Abstract
Even though they would have great benefit across research and clinical fields, currently there are no accepted biomarkers of frailty. Cross-sectional studies in humans have identified promising candidates including inflammatory markers such as IL-6, immune markers such as WBC count, clinical markers such as albumin, endocrine markers such as vitamin D, oxidative stress markers such as isoprostanes, proteins such as BDNF and epigenetic markers such as DNA methylation, but there are limitations to the current state of the research. Future approaches to the identification of frailty biomarkers should include longitudinal studies, studies using animal models of frailty, studies incorporating novel biomarkers combined into composite panels, and studies investigating sex differences and potential overlap between markers of biological age and frailty.
Keywords: Aging, biological age, animal models, sex differences, epigenetics, deficit accumulation
1. Introduction
Frailty is an age-related condition that increases an individual’s vulnerability to adverse outcomes (Clegg et al. 2013). Those who are frail have a greater risk of institutionalization, hospitalization and mortality (Collard et al. 2012). Frailty is an expression of the heterogeneity of the pace of aging between individuals of the same chronological age and serves as a set of non-invasive assessments of future health and lifespan.
There are two main ways of assessing frailty in humans: the frailty ‘phenotype’ (Fried et al. 2001) and the frailty index (Mitnitski et al. 2001). The frailty phenotype includes exhaustion, weakness, slow walking speed, low physical activity and unintentional weight loss, and using this assessment, frailty is typically defined as three or more of these criteria (Fried et al. 2001). The frailty phenotype is often considered a measure of ‘physical’ frailty (Cesari et al. 2014). The frailty index, by contrast, is considered a measure of overall decline in health with age due to the accumulation of health-related deficits (Rockwood and Mitnitski 2007). These include diseases, abnormal laboratory results, functional assessments or self-reported health status. The number of deficits a person has is divided by the total deficits that were measured to yield a frailty score between 0 and 1, where a higher score indicates a greater degree of frailty (Mitnitski et al. 2001).
The definition of a biomarker is ‘a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention’ (Strimbu and Tavel 2010) (Strimbu 2010). Even though blood or urine biomarkers of frailty would have widespread use in both the clinical and research fields, no biomarkers are currently agreed upon by regulatory authorities, clinicians or academics (Calvani et al. 2015). Frailty biomarkers would enable the identification of those who are frail earlier, and allow for greater tracking of frailty over time, better optimization of treatments and interventions, and easier monitoring of whether interventions are working (Figure 1). There are some promising candidates for frailty biomarkers including inflammatory markers such as interleukin (IL)-6 and C reactive protein (CRP), immune markers such as white blood cell (WBC) count, clinical markers such as albumin and fibrinogen, hormone markers such as testosterone and vitamin D, markers of oxidative damage such as isoprostanes, proteins such as brain-derived neurotrophic factor (BDNF) and Sirtuin 1 (SIRT1), and epigenetic and genetic markers such as DNA methylation, telomere length and miRNAs.
Given work in this area began more than 20 years ago, why don’t we have any clinically accepted biomarkers? Possible reasons include the complex pathophysiology of frailty, the lack of a solid definition of frailty and the clinical heterogeneity of aging (Calvani et al. 2015). Additionally, almost no studies have used frailty biomarkers to measure the response to interventions, and very few have done longitudinal biomarker studies. In addition, the reproducibility between studies is low. Potential solutions to these problems include the use of panels of novel biomarkers to address complexity, performing additional longitudinal interventional studies using frailty biomarkers and the frailty index to improve reproducibility and validation, the use of animal models of frailty to optimize approaches and find novel biomarkers, a greater consideration of sex differences, and a focus on the intersection of biological age and frailty.
2. Biomarkers of frailty in human studies
The term “frailty” was used throughout the 20th century to describe people with low resilience, but only since the early 2000s has a concerted effort been placed on identifying universal frailty biomarkers (Ferrucci et al. 2002). Many have now been proposed and some of them explored in large clinical studies (Tables 1 and 2). Many of these show positive associations with frailty, including markers across the inflammatory system (eg. CRP and IL-6), immune system (eg. WBC count), endocrine system (eg. vitamin D), clinical blood markers (eg. albumin), proteins (eg. BDNF), markers of oxidative damage (eg. isoprostanes) and epigenetic and genetic markers (eg. CpG island methylation). This review is limited to the discussion of molecular biomarkers of frailty that can be detected in blood, although there is also evidence for associations with frailty of functional biomarkers such as six-minute walk time and grip strength (Boxer et al. 2010).
Table 1.
Proposed inflammatory, immune and endocrine blood biomarkers of frailty studied in humans
Biomarker group | Biomarker | Frailty assessment tool | Populations | Study design | Direction of association | Comments | References |
---|---|---|---|---|---|---|---|
Inflammatory markers | CRP | Phenotype Frailty Index FRAIL scale |
China, Germany, Wales, Spain, USA, Australia, Norway, UK, Netherlands, HIV+ men, CVD (n=90–3778) | Cross-sectional Longitudinal (3–9 years) |
↑ | Longitudinal association seen for women only not seen (Reiner 2009, Baylis 2013). | (Walston et al. 2002; Puts, Visser, et al. 2005; Barzilay et al. 2007b; Wu et al. 2009; Hubbard et al. 2009; Ronning et al. 2010; Almeida et al. 2012; Carcaillon et al. 2012; Collerton et al. 2012; Saum et al. 2015; Erlandson et al. 2017; Margolick et al. 2017; Marcos-Perez et al. 2018; Zhu et al. 2019) |
TNF-alpha | Phenotype Frailty Index |
Italy, Wales, Norway, Spain, UK, AIDs cohort, MCI and AD (n=62–1326) | Cross-sectional Longitudinal (1 year) |
↑ | (Hubbard et al. 2009; Serviddio et al. 2009; Ronning et al. 2010; Collerton et al. 2012; Piggott et al. 2015; Tay et al. 2016; Marcos-Perez et al. 2018) | ||
IL-6 | Phenotype Frailty Index Leuven Oncogeriatric Frailty Score |
USA, Wales, Norway, Taiwan, Spain, UK, China, Australia, Breast cancer patients, AIDS cohort, HIV+ men (n=110–1919) | Cross-sectional | ↑ | Not seen by Puts et al 2015, Rusanova et al 2018 or Hsu et al 2017 |
(Leng et al. 2007, 2011; Hubbard et al. 2009; Ronning et al. 2010; Collerton et al. 2012; Darvin et al. 2014; Brouwers et al. 2015; Piggott et al. 2015; Lu et al. 2016; Lee et al. 2016; Liu et al. 2016; Erlandson et al. 2017; Hsu et al. 2017; Ma et al. 2018; Marcos-Perez et al. 2018) | |
CXCL10 | Phenotype | USA (n=32) | Cross-sectional | ↑ | (Qu et al. 2009) | ||
sICAM-1 | Phenotype | Taiwan (n=946) | Cross sectional | ↑ | (Lee et al. 2016) | ||
IL-8 | Phenotype Frailty index |
Australia (n=750) | Cross sectional Longitudinal (3 years) |
↑ | Not seen by Rusanova et al 2018 | (Hsu et al. 2017) | |
MCP-1 | Phenotype Frailty index |
China (n=63–306) | Cross sectional | ↑ | (Lu et al. 2016; Su et al. 2017; Yousefzadeh et al. 2018) | ||
RANTES | Phenotype Frailty Index |
China (n=76) | Cross sectional | ↑ | (Lu et al. 2016) | ||
Erythrocyte sedimentation rate | Frailty Index | Italy (n=594) | Cross sectional | ↑ | (Fontana et al. 2013) | ||
Immune markers | WBC | Phenotype Frailty Index |
USA, Italy, UK (n=254–1106) | Cross sectional Longitudinal (10 years) |
↑ | (Leng et al. 2007; Baylis et al. 2013; Fontana et al. 2013) | |
Lymphocyte count | Frailty Index | Italy (n=594) | Cross sectional | ↓ | (Fontana et al. 2013) | ||
Neutrophil count | Phenotype Frailty Index |
UK, USA (n=845–1106) | Cross sectional | ↑ | (Leng et al. 2009; Collerton et al. 2012) | ||
Monocyte count | Phenotype | USA (n=1106) | Cross sectional | ↑ | (Leng et al. 2009) | ||
PBMC proliferation | Phenotype | USA (n=22) | Cross sectional | ↓ | (Leng et al. 2004) | ||
Neopterin | Phenotype | USA (n=133) | Cross sectional | ↑ | (Leng et al. 2011) | ||
CD4+/CD8 + ratio | Phenotype Frailty Index |
China, USA, Spain (n=52–259) | Cross sectional | ↓ or ↑ | Not seen by Erlandson et al 2017 or Collerton 2012. | (Semba et al. 2005; De Fanis et al. 2008; Lu et al. 2016; Marcos-Perez et al. 2018) | |
%CD19+ cells | Phenotype | Spain (n=259) | Cross-sectional | ↓ | (Marcos-Perez et al. 2018) | ||
Hormones/endocrine markers | Adiponectin | Phenotype | Taiwan, China (n=130–189) | Cross-sectional | ↑ | Stronger association in males than females | (Tsai et al. 2013; Ma et al. 2018) |
Testosterone | Phenotype Frailty Index |
Italy HIV+ men (n=310–594) | Cross-sectional | ↓ | Only seen in men (Fontana et al 2013) | (Fontana et al. 2013; Erlandson et al. 2017) | |
DHEA-S | Phenotype Frailty Index |
Italy HIV+ men UK (n=254–594) | Cross-sectional Longitudinal (10 years) |
↓ | Only seen in men (Fontana et al 2013) | (Baylis et al. 2013; Fontana et al. 2013; Erlandson et al. 2017) | |
Triiodothyronine | Frailty Index | Italy (n=112–594) | Cross-sectional | ↓ | (Fontana et al. 2013; Bertoli et al. 2017) | ||
IGF-1 | Frailty Index | Italy (n=594) | Cross-sectional | ↓ | Not seen for Brouwers et al 2015 or Puts 2005. | (Fontana et al. 2013) | |
Estradiol | Phenotype | Spain (n=702) | Cross-sectional | ↑ | (Carcaillon et al. 2012) | ||
HOMA-IR | Phenotype | USA, HIV+ men (n=310–3141) | Longitudinal (9 years) | ↑ | (Barzilay et al. 2007b; Erlandson et al. 2017) | ||
Leptin | Phenotype Frailty Index |
Diabetes, China (n=76–1573) | Cross-sectional Longitudinal (4 years) |
↑ | (Lu et al. 2016; Lana et al. 2017) | ||
Vitamin D | Phenotype Frailty Index |
Spain, Germany ACS Netherlands (n=342–1271) | Cross-sectional Longitudinal (3 years) |
↓ | (Puts, Visser, et al. 2005; Alvarez-ríos et al. 2015; Pabst et al. 2015; Sanchis et al. 2015) |
CRP, C reactive protein; CXCL10, C-X-C Motif Chemokine Ligand 10; sIACM1, soluble intercellular adhesion molecule-1; MCP-1, monocyte chemoattractant protein 1; IL, interleukin; TNF, tumor necrosis factor; RANTES, regulated on activation, normal T cell expressed and secreted; WBC, white blood cells; DHEA-S, dehydroepiandrosterone; HOMA-IR, Homeostatic model assessment of insulin resistance; HIV+, human immunodeficiency virus positive; AD, Alzheimers disease; CVD, cardiovascular disease; MCI, mild cognitive impairment.
Table 2.
Proposed clinical, protein, oxidative stress and genetic/epigenetic blood biomarkers of frailty studied in humans
Biomarker group | Biomarker | Frailty assessment tool | Populations | Study design | Direction association | Comments | References | |
---|---|---|---|---|---|---|---|---|
Clinical markers | t-PA | Phenotype | USA (n=1800) | Longitudinal (3 years) | ↑ | (Reiner et al. 2009) | ||
Factor VIII | Phenotype | USA (n=4735) | Cross sectional | ↑ | (Walston et al. 2002) | |||
D-dimer | Phenotype | USA, Norway
ACS (n=187–4735) |
Cross sectional Longitudinal (3 years) |
↑ | (Walston et al. 2002; Reiner et al. 2009; Ronning et al. 2010; Sanchis et al. 2015) | |||
Albumin | Phenotype FRAIL scale Frailty Index | China, Wales, Italy, UK, Dialysis (n=46–594) | Cross sectional | ↓ | (Walston et al. 2002; Hubbard et al. 2009; Wu et al. 2009; Collerton et al. 2012; Fontana et al. 2013; Chao et al. 2015; Sanchis et al. 2015) | |||
Iron/Ferritin | Frailty Index FRAIL scale | Italy, Dialysis (n=46–594) | Cross sectional | ↓or ↑ | (Fontana et al. 2013; Chao et al. 2015) | |||
Total cholesterol, LDL-c | Frailty Index | Italy (n=594) | Cross sectional | ↓ | (Fontana et al. 2013) | |||
Creatinine | FRAIL scale Phenotype | Dialysis, ACS (n=46–342) | Cross sectional | ↓ | (Chao et al. 2015; Sanchis et al. 2015) | |||
Haemoglobin | Phenotype | ACS (n=342) | Cross sectional | ↓ | (Sanchis et al. 2015) | |||
Fibrinogen | Phenotype | UK USA (n=2146–4735) | Longitudinal (4 years) Cross sectional |
↑ | Not seen for Sanchis et al 2015, or only seen for women (Gale 2013) | (Walston et al. 2002; Shamsi et al. 2012; Gale et al. 2013; Darvin et al. 2014) | ||
Haptoglobin | Phenotype | USA (n=73) | Cross sectional | ↑ | Not seen for Darvin et al 2014 | (Shamsi et al. 2012) | ||
Transferrin | Phenotype | USA (n=65–73) | Cross sectional | ↑ | (Shamsi et al. 2012; Darvin et al. 2014) | |||
ALT | Phenotype | Australia (n=1673) | Cross sectional | ↓ | (Le Couteur et al. 2010) | |||
Proteins/peptides | Proteomics | Phenotype | Taiwan (n=12) | Cross sectional | ↑ & ↓ | (Lin et al. 2017) | ||
PINP | Phenotype | Spain (n=592) | Cross sectional | ↑ | (Alvarez-ríos et al. 2015) | |||
LpPLA2 mass | Phenotype | UK (n=1919) | Cross sectional | ↑ | (Liu et al. 2016) | |||
Cystatin-C | Phenotype | ACS (n=342) | Cross sectional | ↓ | (Sanchis et al. 2015) | |||
CML | Phenotype | USA (n=3373) | Cross sectional | ↑ | Only seen in men. | (Whitson et al. 2014) | ||
BDNF | Phenotype | Brazil (n=48) | Cross sectional | ↓ | (Coelho et al. 2019) | |||
Sirt 1 and 3 | Phenotype | India (n=200) | Cross sectional | ↓ | (Kumar et al. 2016) | |||
Klotho | Phenotype | Italy (n=774) | Longitudinal (3 years) | ↓ | (Shardell et al. 2017) | |||
HtrA1 | Phenotype Frailty Index | Italy (n=120) | Cross sectional | ↑ | (Lorenzi et al. 2016) | |||
Oxidative stress | 8-OHdG | Phenotype | China (n=90) | Cross sectional | ↑ | (Wu et al. 2009) | ||
d-ROM | Phenotype | Germany (n=3124) | Cross sectional | ↑ | (Saum et al. 2015) | |||
TTL | Phenotype | Germany (n=3124) | Cross sectional | ↓ | (Saum et al. 2015) | |||
GSSG | Phenotype | Italy (n=62) | Cross sectional | ↑ | (Serviddio et al. 2009) | |||
NHE- adducts | Phenotype | Italy (n=62) | Cross sectional | ↑ | (Serviddio et al. 2009) | |||
MDA | Phenotype | Italy, Spain (n=62–742) | Cross sectional | ↑ | (Serviddio et al. 2009; Inglés et al. 2014) | |||
Protein carbonylation | Phenotype | Spain (n=742) | Cross sectional | ↑ | (Inglés et al. 2014) | |||
Isoprostanes | Phenotype | UK (n=1919) | Cross sectional | ↑ | Not seen by Collerton et al 2012. | (Liu et al. 2016) | ||
Genetic and epigenetic markers | SNPs | FRAIL scale Phenotype Frailty Index |
Australia, UK, USA (n=326–3778) | Cross sectional | ↑ | (Matteini et al. 2010; Almeida et al. 2012; Mekli et al. 2015, 2016) | ||
Telomere length | FRAIL scale Phenotype Frailty Index |
Finland (n=1078) | Cross sectional Longitudinal (10 years) |
↓ | Not seen for Zhou et al 2018 or Collerton et al 2012 | (Haapanen et al. 2018; Zhou et al. 2018) | ||
cfDNA | Phenotype | Finland (n=144) | Cross sectional | ↑ | (Jylhava et al. 2013) | |||
mtDNA copy number | Phenotype | Finland (n=144) | Cross sectional | ↑ | (Jylhava et al. 2013) | |||
DNA methylation | Phenotype | UK, Italy (n=318–321) | Cross sectional Longitudinal (7 years) |
↓ | (Bellizzi et al. 2012; Collerton et al. 2014) | |||
DNA methylation clock | Phenotype Frailty Index | Germany, UK (n=791–1820) | Cross sectional | ↑ | (Breitling et al. 2016; Gale et al. 2018) | |||
γH2AX | Phenotype | Spain (n=250) | Cross sectional | ↑ | (Valdiglesias et al. 2019) |
tPA, tissue plasminogen activator; ALT, alanine aminotransferase; LDL-c, low-density lipoprotein cholesterol; PINP, N-Terminal Propeptide of Type I Collagen; LpPLA2, Lipoprotein-associated phospholipase A2; HtrA1, high temperature requirement serine peptidase 1; 8-OHdG, 8-hydroxy-2’ -deoxyguanosine; d-ROM, diacron reactive oxygen metabolites; TTL, total thiol levels; GSSG, Glutathione disulfide; NHE, sodium hydrogen exchanger; MDA, malondialdehyde; SNP, single nucleotide polymorphism ;cfDNA, cell-free DNA; mtDNA, mitochondrial DNA; γH2AX, phosphorylated H2A histone family member X, ACS, acute coronary syndrome.
2.1. Inflammatory biomarkers of frailty
The most extensively studied biomarkers of frailty have been those involved with inflammation. Increased inflammation is widely considered a cause of frailty and the pathophysiology of both frailty and inflammation are closely aligned (Hubbard and Woodhouse 2010). Robust correlates of frailty include increased CRP, IL-6 and tumor necrosis-factor (TNF)- α, but some studies have also shown correlations between frailty and other inflammatory markers including C-X-C motif chemokine 10 (CXCL10), neopterin, IL-8 and monocyte chemoattractant protein 1 (MCP-1).
CRP is synthesized by the liver in response to inflammatory factors released by macrophages and T-cells (Pepys and Hirschfield 2003). CRP is associated with frailty in a variety of cross-sectional population studies (Table 1), including in HIV positive men and patients with cardiovascular disease (Walston et al. 2002; Wu et al. 2009; Hubbard et al. 2009; Ronning et al. 2010; Almeida et al. 2012; Carcaillon et al. 2012; Collerton et al. 2012; Saum et al. 2015; Erlandson et al. 2017; Margolick et al. 2017; Marcos-Perez et al. 2018; Zhu et al. 2019). These results are consistent across studies in many different countries and with both the frailty phenotype and frailty index approaches (Table 1).
In longitudinal studies, however, the association between CRP and frailty is less clear. A meta-analysis of inflammatory biomarkers and frailty found that although increased CRP levels were associated with frailty cross-sectionally, in longitudinal studies there was no association (Soysal et al. 2016). This study included three longitudinal studies, one of which found a modest positive association between serum CRP and the onset of frailty with a three year follow up (Puts, Visser, et al. 2005). Two additional longitudinal studies have also found an association between serum CRP at baseline and incident frailty (Barzilay et al. 2007a; Gale et al. 2013), though in one of these studies the association was only seen in women (Gale et al. 2013). All of these studies were completed in similarly aged cohorts (60–70+ years) and all but one (Puts, Visser, et al. 2005) used the frailty phenotype approach. The reasons for these different findings, therefore, are unclear. Although serum CRP levels have a clear association with frailty, determination of the value of CRP as a predictive biomarker of frailty will require further longitudinal studies, perhaps using the frailty index approach.
IL-6 is a pro-inflammatory cytokine that has also been extensively studied as a serum biomarker of frailty. Similarly to CRP, many studies across a variety of countries and in different populations identify cross-sectional associations between increased serum IL-6 levels and frailty (Leng et al. 2007, 2011; Hubbard et al. 2009; Ronning et al. 2010; Collerton et al. 2012; Darvin et al. 2014; Brouwers et al. 2015; Piggott et al. 2015; Lu et al. 2016; Lee et al. 2016; Liu et al. 2016; Erlandson et al. 2017; Hsu et al. 2017; Ma et al. 2018; Marcos-Perez et al. 2018). However, not all studies observe this same association (Tay et al. 2016; Rusanova et al. 2018), and importantly the few longitudinal studies completed do not observe a relationship between frailty and IL-6 (Puts, Visser, et al. 2005; Soysal et al. 2016; Hsu et al. 2017). Hsu et al (2017) completed both a cross-sectional and then prospective study, and although they found an association between frailty status and IL-6 levels at baseline, there was no predictive association found between IL-6 levels and incident frailty over three years (Hsu et al. 2017). One study explored the effect of L-cartinine supplementation on IL-6 as a biomarker of frailty in a small clinical trial but found no significant effect (Press 2016).
TNF-α is a pro-inflammatory cytokine that is primarily produced by activated macrophages (Parameswaran and Patial 2010). Cross-sectional studies in a range of populations also show an association between increased serum TNF-α levels and frailty, assessed using both the phenotype and frailty index (Hubbard et al. 2009; Serviddio et al. 2009; Ronning et al. 2010; Collerton et al. 2012; Piggott et al. 2015; Tay et al. 2016; Marcos-Perez et al. 2018). Rusanova et al (2018) did not see an association between frailty status and serum TNF-α levels, although they did see an association between the TNF-α/IL-10 ratio and frailty (Rusanova et al. 2018). One longitudinal study found that although no individual inflammatory markers predicted onset of frailty one year later, a general pro-inflammatory state was predictive (Tay et al. 2016).
Associations between frailty status and other inflammatory markers have also been observed but these have been less well studied. In a small case-control study, the expression levels of CXC chemokine ligand 10 (CXCL-10), a pro-inflammatory mediator, were increased in those who were frail compared to non-frail (Qu et al. 2009). Levels of soluble intercellular adhesive molecule 1 (sICAM-1), which has been implicated in vascular inflammation, are increased with frailty in a Taiwanese community-dwelling population (Lee et al. 2016), and the chemokine RANTES (regulated on activation, normal T cell expressed and secreted) was increased with frailty in a small cross-sectional study of Chinese adults assessed with both the phenotype and frailty index approaches (Lu et al. 2016). Hsu et al (2017) found increased IL-8 levels were associated with incident frailty as assessed with both the phenotype and index, in a longitudinal study of older Australian men (Hsu et al. 2017), although Rusanova et al (2018) did not see a cross-sectional relationship between IL-8 and frailty. Monocyte chemoattractant protein 1 (MCP-1) has been recently identified as a potential inflammatory biomarker of frailty, and three cross-sectional studies have shown associations between increased MCP-1 levels and frailty (Lu et al. 2016; Su et al. 2017; Yousefzadeh et al. 2018). Erythrocyte sedimentation rate is a commonly used clinical marker of inflammation, that was also associated with frailty in a large hospitalized Italian population (Fontana et al. 2013).
Overall, there appears to be a strong relationship between frailty and inflammatory markers, especially IL-6, CRP and TNF-α, but more longitudinal studies, interventional studies and studies using the frailty index are needed to determine their potential as clinical biomarkers.
2.2. Immune biomarkers of frailty
Dysfunction and dysregulation of the immune system has also been implicated as a potential mechanism related to the development of frailty (Li et al. 2011). For this reason, immune measures have been explored as frailty biomarkers. Positive associations between frailty and white blood cell (WBC) counts have been observed in many cross-sectional studies (Table 1) and one longitudinal study (Leng et al. 2007; Baylis et al. 2013; Fontana et al. 2013). A meta-analysis also concluded that there was an association between higher WBC counts and frailty, although it made no conclusion about the predictive value of WBC counts (Soysal et al. 2016). Increased neutrophil count has also been cross-sectionally associated with frailty (Leng et al. 2009; Collerton et al. 2012), as has increased monocyte count (Leng et al. 2009), although this study was only completed in a female population. Neopterin is a metabolite primarily produced by monocytes and macrophages which is considered a marker of immune activation. Increased neopterin levels were associated with frailty in a small study (n=133) of community dwelling people aged 72 or older (Leng et al. 2011), and higher neopterin has also been associated with higher mortality risk (Oxenkrug et al. 2011).
Changes in the adaptive immune system with frailty seem less clear. An association between decreased lymphocyte count and frailty has been demonstrated (Fontana et al. 2013), as has a reduction in peripheral blood mononuclear cell (PBMC) proliferation after lipopolysaccharide (LPS) stimulation (Leng et al. 2004) and a decrease in the percent of CD19+ B cells in frail patients (Marcos-Perez et al. 2018). Studies have demonstrated either an increase (Lu et al. 2016; Marcos-Perez et al. 2018), decrease (Semba et al. 2005; De Fanis et al. 2008) or no change in the ratio of CD4+ to CD8+ cells in frailty (Collerton et al. 2012; Erlandson et al. 2017). Clearly more studies are needed to understand the potential for adaptive immune system markers as biomarkers of frailty.
2.3. General clinical biomarkers of frailty
Several factors that are analyzed in routine clinical blood tests have been proposed as biomarkers of frailty such as low levels of albumin, dysregulated blood coagulation and thrombosis markers such as D-dimer and fibrinogen (Table 2). Other blood markers, such as those related to anemia, also show promise as biomarkers, but have been less extensively studied.
Cross-sectional studies in a range of populations using both the phenotype and deficit accumulation approaches, have shown that low levels of albumin are associated with frailty (Walston et al. 2002; Hubbard et al. 2009; Wu et al. 2009; Collerton et al. 2012; Fontana et al. 2013; Chao et al. 2015; Sanchis et al. 2015). As far as we are aware, the longitudinal predictive value of albumin for frailty has not yet been investigated.
Frailty is also associated with increases in blood markers of thrombosis, including tissue plasminogen activator (t-PA), fibrinogen, D-dimer and factor VIII. Several cross-sectional studies have shown an association with increased fibrinogen levels (Walston et al. 2002; Shamsi et al. 2012; Darvin et al. 2014), although not all studies have seen this association (Sanchis et al. 2015). One longitudinal study found an association between baseline increased fibrinogen and incident frailty after four years, but only in women (Gale et al. 2013), although another longitudinal study saw no association (Reiner et al. 2009). Increased levels of D-dimer, a fibrin degradation product, have also been associated with frailty in cross-sectional studies (Walston et al. 2002; Ronning et al. 2010; Sanchis et al. 2015) and one longitudinal study (Reiner et al. 2009). Levels of the coagulation protein factor VIII and t-PA also appear to increase with frailty (Walston et al. 2002; Reiner et al. 2009).
Proteins related to anemia have been identified as possible frailty biomarkers. Low levels of hemoglobin and iron are associated with frailty in cross-sectional studies (Fontana et al. 2013; Sanchis et al. 2015), although another study in dialysis patients found that higher levels of ferritin were associated with frailty (Chao et al. 2015). Two small studies of community-dwelling adults explored the association between frailty and haptoglobin, which binds free hemoglobin and indicates inflammation, and transferrin, which binds and transports iron and indicates anemia (Shamsi et al. 2012; Darvin et al. 2014). Increased transferrin was associated with frailty in both studies, while haptoglobin was only associated with frailty in one study (Shamsi et al. 2012).
Other clinical blood markers may also be associated with frailty. Studies have found associations between frailty and low alanine aminotransferase (ALT) levels (Le Couteur et al. 2010), low total cholesterol and LDL-c levels (Fontana et al. 2013) and decreased levels of creatinine, a marker of low muscle mass (Chao et al. 2015; Sanchis et al. 2015).
Routine clinical blood tests including albumin and markers of thrombosis show promise as biomarkers of frailty, especially given the ease of assessment and convenience of incorporating these frailty markers into clinical practice. However, given the non-specific nature of these markers, they will likely be most informative as part of a panel of frailty biomarkers (Section 3.4).
2.4. Hormone and endocrine biomarkers of frailty
Another area of focus in the field of frailty biomarkers is the endocrine system (Table 1). Hormones such as vitamin D and testosterone have been studied as both potential biomarkers, and also treatments or interventions for frailty. As interventions they have shown limited success (Cherniack et al. 2007; Kenny et al. 2010), however many studies have shown an association between low levels of vitamin D and frailty (Puts, Visser, et al. 2005; Alvarez-ríos et al. 2015; Pabst et al. 2015; Sanchis et al. 2015). Although the majority of these studies were cross-sectional, Puts et al (2015) showed a longitudinal relationship between low vitamin D and incident frailty over three years. A cross-sectional relationship between low testosterone and frailty has also been shown in males (Fontana et al. 2013; Erlandson et al. 2017).
Other endocrine biomarkers of frailty have also been explored, but less extensively. Increased adiponectin, a hormone involved in glucose regulation, is associated with frailty (Tsai et al. 2013; Ma et al. 2018), although this association appears to be stronger in males than females (Tsai et al. 2013). Increased levels of leptin, a hormone that inhibits hunger to maintain energy balance, are associated with frailty in both cross-sectional and longitudinal studies (Lu et al. 2016; Lana et al. 2017). Insulin-like growth factor 1 (IGF1) was shown to be decreased with frailty in one study (Fontana et al. 2013), although this association wasn’t seen in two other studies (Puts, Lips, et al. 2005; Brouwers et al. 2015).
The homeostatic model assessment of insulin resistance (HOMA-IR), which is calculated from fasting glucose and insulin levels, is increased in frailty (Barzilay et al. 2007a; Erlandson et al. 2017) as are fasting insulin levels (Walston et al. 2002). Decreased levels of triiodothyronine (T3), a thyroid hormone with roles in physiological maintenance, are associated with frailty (Fontana et al. 2013; Bertoli et al. 2017). Estradiol levels are increased with frailty in women (Carcaillon et al. 2012), and levels of the steroid hormone dehydroepiandrosterone sulfate (DHEA-s) are decreased in frailty (Baylis et al. 2013; Erlandson et al. 2017), although for one study this association was only seen in males (Fontana et al. 2013).
Hormone and endocrine markers, particularly Vitamin D levels, show promise as potential biomarkers of frailty, although as with other markers, more longitudinal studies would be needed to confirm their predictive and diagnostic value. Sex-specific associations of some of these endocrine markers with frailty should also be further explored.
2.5. Protein and peptide biomarkers of frailty
There has been growing focus recently on the characterization of secreted proteins and peptides that change during aging and in age-specific disease states (Morimoto and Cuervo 2014; Tanaka et al. 2018). So far, however, only one study has looked for correlations between changes to the secretome and frailty. This small study (n=12) found 31 proteins increased with frailty, including angiotensinogen (ANGT), kininogen-1 (KG) and antithrombin III (AT). Three proteins decreased, including albumin (Lin et al. 2017). A larger scale study to confirm the changing secreted proteome with frailty would be of great interest.
Several studies have explored the association between levels of specific blood-based proteins and frailty and some potential biomarkers have been identified (Table 2). A study using data from women enrolled in the Toledo Study for Healthy Aging found that levels of N-terminal pro-peptide of type I procollagen (PINP), a measure of collagen and bone turnover, were increased with frailty (Alvarez-ríos et al. 2015). Lipoprotein phospholipase A2 (LpPLA2), a marker of cardiovascular risk, was increased with frailty in a large study of older adults (Liu et al. 2016) and cystatin C, a marker of renal function, was associated with frailty in acute coronary syndrome (ACS) patients (Sanchis et al. 2015). High-temperature requirement serine protease A1 (HtrA1), an enzyme that regulates IGF1 levels and is involved in inflammatory processes, is increased with frailty (Lorenzi et al. 2016). Levels of carboxymethyl-lysine (CML), an advanced glycation end-product, are increased with frailty, but only in males (Whitson et al. 2014). Brain-derived neurotrophic factor (BDNF) levels were decreased with frailty in a small study of Brazilian women (Coelho et al. 2019), as were Klotho levels in a large longitudinal study in Italy (Shardell et al. 2017) and Sirt1 and Sirt3 levels in a cross-sectional study in India (Kumar et al. 2016). Levels of BDNF, klotho and Sirt1 have also been shown to decrease in aging (Kuro-o 2009; Erickson et al. 2010; Braidy 2011).
2.6. Oxidative stress biomarkers of frailty
Markers of oxidative damage and stress have also been associated with frailty and suggested as potential biomarkers (Table 2). Clinically used oxidative stress markers including 8-hydroxy-2’-deoxyguanosine (8-OHdG), a marker of oxidative damage to DNA; protein carbonylation, a general measure of oxidative stress; and diacron reactive oxygen metabolites (d-ROMs), a test that measures hydroperoxides in serum, are all increased with frailty (Wu et al. 2009; Inglés et al. 2014; Saum et al. 2015). Additionally, increased levels of markers and products of fatty acid peroxidation including isoprostanes, malondialdehyde (MDA) and 4-hydroxynonenal (HNE) are also associated with frailty (Serviddio et al. 2009; Inglés et al. 2014; Liu et al. 2016). Total thiol levels (TTL), a marker of redox status, are reduced in frailty (Saum et al. 2015), and glutathione disulfide (GSSG), the oxidised form of glutathione, is increased, but without any change to glutathione levels (GSH) (Serviddio et al. 2009).
Further confirmation of these potential oxidative stress markers of frailty is needed, especially as all studies were cross-sectional, and none were completed using the frailty index approach.
2.7. Epigenetic and genetic markers
Genetic markers of frailty have been identified and proposed as potential biomarkers (Table 2). Single nucleotide polymorphisms (SNPs) of genes including CRP, IL-18 TCN2 and IL-12 have been associated with frailty (Matteini et al. 2010; Almeida et al. 2012; Mekli et al. 2015, 2016) but SNPs are limited as biomarkers for anything other than identifying increased risk.
Telomere length has been proposed as a potential frailty biomarker. The association between telomere length and frailty, however, is controversial. Some studies have found an association between frailty and leukocyte telomere length (Collerton et al. 2012; Pathai et al. 2013; Brault et al. 2014), including one longitudinal study (Haapanen et al. 2018), while others have seen no association (Woo et al. 2008; Marzetti et al. 2014; Saum et al. 2014; Yu et al. 2015). A recent meta-analysis determined that there was no association between telomere length and frailty (Zhou et al. 2018). The authors suggest that since an association with telomere length was only observed in two studies using the frailty phenotype, that “telomere length may reflect reduction in muscle mass and muscle performance better than frailty” (Zhou et al. 2018). Total plasma cell-free DNA (cfDNA) levels and mitochondrial DNA (mtDNA) copy number have also been associated with frailty in a cross-sectional study of nonagenarians (Jylhava et al. 2013). Prospective studies of these potential genetic markers are needed before they could be considered as clinical biomarkers.
Epigenetic changes are increasingly appreciated as a correlate and a possible driver of the aging process (Kane and Sinclair 2019). Epigenetic markers of biological age based on specific GpG island DNA methylation patterns have provided a clock by which to estimate chronological and biological age in animals and humans (Horvath 2013; Horvath and Raj 2018). Global downregulation of DNA methylation is associated with frailty both cross-sectionally and longitudinally (Bellizzi et al. 2012) and decreased CpG island methylation is also associated with frailty (Collerton et al. 2014). Two studies have reported an association between the DNA methylation clock developed by Horvath and frailty (Breitling et al. 2016; Gale et al. 2018). Furthermore phosphorylated H2A histone family member X (γH2AX), a marker of DNA damage, was also shown to be related to frailty in a recent cross-sectional study (Valdiglesias et al. 2019).
Research into the epigenetics of frailty will continue to grow over the next few years, not just for the identification of potential frailty biomarkers but also to help us understand potential underlying mechanisms of frailty and aging.
2.8. Limitations of frailty biomarker studies
Despite progress in identifying biomarkers of frailty in recent years, some major limitations remain. Some of the biggest problems in the field are that associations are not always reproduced across different populations, rarely is the predictive value of biomarkers explored in longitudinal clinical studies, and even more rarely are they investigated in regards to their ability to evaluate the response to an intervention. This is not specific to frailty biomarkers, however, and is a feature of the entire biomarkers field, with the US National Institutes of Health (NIH) recently highlighting that biomarker development lags behind therapeutics in all areas (Brady 2014).
The majority of the proposed frailty biomarker have been investigated using the ‘physical’ frailty phenotype definition, which mostly measures reduced physical function and sarcopenia with age, rather than the deficit accumulation approach, which measures overall health decline in aging. Indeed, Ramakrishnan and colleagues completed a systematic review of frailty biomarker studies, and found that 33 out of 40 identified studies used the physical frailty phenotype definition (Ramakrishnan et al. 2017). Hubbard et al (2009) suggest that the associations between inflammatory markers and the frailty phenotype may be primarily related to sarcopenia and reduced muscle strength in these patients (Hubbard et al. 2009). More clinical studies exploring frailty biomarkers using the frailty index and more studies exploring the longitudinal predictive value of frailty biomarkers, especially in response to frailty-attenuating interventions, would be of great value in identifying viable biomarkers of frailty.
Other considerations to accelerate the discovery of frailty biomarkers are discussed below, and include using animal models of frailty to reduce the complexity and heterogeneity of clinical studies, developing composite biomarker panels, considering that markers for biological age and frailty may overlap, investigating sex differences in biomarkers and identifying novel biomarkers.
3. Considerations for the development of frailty biomarkers
3.1. Animal studies of frailty biomarkers
To complement and predict human studies, animal models of frailty have been developed. The first proposed mouse frailty model was the IL-10 knockout mouse (Walston et al. 2008), which was originally developed as a model of colitis, but found to display many characteristics seen in human frailty such as increased inflammation and decreased strength (Walston et al. 2008). Recently Cu/Zn superoxide dismutase (Sod1) knock-out mice have also been proposed as a model of frailty. These mice display weight loss, weakness, reduced activity and increased inflammation (Deepa et al. 2017). Unfortunately, these transgenic models are somewhat limited in their use, as they do not model the development of frailty in natural aging.
Both the frailty index and frailty phenotype assessment tools have been adapted for and validated in mice (Banga et al. 2019). Whitehead et al (2014) developed a non-invasive clinical frailty index for use in mice, which increases with age, is associated with increased mortality risk and has many of the same characteristics as human frailty indices (Whitehead et al. 2014; Rockwood et al. 2017). About the same time, Liu et al (2014) adapted the frailty phenotype approach into mice based on walking speed, endurance, grip strength and activity assessments (Liu et al. 2014). These tools provide the opportunity to study the development and mechanisms of frailty, and test interventions to delay frailty as well as to explore potential biomarkers of frailty in preclinical models.
Some studies have already used these frailty models and tools to explore potential biomarkers of frailty (Table 3). Levels of several inflammatory cytokines including IL-6, IL-1β, TNF-α, interferon (IFN)-γ and keratinocyte chemoattractant (KC) are increased in IL-10 knockout mice which are considered frail, compared to control mice, which are considered not-frail (Walston et al. 2008; Ko et al. 2012). Sod1 knockout mice have increased levels of eotaxin and Granulocyte-colony stimulating factor (G-CSF) (Deepa et al. 2017). Yousefzadeh et al (2018) also propose the progeria mouse models Ercc1−/Δ and Bubr1H/H as possible models of frailty, although their frailty phenotype is not characterised, and find that these mice have elevated MCP-1 compared to controls (Yousefzadeh et al. 2018). Several studies have used the mouse clinical frailty index to explore possible biomarkers of frailty in aging C57BL/6 mice (Kane et al. 2016, 2017, 2018). Low levels of serum ALT, alkaline phosphatase (ALP), albumin and total protein were all associated with increasing frailty index score in 19 to 24 month-old male mice (Kane et al. 2016, 2017). Although not a blood biomarker, reduced heart rate and heart rate variability were also associated with increasing frailty score in old male C57BL/6 mice (Kane et al. 2017). Only one of these studies included female mice, and interestingly found a different profile of inflammatory biomarkers in female mice compared to male (Kane et al. 2018). Female mice have increased levels of IL-6, IL-9 and IFN-γ with increasing frailty index score, whilst males have only increasing levels of IL-12p40 with frailty (Kane et al. 2018). The mouse frailty phenotype assessment has not yet been used to explore frailty biomarkers.
Table 3.
Frailty biomarker studies in rodents
Biomarker | Animal model | Frailty assessment/model | Findings | Type of biomarker | Reference |
---|---|---|---|---|---|
MCP-1 | Wild-type and Ercc1−/Δ and Bubr1H/H mouse models | Frailty not assessed | Increased with age and accelerated in progeria models | Serum | (Yousefzadeh et al. 2018) |
ALT, ALP | Male C57BL/6 mice 24 months | Mouse Clinical Frailty Index | Lower levels associated with increasing FI Score | Serum | (Kane et al. 2016, 2017) |
HR, HR variability | Male C57BL/6 mice 24 months | Mouse Clinical Frailty Index | Lower HR and lower variability associated with increasing FI Score | Non-invasive cuff assessment | (Kane et al. 2017) |
Total protein, albumin | Male C57BL/6 mice 19–23 months | Mouse Clinical Frailty Index | Lower levels with increased FI score | Serum | (Kane et al. 2016) |
IL-6, IL-12p40, IL-9, IFN-γ | Male and female C57BL/6 mice 17–23 months | Mouse Clinical Frailty Index | Sex differences: IL-6, IL-9 and IFN-γ increased with FI score in females and IL-12p40 increased with FI score in males. | Serum | (Kane et al. 2018) |
IL-6, IL-lβ, TNF-α, IFN-γ, KC | IL-10 knock-out mice | Frailty not assessed | Higher in knock-out than in wild-type mice. | Serum | (Walston et al. 2008; Ko et al. 2012) |
G-CSF, Eotaxin | Sodi knock-out mice | Frailty not assessed | Higher in knock-out than in wild-type mice. | Serum | (Deepa et al. 2017) |
CML, ; BDNF, ; MCP-1, monocyte chemoattractant protein 1; ALT, alanine aminotransferase; ALP, alkaline phosphatase; HR, heart rate; IL, interleukin; IFN, interferon; TNF, tumor necrosis factor; KC, keratinocyte chemoattractant; G-CSF, granulocyte colony-stimulating factor; sod1, superoxide dismutase 1; FI, frailty index.
It is interesting, and encouraging, that many of the frailty biomarkers identified in humans are also seen in animal models, including IL-6, albumin, ALT and MCP-1. Research using these animal tools should accelerate the discovery of clinical frailty biomarkers, and this area will doubtless continue to grow over the next few years.
3.2. Other possible/novel biomarker options for inclusion
Cardoso and colleagues recently proposed a series of possible frailty biomarkers that were identified based on their associations with aging, age-related diseases or longevity (Cardoso et al. 2018). They identified 44 potential biomarkers across each of the hallmarks of aging pathways (Lopez-Otin et al. 2013). Nineteen of these were considered “high priority”: CXCL10, IL-6, CX3CL1, growth differentiation factor 15 (GDF15), fibronectin type III domain-containing protein 5 (FNDC5), vimentin, regucalcin, calreticulin, urokinase, angiotensin II, BDNF, progranulin, klotho, fibroblast growth factor (FGF)23, FGF21, leptin, adenosylhomocysteinase, keratin 18 and an miRNA panel. Some of these markers have already been shown to be associated with frailty (Section 2), but investigation of each of these biomarkers, both singly and in combination with frailty outcomes would be informative.
The European FRAILOMIC initiative is a large-scale project focused on the identification of novel frailty biomarkers. They have also identified potential genomic, proteomic and transcriptomic markers from their association with age-related outcomes such as grip-strength. Using the frailty phenotype approach, they will assess the association of frailty with their initial proposed biomarkers of frailty including angiotensin converting enzyme, alpha-actinin-3, ciliary neurotrophic factor, GDF8, IL-6, mitochondrial DNA (mtDNA), vitamin D receptor, protein kinase B, FoxOs, mammalian target of rapamycin (mTOR), Hypoxia-inducible factor 1, Peroxisome proliferator-activated receptor gamma coactivator 1 (PGC1), Sirt1, superoxide dismutase 2, p53, sestrin-2, angiotensin II, nitric oxide synthase 3, advanced glycation end products, soluble receptor for advanced glycation end products (sRAGE), C-C motif chemokine 11 (CCL11), lectin galactoside-binding soluble 3, Jagged-1, versican, IGFBP6, telomere length and a panel of 14 mRNAs (Erusalimsky 2016). It will be interesting to see results from this initiative in the next few years.
Other possible novel frailty biomarkers could be identified from an examination of the hallmarks of aging (Lopez-Otin et al. 2013), for example markers of cellular senescence such as p16 (REF). MicroRNAs (miRNAs) are also a growing area of interest in the aging field, and a recent study found an association of miR-21 with frailty (Rusanova et al. 2018). Additionally, growing focus on characterizing the metabolomic, proteomic and transcriptomic signatures of aging (Kirkwood 2011) will likely identify new, innovative biomarkers of aging and frailty.
3.3. Sex differences in biomarkers
There are clear sex differences in the prevalence and progression of age-related diseases (Austad 2006; Austad and Fischer 2016) and in the response to interventions that target lifespan and healthspan (Austad and Bartke 2016). Females have lower mortality than males at all ages despite having greater frailty and morbidity, a situation known as the morbidity-mortality sex paradox. Thus, as biomarkers of frailty are developed, it will be important to be aware of possible sex differences. Differences in frailty biomarkers between the sexes have already been observed. For example fibrinogen is only associated with frailty in women (Gale et al. 2013) and adiponectin has a stronger association with frailty in males than females (Tsai et al. 2013).
Sex was almost always adjusted for in the clinical cross-sectional and longitudinal studies summarized in Section 2, but it would be interesting for more studies to directly compare the association of biomarkers with frailty in both men and women. Studies using animal models of frailty will likely contribute to research into sex differences in frailty, and will enable the separation of gender and sex influences. It is interesting that the one animal study thus far exploring frailty biomarkers in both sexes observed sex-differences in inflammatory markers (Kane et al. 2018).
3.4. Composite biomarker measures
Given the complexity and heterogeneity of aging it is unlikely that one single biomarker of frailty will accurately predict its onset and progression. A panel of biomarkers assessing changes across many systems and at many levels will likely be more informative. Studies are beginning to explore the association of panels of biomarkers with frailty as well as other age-related outcomes.
One study of older patients that had experienced acute coronary syndrome (ACS) identified seven variables that predicted frailty. These were an age greater than 75 years, female sex, prior ischemic heart disease, admission for heart failure, haemoglobin below 12.5 g/dL, vitamin D below 9 ng/mL, and cystatin-C above 1.2 mg/L (Sanchis et al. 2015). Although each of these items predicted frailty, the combination of all seven variables did so with greater accuracy. Interestingly, this biomarker panel was a better predictor of mortality over the 2.5 year follow up than the frailty phenotype score (Sanchis et al. 2015), demonstrating the potential for a panel of markers to be more informative than a single biomarker. Another recent cross-sectional study using the frailty index approach found that, although there was an association of frailty with individual biomarkers including hemoglobin and vitamin D, the presence of four or more abnormal biomarker results also predicted frailty (Perez-Zepeda et al 2019). This supports the deficit accumulation theory of the frailty index, whereby it is the number of health-related deficits that is important, rather than their specific nature (Rockwood and Mitnitski 2007).
Howlett and colleagues (2014) developed a frailty index based on standard laboratory tests, called the “FI-lab”. The FI-lab is made up of 23 items including albumin, aspartate aminotransferase (AST), blood pressure, calcium, creatinine, folate, glucose, hemoglobin, mean corpuscular volume (MCV), phosphatase, potassium and total protein. High and low cut-offs are determined for each item, and for each item a score of 0 is given if results were within the cut-offs and a score of one given if the result was outside the cut-off range. The scores across all items are then summed and divided by 23 to give an FI score between zero and one. Increased FI-lab scores were associated with increased mortality risk over a follow-up period of six years. There is a strong linear association between FI-lab scores and frailty determined using a more traditional clinical frailty index (r2 = 0.81), implying that the FI-lab could be considered a biomarker panel for frailty (Howlett et al. 2014). This work was continued by Mitnitski et al (2015), who developed a biomarker-based frailty index (FI-B) from data collected for the Newcastle 85+ study (Mitnitski et al. 2015). The panel includes 40 biomarkers such as IL-6, TNF-α, leptin, adiponectin, hemoglobin, WBC count, CD4+ T cells, telomere length and CpG island DNA methylation, and these outcomes are coded according to cut-offs as described above (Howlett et al. 2014). Increased FI-B scores are associated with increased risk of mortality, and this association was stronger for the combined panel than for any individual biomarker (Mitnitski et al. 2015). Interestingly, similar approaches have been used in mouse studies, where frailty indices based on deficits in ‘laboratory’ outcomes including blood pressure and blood markers have been developed (Antoch et al. 2017; Kane et al. 2018). These lab-based frailty indices are sensitive to interventions thought to delay or accelerate aging (Antoch et al. 2017) and associated with increased cytokine levels (Kane et al. 2018).
Over the last 30 years, many studies have focused on the development and modelling of biomarker panels for the prediction of biological age rather than frailty (Hochschild 1989; Klemera and Doubal 2006; Levine 2013; Jia et al. 2017). Some of the biomarkers that have been included in these modelling approaches are urea nitrogen, creatinine, ferritin, cholesterol and albumin as well as non-blood markers such as systolic blood pressure, forced expiratory volume in one second, highest audible pitch and visual reaction time (Jia et al. 2017). Various approaches to modelling these biomarkers, including multiple linear regression, principal component analysis, Hochschild’s method, and the Klemera and Doubal’s method have shown strong predictive value for increased mortality risk (Jia et al. 2017). Whether or not chronological age should be included in these models of biological age is a matter of some controversy (Mitnitski et al. 2017).
The biological age measure with the most promising evidence is the recently developed DNA methylation clock (Horvath and Raj 2018). The clock is based on patterns of DNA methylation at specific CpG sites and can very accurately predict chronological age, mortality risk and risk of age-related diseases. Recently, two DNA methylation clocks based on biomarker panels were proposed. The first is based on the development of a composite biomarker score of healthspan that is associated with chronological age and increased risk of mortality (Levine et al. 2018). This biomarker panel is made up of albumin, creatinine, glucose, CRP, lymphocyte %, MCV, red blood cell distribution width (RCDW), alkaline phosphatase, WBC count and chronological age modelled using a parametric proportional hazards model. A DNA methylation clock for biological age prediction was then developed based on this biomarker panel, and termed PhenoAge. This clock accurately predicts age-related outcomes including mortality and age-related diseases such as Alzheimer’s disease. Interestingly, however, the composite biomarker panel used to develop this DNA methylation-based measure, was actually a better predictor of mortality and morbidity that the PhenoAge measure itself, and the authors suggest that clinical measures of biological age may be superior to epigenetic measures for risk assessment purposes (Levine et al. 2018).
Similarly, Lu and colleagues (2019) identified seven protein markers of aging, adrenomedullin (ADM), beta-2-microglobulim (B2M), cystatin C (Cystatin C), GDF-15, leptin, plasminogen activator inhibitor-1 (PAI-1), and tissue inhibitor metalloproteinases 1 (TIMP-1), and developed DNA methylation based surrogate markers of these proteins. The resulting clock, called DNAm GrimAge, is able to predict time to death with high accuracy and is also associated with other age-related outcomes such as telomere length and blood cell composition (Lu et al. 2019). Given the demonstrated relationship between DNA methylation and frailty (Bellizzi et al. 2012; Collerton et al. 2014; Breitling et al. 2016; Gale et al. 2018), an investigation of the relationship between these two biomarker-derived DNA methylation outcomes and frailty would be of considerable interest.
Most of these studies of composite biomarkers are not identifying frailty biomarkers per se, but biomarkers of biological age or increased mortality risk. Although it may be that these outcomes are all in fact identifying the same thing – those who are aging faster than others of the same chronological age (see Section 3.5 below) - it would be informative to explore the association of these proposed biomarker panels directly with frailty as assessed by the deficit accumulation or physical frailty phenotype approaches.
3.5. Biological age vs frailty biomarkers
One question to consider in the development of biomarkers of frailty is: Are frailty and biological age measures of the same thing? Frailty, as assessed by the frailty index approach, is an overall measure of the accumulation of health-related deficits with age that explains some of the heterogeneity in health of people of the same chronological age (Mitnitski et al. 2001). Biological age is also a measure of the age of a person based on their overall health and risk, rather than their chronological age (Mitnitski 2018). Both frailty and biological age are also measures of increased mortality risk, and this outcome is often how the value of these measures is validated. Additionally, the underlying biological process and biomarkers known to be associated with aging overlap with those associated with frailty (Fedarko 2011; Kane et al. 2019). Further examination of the association and overlap between markers of biological age and markers of frailty would be very interesting.
4. Conclusions
Cross-sectional studies have identified a number of promising frailty biomarkers including fibrinogen, albumin, D-dimer, WBC count, IL-6, CRP and TNF-α. More longitudinal, interventional studies using the frailty index approach will confirm whether these are predictive biomarkers of frailty. The discovery and validation of frailty biomarkers would be accelerated by additional studies using animals as models of human frailty, by incorporating novel and new potential biomarkers, and by carefully considering sex differences and combining biomarkers into composite panels. Finally, biomarkers of frailty and biological age are likely measuring the same outcomes, and further investigation of whether biomarkers of biological age are also biomarkers of frailty would be of value to the field.
Highlights (3–5):
There are no currently accepted biomarkers of frailty
Cross-sectional studies have identified potential frailty biomarkers including IL-6 and WBC
Future studies should be longitudinal, include biomarker panels and explore biological age
Animal models of frailty provide opportunities to identify and investigate frailty biomarkers
Declaration of Interest:
This work was supported by the Glenn Foundation for Medical Research and grants from the NIH (R37 AG028730, R01 AG019719 and R01 DK100263), and Epigenetics Seed Grant (601139_2018) from Department of Genetics, Harvard Medical School. A.E.K is supported by an NHMRC CJ Martin biomedical fellowship (GNT1122542). D.A.S. is a founder, equity owner, advisor to, director of, consultant to, investor in and/or inventor on patents licensed to Vium, Jupiter Orphan Therapeutics, Cohbar, Galilei Biosciences, GlaxoSmithKline, OvaScience, EMD Millipore, Wellomics, Inside Tracker, Caudalie, Bayer Crop Science, Longwood Fund, Zymo Research, EdenRoc Sciences (and affiliates Arc-Bio, Dovetail Genomics, Claret Bioscience, Revere Biosensors, UpRNA and MetroBiotech (an NAD booster company), Liberty Biosecurity). Life Biosciences (and affiliates Selphagy, Senolytic Therapeutics, Spotlight Biosciences, Animal Biosciences, Iduna, Immetas, Continuum Biosciences, Jumpstart Fertility (an NAD booster company), and Lua Communications). Iduna is a cellular reprogramming company, partially owned by Life Biosciences. DS sits on the board of directors of both companies. D.A.S. is an inventor on a patent application filed by Mayo Clinic and Harvard Medical School that has been licensed to Elysium Health; his personal share is directed to the Sinclair lab. For more information see https://genetics.med.harvard.edu/sinclair-test/people/sinclair-other.php. A.E.K. has no conflicts to declare.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References:
- Almeida OP, Norman PE, Bockxmeer FM, Hankey GJ, Flicker L 2012. CRP 1846G > A polymorphism increases risk of frailty. Maturitas. 71:261–266. [DOI] [PubMed] [Google Scholar]
- Alvarez-ríos AI, Guerrero JM, García-garcía FJ, Rodríguez-mañas L, Medrano-campillo P, De MA, Lanza T, Alvarez-sánchez N, Carrillo-vico A. 2015. Associations between frailty and serum N-terminal propeptide of type I procollagen and 25-hydroxyvitamin D in older Spanish women : The Toledo Study for Healthy Aging. EXG. 69:79–84. [DOI] [PubMed] [Google Scholar]
- Antoch MP, Wrobel M, Kuropatwinski KK, Gitlin I, Leonova KI, Toshkov I, Gleiberman AS, Hutson AD, Chernova OB, Gudkov AV. 2017. Physiological frailty index (PFI):quantitative in ‐ life estimate of individual biological age in mice. Aging (Albany NY). 9:615–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Austad SN. 2006. Why Women Live Longer Than Men:Sex Differences in Longevity. Gend Med. 3:79–92. [DOI] [PubMed] [Google Scholar]
- Austad SN, Bartke A. 2016. Sex Differences in Longevity and in Responses to Anti-Aging Interventions: A Mini-Review. Gerontology. 62:40–46. [DOI] [PubMed] [Google Scholar]
- Austad SN, Fischer KE. 2016. Perspective sex differences in lifespan. Cell Metab. 23:1022–1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banga S, Heinze-milne SD, Howlett SE. 2019. Rodent models of frailty and their application in preclinical research. Mech Ageing Dev. 179:1–10. [DOI] [PubMed] [Google Scholar]
- Barzilay JI, Blaum C, Moore T, Xue QL, Hirsch CH, Walston JD, Fried LP. 2007a. Insulin Resistance and Inflammation as Precursors of Frailty. 167. [DOI] [PubMed] [Google Scholar]
- Barzilay JI, Blaum C, Moore T, Xue QL, Hirsch CH, Walston JD, Fried LP. 2007b. Insulin resistance and inflammation as precursors of frailty: the Cardiovascular Health Study. Arch Intern Med. 167:635–641. [DOI] [PubMed] [Google Scholar]
- Baylis D, Bartlett DB, Syddall HE, Ntani G, Gale CR, Cooper C, Lord JM, Sayer AA. 2013. Immune-endocrine biomarkers as predictors of frailty and mortality: A 10-year longitudinal study in community-dwelling older people. Age (Omaha). 35:963–971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bellizzi D, D’Aquila P, Montesanto A, Corsonello A, Mari V, Mazzei B, Lattanzio F, Passarino G. 2012. Global DNA methylation in old subjects is correlated with frailty. Age (Omaha). 34:169–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bertoli A, Valentini A, Cianfarani MA, Gasbarra E, Tarantino U, Federici M. 2017. Low FT3 : a possible marker of frailty in the elderly. Clin Interv Aging. 12:335–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boxer R, Kleppinger A, Ahmad A, Annis K, Hager D, Kenny A, Boxer R, Heart HM. 2010. The 6-Minute Walk Is Associated With Frailty and Predicts. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brady L 2014. Biomarker Development and Validation: Establishing Standards of Evidence for their Context of Use in Clinical Trials. NAMHC Concept Clear. May 22. [Google Scholar]
- Braidy N 2011. Age related changes in NAD+ metabolism oxidative stress and Sirt1 activity in Wistar rats. PLoS One. 6:e19194. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Brault M, Ohayon S, Kwan R, Bergman H, Eisenberg M, Boivin J, Morin J, Langlois Y, Autexier C, Afilalo J. 2014. Telomere Length and the Clinical Phenotype of Frailty in Older Adults Undergoing Cardiac Surgery. J Am Geriatr Soc. 62:2205–2207. [DOI] [PubMed] [Google Scholar]
- Breitling LP, Saum KU, Perna L, Schöttker B, Holleczek B, Brenner H. 2016. Frailty is associated with the epigenetic clock but not with telomere length in a German cohort. Clin Epigenetics. 8:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brouwers B, Dalmasso B, Hatse S, Laenen A, Kenis C, Swerts E, Neven P, Smeets A, Schöffski P, Wildiers H. 2015. Biological ageing and frailty markers in breast cancer patients. 7:319–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calvani R, Marini F, Cesari M, Tosato M, Anker SD, Von Haehling S, Miller RR, Bernabei R, Landi F, Marzetti E. 2015. Biomarkers for physical frailty and sarcopenia: State of the science and future developments. J Cachexia Sarcopenia Muscle. 6:278–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carcaillon L, Tresguerres JAF, Avila GG, Kireev R. 2012. Higher Levels of Endogenous Estradiol are Associated with Frailty in Postmenopausal Women from the Toledo Study for Healthy Aging. 97:2898–2906. [DOI] [PubMed] [Google Scholar]
- Cardoso AL, Fernandes A, Aguilar-Pimentel JA, de Angelis MH, Guedes JR, Brito MA, Ortolano S, Pani G, Athanasopoulou S, Gonos ES, Schosserer M, Grillari J, Peterson P, Tuna BG, Dogan S, Meyer A, van Os R, Trendelenburg A-U. 2018. Towards frailty biomarkers: Candidates from genes and pathways regulated in aging and age-related diseases. Ageing Res Rev [DOI] [PubMed] [Google Scholar]
- Cesari MAC, Gambassi GIG, van Kan GAAB, Vellas BR V 2014. The frailty phenotype and the frailty index : different instruments for different purposes. 10–12. [DOI] [PubMed] [Google Scholar]
- Chao C, Hsu Y, Chang P, He Y, Ueng R, Lai C, Chiang C, Huang J, Huang S. 2015. Simple self-report FRAIL scale might be more closely associated with dialysis complications than other frailty screening instruments in rural chronic dialysis patients Correspondence : ABSTRACT : Aim : Despite the perceived importance of frailty, few stu. 20:321–328. [DOI] [PubMed] [Google Scholar]
- Cherniack EP, Florez HJ, Troen BR. 2007. Emerging Therapies to Treat Frailty Syndrome in the Elderly. 12. [PubMed] [Google Scholar]
- Clegg A, Young J, Iliffe S, Rikkert MO, Rockwood K. 2013. Frailty in elderly people. Lancet. 381:752–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coelho FM, Pereira DS, Lustosa LP, Silva JP, Dias JMD, Dias RCD, Queiroz BZ, Teixeira AL, Teixeira MM, Pereira LSM. 2019. Physical therapy intervention ( PTI ) increases plasma brain-derived neurotrophic factor ( BDNF ) levels in non-frail and pre-frail elderly women. 54:415–420. [DOI] [PubMed] [Google Scholar]
- Collard RM, Boter H, Schoevers RA, Oude Voshaar RC. 2012. Prevalence of frailty in community-dwelling older persons: A systematic review. J Am Geriatr Soc. 60:1487–1492. [DOI] [PubMed] [Google Scholar]
- Collerton J, Gautrey HE, Van Otterdijk SD, Strathdee G. 2014. Acquisition of aberrant DNA methylation is associated with frailty in the very old : findings from the Newcastle. 317–328. [DOI] [PubMed]
- Collerton J, Martin-Ruiz C, Davies K, Hilkens CM, Isaacs J, Kolenda C, Parker C, Dunn M, Catt M, Jagger C, von Zglinicki T, Kirkwood TBL. 2012. Frailty and the role of inflammation, immunosenescence and cellular ageing in the very old: Cross-sectional findings from the Newcastle 85+ Study. Mech Ageing Dev. 133:456–466. [DOI] [PubMed] [Google Scholar]
- Darvin K, Randolph A, Ovalles S, Halade D, Breeding L, Richardson A, Espinoza SE. 2014. Plasma protein biomarkers of the geriatric syndrome of frailty. Journals Gerontol Ser A, Biol Sci Med Sci. 69:182–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Fanis U, Wang G, Fedarko N, Walston J, Casolaro V, Leng SX. 2008. T-Lymphocytes Expressing CC Chemokine Receptor-5 Are Increased in Frail Older Adults. J Am Geriatr Soc. 56:904–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deepa SS, Bhaskaran S, Espinoza S, Brooks SV., McArdle A, Jackson MJ, Van Remmen H, Richardson A 2017. A new mouse model of frailty: the Cu/Zn superoxide dismutase knockout mouse. GeroScience. 39:187–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erickson KI, Prakash RS, Voss MW, Chaddock L, Heo S, Mclaren M, Pence BD, Martin SA, Vieira VJ, Woods JA, Mcauley E, Kramer AF. 2010. Brain-Derived Neurotrophic Factor Is Associated with Age-Related Decline in Hippocampal Volume. i:5368–5375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erlandson KM, Ng DK, Jacobson LP, Margolick JB, Dobs AS, Jr FJP, Lake JE, Bui H, Kingsley L, Brown TT. 2017. In fl ammation, Immune Activation, Immunosenescence, and Hormonal Biomarkers in the Frailty-Related Phenotype of Men With or at Risk for HIV Infection. 215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erusalimsky JD. 2016. In Search of ‘ Omics ’ -Based Biomarkers to Predict Risk of Frailty and Its Consequences in Older Individuals : The FRAILOMIC Initiative. 182–190. [DOI] [PubMed]
- Fedarko N 2011. The Biology of Aging and Frailty. Clin Geriatr Med. 27:27–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferrucci L, Cavazzini C, Corsi A, Bartali B, Russo C, Lauretani F, Ferrucci L, Cavazzini C, Corsi A, Bartali B, Russo C, Lauretani F, Bandinelli S, Bandinelli S, Guralnik J. 2002. Biomarkers of frailty in older persons. J Endocrinol Invest. 25:10–15. [PubMed] [Google Scholar]
- Fontana L, Addante F, Copetti M, Paroni G, Fontana A, Sancarlo D, Pellegrini F, Ferrucci L, Pilotto A. 2013. Identification of a metabolic signature for multidimensional impairment and mortality risk in hospitalized older patients. Aging Cell. 12:459–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fried LP, Tangen CM, Walston J, Newman a B, Hirsch C, Gottdiener J, Seeman T, Tracy R, Kop WJ, Burke G, McBurnie M a. 2001. Frailty in older adults: evidence for a phenotype. Journals Gerontol Ser A Biol Sci Med Sci. 56:M146–56. [DOI] [PubMed] [Google Scholar]
- Gale CR, Baylis D, Cooper C. 2013. Inflammatory markers and incident frailty in men and women : the English Longitudinal Study of Ageing. Age (Omaha). 35:2493–2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gale CR, Marioni RE, Harris SE, Starr JM, Deary IJ. 2018. DNA methylation and the epigenetic clock in relation to physical frailty in older people : the Lothian Birth Cohort 1936. 1–8. [DOI] [PMC free article] [PubMed]
- Haapanen MJ, Perälä M, Salonen MK, Guzzardi MA, Iozzo P, Kajantie E. 2018. Telomere Length and Frailty : The Helsinki Birth Cohort Study. J Am Med Dir Assoc. 19:658–662. [DOI] [PubMed] [Google Scholar]
- Hochschild R 1989. Improving the precision of biological age determinations. Part 1: A new approach to calculating biological age. Exp Gerontol. 24:289–300. [DOI] [PubMed] [Google Scholar]
- Horvath S 2013. DNA methylation age of human tissues and cell types. Genome Biol. 14:R115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horvath S, Raj K. 2018. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat Rev Genet. [DOI] [PubMed] [Google Scholar]
- Howlett SE, Rockwood MRH, Mitnitski A, Rockwood K. 2014. Standard laboratory tests to identify older adults at increased risk of death. BMC Med. 12:171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsu B, Hirani V, CUmming R, Naganathan V, Blyth F, Wright F, Waite L, Seibel M, Handelsman D, Le Couteur D. 2017. Cross-Sectional and Longitudinal Relationships Between Inflammatory Biomarkers and Frailty in Community-dwelling Older Men: The Concord Health and Ageing in Men Project. Journals Gerontol Ser A Biol Sci. In press. [DOI] [PubMed] [Google Scholar]
- Hubbard RE, O’Mahony MS, Savva GM, Calver BL, Woodhouse KW. 2009. Inflammation and frailty measures in older people. J Cell Mol Med. 13:3103–3109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hubbard RE, Woodhouse KW. 2010. Frailty, inflammation and the elderly. Biogerontology.11:635–641. [DOI] [PubMed] [Google Scholar]
- Inglés M, Gambini J, Carnicero JA, García-García FJ, Rodríguez-Mañas L, Olaso-González G, Dromant M, Borrás C, Viña J. 2014. Oxidative stress is related to frailty, not to age or sex, in a geriatric population: Lipid and protein oxidation as biomarkers of frailty. J Am Geriatr Soc. 62:1324–1328. [DOI] [PubMed] [Google Scholar]
- Jia L, Zhang W, Chen X. 2017. Common methods of biological age estimation. Clin Interv Aging. 12:759–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jylhava J, Nevalainen T, Marttila S, Jylha M, Hervonen A, Hurme M. 2013. Characterization of the role of distinct plasma cell-free DNA species in age-associated inflammation and frailty. Aging Cell. 12:388–397. [DOI] [PubMed] [Google Scholar]
- Kane A, Bisset E, Howlett S. 2019. Biology of Frailty. In: Encyclopedia of Gerontology and Population Aging. p. In press.
- Kane A, Keller K, Heinze-Milne S, Grandy S, Howlett S. 2018. A murine FI based on clinical and laboratory measurements: links between frailty and pro-inflammatory cytokines differ in a sex-specific manner. Journals Gerontol - Ser A Biol Sci Med Sci. Epub ahead. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kane A, Sinclair D. 2019. Epigenetic changes during aging and their reprogramming potential. Crit Rev Biochem Mol Biol. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kane AAE, Mitchell SJSJ, Mach J, Huizer-Pajkos A, McKenzie C, Jones B, Cogger V, Le Couteur DGDG, de Cabo R, Hilmer SNSN. 2016. Acetaminophen hepatotoxicity in mice: effect of age, frailty and exposure type. Exp Gerontol. 73:95–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kane AE, Huizer-Pajkos A, MacH J, Mitchell SJ, De Cabo R, Le Couteur DG, Howlett SE, Hilmer SN. 2017. A Comparison of Two Mouse Frailty Assessment Tools.Journals Gerontol - Ser A Biol Sci Med Sci. 72:904–910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kenny AM, Boxer RS, Kleppinger A, Brindisi J, Feinn R, Burleson J a. 2010. Dehydroepiandrosterone combined with exercise improves muscle strength and physical function in frail older women. J Am Geriatr Soc. 58:1707–1714. [DOI] [PubMed] [Google Scholar]
- Kirkwood TBL. 2011. Systems biology of ageing and longevity. 64–70. [DOI] [PMC free article] [PubMed]
- Klemera P, Doubal S. 2006. A new approach to the concept and computation of biological age. Mech Ageing Dev. 127:240–248. [DOI] [PubMed] [Google Scholar]
- Ko F, Yu Q, Xue QL, Yao W, Brayton C, Yang H, Fedarko N, Walston J. 2012. Inflammation and mortality in a frail mouse model. Age (Omaha). 34:705–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar R, Pradhan R, Ambashtha AK, Jathar S, Dey AB. 2016. Comparative evaluation of seven isoforms of serum sirtuins as protein marker for frailty. J Proteins Proteomics. 7:101–106. [Google Scholar]
- Kuro-o M 2009. Klotho and aging. Biochim Biophys Acta. 1790:1049–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lana A, Valdé-Becars A, Buño A, Rodríguez-Artajelo F, Lopez-Garcia E. 2017. Serum Leptin Concentration is Associated with Incident Frailty in Older Adults. Aging Dis. 8:240–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Couteur DG, Blyth FM, Creasey HM, Handelsman DJ, Naganathan V, Sambrook PN, Seibel MJ, Waite LM, Cumming RG. 2010. The association of alanine transaminase with aging, frailty, and mortality. Journals Gerontol - Ser A Biol Sci Med Sci. 65 A:712–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee WJ, Chen LK, Liang CK, Peng LN, Chiou ST, Chou P. 2016. Soluble ICAM-1, independent of IL-6, is associated with prevalent frailty in community-dwelling elderly Taiwanese people. PLoS One. 11:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leng S, Tian X, Matteini A, Li H, Hughes J, Jain A, Walston J, Fedarko N. 2011. IL-6-independent association of elevated serum neopterin levels with prevalent frailty in community-dwelling older adults. Age Ageing. 40:475–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leng SX, Xue QL, Tian J, Huang Y, Yeh SH, Fried LP. 2009. Associations of neutrophil and monocyte counts with frailty in community-dwelling disabled older women: Results from the Women’s Health and Aging Studies I. Exp Gerontol. 44:511–516. [DOI] [PubMed] [Google Scholar]
- Leng SX, Xue QL, Tian J, Walston JD, Fried LP. 2007. Inflammation and frailty in older women. J Am Geriatr Soc. 55:864–871. [DOI] [PubMed] [Google Scholar]
- Leng SX, Yang H, Walston JD. 2004. Decreased cell proliferation and altered cytokine production in frail older adults. Aging - Clin Exp Res. 16:249–252. [DOI] [PubMed] [Google Scholar]
- Levine ME. 2013. Modeling the rate of senescence: Can estimated biological age predict mortality more accurately than chronological age? Journals Gerontol - Ser A Biol Sci Med Sci. 68:667–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levine ME, Lu AT, Quach A, Chen BH, Assimes TL, Bandinelli S, Hou L, Baccarelli AA, Stewart JD, Li Y, Whitsel EA, Wilson JG, Reiner AP, Aviv A, Lohman K, Liu Y, Ferrucci L, Horvath S. 2018. An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY). 10:573–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li H, Manwani B, Leng SX. 2011. Frailty, inflammation, and immunity. Aging Dis. 2:466–473. [PMC free article] [PubMed] [Google Scholar]
- Lin CH, Liao CC, Huang CH, Tung YT, Chang HC, Hsu MC, Huang CC. 2017. Proteomics analysis to identify and characterize the biomarkers and physical activities of non-frail and frail older adults. Int J Med Sci. 14:231–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu CK, Lyass A, Larson MG, Massaro JM, Wang N, DA RB, Benjamin EJ, Murabito JM. 2016. Biomarkers of oxidative stress are associated with frailty : the Framingham Offspring Study. [DOI] [PMC free article] [PubMed]
- Liu H, Graber TG, Ferguson-Stegall L, Thompson LV. 2014. Clinically relevant frailty index for mice. Journals Gerontol Ser A Biol Sci Med Sci. 69:1485–1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G, López-otín C, Blasco MA, Partridge L, Serrano M. 2013. The Hallmarks of Aging. Cell. 153:1194–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lorenzi M, Lorenzi T, Marzetti E, Landi F, Vetrano DL, Settanni S, Antocicco M, Bonassi S, Valdiglesias V, Bernabei R, Onder G. 2016. Association of frailty with the serine protease HtrA1 in older adults. EXG. 81:8–12. [DOI] [PubMed] [Google Scholar]
- Lu AT, Quach A, Wilson JG, Reiner AP, Aviv A, Raj K, Hou L, Baccarelli AA, Li Y, Stewart JD, Whitsel EA, Assimes TL, Ferrucci L, Horvath S. 2019. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY). 11:303–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu Y, Tze Ying Tan C, Nyunt MSZ, Hei Mok EW, Camous X, Kared H, Fulop T, Feng L, Pin Ng T, Larbi A. 2016. Inflammatory and immune markers associated with physical frailty syndrome: findings from Singapore longitudinal aging studies. Oncotarget. 7:28783–28795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma L, Sha G, Zhang Y, Li Y. 2018. Elevated serum IL-6 and adiponectin levels are associated with frailty and physical function in Chinese older adults. Clin Interv Aging. 13:2013–2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marcos-Perez D, Sanchez-Flores M, Maseda A, Lorenzo-Lopez L, Millan-Calenti J, Gostner J, Fuchs D, Pasaro E, Laffon B, Valdiglesias V. 2018. Frailty in Older Adults Is Associated With Plasma Concentrations of Inflammatory Mediators but Not With Lymphocyte Subpopulations. Front Immunol. 9:1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Margolick JB, Bream JH, Martínez-maza O, Lopez J, Li X, Phair JP, Koletar SL, Jacobson LP. 2017. Frailty and Circulating Markers of Inflammation in HIV + and HIV 2 Men in the Multicenter AIDS Cohort Study. J Acquir Immune Defic Syndr. 74:13–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marzetti E, Lorenzi M, Antocicco M, Bonassi S, Celi M, Mastropaolo S, Settanni S, Valdiglesias V, Landi F, Bernabei R, Onder G. 2014. Shorter telomeres in peripheral blood mononuclear cells from older persons with sarcopenia : results from an exploratory study. Front Aging Neurosci. 6:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matteini A, Walston J, Bandeen-Roche K, Arking D, Allen R, Fried LP, Chakravarti A, Stabler SP, Fallin MD. 2010. TRANSCOBALAMIN-II VARIANTS, DECREASED VITAMIN B12 AVAILABILITY AND INCREASED RISK OF FRAILTY. J Nutr Heal Aging. 14:73–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mekli K, Marshall A, Nazroo J, Vanhoutte B, Pendleton N. 2015. Genetic variant of Interleukin-18 gene is associated with the Frailty Index in the English Longitudinal Study of Ageing. Age Ageing. 44:938–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mekli K, Nazroo JY, Marshall AD, Kumari M, Pendleton N. 2016. Proinflammatory genotype is associated with the frailty phenotype in the English Longitudinal Study of Ageing. Aging Clin Exp Res. 28:413–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitnitski A, Collerton J, Martin-Ruiz C, Jagger C, von Zglinicki T, Rockwood K, Kirkwood TBL. 2015. Age-related frailty and its association with biological markers of ageing. BMC Med. 13:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitnitski A, Howlett SE, Rockwood K. 2017. Heterogeneity of Human Aging and Its Assessment. Journals Gerontol - Ser A Biol Sci Med Sci. 72:877–884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitnitski AB. 2018. Chapter 7 - Epigenetic Biomarkers for Biological Age, Epigenetics of Aging and Longevity. Elsevier Inc.
- Mitnitski AB, Mogilner AJ, Rockwood K. 2001. Accumulation of Deficits as a Proxy Measure of Aging. Sci World J. 1:323–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morimoto RI, Cuervo AM. 2014. Proteostasis and the aging proteome in health and disease. Journals Gerontol - Ser A Biol Sci Med Sci. 69:S33–S38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oxenkrug G, Tucker K, Requintina P, Summergrad P. 2011. Neopterin, a Marker of Interferon-Gamma-Inducible Inflammation, Correlates with Pyridoxal-5′-Phosphate, Waist Circumference, HDL-Cholesterol, Insulin Resistance and Mortality Risk in Adult Boston Community Dwellers of Puerto Rican Origin. Am J Neuroprot Neuroregen. 3:48–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pabst G, Huth C, Koenig W, Ludwig T, Zierer A, Peters A, Thorand B. 2015. ASSOCIATION OF LOW 25-HYDROXYVITAMIN D LEVELS WITH THE FRAILTY SYNDROME IN AN AGED POPULATION : RESULTS FROM THE KORA-AGE AUGSBURG STUDY. 19. [DOI] [PubMed] [Google Scholar]
- Parameswaran N, Patial S. 2010. Tumor Necrosis Factor-α Signaling in Macrophages. Crit Rev Eukaryot Gene Expr. 20:87–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pathai S, Lawn SD, Gilbert CE, Mcguinness D, Mcglynn L, Weiss HA, Port J, Christ T, Barclay K, Wood R, Bekker L, Shiels PG. 2013. Accelerated biological ageing in HIV-infected individuals in South Africa : a case – control study. 27:2375–2384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pepys MB, Hirschfield GM. 2003. C-reactive protein : a critical update. 111:1805–1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piggott DA, Varadhan R, Mehta SH, Brown TT, Li H, Walston JD, Leng SX, Kirk GD. 2015. Frailty, Inflammation, and Mortality Among Persons Aging With HIV Infection and Injection Drug Use. 70:1542–1547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Press D 2016. Efficacy of L-carnitine supplementation on frailty status and its biomarkers, nutritional status, and physical and cognitive function among prefrail older adults : a double-blind, randomized, placebo-controlled clinical trial. 1675–1686. [DOI] [PMC free article] [PubMed]
- Puts MTE, Lips P, Deeg DJH. 2005. Sex differences in the risk of frailty for mortality independent of disability and chronic diseases. J Am Geriatr Soc. 53:40–47. [DOI] [PubMed] [Google Scholar]
- Puts MTE, Visser M, Twisk J, Deeg DJH, Lips P. 2005. Endocrine and inflammatory markers as predictors of frailty. Clin Enocrinology. 63:403–411. [DOI] [PubMed] [Google Scholar]
- Qu T, Yang H, Walston JD, Fedarko NS, Leng SX. 2009. Upregulated monocytic expression of CXC chemokine ligand 10 (CXCL-10) and its relationship with serum interleukin-6 levels in the syndrome of frailty. Cytokine 46:319–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramakrishnan P, Alyousefi N, Abdul-Rahman PS, Kamaruzzaman SB, Chin AV., Tan MP. 2017. A systematic review of studies comparing potential biochemical biomarkers of frailty with frailty assessments. Eur Geriatr Med. 8:397–407. [Google Scholar]
- Reiner A, Aragaki A, Gray S, Wactawski-Wende J, Cauley J, Cochrane B, Kooperberg C, Woods N, LaCroix A. 2009. Inflammation and Thrombosis Biomarkers and Incident Frailty in Postmenopausal Women. Am J Med. 122:947–954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rockwood K, Blodgett JM, Theou O, Sun MH, Feridooni HA, Mitnitsi A, Rose RA, Godin J,Gregson E, Howlett SE, Blodge JM, Theou O, Sun MH, F HA, Mitnitski A. 2017. A frailty index based on deficit accumulation quantifies mortality risk in humans and in mice. Sci Rep. 7:43068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rockwood K, Mitnitski A. 2007. Frailty in relation to the accumulation of deficits. Journals Gerontol Biol Sci Med Sci. 62:722–727. [DOI] [PubMed] [Google Scholar]
- Ronning B, Wyler T, Seljeflot I, Jordhoy M, Skovlund E, Nesbakken A, Kristjansson S. 2010. Frailty measures, inflammatory biomarkers and post-operative complications in older surgical patients. Age Ageing. 39:758–761. [DOI] [PubMed] [Google Scholar]
- Rusanova I, Diaz-Casado ME, Fernández-Ortiz M, Aranda-Martínez P, Guerra-Librero A, García-García FJ, Escames G, Mañas L, Acuña-Castroviejo D. 2018. Analysis of Plasma MicroRNAs as Predictors and Biomarkers of Aging and Frailty in Humans. Oxid Med Cell Longev. 2018:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchis J, Núñez E, Ruiz V, Bonanad C, Fernández J, Cauli O, García-blas S, Mainar L, Valero E, Rodríguez-borja E, Chorro FJ, Hermenegildo C, Núñez J. 2015. Usefulness of Clinical Data and Biomarkers for the Identi fi cation of Frailty After Acute Coronary Syndromes. Can J Cardiol. 31:1462–1468. [DOI] [PubMed] [Google Scholar]
- Saum K, Karina A, Jansen HJM. 2015. Association between Oxidative Stress and Frailty in an Elderly German Population : Results from the ESTHER Cohort Study. 407–415. [DOI] [PubMed]
- Saum KU, Dieffenbach AK, Müezzinler A, Müller H, Holleczek B, Stegmaier C, Butterbach K, Schick M, Canzian F, Stammer H, Boukamp P, Hauer K, Brenner H. 2014. Frailty and telomere length: Cross-sectional analysis in 3537 older adults from the ESTHER cohort. Exp Gerontol. 58:250–255. [DOI] [PubMed] [Google Scholar]
- Semba RD, Margolick JB, Leng S, Walston J, Ricks MO, Fried LP. 2005. T cell subsets and mortality in older community-dwelling women. Exp Gerontol. 40:81–87. [DOI] [PubMed] [Google Scholar]
- Serviddio G, Romano AD, Greco A, Rollo T, Bellanti F, Altomare EI, Vendemiale G, Serviddiol G, Romanol AD, Grec A, Rolloi T, Bellantp F, Altomare EI, Vendemialel G, Sciences O, Sofjerenza S, Rotondo SG, Serviddio G, Romano AD, Greco A, Rollo T, Bellanti F, Altomare EI, Vendemiale G. 2009. Frailty Syndrome Is Associated With Altered Circulating Redox Balance and Increased Markers of Oxidative Stress. Int J Immunopathol Pharmacol. 22:819–827. [DOI] [PubMed] [Google Scholar]
- Shamsi KS, Pierce A, Ashton AS, Halade DG, Richardson A, Espinoza SE. 2012. Proteomic screening of glycoproteins in human plasma for frailty biomarkers. Journals Gerontol -Ser A Biol Sci Med Sci. 67 A:853–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shardell M, Semba RD, Kalyani RR, Bandinelli S, Prather AA, Chia CW, Ferrucci L. 2017. Plasma Klotho and Frailty in Older Adults: Findings From the InCHIANTI Study. Journals Gerontol Ser A. 00:1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soysal P, Stubbs B, Lucato P, Luchini C, Solmi M, Peluso R, Sergi G, Isik AT, Manzato E, Maggi S, Maggio M, Prina AM, Cosco TD, Wu YT, Veronese N. 2016. Inflammation and frailty in the elderly: A systematic review and meta-analysis. Ageing Res Rev. 31:1–8. [DOI] [PubMed] [Google Scholar]
- Strimbu K, Tavel JA. 2010. What are Biomarkers? Kyle. Curr Opin HIV AIDS. 5:463–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su L, Hao Q-KK, Liu S, Dong B-RR. 2017. Monocytes Related Inflammatory Biomarkers are Associated With Frailty Syndrome. Int J Gerontol. 11:225–229. [Google Scholar]
- Tanaka T, Biancotto A, Moaddel R, Moore AZ, Gonzalez-Freire M, Aon MA, Candia J, Zhang P, Cheung F, Fantoni G, Semba RD, Ferrucci L. 2018. Plasma proteomic signature of age in healthy humans. Aging Cell. e12799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tay L, Lim WS, Chan M, Ye RJ, Chong MS. 2016. THE INDEPENDENT ROLE OF INFLAMMATION IN PHYSICAL FRAILTY AMONG OLDER ADULTS WITH MILD COGNITIVE IMPAIRMENT AND MILD-TO-MODERATE ALZHEIMER ‘ S DISEASE. J Nutr Heal Aging. 20:288–299. [DOI] [PubMed] [Google Scholar]
- Tsai J, Wu C, Chen S, Huang K, Chen C, Chang C, Chuang L, Chen C. 2013. Plasma Adiponectin Levels Correlate Positively with an Increasing Number of Components of Frailty in Male Elders. 8:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Valdiglesias V, Sanchez-flores M, Marcos-pérez D, Lorenzo-lópez L, Maseda A, Milláncalenti JC, Pásaro E, Laffon B. 2019. Exploring Genetic Outcomes as Frailty Biomarkers. Journals Gerontol - Ser A Biol Sci Med Sci. 74:168–175. [DOI] [PubMed] [Google Scholar]
- Walston J, Fedarko N, Yang H, Leng S, Beamer B, Espinoza S, Lipton A, Zheng H, Becker K. 2008. The physical and biological characterization of a frail mouse model. Journals Gerontol Ser A, Biol Sci Med Sci. 63:391–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walston J, McBurnie MA, Newman A, Tracy RP, Kop WJ, Hirsch CH, Gottdiener J, Fried LP. 2002. Frailty and activation of the inflammation and coagulation systems with and without clinical comorbidities: results from the Cardiovascular Health Study. Arch Intern Med. 162:2333–2341. [DOI] [PubMed] [Google Scholar]
- Whitehead JC, Hildebrand B a, Sun M, Rockwood MR, Rose R a, Rockwood, Howlett SE. 2014. A clinical frailty index in aging mice: comparisons with frailty index data in humans. Journals Gerontol Biol Sci Med Sci. 69:621–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitson HE, Arnold AM, Yee LM, Mukamal KJ, Kizer JR, Djousse L, Ix JH, Siscovick D, Tracy RP, Thielke SM, Hirsch C, Newman AB, Zieman S. 2014. Serum Carboxymethyl-Lysine , Disability , and Frailty in Older Persons : The Cardiovascular Health Study. Journals Gerontol - Med Sci. 69:710–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woo J, Tang NLS, Suen E, Leung JCS, Leung PC. 2008. Telomeres and frailty. Mech Ageing Dev. 129:642–648. [DOI] [PubMed] [Google Scholar]
- Wu I, Shiesh ÃS, Kuo P, Lin X. 2009. High Oxidative Stress Is Correlated with Frailty in Elderly Chinese. 1666–1671. [DOI] [PubMed]
- Yousefzadeh MJ, Schafer MJ, Noren Hooten N, Atkinson EJ, Evans MK, Baker DJ, Quarles EK, Robbins PD, Ladiges WC, Lebrasseur NK, Niedernhofer LJ. 2018. Circulating levels of monocyte chemoattractant protein-1 as a potential measure of biological age in mice and frailty in humans. Aging Cell. 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu R, Tang N, Leung J, Woo J. 2015. Telomere length is not associated with frailty in older Chinese elderly : Cross-sectional and longitudinal analysis. Mech Ageing Dev. 152:74–79. [DOI] [PubMed] [Google Scholar]
- Zhou J, Wang J, Shen Y, Yang Y, Huang P, Chen S. 2018. The association between telomere length and frailty : A systematic review and meta-analysis. Exp Gerontol. 106:16–20. [DOI] [PubMed] [Google Scholar]
- Zhu Y, Liu Z, Wang Y, Wang Z, Shi J, Xie X, Jin L, Chu X, Wang X. 2019. C-reactive protein, frailty and overnight hospital admission in elderly individuals : A population-based study. Arch Gerontol Geriatr. 64:1–5. [DOI] [PubMed] [Google Scholar]