aged DNA Methylation States to Combat Ageing and Age-Related Diseases. Mechanisms of Ageing and Development, 206, Article ID: 111695.
https://doi.org/10.1016/j.mad.2022.111695
[71] Fang, Z., Wang, X., Sun, X., Hu, W. and Miao, Q.R. (2021) The Role of Histone Protein Acetylation in Regulating Endothelial Function. Frontiers in Cell and Developmental Biology, 9, Article ID: 672447.
https://doi.org/10.3389/fcell.2021.672447
[72] Eberharter, A. and Becker, B. (2002) Histone Acetylation: A Switch between Repressive and Permissive Chromatin. EMBO Reports, 3, 224-229.
https://doi.org/10.1093/embo-reports/kvf053
[73] Yi, S.-J. and Kim, K. (2020) New Insights into the Role of Histone Changes in Aging. International Journal of Molecular Sciences, 21, 8241.
https://doi.org/10.3390/ijms21218241
[74] Stilling, R.M. and Fischer, A. (2011) The Role of Histone Acetylation in Age-Associated Memory Impairment and Alzheimer’s Disease. Neurobiology of Learning and Memory, 96, 19-26.
https://doi.org/10.1016/j.nlm.2011.04.002
[75] Yu, R., Cao, X., Sun, L., Zhu, J., Wasko, B.M., Liu, W., Crutcher, E., Liu, H., Jo, M.C., Qin, L., Kaeberlein, M., Han, Z. and Dang, W. (2021) Inactivating Histone Deacetylase HDA Promotes Longevity by Mobilizing Trehalose Metabolism. Nature Communications, 12, Article No. 1981.
https://doi.org/10.1038/s41467-021-22257-2
[76] Pasyukova, E.G. and Vaiserman, A.M. (2017) HDAC Inhibitors: A New Promising Drug Class in Anti-Aging Research. Mechanisms of Ageing and Development, 166, 6-15.
https://doi.org/10.1016/j.mad.2017.08.008
[77] Shukla, S. and Tekwani, B.L. (2020) Histone Deacetylases Inhibitors in Neurodegenerative Diseases, Neuroprotection and Neuronal Differentiation. Frontiers in Pharmacology, 11, Article No. 537.
https://doi.org/10.3389/fphar.2020.00537
[78] Eckschlager, T., Plch, J., Stiborova, M. and Hrabeta, J. (2017) Histone Deacetylase Inhibitors as Anticancer Drugs. International Journal of Molecular Sciences, 18, 1414.
https://doi.org/10.3390/ijms18071414
[79] Jiang, H., Ju, Z. and Rudolph, K.L. (2007) Telomere Shortening and Ageing. Zeitschrift für Gerontologie und Geriatrie, 40, 314-324.
https://doi.org/10.1007/s00391-007-0480-0
[80] Vaiserman, A. and Krasnienkov, D. (2021) Telomere Length as a Marker of Biological Age: State-of-the-Art, Open Issues, and Future Perspectives. Frontiers in Genetics, 11, Article ID: 630186.
https://doi.org/10.3389/fgene.2020.630186
[81] Zvereva, M.I., Shcherbakova, D.M. and Dontsova, O.A. (2010) Telomerase: Structure, Functions, and Activity Regulation. Biochemistry (Moscow), 75, 1563-1583.
https://doi.org/10.1134/S0006297910130055
[82] Roake, C.M. and Artandi, S.E. (2020) Regulation of Human Telomerase in Homeostasis and Disease. Nature Reviews Molecular Cell Biology, 21, 384-397.
https://doi.org/10.1038/s41580-020-0234-z
[83] Logeswaran, D. and Chen, J.J.-L. (2019) Effects of Telomerase Activation. In: Gu, D. and Dupre, M.E., Eds., Encyclopedia of Gerontology and Population Aging, Springer, Berlin, 1-8.
https://doi.org/10.1007/978-3-319-69892-2_42-1
[84] de Jesus, B.B. and Blasco, M.A. (2012) Potential of Telomerase Activation in Extending Health Span and Longevity. Current Opinion in Cell Biology, 24, 739-743.
https://doi.org/10.1016/j.ceb.2012.09.004
[85] Guterres, A.N. and Villanueva, J. (2020) Targeting Telomerase for Cancer Therapy. Oncogene, 39, 5811-5824.
https://doi.org/10.1038/s41388-020-01405-w
[86] Jafri, M.A., Ansari, S.A., Alqahtani, M.H. and Shay, J.W. (2016) Roles of Telomeres and Telomerase in Cancer, and Advances in Telomerase-Targeted Therapies. Genome Medicine, 8, Article No. 69.
https://doi.org/10.1186/s13073-016-0324-x
[87] Robinson, N.J. and Schiemann, W.P. (2022) Telomerase in Cancer: Function, Regulation, and Clinical Translation. Cancers, 14, 808.
https://doi.org/10.3390/cancers14030808
[88] Shi, Y., Zhang, Y., Zhang, L., Ma, J.-L., Zhou, T., Li, Z.-X., Liu, W.-D., Li, W.-Q., Deng, D.-J., You, W.-C. and Pan, K.-F. (2019) Telomere Length of Circulating Cell-Free DNA and Gastric Cancer in a Chinese Population at High-Risk. Frontiers in Oncology, 9, Article No. 1434.
https://doi.org/10.3389/fonc.2019.01434
[89] Wu, X. and Tanaka, H. (2015) Aberrant Reduction of Telomere Repetitive Sequences in Plasma Cell-Free DNA for Early Breast Cancer Detection. Oncotarget, 6, 29795-29807.
https://doi.org/10.18632/oncotarget.5083
[90] van Deursen, J.M. (2014) The Role of Senescent Cells in Ageing. Nature, 509, 439-446.
https://doi.org/10.1038/nature13193
[91] McHugh, D. and Gil, J. (2017) Senescence and Aging: Causes, Consequences, and Therapeutic Avenues. Journal of Cell Biology, 217, 65-77.
https://doi.org/10.1083/jcb.201708092
[92] Chaib, S., Tchkonia, T. and Kirkland, J.L. (2022) Cellular Senescence and Senolytics: The Path to the Clinic. Nature Medicine, 28, 1556-1568.
https://doi.org/10.1038/s41591-022-01923-y
[93] Novais, E.J., Tran, V.A., Johnston, S.N., Darris, K.R., Roupas, A.J., Sessions, G.A., Shapiro, I.M., Diekman, B.O. and Risbud, M.V. (2021) Long-Term Treatment with Senolytic Drugs Dasatinib and Quercetin Ameliorates Age-Dependent Intervertebral Disc Degeneration in Mice. Nature Communications, 12, Article No. 5213.
https://doi.org/10.1038/s41467-021-25453-2
[94] Mylonas, A. and O’Loghlen, A. (2022) Cellular Senescence and Ageing: Mechanisms and Interventions. Frontiers in Aging, 3, Article ID: 866718.
https://doi.org/10.3389/fragi.2022.866718
[95] Hofer, S.J., Davinelli, S., Bergmann, M., Scapagnini, G. and Madeo, F. (2021) Caloric Restriction Mimetics in Nutrition and Clinical Trials. Frontiers in Nutrition, 8, Article ID: 717343.
https://doi.org/10.3389/fnut.2021.717343
[96] Schally, A.V., Zhang, X., Cai, R., Hare, J.M., Granata, R. and Bartoli, M. (2019) Actions and Potential Therapeutic Applications of Growth Hormone-Releasing Hormone Agonists. Endocrinology, 160, 1600-1612.
https://doi.org/10.1210/en.2019-00111
[97] Zemel, M.B. (2021) Modulation of Energy Sensing by Leucine Synergy with Natural Sirtuin Activators: Effects on Health Span. Journal of Medicinal Food, 23, 1129-1135.
[98] Dumas, S.N. and Lamming, D.W. (2019) Next Generation Strategies for Geroprotection via mTORC1 Inhibition. The Journals of Gerontology: Series A, 75, 14-23.
https://doi.org/10.1093/gerona/glz056
[99] Balasubramanian, P., Howell, R. and Anderson, R.M. (2017) Aging and Caloric Restriction Research: A Biological Perspective with Translational Potential. EBioMedicine, 21, 37-44.
https://doi.org/10.1016/j.ebiom.2017.06.015
[100] Redmancorresponding, L.M. and Ravussin, E. (2021) Caloric Restriction in Humans: Impact on Physiological, Psychological, and Behavioral Outcomes. Antioxidants & Redox Signaling, 14, 275-287.
[101] Duran-Ortiz, S., List, E.O., Basu, R. and Kopchick, J.J. (2021) Extending Lifespan by Modulating the Growth Hormone/Insulin-Like Growth Factor-1 Axis: Coming of Age. Pituitary, 24, 438-456.
https://doi.org/10.1007/s11102-020-01117-0
[102] Salehi, B., Mishra, A., Nigam, M., Sener, B., Kilic, M., Sharifi-Rad, M., Fokou, P., Martins, N. and Sharifi-Rad, J. (2018) Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines, 6, 91.
https://doi.org/10.3390/biomedicines6030091
[103] Zhang, L.-X., Li, C.-X., Kakar, M.U., Khan, M.S., Wu, F., Amir, R.M., Dai, D.-F., Naveed, M., Li, Q.-Y., Saeed, M., Shen, J.-Q., Rajput, S.A. and Li, J.-H. (2021) Resveratrol (RV): A Pharmacological Review and Call for Further Research. Biomedicine & Pharmacotherapy, 143, Article ID: 112164.
https://doi.org/10.1016/j.biopha.2021.112164
[104] Wallerath, T., Deckert, G., Ternes, T., Anderson, H., Li, H., Witte, K. and Förstermann, U. (2002) Resveratrol, a Polyphenolic Phytoalexin Present in Red Wine, Enhances Expression and Activity of Endothelial Nitric Oxide Synthase. Circulation, 106, 1652-1658.
https://doi.org/10.1161/01.CIR.0000029925.18593.5C
[105] Low, K.C. and Tergaonkar, V. (2013) Telomerase: Central Regulator of All of the Hallmarks of Cancer. Trends in Biochemical Sciences, 38, 426-434.
https://doi.org/10.1016/j.tibs.2013.07.001
[106] Zhou, L., Li, M., Chai, Z., Zhang, J., Cao, K., Deng, L., Liu, Y., Jiao, C., Zou, G.-M., Wu, J. and Han, F. (2022) Anticancer Effects and Mechanisms of Astragaloside IV (Review). Oncology Reports, 49, Article No. 5.
https://doi.org/10.3892/or.2022.8442
[107] Tsai, S.-J. (2019) Huperzine-A, a Versatile Herb, for the Treatment of Alzheimer’s Disease. Journal of the Chinese Medical Association, 82, 750-751.
https://doi.org/10.1097/JCMA.0000000000000151
[108] Yu, Y., Zhou, L., Yang, Y. and Liu, Y. (2018) Cycloastragenol: An Exciting Novel Candidate for Age Associated Diseases. Experimental and Therapeutic Medicine, 16, 2175-2182.
https://doi.org/10.3892/etm.2018.6501
[109] Alshinnawy, A.S., El-Sayed, W.M., Sayed, A.A., Salem, A.M. and Taha, A.M. (2021) Telomerase Activator-65 and Pomegranate Peel Improved the Health Status of the Liver in Aged Rats; Multi-Targets Involved. Iranian Journal of Basic Medical Sciences, 24, 842-850.
[110] Sharma, A.K., Roberts, R.L., Benson, R.D., Pierce, J.L., Yu, K., Hamrick, M.W. and McGee-Lawrence, M.E. (2020) The Senolytic Drug Navitoclax (ABT-263) Causes Trabecular Bone Loss and Impaired Osteoprogenitor Function in Aged Mice. Frontiers in Cell and Developmental Biology, 8, 354.
https://doi.org/10.3389/fcell.2020.00354
[111] Krzystyniak, A., Wesierska, M., Petrazzo, G., Gadecka, A., Dudkowska, M., Bielak-Zmijewska, A., Mosieniak, G., Figiel, I., Wlodarczyk, J. and Sikora, E. (2022) Combination of Dasatinib and Quercetin Improves Cognitive Abilities in Aged Male Wistar Rats, Alleviates Inflammation and Changes Hippocampal Synaptic Plasticity and Histone H3 Methylation Profile. Aging, 14, 572-595.
https://doi.org/10.18632/aging.203835
[112] Chang, J., Wang, Y., Shao, L., Laberge, R.-M., Demaria, M., Campisi, J., Janakiraman, K., Sharpless, N.E., Ding, S., Feng, W., Luo, Y., Wang, X., Aykin-Burns, N., Krager, K., Ponnappan, U., Hauer-Jensen, M., Meng, A. and Zhou, D. (2015) Clearance of Senescent Cells by ABT263 Rejuvenates Aged Hematopoietic Stem Cells in Mice. Nature Medicine, 22, 78-83.
https://doi.org/10.1038/nm.4010
[113] Zhang, C., Xie, Y., Chen, H., Lv, L., Yao, J., Zhang, M., Xia, K., Feng, X., Li, Y., Liang, X., Sun, X., Deng, C. and Liu, G. (2020) FOXO4-DRI Alleviates Age-Related Testosterone Secretion Insufficiency by Targeting Senescent Leydig Cells in Aged Mice. Aging, 12, 1272-1284.
https://doi.org/10.18632/aging.102682
[114] Reiten, O.K., Wilvang, M.A., Mitchell, S.J., Hu, Z. and Fang, E.F. (2021) Preclinical and Clinical Evidence of NAD+ Precursors in Health, Disease, and Ageing. Mechanisms of Ageing and Development, 199, Article ID: 111567.
https://doi.org/10.1016/j.mad.2021.111567
[115] Cheng, Y., et al. (2019) Targeting Epigenetic Regulators for Cancer Therapy: Mechanisms and Advances in Clinical Trials. Signal Transduction and Targeted Therapy, 4, 1-39.
https://doi.org/10.1038/s41392-019-0095-0
[116] Giri, A.K. and Aittokallio, T. (2019) DNMT Inhibitors Increase Methylation in the Cancer Genome. Frontiers in Pharmacology, 10, Article No. 385.
https://www.frontiersin.org/articles/10.3389/fphar.2019.00385
https://doi.org/10.3389/fphar.2019.00385
[117] Xu, W.S., Parmigiani, R.B. and Marks, A. (2007) Histone Deacetylase Inhibitors: Molecular Mechanisms of Action. Oncogene, 26, 5541-5552.
https://doi.org/10.1038/sj.onc.1210620
[118] Reuter, J.A., Spacek, D.V. and Snyder, M.P. (2015) High-Throughput Sequencing Technologies. Molecular Cell, 58, 586-597.
https://doi.org/10.1016/j.molcel.2015.05.004
[119] Wu, D.-Y., Bittencourt, D., Stallcup, M.R. and Siegmund, K.D. (2015) Identifying Differential Transcription Factor Binding in ChIP-seq. Frontiers in Genetics, 6, Article No. 169.
https://doi.org/10.3389/fgene.2015.00169
[120] Muhammad, I.I., Kong, S.L., Akmar Abdullah, S.N. and Munusamy, U. (2019) RNA-seq and ChIP-seq as Complementary Approaches for Comprehension of Plant Transcriptional Regulatory Mechanism. International Journal of Molecular Sciences, 21, 167.
https://doi.org/10.3390/ijms21010167
[121] Hong, M., Tao, S., Zhang, L., Diao, L.-T., Huang, X., Huang, S., Xie, S.-J., Xiao, Z.-D. and Zhang, H. (2020) RNA Sequencing: New Technologies and Applications in Cancer Research. Journal of Hematology & Oncology, 13, Article No. 166.
https://doi.org/10.1186/s13045-020-01005-x
[122] Yan, F., Powell, D.R., Curtis, D.J. and Wong, N.C. (2020) From Reads to Insight: A Hitchhiker’s Guide to ATAC-seq Data Analysis. Genome Biology, 21, Article No. 22.
https://doi.org/10.1186/s13059-020-1929-3
[123] Zhou, L., Ng, H.K., Drautz-Moses, D.I., Schuster, S.C., Beck, S., Kim, C., Chambers, J.C. and Loh, M. (2019) Systematic Evaluation of Library Preparation Methods and Sequencing Platforms for High-Throughput Whole Genome Bisulfite Sequencing. Scientific Reports, 9, Article No. 10383.
https://doi.org/10.1038/s41598-019-46875-5
[124] Bonora, G., Rubbi, L., Morselli, M., Ma, F., Chronis, C., Plath, K. and Pellegrini, M. (2019) DNA Methylation Estimation Using Methylation-Sensitive Restriction Enzyme Bisulfite Sequencing (MREBS). PLOS ONE, 14, e0214368.
https://doi.org/10.1371/journal.pone.0214368
[125] Ma, F., Jiang, S. and Zhang, C.-Y. (2019) Recent Advances in Histone Modification and Histone Modifying Enzyme Assays. Expert Review of Molecular Diagnostics, 19, 27-36.
[126] Lafontaine, D.L., Yang, L., Dekker, J. and Gibcus, J.H. (2021) Hi-C 3.0: Improved Protocol for Genome-Wide Chromosome Conformation Capture. Current Protocols, 1, e198.
https://doi.org/10.1002/cpz1.198
[127] Arneson, A., Haghani, A., Thompson, M.J., Pellegrini, M., Kwon, S.B., Vu, H., Maciejewski, E., Yao, M., Li, C.Z., Lu, A.T., Morselli, M., Rubbi, L., Barnes, B., Hansen, K.D., Zhou, W., Breeze, C.E., Ernst, J. and Horvath, S. (2022) A Mammalian Methylation Array for Profiling Methylation Levels at Conserved Sequences. Nature Communications, 13, Article No. 783.
https://doi.org/10.1038/s41467-022-28355-z
[128] Liu, Y., Weng, W., Gao, R. and Liu, Y. (2019) New Insights for Cellular and Molecular Mechanisms of Aging and Aging-Related Diseases: Herbal Medicine as Potential Therapeutic Approach. Oxidative Medicine and Cellular Longevity, 2019, Article ID: 4598167.
https://doi.org/10.1155/2019/4598167
[129] Nanayakkara, N., Curtis, A.J., Heritier, S., Gadowski, A.M., Pavkov, M.E., Kenealy, T., Owens, D.R., Thomas, R.L., Song, S., Wong, J., Chan, J.C.-N., Luk, A.O.-Y., Penno, G., Ji, L., Mohan, V., Amutha, A., Romero-Aroca, P., Gasevic, D., Magliano, D.J. and Teede, H.J. (2020) Impact of Age at Type 2 Diabetes Mellitus Diagnosis on Mortality and Vascular Complications: Systematic Review and Meta-Analyses. Diabetologia, 64, 275-287.
https://doi.org/10.1007/s00125-020-05319-w
[130] Driban, J.B., Harkey, M.S., Liu, S.-H., Salzler, M. and McAlindon, T.E. (2020) Osteoarthritis and Aging: Young Adults with Osteoarthritis. Current Epidemiology Reports, 7, 9-15.
https://doi.org/10.1007/s40471-020-00224-7
[131] Breijyeh, Z. and Karaman, R. (2020) Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules, 25, 5789.
https://doi.org/10.3390/molecules25245789
[132] Sengoku, R. (2019) Aging and Alzheimer’s Disease Pathology. Neuropathology, 40, 22-29.
https://doi.org/10.1111/neup.12626
[133] Saddiki, H., Fayosse, A., Cognat, E., Sabia, S., Engelborghs, S., Wallon, D., Alexopoulos, P., Blennow, K., Zetterberg, H., Parnetti, L., Zerr, I., Hermann, P., Gabelle, A., Boada, M., Orellana, A., de Rojas, I., Lilamand, M., Bjerke, M., Van Broeckhoven, C. and Farotti, L. (2020) Age and the Association between Apolipoprotein E Genotype and Alzheimer Disease: A Cerebrospinal Fluid Biomarker-Based Case-Control Study. PLOS Medicine, 17, e1003289.
https://doi.org/10.1371/journal.pmed.1003289
[134] Han, J., Park, H., Maharana, C., Gwon, A-Ryeong., Park, J., Baek, S.H., Bae, H.-G., Cho, Y., Kim, H.K., Sul, J.H., Lee, J., Kim, E., Kim, J., Cho, Y., Park, S., Palomera, L.F., Arumugam, T.V., Mattson, M.P. and Jo, D.-G. (2021) Alzheimer’s Disease-Causing Presenilin-1 Mutations Have Deleterious Effects on Mitochondrial Function. Theranostics, 11, 8855-8873.
https://doi.org/10.7150/thno.59776
[135] Galla, L., Redolfi, N., Pozzan, T., Pizzo, P. and Greotti, E. (2020) Intracellular Calcium Dysregulation by the Alzheimer’s Disease-Linked Protein Presenilin 2. International Journal of Molecular Sciences, 21, 770.
https://doi.org/10.3390/ijms21030770
[136] Mitsumori, R., Sakaguchi, K., Shigemizu, D., Mori, T., Akiyama, S., Ozaki, K., Niida, S. and Shimoda, N. (2020) Lower DNA Methylation Levels in CpG Island Shores of CR1, CLU, and PICALM in the Blood of Japanese Alzheimer’s Disease Patients. PLOS ONE, 15, e0239196.
https://doi.org/10.1371/journal.pone.0239196
[137] Fratiglioni, L., Marseglia, A. and Dekhtyar, S. (2020) Ageing without Dementia: Can Stimulating Psychosocial and Lifestyle Experiences Make a Difference? The Lancet Neurology, 19, 533-543.
https://doi.org/10.1016/S1474-4422(20)30039-9
[138] McGrattan, A.M., McGuinness, B., McKinley, M.C., Kee, F., Passmore, P., Woodside, J.V. and McEvoy, C.T. (2019) Diet and Inflammation in Cognitive Ageing and Alzheimer’s Disease. Current Nutrition Reports, 8, 53-65.
https://doi.org/10.1007/s13668-019-0271-4
[139] Bray, F., Laversanne, M., Weiderpass, E. and Soerjomataram, I. (2021) The Ever-Increasing Importance of Cancer as a Leading Cause of Premature Death Worldwide. Cancer, 127, 3029-3030.
https://doi.org/10.1002/cncr.33587
[140] Laconi, E., Marongiu, F. and DeGregori, J. (2020) Cancer as a Disease of Old Age: Changing Mutational and Microenvironmental Landscapes. British Journal of Cancer, 122, 943-952.
https://doi.org/10.1038/s41416-019-0721-1
[141] Marbun, V.M.G., Erlina, L. and Lalisang, T.J.M. (2022) Genomic Landscape of Pathogenic Mutation of APC, KRAS, TP53, PIK3CA, and MLH1 in Indonesian Colorectal Cancer. PLOS ONE, 17, e0267090.
https://doi.org/10.1371/journal.pone.0267090
[142] Madakashira, B.P. and Sadler, K.C. (2017) DNA Methylation, Nuclear Organization, and Cancer. Frontiers in Genetics, 8, Article No. 76.
https://www.frontiersin.org/articles/10.3389/fgene.2017.00076
https://doi.org/10.3389/fgene.2017.00076
[143] Pfeifer, G.P. (2018) Defining Driver DNA Methylation Changes in Human Cancer. International Journal of Molecular Sciences, 19, 1166.
https://doi.org/10.3390/ijms19041166
[144] Tang, Q., Cheng, J., et al. (2016) Blood-Based DNA Methylation as Biomarker for Breast Cancer: A Systematic Review. Clinical Epigenetics, 8, 115.
https://doi.org/10.1186/s13148-016-0282-6
[145] Nassar, F.J., et al. (2021) Methylated Circulating Tumor DNA as a Biomarker for Colorectal Cancer Diagnosis, Prognosis, and Prediction. Clinical Epigenetics, 13, 111.
https://doi.org/10.1186/s13148-021-01095-5
[146] Wang, Y., et al. (2007) Identification of Epigenetic Aberrant Promoter Methylation of RASSF1A in Serum DNA and Its Clinicopathological Significance in Lung Cancer. Lung Cancer, 56, 289-294.
https://doi.org/10.1016/j.lungcan.2006.12.007
[147] Krittanawong, C., Khawaja, M., Rosenson, R.S., Amos, C.I., Nambi, V., Lavie, C.J. and Virani, S.S. (2022) Association of PCSK9 Variants with the Risk of Atherosclerotic Cardiovascular Disease and Variable Responses to PCSK9 Inhibitor Therapy. Current Problems in Cardiology, 47, Article ID: 101043.
https://doi.org/10.1016/j.cpcardiol.2021.101043
[148] Wagner, N. and Wagner, K.-D. (2023) Pharmacological Utility of PPAR Modulation for Angiogenesis in Cardiovascular Disease. International Journal of Molecular Sciences, 24, 2345.
https://doi.org/10.3390/ijms24032345
[149] Balcerzyk-Matić, A., Nowak, T., Mizia-Stec, K., Iwanicka, J., Iwanicki, T., Bańka, P., Jarosz, A., Filipecki, A., Żak, I., Krauze, J. and Niemiec, P. (2022) Polymorphic Variants of AGT, ABCA1, and CYBA Genes Influence the Survival of Patients with Coronary Artery Disease: A Prospective Cohort Study. Genes, 13, 2148.
https://doi.org/10.3390/genes13112148

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.