Sarcopenia en Adultos

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Ageing Research Reviews 73 (2022) 101530

Contents lists available at ScienceDirect

Ageing Research Reviews


journal homepage: www.elsevier.com/locate/arr

Review

Biomarkers shared by frailty and sarcopenia in older adults: A systematic


review and meta-analysis
Anna Picca a, b, Hélio José Coelho-Junior c, Riccardo Calvani a, b, *, Emanuele Marzetti a, c,
Davide Liborio Vetrano a, b
a
Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
b
Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
c
Università Cattolica del Sacro Cuore, Department of Geriatrics and Orthopedics, Rome, Italy

A R T I C L E I N F O A B S T R A C T

Keywords: Background: Physical frailty and sarcopenia show extensive clinical similarities. Whether biomarkers exist that
Amino acids are shared by the two conditions is presently unclear.
Cytokines Methods: We conducted a systematic review and meta-analysis of cross-sectional and longitudinal studies that
Hematologic markers
investigated the association of frailty and/or sarcopenia with biomarkers as a primary or secondary outcome in
Inflammation
Multivariate
adults aged 60 years and older. Only studies published in English that defined frailty using a validated scale and/
Muscle or questionnaire and diagnosed sarcopenia according to the presence of muscle atrophy plus dynapenia or low
physical function were included. Studies were identified from a systematic search of MEDLINE and SCOPUS
databases from inception through August 2020. The quality of reporting of each study was assessed by using the
Quality Assessment Tool for Observational Cohort, Cross-Sectional and Case-Control studies of the National
Institute of Health. A meta-analysis was conducted when at least three studies investigated the same biomarker in
both frailty and sarcopenia. Pooled effect size was calculated based on standard mean differences and random-
effect models. Sensitivity analysis was performed based on age and the setting where the study was conducted.
Results: Eighty studies (58 on frailty and 22 on sarcopenia) met the inclusion criteria and were included in the
qualitative analysis. Studies on frailty included 33,160 community-dwellers, hospitalized, or institutionalized
older adults (60–88 years) from 21 countries. Studies on sarcopenia involved 4904 community-living and
institutionalized older adults (68–87.6 years) from 9 countries. Several metabolic, inflammatory, and hemato­
logic markers were found to be shared between the two conditions. Albumin and hemoglobin were negatively
associated with both frailty and sarcopenia. Interleukin 6 was associated with frailty and sarcopenia only in
people aged < 75. Community-dwelling older adults with frailty and sarcopenia had higher levels of tumor
necrosis factor alpha compared with their robust and non-sarcopenic counterparts.
Conclusions: A set of metabolic, hematologic, and inflammatory biomarkers was found to be shared by frailty and
sarcopenia. These findings fill a knowledge gap in the quest of biomarkers for these conditions and provide a
rationale for biomarker selection in studies on frailty and sarcopenia.

1. Introduction exists on the theoretical framework of frailty, its clinical identification is


hampered by the complex pathophysiology, heterogeneous phenotypic
Frailty, a decline in physiologic reserve that is commonly found in manifestations, intra-individual fluctuations across severity states, and
older adults, refers to a state of increased vulnerability to stressors that the existence of multiple operational definitions (Cesari et al., 2017;
develops as a multisystemic derangement independent of chronological Chen et al., 2014; Looman et al., 2018; Stolz et al., 2019). Several
aging or specific diseases and predisposes to numerous negative health- operational definitions of frailty have been proposed over time (Morley
related events (e.g., falls, morbidity, disability, hospitalization, institu­ et al., 2013) that are mostly inspired by two seminal models: the frailty
tionalization, death) (Clegg et al., 2013). Although large agreement phenotype by Fried et al. (2001) and the frailty index by Rockwood et al.

* Correspondence to: Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L. go F. Vito 1, 00168 Rome, Italy.
E-mail addresses: riccardo.calvani@guest.policlinicogemelli.it, riccardo.calvani@policlinicogemelli.it, riccardo.calvani@ki.se (R. Calvani).

https://doi.org/10.1016/j.arr.2021.101530
Received 28 May 2021; Received in revised form 4 November 2021; Accepted 21 November 2021
Available online 25 November 2021
1568-1637/© 2021 Elsevier B.V. All rights reserved.
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

(2005). The phenotypic model of frailty is based on five elements: 1) We performed a systematic review and meta-analysis to provide a
unintentional weight loss, 2) muscle weakness, 3) exhaustion, 4) slow state-of-the art appraisal of existing biomarkers associated with both
walking speed, and 5) inactive lifestyle (Fried et al., 2001). The frailty frailty and sarcopenia and to identify potential signaling pathways
index is based on a cumulative deficit paradigm and includes health explaining their clinical overlap. Our work may help address knowledge
deficits spanning multiple domains (Rockwood et al., 2005). As such, the gaps in the quest of biomarkers for these conditions and build a solid
frailty index may be considered to serve as a proxy measure of aging rationale to guide biomarker selection for future studies on frailty and
(Mitnitski et al., 2001). sarcopenia.
However, when comparing the ability of available tools at identi­
fying frailty, only little-to-moderate overlap has been observed (Apra­ 2. Material and methods
hamian et al., 2017; Theou et al., 2013). Therefore, subtle deficits that
are intrinsic to the condition may easily be missed, which impacts the We conducted a systematic review and meta-analysis of observa­
prediction of adverse outcomes associated with frailty (Coelho-Júnior tional studies to investigate biomarkers shared between frailty and
et al., 2020; Theou et al., 2013). sarcopenia as a phenotypic expression of common pathways. Cross-
Loss of physical function is a cardinal manifestation of frailty (Fried sectional, case-control, and baseline data of longitudinal studies that
et al., 2001; Hoogendijk et al., 2019), which explains why most frailty examined the association of serum, plasma, saliva, urine, feces, and
assessment tools are built upon the physical frailty model (Faller et al., cerebrospinal fluid biomarkers with frailty and/or sarcopenia were
2019). While being influenced by multisystemic alterations, the selected. Data from each measured molecule were retrieved and allo­
conceptualization and the clinical identification of sarcopenia – a cated in spreadsheets. Then, molecules that were measured in at least
neuromuscular disease characterized by quantitative (i.e., mass) and three studies on frailty and three on sarcopenia were identified as
qualitative (i.e., strength and/or function) muscle declines – are based candidate biomarkers shared between the two conditions and included
on physical deficits (Landi et al., 2018). Muscle atrophy, dynapenia, and in the final qualitative and quantitative analysis. The study was fully
poor physical function are shared between the phenotypic model of performed by investigators and no librarian was part of the team. The
frailty and sarcopenia, with physically inactive lifestyle and fatigue study complies with the criteria of the Preferred Reporting Items for
commonly being associated with the progression of both conditions Systematic Reviews and Meta-Analyses (PRISMA) Statement (Page
(Coelho-Junior et al., 2020; Landi et al., 2015). This scenario has led to et al., 2021) and the Cochrane Handbook for Systematic Reviews and
propose sarcopenia as the biological substratum of frailty (Landi et al., Interventions (Higgins et al., 2019).
2015) and to merge the two conditions into a new clinical entity called
physical frailty-and-sarcopenia (Marzetti et al., 2016). 2.1. Selection criteria
The pathophysiology of frailty and sarcopenia resembles that of
other prototypical geriatric conditions, for which a multifactorial eti­ Inclusion criteria were: (a) observational studies, including cross-
ology is at play involving many of the biological hallmarks of aging (i.e., sectional, case-control, and longitudinal studies which investigated the
genomic and epigenetic instability, loss of proteostasis, mitochondrial association of frailty and/or sarcopenia with biomarkers in older adults
dysfunction, telomere shortening, dysregulated nutrient signaling, stem as a primary or secondary outcome; (b) participant age 60 or greater; (c)
cell exhaustion, cellular senescence, and altered intercellular signaling) frailty defined by a validated scale and/or questionnaires; (d) sarcopenia
(Justice et al., 2018; López-Otín et al., 2013; Sierra, 2016). Currently diagnosed according to the presence of muscle atrophy plus dynapenia
available biomarkers of biological aging might not capture the multitude and/or low physical function (Chen et al., 2020; Cruz-Jentoft et al.,
of intrinsic and extrinsic factors underlying the decline in physical 2019); (e) studies published in the English language; and (f) availability
function that characterizes frailty and sarcopenia (El Assar et al., 2021; of means and standard deviations (SDs) for biomarker levels in cases (i.
Marzetti et al., 2014a, 2014b; Rodríguez-Mañas et al., 2021). However, e., frailty/sarcopenia) and non-cases (i.e., robust/non-sarcopenia). We
perturbations in the above-mentioned aging pillars have been proposed excluded meta-analyses, experimental, quasi-experimental, and
as an important contributors to frailty and sarcopenia (Sierra, 2016). It cross-over investigations that examined intervention strategies. Studies
follows that multi-platform biomarker analyses are probably more that classified participants as frail according to reduced physical/or
suitable strategies for characterizing the pathogenetic pathways of these cognitive function as well as sarcopenic according to a single parameter
dynamic and complex conditions than single-marker approaches (Cal­ (e.g., muscle atrophy) were excluded. Studies that identified genetic
vani et al., 2015; Curcio et al., 2016; Marzetti et al., 2019; Picca et al., markers were also excluded.
2019a). The principle underlying multimarker paradigm is rooted into
the concept of allostatic load, i.e., the cumulative biological burden to 2.2. Search strategy and selection criteria
which cells and body systems are exposed during recurrent or chronic
stresses (Seeman et al., 2001). Biomarkers are, therefore, endopheno­ Studies published on or before 31 August 2020 were retrieved by two
types of physiological dysregulation that may support the diagnosis and investigators (AP, HJCJ) from MEDLINE (PubMed interface) and SCO­
monitoring of a given condition as well as clinical decision-making and PUS (EBSCO interface). The reference lists obtained were checked for
verification of intervention efficacy. Multimarker analyses coupled with reviews, and Google Scholar and ResearchGate were interrogated for
ad hoc multivariate statistics may also identify patterns of mediators additional studies and citation searches of key articles. The initial search
reflecting the individual’s departure from their "normal operating con­ strategy was performed using keywords, MeSH terms, and free text
ditions" (Milot et al., 2014). These results may, therefore, complement words such as frailty, sarcopenia, biomarkers. Moreover, keywords and
clinical appraisals and indices derived from routine clinical evaluations subject headings were exhaustively combined using Boolean operators.
(Howlett et al., 2014; Muscedere et al., 2019). The search strategy is detailed in the Supplementary File 1.
Innovative multimarker analytical strategies have already been
applied to identify candidate biomarkers of frailty and sarcopenia 2.3. Data extraction and quality assessment
(Calvani et al., 2020; Marzetti et al., 2020). However, even more
informative would be the exploitation of this information to understand Titles and abstracts of retrieved articles were screened for eligibility
the pathways that are shared between the two conditions. This would by two researchers (AP, HJCJ). The full-text was consulted if the abstract
allow molecular targets and modifiable risk factors to be identified, did not provide enough information for final evaluation. Two reviewers
which may support the development of new tools for a more accurate (AP, HJCJ) extracted coded variables (i.e., methodological quality and
identification of frailty and sarcopenia and the design of interventions to the characteristics of the studies) using a standardized coding form. A
delay, prevent, or alleviate the associated negative outcomes. third researcher (EM) was consulted to solve disagreements. The quality

2
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

of reporting for each study was performed by two researchers (AP, the Plot Digitizer 2.4.4 software (De Souza et al., 2015). A single pair­
HJCJ) using the Quality Assessment Tool for Observational Cohort and wise comparison was created when multiple studies referred to the same
Cross-Sectional of the National Institute of Health (NIH) and the Quality database using the formulas proposed by the Cochrane group. In
Assessment Tool for Case-Control Studies of the NIH (NIH, 2014). These particular, data from the original database were used for the "BIO­
tools contain 12–14 questions that assess several aspects associated with markers associated with Sarcopenia and PHysical frailty in EldeRly
risk of bias, type I and type II errors, transparency, and confounding pErsons" (BIOSPHERE) study (Calvani et al., 2020), which was available
factors. Items 6, 7, 8, and 10 of the Quality Assessment Tool for to the assessors. Pooled ES was calculated based on standard mean
Observational Cohort and Cross-Sectional do not refer to cross-sectional differences (SMDs), given that different techniques (e.g., ELISA, multi­
studies and were not considered for the quality analysis. Similarly, items plex assays) and/or biospecimens (e.g., serum, plasma) were used for
7 and 9 of the Quality Assessment Tool for Case-Control Studies of the biomarkers quantification. Due to the variability of sample character­
NIH were excluded from the analyses of case-control studies. At the end istics, study design, and assessment instruments for frailty and sarco­
of the evaluation, each study was assigned to one of three categories penia, a random-effect model was used to calculate the pooled ES.
according to the following scores: 0–4: poor, 5–6: fair, and 7–8: good. Sensitivity analysis was performed based on the stratification method
The rate of agreement on quality assessment between reviewers was using binary data (i.e., age: <75 and ≥75 years; setting: community,
99%. hospital, and long-term care).

3. Results
2.4. Statistical analysis
3.1. Literature search
The meta-analysis was conducted using STATA 13 (StataCorp, Col­
lege Station, TX, USA). Pooled analyses comparing frailty vs. robustness A PRISMA flow diagram depicting the different phases of the sys­
and sarcopenia vs. non-sarcopenia were performed when at least three tematic review is reported in Fig. 1. Thirty-six thousand seven-hundred
studies investigated the same biomarker in both conditions. Effect sizes and thirteen records were identified through database searching. Eleven
(ESs) were measured using means and SDs. When data were not made thousand four-hundred and sixty-nine records were removed as dupli­
available by authors as means and SDs, they were calculated according cates, and 24,933 articles were excluded by title and abstract, leaving
to the Cochrane guidelines (Higgins et al., 2019). Specifically, SDs were 311 articles to be assessed for eligibility. After removing 231 studies
calculated from confidence intervals or standard errors, and means were based on eligibility criteria, 80 were included in the qualitative analysis
converted from medians. Data from studies reporting measures of cen­ and 75 investigations provided data for the pooled analysis.
tral tendency and dispersion in graphical formats were extracted using

Fig. 1. PRISMA flow diagram.

3
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

3.2. Characteristics of included studies candidates was of at least 50% (item 3).
As for case-control studies, all investigations clearly stated the
The general characteristics of the included studies are shown in research question (item 1), specified and defined the studied population
Table 1. Eighty investigations, 58 on frailty (Aarts et al., 2015; Al Saedi (item 2), used concurrent controls (item 8), and used valid and reliable
et al., 2018; Alvarez-Ríos et al., 2015; Álvarez-Sánchez et al., 2020; instruments to assess frailty or sarcopenia (item 10). Nine studies
Arauna et al., 2020; Arts et al., 2015; Badrasawi et al., 2019; Brouwers recruited controls from the same or a similar population of cases (item
et al., 2015; Butcher et al., 2019; Darvin et al., 2014; Fried et al., 2009; 4), used the same eligibility criteria to select cases and controls (item 5),
Furtado et al., 2020; Gale et al., 2017; García-García et al., 2014; and clearly defined and differentiated both groups (item 6). None of the
Gunawardene et al., 2016; Hou et al., 2018; Hubbard et al., 2009; Inglés studies justified the sample size (item 3), adjusted data by potential
et al., 2014; Jiang et al., 2020; Kalyani et al., 2012; Kiss et al., 2019; cofactors (item 12), or described if assessors were blinded to the case or
Kochlik et al., 2019; Komici et al., 2020; Krams et al., 2016; Leng et al., control status of participants (item 11).
2002, 2011, 2009, 2007; Liang et al., 2020; Lin et al., 2017; Liu et al.,
2016; Ma et al., 2019; Mahmoudi et al., 2019; Marcos-Pérez et al., 2019, 3.4. Biomarkers shared between frailty and sarcopenia
2018; Margiotta et al., 2020; Mohamad et al., 2018; Munguia et al.,
2018; Namioka et al., 2017; Nascimento et al., 2018; Parsons et al., Table 2 lists the biomarkers that were measured for both frailty and
2019; Qu et al., 2009b, 2009a; Ruangsuriya et al., 2019; Rusanova et al., sarcopenia in at least three studies. Metabolic, hematologic, and in­
2018; Saum et al., 2015; Semmarath et al., 2019; Serviddio et al., 2009; flammatory biomarkers were identified. Albumin was negatively asso­
Su et al., 2017; Szanton et al., 2009; Tajar et al., 2013; Valdiglesias et al., ciated with frailty and sarcopenia regardless of participant’s age and
2018; Valentini et al., 2019; Walston et al., 2002; Wu et al., 2009; setting. A similar pattern was observed for hemoglobin, although studies
Yanagita et al., 2020; Yang et al., 2018; Yeap et al., 2013) and 22 on on sarcopenia did not provide enough data for age-stratified analysis.
sarcopenia (Ates Bulut et al., 2017; Bellanti et al., 2018; Bian et al., Two inflammatory molecules were significantly associated with both
2017; Cacciatore et al., 2015; Calvani et al., 2020, 2018; Can et al., frailty and sarcopenia. Interleukin (IL) 6 was associated with frailty and
2017; Coto Montes et al., 2017; Kuo et al., 2019; Landi et al., 2016; Lim sarcopenia in people aged < 75 years, whereas no associations were
et al., 2015; Lu et al., 2020; Lustosa et al., 2017; Marzetti et al., 2014a, observed in those older than 75. Circulating IL-6 levels were signifi­
2014b; Mochizuki et al., 2019; Rong et al., 2018; Sánchez-Castellano cantly higher in frail community-dwelling and hospitalized individuals
et al., 2020; Tay et al., 2018; Ter Borg et al., 2016; Vettoretti et al., 2019; compared with their robust counterparts. However, IL-6 was signifi­
Yalcin et al., 2018; Yoo et al., 2018) were included in the qualitative cantly lower in frail institutionalized individuals compared with robust
analysis. Molecules pertaining to inflammatory, metabolic, hematolog­ peers. Community-dwelling older adults with frailty and sarcopenia had
ic, and hormonal domains were investigated in the retrieved studies and also higher circulating levels of tumor necrosis factor alpha (TNF-α)
reported accordingly. Studies on frailty included 33,160 compared with their robust and non-sarcopenic counterparts, respec­
community-dwelling, hospitalized, and institutionalized older adults tively. The association of TNF-α and frailty was not affected by age, but
from Australia, Belgium, Brazil, Chile, China, Egypt, France, Germany, was significant only among community-dwelling participants. A nega­
Hungary, Iceland, Italy, Japan, Malaysia, Mexico, Portugal, Scotland, tive association was found between TNF-α and sarcopenia in hospital­
Spain, Taiwan, Thailand, United Kingdom, and United States of Amer­ ized patients. Glucose was positively associated with frailty only in older
ica. The mean age of study participants ranged from 60 to 88 years. Most participants. Triglycerides were associated negatively with sarcopenia
studies assessed frailty using the Frailty Phenotype (FP) (82.8%). Of independently of age and clinical setting. However, no information was
these, 51.7% used a modified FP, 24.2% did not specified cut-off values, available regarding the association with frailty. Plasmatic low-density
while 6.9% provided cut-offs and criteria information. The FRAIL scale lipoprotein (LDL) levels were negatively associated only with frailty in
(3.4%), the Clinical Frailty Scale (CFS) (1.7%), the Edmonton Frailty younger participants. Circulating C-reactive protein (CRP) was posi­
Scale (1.7%), the Frailty Trait Scale (1.7%), the Survey of the Health, tively associated with frailty among the youngest and hospitalized in­
Ageing and Retirement in Europe (SHARE) Frailty Instrument (1.7%), dividuals, but not with sarcopenia. Circulating insulin-like growth factor
the Obu Study Health Promotion for The Elderly index (1.7%), and the 1 (IGF-1) was negatively associated with frailty, regardless of age and
Leuven Oncogeriatric Frailty Score (1.7%) were used in the remaining clinical setting, but not with sarcopenia. No significant associations were
studies. Studies that investigated sarcopenia included 4904 found between total cholesterol, high-density lipoprotein (HDL),
community-living, hospitalized, and institutionalized older adults with a vitamin D and either frailty or sarcopenia.
mean age ranging from 68 to 87.6 years from Brazil, China, Holland,
Italy, Japan, Korea, Singapore, Spain, and Turkey. As per the study 4. Discussion
protocol, all investigations identified sarcopenia according to
co-occurrence of muscle atrophy and dynapenia and/or low physical The main findings of the present study indicate that selected meta­
function. bolic, hematologic, and inflammatory biomarkers are significantly
associated with both frailty and sarcopenia. Specifically, older adults
3.3. Methodology quality appraisal with frailty and/or sarcopenia had lower levels of albumin and hemo­
globin compared with their robust and non-sarcopenic counterparts,
The methodological quality of the retrieved studies ranged from 5 to regardless of age and setting. Inflammatory molecules showed a distinct
8 (Supplementary Table 1). Sixty studies were classified as fair, 17 as pattern of associations: IL-6 was higher in community-dwellers and
good, and four as poor. All cohort and cross-sectional studies clearly hospitalized frail older adults, while it was significantly associated with
stated the research question (item 1), described the study population sarcopenia only in people younger than 75. A negative association be­
(item 2), and used valid and reliable instruments to assess frailty or tween IL-6 and frailty was found in older nursing-home residents.
sarcopenia (item 11). Participants were recruited from the same or Finally, higher TNF-α levels were identified in both frail and sarcopenic
similar populations and were evaluated according to the same eligibility community-dwellers, while a negative association was identified in
criteria in 68 studies (item 4). Sixty-five studies used valid and reliable hospitalized older people with sarcopenia.
instruments, and provided a clear description of biomarker assessment The existence of a core biomarker set shared by frailty and sarco­
(item 9), while 33 studies adjusted results according to potential con­ penia is not unexpected. Indeed, most studies that explored biomarkers
founding variables (item 14). Only 8 studies justified the sample size of frailty were based on the phenotypic model of frailty. This oper­
(item 5) and reported if variables were evaluated by a blinded assessor ationalization of frailty is centered around the physical domain of the
(item 8). Only three studies indicated if the participation rate of eligible condition and encompasses parameters such as muscle weakness and

4
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

Table 1
General description of the studies included in the systematic review.
Frailty

Year Authors Country Study Sample Sample Mean age Assessment Biomarkers
design size (n) (years)

2020 Álvarez-Sánchez Spain Cohort Toledo study for 1211 65–98 Modified Fried Frailty Metabolic and
et al. healthy aging Phenotype inflammatory
2020 Arauna et al. Chile Case- Community-dwelling 58 ~73 Modified Fried Frailty Metabolic and
Control Phenotype inflammatory
2020 Furtado et al. Portugal Cross- Nursing home residents 358 88.0 Modified Fried Frailty Hormonal and
Sectional Phenotype inflammatory
2020 Jiang et al. China Cross- Senior community 230 83.9 Modified Fried Frailty Urinary RNA oxidative
Sectional Phenotype damage
2020 Komici et al. Italy Cross- Hospitalized 128 69.2 CFS Metabolic
Sectional
2020 Liang et al. China Cross- Hospitalized 508 75.0 Modified Fried Frailty Metabolic
Sectional Phenotype
2020 Margiotta et al. Italy Case- Community-dwelling 79 ~77.2 Fried Frailty Phenotype Metabolic and
Control inflammatory
2020 Yanagita et al. Japan Cross- Community-dwelling 148 76.9 CFS Metabolic
Sectional
2019 Badrasawi et al. Malaysia Cross- Community-dwelling 382 60 + Fried Frailty Phenotype Oxidative,
Sectional inflammatory,
hormonal and
metabolic
2019 Butcher et al. Spain Cohort Toledo study for 691 75.0 Modified Fried Frailty Metabolic
healthy aging Phenotype
2019 Kiss et al. Hungary Cross- Elective surgery 313 65 + Edmong Frailty Scale Inflammatory
Sectional
2019 Kochlik et al. Germany Cross- FRAILOMIC database 360 78.8 Modified Fried Frailty Metabolic
Sectional Phenotype
2019 Ma et al. China Cross- Community-dwelling 130 60 + Modified Fried Frailty Metabolic
Sectional Phenotype
2018 Mahmoudi et al. France Cohort MArkers of FRAilty in 250 84.7 Fried Frailty Phenotype Metabolic and
elderly subjects hormonal
2019 Marcos-Pérez Spain Cross- Community-dwelling 252 79.3 Fried Frailty Phenotype Oxidative and
et al. Sectional hormonal
2019 Parsons et al. UK Cohort British regional heart 1660 ~79.2 Modified Fried Frailty Inflammation
study Phenotype
2019 Ruangsuriya Thailand Cross- Community-Dwelling 56 ~71.5 Fried Frailty Phenotype Oxidative and
et al. Sectional metabolic
2019 Semmarath et al. Thailand Cross- Community-dwelling 118 68.1 Modified Fried Frailty Metabolic
Sectional Phenotype
2019 Valentini et al. Italy Cross- Hospitalized 172 79.5 Survey of Health, Ageing Hormonal and
Sectional and Retirement in Europe inflammatory
Frailty
Instrument
2018 Al Saedi et al. Australia Cross- Community-dwelling 66 74 Fried Frailty Phenotype Hormonal,
Sectional inflammatory and
metabolic
2018 Hou et al. China Cross- Hospitalized 345 ~71.3 Modified Fried Frailty Metabolic and
Sectional Phenotype inflammatory
2018 Marcos-Pérez et Spain Cross- Community-dwelling 259 65 + Modified Fried Frailty Inflammatory
al Sectional Phenotype
2018 Mohamad et al. Egypt Cross- Community-dwelling 80 65 Fried Frailty Phenotype Oxidative and
Sectional inflammatory
2018 Mungia et al Mexico Cross- Community-dwelling 170 65 + Modified Fried Frailty Oxidative and
Sectional Phenotype metabolic
2018 Nascimento et al. Brazil Cross- Community-dwelling 347 70.1 Modified Fried Frailty Metabolic and
Sectional Phenotype inflammatory
2018 Rusanova et al. Spain Case- Toledo study for 96 80.5 Modified Fried Frailty Oxidative,
Control healthy aging Phenotype inflammatory and
metabolic
2018 Valdiglesias et al. Spain Cross- Community-dwelling 180 74.9 Modified Fried Frailty Metabolic and
Sectional Phenotype inflammatory
2018 Yang et al. China Cross- Hospitalized 435 81.6 FRAIL scale Inflammatory
Sectional
2017 Gale et al. Scotland Cross- Community-dwelling 594 ~69.5 Modified Fried Frailty Inflammatory
Sectional Phenotype
2017 Lin et al. China Cross- Community-dwelling 12 77.6 Chinese-Canadian Study of Metabolic
Sectional Health and Aging Clinical
Frailty Scale
2017 Namioka et al. Japan Cross- Patients with AD 140 ~80.3 Obu study health Metabolic
Sectional promotion for the elderly
2017 Su et al. China Cross- Community-dwelling 306 70.5 Modified Fried Frailty Inflammatory
Sectional Phenotype
2016 Krams et al. France Community-dwelling 321 82.9 Hormonal
(continued on next page)

5
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

Table 1 (continued )
Frailty

Year Authors Country Study Sample Sample Mean age Assessment Biomarkers
design size (n) (years)

Cross- Modified Fried Frailty


Sectional Phenotype
2016 Liu et al. USA Cross- Framingham heart 1919 ~72.7 Fried Frailty Phenotype Inflammation
Sectional study original
2015 Aarts et al. Iceland Cohort AGES-Reykjavik 4.414 75.4 Modified Fried Frailty Metabolic and
Phenotype inflammatory
2015 Alvarez-Ríos Spain Cross- Toledo study for 592 74 Modified Fried Frailty Hormonal
et al. Sectional healthy aging Phenotype
2015 Arts et al. Iceland Cohort Netherlands study of 378 ~71.5 Modified Fried Frailty Inflammatory
depression in older Phenotype
persons
2015 Browers et al. Belgium Cross- Cancer patients 162 76.0 Leuven Oncogeriatric Inflammatory
Sectional Frailty Score
2015 Gunawardene Australia Cross- Community-dwelling 77 ~75.9 Fried Frailty Phenotype Hormonal,
et al. Sectional inflammatory and
metabolic
2015 Saum et al. Germany Cross- ESTHER cohort study 2.518 ~70.6 Modified Fried Frailty Oxidative and
Sectional Phenotype inflammatory
2014 Darvin et al. USA Cross- Community-dwelling 65 80.6 Fried Frailty Phenotype Metabolic and
Sectional Inflammatory
2014 García-García Spain Cohort Toledo study for 1972 75.1 Frailty Trait Scale Hormonal,
et al. healthy aging inflammatory and
metabolic
2014 Inglés et al. Spain Cohort Toledo study for 742 ~74.3 Modified Fried Frailty Oxidative and
healthy aging Phenotype metabolic
2013 Tajar et al. Europe and Cross- Community-dwelling 1504 69.5 Modified Fried Frailty Metabolic
South Sectional Phenotype
America
2013 Yeap et al. Australia Cross- The health in men study 527 70–89 FRAIL scale Hormonal
Sectional
2012 Kalyani et al. USA Cross- Women’s health and 73 84–95 Modified Fried Frailty Inflammatory and
Sectional aging studies Phenotype hormonal
2011 Leng et al. USA Cross- Community-dwelling 133 84.0 Fried Frailty Phenotype Inflammatory
Sectional
2009 Fried et al. USA Cross- Cardiovascular health 704 70–79 Fried Frailty Phenotype Inflammatory and
Sectional study metabolic
2009 Hubbard et al. England Case- Nursing home residents 110 ~82.4 Modified Fried Frailty Inflammatory
Control Phenotype
2009 Leng et al. USA Cross- Women’s health and 558 ~76.6 Fried Frailty Phenotype Inflammatory
Sectional aging studies
2009 Qu et al. USA Cross- Community-dwelling 32 83 Fried Frailty Phenotype Oxidative and
Sectional metabolic
2009 Qu et al. USA Cross- Community-dwelling 32 83 Fried Frailty Phenotype Inflammation
Sectional
2009 Serviddio et al. Italy Cross- Hospitalized 62 76.7 Modified Fried Frailty Oxidative
Sectional Phenotype
2009 Szanton et al. USA Cohort Women’s health and 728 74.2 Modified Fried Frailty Inflammatory and
aging studies Phenotype metabolic
2009 Wu et al. Taiwan Cross- Community-dwelling 90 ~76.6 Modified Fried Frailty Inflammatory and
Sectional and hospitalized Phenotype metabolic
2007 Leng et al. USA Cohort Women’s health and 548 73.9 Fried Frailty Phenotype Inflammatory
aging studies
2002 Walston et al. USA Cohort Cardiovascular health 4735 65 + Fried Frailty Phenotype Inflammatory and
study metabolic
2002 Leng et al. USA Cross- Community-dwelling 30 ~83.1 Fried Frailty Phenotype Inflammatory
Sectional and hospitalized
Sarcopenia
2020 Lu et al. Singapore Cross- Community-dwelling 189 73.2 Muscle mass plus IHG or Serum amino acids and
Sectional WS metabolic
2020 Sánchez- Spain Cross- Hospitalized 150 87.6 ASMI plus IHG Oxidative and
Castellano et al. Sectional inflammatory
2019 Kuo et al. China Cross- I-Lan longitudinal aging 731 ~74.9 Muscle mass plus IHG or Metabolic and
Sectional study WS hormonal
2019 Mochizuki et al. Japan Cross- Patients with OA 240 75.0 Muscle mass plus IHG or Hormonal, metabolic,
Sectional WS and inflammatory
2019 Vettoretti et al. Italy Cross- Patients with CKD 113 80.6 Muscle mass plus IHG or Metabolic and
Sectional WS Inflammatory
2018 Bellanti et al. Italy Cross- Community-dwelling 115 ~77.1 Muscle mass plus IHG or Metabolic, oxidative
Sectional WS and inflammatory
2018 Rong et al. China Cross- Community-dwelling 421 ~74.5 Muscle mass plus IHG or Inflammation
Sectional WS
2018 Tay et al. Singapore Cross- Community-dwelling 108 76.7 Muscle mass plus IHG or
Sectional WS
(continued on next page)

6
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

Table 1 (continued )
Frailty

Year Authors Country Study Sample Sample Mean age Assessment Biomarkers
design size (n) (years)

Hormonal,
inflammatory and
apolipoprotein
2018 Yalcin et al. Turkey Case- Community-dwelling 160 ~79 Muscle mass plus IHG or Hormonal and
Control WS inflammatory
2018 Yoo et al. Korea Cross- Hospitalized 327 77.6 Muscle mass plus IHG or Inflammatory and
Sectional WS metabolic
2017 Ates Bulut et al. Turkey Cross- Community-dwelling 403 ~74.0 Muscle mass plus IHG or Metabolic
Sectional WS
2017 Bian et al. China Cross- Community-dwelling 441 75.3 ASMI plus IHG Inflammatory and
Sectional metabolic
2017 Can et al. Turkey Cross- Hospitalized 72 ~78.6 Muscle mass plus IHG or Metabolic
Sectional WS
2017 Lustosa et al. Brazil Cross- Community-dwelling 63 ~77.1 Muscle mass plus IHG or Inflammatory
Sectional WS
2017 Coto Montes Spain Cross- Community-dwelling 200 ~77.1 Muscle mass plus IHG or Metabolic, oxidative
et al. Sectional WS and inflammatory
2016 Landi et al. Italy Cross- ilSIRENTE 332 ~86.3 Muscle mass plus IHG or Metabolic and
Sectional WS inflammatory
2016 Ter Borg et al. Holland Cross- MaSS 227 74.0 Muscle mass plus IHG or Metabolic and
Sectional WS hormonal
2015 Cacciatore et al. Italy Cross- Community-dwelling 337 76.8 Muscle mass plus IHG Inflammatory and
Sectional metabolic
2015 Lim et al. Singapore Cross- Community-dwelling 143 68.0 Muscle mass plus IHG or Inflammatory
Sectional WS
2014 Marzetti et al. Italy Cross- Hospitalized 42 83.7 Muscle mass plus IHG or Metabolic
Sectional WS
2018–2020 BIOSPHERE Italy Case- Community-dwelling 90 77.6 Muscle mass and SPPB Metabolic, oxidative
Control and inflammatory

Abbreviations: AD, Alzheimer’s disease; AGES-Reykjavik, Age Gene/Environment Susceptibility-Reykjavik; ASMI, appendicular skeletal muscle index; BIOSPHERE,
BIOmarkers associated with Sarcopenia and PHysical frailty in EldeRly pErsons; CFS, clinical frailty scale; CKD, chronic kidney disease; ESTHER, Epidemiological
Investigations of the Chances of Preventing, Recognizing Early and Optimally Treating Chronic Diseases in an Elderly Population; IHG, isometric handgrip; MaSS,
Maastricht Sarcopenia Study; OA, osteoarthritis; WS, walking speed

slow gait speed which are also features of sarcopenia. activity of other inflammatory mediators such as TNF-α may create an
A dysregulation of the cytokine network has been indicated as a environment favoring muscle atrophy (Belizário et al., 2016; Tuttle
converging point of mechanistic pillars of aging and a major driver of et al., 2020). TNF-α is a pro-inflammatory cytokine released by diseased
age-associated conditions (Cesari et al., 2004; Franceschi et al., 2018; tissues that is able to exert endocrine effects on skeletal muscle (Powers
Michaud et al., 2013; Varadhan et al., 2014). A chronic state of et al., 2016). TNF-α has also been shown to depress contractility by
low-grade inflammation, often referred to as inflamm-aging, is sustained acting upon skeletal muscle both in vitro and in vivo (Alloatti et al.,
by a variety of stimuli, including pathogens, end products of gut mi­ 2000; Wilcox et al., 1994). Hence, the higher systemic TNF-α levels
crobial activity, endogenous cellular debris, or misplaced intracellular found in community-dwelling older adults with frailty and sarcopenia
components (i.e., damage-associated molecular patterns) (Franceschi supports the hypothesis that IL-6 and TNF-α may contribute to muscle
et al., 2018). In response to these stimuli, local inflammatory cells (e.g., mass and strength loss during aging (Wilcox et al., 1994).
neutrophils and macrophages) secrete several cytokines, among which In the context of chronic inflammation, the liver responds by pro­
IL-1, IL-6, and TNF-α are especially relevant (Franceschi et al., 2018). ducing several acute-phase reactants, while the synthesis of a number of
From this perspective, the association of IL-6 and TNF-α with frailty and other proteins is depressed. Albumin is one of these "negative" acute-
sarcopenia may reflect an endophenotypic expression of inflamm-aging. phase proteins and its decrease is used as a marker of inflammation.
However, we found that IL-6 was associated with frailty and sarcopenia Decreased protein synthesis of during inflammation may serve to save
in people aged < 75 years, whereas no associations were observed in amino acids for producing "positive" acute-phase proteins more effi­
those older than 75. IL-6 is a "double-edged sword" mediator able to ciently. This may explain the negative association of albumin with
promote muscle anabolism or catabolism depending on the tissular frailty and sarcopenia, regardless of participant’s age and setting
milieu (Belizário et al., 2016). Following tissue infection and/or dam­ (Table 2).
age, local immune cells release IL-6 as part of the inflammatory secre­ Finally, a negative association with frailty and sarcopenia was
tome, and chronic low-grade inflammation ensues in the setting of identified for hemoglobin, although no data were obtained in age-
prolonged exposure to IL-6 signaling. IL-6 may also act as a myokine stratified analysis for sarcopenia. This association, while confirming
when is produced by contractile skeletal myocytes especially during the role of hemoglobin as a biomarker of frailty, also opens to the
exercise to promote glucose tolerance and insulin sensitivity (Coelho-­ implication of reduced hemopoiesis as a pathway to sarcopenia possibly
Junior et al., 2019; Pedersen and Febbraio, 2006). Muscle catabolism through the mediation of inflammation. Anemia is responsible for
and atrophy have also been reported in the setting of prolonged expo­ reduced tissue oxygenation and consequent fatigue, weakness, and
sure to IL-6 via blunting muscle anabolism and perturbing energy ho­ increased functional impairment, and low hemoglobin concentrations
meostasis (Belizário et al., 2016). However, a synergistic interaction of have been associated with decline of self-reported physical activity and
IL-6 with other inflammatory mediators is needed to deploy its catabolic muscle strength in community-dwellers (Cecchi et al., 2017).
effect and trigger muscle wasting (Belizário et al., 2016). The Taken as a whole, our results indicate that common pathways exist
age-related higher levels of IL-6 may not suffice to induce the catabolic between frailty and sarcopenia and that several mediators may be
effect required for muscle wasting. Though, concomitant release and involved in the way to the progression from one (frailty) to the other

7
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

Table 2
Meta-analysis stratified by age and setting.
Frailty (n = 52) Sarcopenia (n = 23)

SMD 95% CI P-value SMD 95% CI P-value

Metabolic markers
Glucose (6/4) Overall 0.205 -0.107, 0.516 0.198 -0.298 -1.404, 0.808 0.597
Age
< 75 years 0.176 -0.152, 0.503 0.293 — — —
≥ 75 years 0.700 0.472, 0.929 0.0001 — — —

Cholesterol (11/6) Overall -0.801 -1.841, 0.238 0.131 0.159 -0.793, 1.111 0.743
Age
< 75 years — — — 0.133 -2.855, 3.121 0.930
≥ 75 years — — — 0.178 -0.394, 0.751 0.542
Setting
Community-dwelling — — — 0.221 -0.897, 1.338 0.698
Hospitalized — — — -0.153 -0.607, 0.301 0.509

Triglycerides (12/6) Overall 0.407 -0.209, 1.023 0.196 -0.604 -1.022, 0.187 0.005
Age
< 75 years — — — -0.453 -0.763, − 0.143 0.004
≥ 75 years — — — -0.635 -1.155, − 0.114 0.017
Setting
Community-dwelling — — — -0.610 -1.099, − 0.121 0.014
Hospitalized — — — -0.570 -1.042, − 0.098 0.018

LDL (10/7) Overall -0.180 -0.264, − 0.097 0.0001 0.073 -0.414, 0.559 0.770
Age
< 75 years -0.180 -0.272, − 0.089 0.0001 0.443 -0.264, 1.149 0.219
≥ 75 years -0.180 -0.384, 0.024 0.084 -0.080 -0.680, 0.521 0.795
Setting
Community-dwelling — — — 0.077 -0.477, 0.631 0.785
Hospitalized — — — 0.043 -0.410, 0.497 0.851

HDL (10/4) Overall -0.130 -0.283, 0.023 0.096 0.593 -0.282, 1.469 0.184

Albumin (12/10) Overall -1.003 -1.421, − 0.586 0.0001 -0.452 -0.726, − 0.179 0.001
Age
< 75 years -0.862 -1.579, − 0.145 0.019 -0.503 -0.987, − 0.020 0.041
≥ 75 years -1.232 -1.832, − 0.633 0.0001 -0.445 -0.810, − 0.079 0.017
Setting
Community-dwelling -0.856 -1.266, − 0.447 0.0001 -0.389 -0.725, − 0.053 0.023
Hospitalized -1.281 -2.464, − 0.098 0.034 -0.620 -1.071, − 0.168 0.007

Inflammatory markers
IL-6 (23/9) Overall 0.665 0.120, 1.209 0.017 0.198 -0.192, 0.589 0.319
Age
< 75 years 1.437 0.619, 2.256 0.001 1.670 1.314, 2.026 0.0001
≥ 75 years 0.017 -0.861, 0.895 0.970 0.023 -0.245, 0.291 0.865
Setting
Community-dwelling 0.947 0.502, 1.393 0.0001 0.216 -0.234, 0.667 0.347
Hospitalized 0.836 0.279, 1.394 0.003 0.198 -0.192, 0.589 0.679
Institutionalized -5.872 -6.486, − 5.258 0.0001 — — —

CRP (21/10) Overall 0.385 -0.070, 0.839 0.097 0.141 -0.445, 0.726 0.637
Age
< 75 years 0.816 0.019, 1.614 0.045 0.500 -0.522,1.521 0.338
≥ 75 years 0.028 -0.375, 0.431 0.891 0.023 -0.445, 0.726 0.892
Setting
Community-dwelling 0.430 -0.149, 1.009 0.146 0.328 -0.022, 0.679 0.066
Hospitalized 0.652 0.283, 1.021 0.001 -0.606 -4.798, 3.585 0.777
Institutionalized -0.491 -1.482, 0.500 0.331 — — —
TNF-α (10/6)
Overall 0.438 0.222, 0.654 0.001 1.033 0.164, 1.903 0.020
Age
< 75 years 0.460 0.244, 0.675 0.0001 — — —
≥ 75 years 0.421 0.069, 0.773 0.019 — — —
Setting
Community-dwelling 0.373 0.200, 0.547 0.0001 1.344 0.268, 2.421 0.014
Hospitalized 0.953 -0.091, 1.997 0.074 -0.280 -0.550, − 0.011 0.042
Institutionalized 0.142 -1.333, 0.418 0.312 — — —

Hormonal markers
Overall -0.636 -1.296, 0.023 0.059 -0.101 -0.352, 0.149 0.427
Age
Vitamin D (9/5) < 75 years -0.551 -1.518, 0.417 0.265 — — —
≥ 75 years -0.215 -1.538, 1.108 0.750 — — —
(continued on next page)

8
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

Table 2 (continued )
Frailty (n = 52) Sarcopenia (n = 23)

SMD 95% CI P-value SMD 95% CI P-value

Overall -1.387 -2.639, − 0.135 0.030 -0.035 -0.606, 0.536 0.904


IGF-1 (8/3) Setting
Community-dwelling -1.513 -2.974, − 0.052 0.042 — — —
Hospitalized -0.553 -0.929, − 0.178 0.004 — — —

Hematologic markers
Hemoglobin (14/5) Overall -1.679 -2.751, − 0.607 0.002 -0.749 -1.065, − 0.433 0.0001
Age
< 75 years -1.018 -1.589, − 0.446 0.0001 — — —
≥ 75 years -2.392 -4.477, − 0.307 0.025 — — —
Setting
Community-dwelling -2.111 -4.197, − 0.025 0.047 -0.753 -1.205, − 0.301 0.014
Hospitalized -1.214 -1.692, − 0.736 0.0001 -0.784 -0.550, − 0.011 0.0001
Institutionalized -0.576 -0.856, − 0.295 0.0001 — — —

Abbreviations: (# of studies referring to frailty/sarcopenia), CI, confidence interval; CRP, C-reactive protein; HDL, high-density lipoprotein; IGF-1, Insulin-like growth
factor 1; IL-6, interleukin 6; LDL, low-density lipoprotein; SMD, standard mean difference; TNF-α, tumor necrosis factor alpha

(sarcopenia). Indeed, according to the main biomarkers identified by lymphocyte counts according to frailty severity in institutionalized older
our systematic review and meta-analysis in both conditions, a mecha­ adults (Fernandez-Garrido et al., 2018). Although the exact mechanism
nistic hypothesis may be envisioned. In particular, the age-related underlying this association is unknown (Fernandez-Garrido et al.,
reduction in hemoglobin levels and the consequent lower tissue 2018), frailty progression has been associated with a lower cellular
oxygenation may have a negative impact on several organs and, espe­ response to stress (El Assar et al., 2017). In addition, long-term treat­
cially, on muscle which is highly reliant on oxidative metabolism. As a ment with glucocorticoid medications, which are frequently prescribed
response to tissue damage, the release of pro-inflammatory cytokines (i. to institutionalized older adults (Tjia et al., 2010), may also induce
e., IL-6) ensues. However, in the setting of persistent inflammatory immunosuppression by inhibiting leukocyte trafficking, modifying im­
stimuli, a synergistic expression of other inflammatory mediators that mune cell’s function, and suppressing the production and actions of
may act also as pro-atrophy factors (e.g., TNF-α) may be triggered and humoral factors involved in the inflammatory process (Strehl et al.,
ultimately lead to muscle wasting. Reduced rates of muscle protein 2019). However, the possibility that only less impaired nursing-home
synthesis paralleled by enhanced protein breakdown in the context of residents were assessed in this study cannot be ruled out. Though, the
overt inflammation may further sustain the decline in muscle mass, since generalizability of this finding is limited as only one study investigated
a metabolic shift towards amino acid saving is promoted to support the association between IL-6 levels and frailty in older adults who lived
acute phase protein synthesis and inflammatory signaling. The regula­ in long-term institutions. Hence, further studies reporting results
tion of inflammatory and metabolic mediators associated with the adjusted for confounding factors are required. Moreover, studies
transition from frailty to sarcopenia is quite intricate. The negative as­ investigating specific populations (e.g., institutionalized older adults)
sociation found between TNF-α and sarcopenia in hospitalized people is and genders with both sarcopenia and frailty are lacking, thus
an example of this complexity. While acting synergistically with IL-6 hampering the conduction of stratified analysis. Other important limi­
during frailty, the possibility exists that clinical conditions such as tations were identified by the quality analysis, including the lack of a
poor nutrition and severe muscle atrophy may impact the function of the clear description of the source of biomarkers (e.g., plasma, serum),
immune system (Compté et al., 2013) and be responsible for the inverse sample size calculations, and whether assessors were blinded. The in­
association between TNF-α and sarcopenia in hospitalized people. formation on biomarker source is particularly relevant as it has been
Of note, a set of molecules were also identified by our analysis as reported to influence the association between levels of several molecules
significantly associated with only one of the two conditions, which and frailty (Furtado et al., 2020). The above-listed limitations might
might indicate that different pathophysiological pathways may also be provide a possible explanation to the counterintuitive negative associ­
associated with frailty and sarcopenia. Indeed, glucose, LDL, CRP and ation between circulating levels of TNF-α and sarcopenia in hospitalized
IGF-1 were exclusively associated with frailty, whereas triglycerides older people.
were only significantly associated with sarcopenia. However, our results As previously mentioned, substantial agreement exists on the theo­
were based on measures of central tendency and dispersion instead of retical construct of frailty and sarcopenia. Yet, their operationalization
adjusted measure estimations, thus limiting inferences about the effects is still a matter of debate. This is reflected by the fact that the two
of important covariates on the analysis. For instance, glucose (Kim et al., conditions were operationalized according to different criteria across
2014; Seok et al., 2007) and IGF-1 levels (Musarò et al., 2001, 1999; the studies included in our investigation. This might have an impact on
Saeki et al., 2020) have a bi-directional relationship with muscle mass the identification of associated biomarkers. The present study included a
and quality. Physical activity/exercise and eating patterns may also large array of biomarkers and indicated a clear set of candidate mole­
influence metabolic (Kelley et al., 2011, 2005; Schwingshackl et al., cules and pathways shared between frailty and sarcopenia. However,
2020; Tosti et al., 2018), hormonal (Amiri et al., 2021; Tosti et al., numerous other molecules that may explain the pathophysiology of both
2018), and inflammatory milieu (Chupel et al., 2017; Nani et al., 2021; conditions have been investigated, but were not included in our report
Schwingshackl et al., 2020). In addition, the prevalence of comorbidities due to the small number of studies (<3 in frailty and sarcopenia). Among
and sleep disturbance may also affect the circadian cycle and conse­ these, gut microbial derivatives and mediators carried within extracel­
quently have an impact on biomarker levels (Montaruli et al., 2021). lular vesicles warrant investigation. Indeed, gut dysbiosis is a pillar of
The stratified analysis of the present study also provided relevant the gut-muscle axis that has been included among the mechanisms
insights. In particular, IL-6 was positively associated with frailty in contributing to physical frailty and sarcopenia (Picca et al., 2019b).
community-living and hospitalized older adults, whereas a negative Along with this, altered intracellular trafficking is among the main
association was found in those living in long-term institutions. These mechanisms underlying the bioenergetic deficit and overall decline in
findings are in line with previous investigations reporting lower cellular quality control mechanisms observed in old age (Picca et al.,

9
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

2020). The small number of studies eligible for inclusion, the hetero­ review because data were obtained from secondary sources.
geneity of assayed biomolecules, and the limited access to raw data
impeded conducting deeper statistical analysis. For instance, principal Appendix A. Supporting information
components analysis (PCA), a promising approach to identify bio­
markers for frailty and sarcopenia in older adults (Calvani et al., 2015), Supplementary data associated with this article can be found in the
was not applicable to our study because of the limited availability of raw online version at doi:10.1016/j.arr.2021.101530.
data. To gather preliminary information, a PCA was run using available
data (Supplementary File 2). PCA returned six biomarkers: albumin, References
cholesterol, CRP, glucose, IL-6, and vitamin D, and with factorial
weights ranging from 0.638 to 0.966 and explaining 94.5% of the Aarts, S., Patel, K.V., Garcia, M.E., Van den Akker, M., Verhey, F.R.J., Metsemakers, J.F.
M., Van Boxtel, M.P.J., Gudnason, V., Jonsdottir, M.K., Siggeirsdottir, K., Jonsson, P.
variance (Supplementary Table 2). These findings corroborate, at least V., Harris, T.B., Launer, L.J., 2015. Co-presence of multimorbidity and disability
partially, our results and deserve further investigation. with frailty: an examination of heterogeneity in the frail older population. J. Frailty
Aging 4, 131–138. https://doi.org/10.14283/jfa.2015.45.
Al Saedi, A., Gunawardene, P., Bermeo, S., Vogrin, S., Boersma, D., Phu, S., Singh, L.,
5. Conclusion Suriyaarachchi, P., Duque, G., 2018. Lamin A expression in circulating
osteoprogenitors as a potential biomarker for frailty: the Nepean Osteoporosis and
A major goal in the process of biomarker discovery is the identifi­ Frailty (NOF) Study. Exp. Gerontol. 102, 69–75. https://doi.org/10.1016/j.
exger.2017.11.015.
cation of a small number of mediators that can reliably be used as Alloatti, G., Penna, C., Mariano, F., Camussi, G., 2000. Role of NO and PAF in the
endophenotypes of physiological dysregulation to support the diagnosis impairment of skeletal muscle contractility induced by TNF-α. Am. J. Physiol. -
and monitoring of a specific condition as well as clinical decision- Regul. Integr. Comp. Physiol. 279, R2156–R2163. https://doi.org/10.1152/
ajpregu.2000.279.6.r2156.
making and verification of intervention efficacy. Changes over time in
Alvarez-Ríos, A.I., Guerrero, J.M., García-García, F.J., Rodríguez-Mañas, L., Medrano-
specific sets of molecules may also support the definition of patterns of Campillo, P., de la Torre Lanza, M.A., Alvarez-Sánchez, N., Carrillo-Vico, A., 2015.
biomarkers reflecting the individual’s departure from their "normal 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
operating conditions". The inclusion of biomarkers of frailty and sarco­
Aging. Exp. Gerontol. 69, 79–84. https://doi.org/10.1016/j.exger.2015.05.011.
penia in daily practice and clinical research is yet to come. However, the Álvarez-Sánchez, N., Álvarez-Ríos, A.I., Guerrero, J.M., García-García, F.J., Rodríguez-
identification of a set of metabolic, hematologic, and inflammatory Mañas, L., Cruz-Chamorro, I., Lardone, P.J., Carrillo-Vico, A., 2020. Homocysteine
biomarkers shared by frailty and sarcopenia through our systematic and C-reactive protein levels are associated with frailty in older spaniards: the toledo
study for healthy aging. J. Gerontol. Biol. Sci. Med. Sci. 75, 1488–1494. https://doi.
review and meta-analysis is a pivotal step towards the achievement of a org/10.1093/gerona/glz168.
solid rationale for biomarker selection in studies investigating these Amiri, N., Fathei, M., Mosaferi Ziaaldini, M., 2021. Effects of resistance training on
conditions. Following validation, these mediators may eventually assist muscle strength, insulin-like growth factor-1, and insulin-like growth factor–binding
protein-3 in healthy elderly subjects: a systematic review and meta-analysis of
in the diagnostic makeup and prognostication of frailty and sarcopenia randomized controlled trials. Hormones 20, 247–257. https://doi.org/10.1007/
in clinical practice. s42000-020-00250-6.
Aprahamian, I., Cezar, N.O., de, C., Izbicki, R., Lin, S.M., Paulo, D.L.V., Fattori, A.,
Biella, M.M., Jacob Filho, W., Yassuda, M.S., 2017. Screening for frailty with the
Declaration of Competing Interest FRAIL scale: a comparison with the phenotype criteria. J. Am. Med. Dir. Assoc. 18,
592–596. https://doi.org/10.1016/j.jamda.2017.01.009.
E.M., and R.C. are partners of the SPRINTT Consortium, which was Arauna, D., García, F., Rodríguez-Mañas, L., Marrugat, J., Sáez, C., Alarcón, M.,
Wehinger, S., Espinosa-Parrilla, Y., Palomo, I., Fuentes, E., 2020. Older adults with
partly funded by the European Federation of Pharmaceutical Industries
frailty syndrome present an altered platelet function and an increased level of
and Associations (EFPIA). The other authors have no conflict interest to circulating oxidative stress and mitochondrial dysfunction biomarker GDF-15. Free
disclose. Radic. Biol. Med. 149, 64–71. https://doi.org/10.1016/j.
freeradbiomed.2020.01.007.
Arts, M.H.L., Collard, R.M., Comijs, H.C., Naude, P.J.W., Risselada, R., Naarding, P.,
Funding Oude Voshaar, R.C., 2015. Relationship between physical frailty and low-grade
inflammation in late-life depression. J. Am. Geriatr. Soc. 63, 1652–1657. https://doi.
This work was supported by an Intramural Research Grant from the org/10.1111/jgs.13528.
Ates Bulut, E., Soysal, P., Aydin, A.E., Dokuzlar, O., Kocyigit, S.E., Isik, A.T., 2017.
Università Cattolica del Sacro Cuore [D1 2020], the nonprofit research Vitamin B12 deficiency might be related to sarcopenia in older adults. Exp. Gerontol.
foundation “Centro Studi Achille e Linda Lorenzon”, and the Swedish 95, 136–140. https://doi.org/10.1016/j.exger.2017.05.017.
Research Council [Ref.: 2017-06088]. Hélio José Coelho-Junior is fun­ Badrasawi, M., Shahar, S., Rajab, N.F., Singh, D.K.A., 2019. Blood inflammatory markers,
anabolic hormone, Vitamin D and L-carnitine status according to frailty status
ded by a scholarship from the Brazilian federal government [Coor­ among older adults. Sains Malays. 48, 613–619. https://doi.org/10.17576/jsm-
denação de Aperfeiçoamento de Pessoal de Nivel Superior; 001]. The 2019-4803-14.
funders had no role in study design, data collection and analysis, deci­ Belizário, J.E., Fontes-Oliveira, C.C., Borges, J.P., Kashiabara, J.A., Vannier, E., 2016.
Skeletal muscle wasting and renewal: a pivotal role of myokine IL-6. Springerplus 5,
sion to publish, or preparation of the manuscript. 619. https://doi.org/10.1186/s40064-016-2197-2.
Bellanti, F., Romano, A.D., Lo Buglio, A., Castriotta, V., Guglielmi, G., Greco, A.,
Authors’ contributions Serviddio, G., Vendemiale, G., 2018. Oxidative stress is increased in sarcopenia and
associated with cardiovascular disease risk in sarcopenic obesity. Maturitas 109,
6–12. https://doi.org/10.1016/j.maturitas.2017.12.002.
AP and HJCJ designed the study question, completed the literature Bian, A.-L., Hu, H.-Y., Rong, Y.-D., Wang, J., Wang, J.-X., Zhou, X.-Z., 2017. A study on
search, selected articles, extracted data, prepared tables and figures, and relationship between elderly sarcopenia and inflammatory factors IL-6 and TNF-α.
Eur. J. Med. Res. 22, 25. https://doi.org/10.1186/s40001-017-0266-9.
wrote the article. EM was consulted to solve disagreements on data
Brouwers, B., Dalmasso, B., Hatse, S., Laenen, A., Kenis, C., Swerts, E., Neven, P.,
extraction and quality assessment and, together with RC and DLV, Smeets, A., Schoffski, P., Wildiers, H., 2015. Biological ageing and frailty markers in
revised the article and provided critical recommendations on structure breast cancer patients. Aging 7, 319–333. https://doi.org/10.18632/aging.100745.
and presentation. All authors approved the final version of the Butcher, L., Carnicero, J.A., Gomez Cabrero, D., Dartigues, J.-F., Pérès, K., Garcia-
Garcia, F.J., Rodriguez-Mañas, L., Erusalimsky, J.D., 2019. Increased levels of
manuscript. soluble Receptor for Advanced Glycation End-products (RAGE) are associated with a
higher risk of mortality in frail older adults. Age Ageing 48, 696–702. https://doi.
Ethic approval org/10.1093/ageing/afz073.
Cacciatore, F., Della-Morte, D., Basile, C., Curcio, F., Liguori, I., Roselli, M., Gargiulo, G.,
Galizia, G., Bonaduce, D., Abete, P., 2015. Butyryl-cholinesterase is related to muscle
Institutional review board approval for data related to the mass and strength. A new biomarker to identify elderly subjects at risk of sarcopenia.
BIOSPHERE study was obtained by the Ethics Committee of the Uni­ Biomark. Med. 9, 669–678. https://doi.org/10.2217/bmm.15.28.
Calvani, R., Marini, F., Cesari, M., Tosato, M., Anker, S.D., von Haehling, S., Miller, R.R.,
versità Cattolica del Sacro Cuore (Rome, Italy; protocol number: 8498/ Bernabei, R., Landi, F., Marzetti, E., SPRINTT consortium, 2015. Biomarkers for
15). No approval was needed for all other studied of the systematic

10
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

physical frailty and sarcopenia: state of the science and future developments. El Assar, M., Angulo, J., Carnicero, J.A., Walter, S., García-García, F.J., López-
J. Cachex. Sarcopenia Muscle 6, 278–286. https://doi.org/10.1002/jcsm.12051. Hernández, E., Sánchez-Puelles, J.-M., Rodríguez-Mañas, L., 2017. Frailty is
Calvani, R., Picca, A., Marini, F., Biancolillo, A., Gervasoni, J., Persichilli, S., associated with lower expression of genes involved in cellular response to stress:
Primiano, A., Coelho-Junior, H.J., Bossola, M., Urbani, A., Landi, F., Bernabei, R., results from the toledo study for healthy aging. J. Am. Med. Dir. Assoc. 18, 734.
Marzetti, E., 2018. A distinct pattern of circulating amino acids characterizes older e1–734.e7. https://doi.org/10.1016/j.jamda.2017.04.019.
persons with physical frailty and sarcopenia: results from the BIOSPHERE study. El Assar, M., Angulo, J., Carnicero, J.A., Walter, S., García-García, F.J., López-
Nutrients 10, 1691. https://doi.org/10.3390/nu10111691. Hernández, E., Sánchez-Puelles, J.-M., Rodríguez-Mañas, L., Rodríguez-Artalejo, F.,
Calvani, R., Picca, A., Marini, F., Biancolillo, A., Gervasoni, J., Persichilli, S., Rodríguez-Mañas, L., 2021. Association between telomere length, frailty and death
Primiano, A., Coelho-Junior, H.J., Cesari, M., Bossola, M., Urbani, A., Onder, G., in older adults. Geroscience 43, 1015–1027. https://doi.org/10.1007/s11357-020-
Landi, F., Bernabei, R., Marzetti, E., 2020. Identification of biomarkers for physical 00291-0.
frailty and sarcopenia through a new multi-marker approach: results from the Faller, J.W., Pereira, D., do, N., de Souza, S., Nampo, F.K., Orlandi, F., de, S.,
BIOSPHERE study. GeroScience 43, 727–740. https://doi.org/10.1007/s11357-020- Matumoto, S., 2019. Instruments for the detection of frailty syndrome in older
00197-x. adults: a systematic review. PLoS One 14, e0216166. https://doi.org/10.1371/
Can, B., Kara, O., Kizilarslanoglu, M.C., Arik, G., Aycicek, G.S., Sumer, F., Civelek, R., journal.pone.0216166.
Demirtas, C., Ulger, Z., 2017. Serum markers of inflammation and oxidative stress in Fernandez-Garrido, J., Ruiz-Ros, V., Navarro-Martínez, R., Buigues, C., Martínez-
sarcopenia. Aging Clin. Exp. Res. 29, 745–752. https://doi.org/10.1007/s40520- Martínez, M., Verdejo, Y., Sanantonio-Camps, L., Mascarós, M.C., Cauli, O., 2018.
016-0626-2. Frailty and leucocyte count are predictors of all-cause mortality and hospitalization
Cecchi, F., Pancani, S., Vannetti, F., Boni, R., Castagnoli, C., Paperini, A., Pasquini, G., length in non-demented institutionalized older women. Exp. Gerontol. 103, 80–86.
Sofi, F., Molino-Lova, R., Macchi, C., Gori, A., Turcan, N., Valecchi, D., Frandi, R., https://doi.org/10.1016/j.exger.2018.01.007.
Razzolini, L., Fabbri, V., Bonaccorsi, G., Lorini, C., 2017. Hemoglobin concentration Franceschi, C., Garagnani, P., Parini, P., Giuliani, C., Santoro, A., 2018. Inflammaging: a
is associated with self-reported disability and reduced physical performance in a new immune–metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 14,
community dwelling population of nonagenarians: the Mugello Study. Intern. 576–590. https://doi.org/10.1038/s41574-018-0059-4.
Emerg. Med. 12, 1167–1173. https://doi.org/10.1007/s11739-017-1762-1. Fried, L.P., Tangen, C.M., Walston, J., Newman, A.B., Hirsch, C., Gottdiener, J.,
Cesari, M., Calvani, R., Marzetti, E., 2017. Frailty in older persons. Clin. Geriatr. Med. 33, Seeman, T., Tracy, R., Kop, W.J., Burke, G., McBurnie, M.A., 2001. Frailty in older
293–303. https://doi.org/10.1016/j.cger.2017.02.002. adults: evidence for a phenotype. J. Gerontol. Ser. Biol. Sci. Med. Sci. 56,
Cesari, M., Penninx, B.W.J.H., Pahor, M., Lauretani, F., Corsi, A.M., Williams, G.R., M146–M157. https://doi.org/10.1093/gerona/56.3.M146.
Guralnik, J.M., Ferrucci, L., 2004. Inflammatory markers and physical performance Fried, L.P., Xue, Q.-L., Cappola, A.R., Ferrucci, L., Chaves, P., Varadhan, R., Guralnik, J.
in older persons: the InCHIANTI study. J. Gerontol. - Ser. Biol. Sci. Med. Sci. 59, M., Leng, S.X., Semba, R.D., Walston, J.D., Blaum, C.S., Bandeen-Roche, K., 2009.
242–248. https://doi.org/10.1093/gerona/59.3.m242. Nonlinear multisystem physiological dysregulation associated with frailty in older
Chen, L.K., Woo, J., Assantachai, P., Auyeung, T.W., Chou, M.Y., Iijima, K., Jang, H.C., women: implications for etiology and treatment. J. Gerontol. Biol. Sci. Med. Sci. 64,
Kang, L., Kim, M., Kim, S., Kojima, T., Kuzuya, M., Lee, J.S.W., Lee, S.Y., Lee, W.J., 1049–1057. https://doi.org/10.1093/gerona/glp076.
Lee, Y., Liang, C.K., Lim, J.Y., Lim, W.S., Peng, L.N., Sugimoto, K., Tanaka, T., Furtado, G.E., Uba Chupel, M., Minuzzi, L., Patrício, M., Loureiro, M., Bandelow, S.,
Won, C.W., Yamada, M., Zhang, T., Akishita, M., Arai, H., 2020. Asian working Hogervorst, E., Ferreira, J.P., Teixeira, A.M., 2020. Exploring the potential of
group for sarcopenia: 2019 consensus update on sarcopenia diagnosis and treatment. salivary and blood immune biomarkers to elucidate physical frailty in
J. Am. Med. Dir. Assoc. 21, 300–307.e2. https://doi.org/10.1016/j. institutionalized older women. Exp. Gerontol. 129, 110759 https://doi.org/
jamda.2019.12.012. 10.1016/j.exger.2019.110759.
Chen, X., Mao, G., Leng, S.X., 2014. Frailty syndrome: an overview. Clin. Interv. Aging 9, Gale, C.R., Ritchie, S.J., Cooper, C., Starr, J.M., Deary, I.J., 2017. Cognitive ability in late
433–441. https://doi.org/10.2147/CIA.S45300. life and onset of physical frailty: the lothian birth cohort 1936. J. Am. Geriatr. Soc.
Chupel, M.U., Direito, F., Furtado, G.E., Minuzzi, L.G., Pedrosa, F.M., Colado, J.C., 65, 1289–1295. https://doi.org/10.1111/jgs.14787.
Ferreira, J.P., Filaire, E., Teixeira, A.M., 2017. Strength training decreases García-García, F.J., Carcaillon, L., Fernandez-Tresguerres, J., Alfaro, A., Larrion, J.L.,
inflammation and increases cognition and physical fitness in older women with Castillo, C., Rodriguez-Mañas, L., 2014. A new operational definition of frailty: the
cognitive impairment. Front. Physiol. 8, 377. https://doi.org/10.3389/ Frailty Trait Scale. J. Am. Med. Dir. Assoc. 15, 371.e7–371.e13. https://doi.org/
fphys.2017.00377. 10.1016/j.jamda.2014.01.004.
Clegg, A., Young, J., Iliffe, S., Rikkert, M.O., Rockwood, K., 2013. Frailty in elderly Gunawardene, P., Bermeo, S., Vidal, C., Al-Saedi, A., Chung, P., Boersma, D., Phu, S.,
people. Lancet 381, 752–762. https://doi.org/10.1016/S0140-6736(12)62167-9. Pokorski, I., Suriyaarachchi, P., Demontiero, O., Duque, G., 2016. Association
Coelho-Júnior, H.J., Calvani, R., Picca, A., Gonçalves, I.O., Landi, F., Bernabei, R., between circulating osteogenic progenitor cells and disability and frailty in older
Cesari, M., Uchida, M.C., Marzetti, E., 2020. Protein-related dietary parameters and persons: the nepean osteoporosis and frailty study. J. Gerontol. Biol. Sci. Med. Sci.
frailty status in older community-dwellers across different frailty instruments. 71, 1124–1130. https://doi.org/10.1093/gerona/glv190.
Nutrients 12, 508. https://doi.org/10.3390/nu12020508. Higgins, J.P.T., Thomas, J., Chandler, J., Cumpston, M., Li, T., Page, M.J., Welch, V.A.,
Coelho-Junior, H.J., Marzetti, E., Picca, A., Cesari, M., Uchida, M.C., Calvani, R., 2020. 2019. Cochrane handbook for systematic reviews of interventions. Cochrane
Protein intake and frailty: a matter of quantity, quality, and timing. Nutrients 12, Handbook for Systematic Reviews of Interventions. https://doi.org/10.1002/
2915. https://doi.org/10.3390/nu12102915. 9781119536604.
Coelho-Junior, H.J., Picca, A., Calvani, R., Uchida, M.C., Marzetti, E., 2019. If my muscle Hoogendijk, E.O., Afilalo, J., Ensrud, K.E., Kowal, P., Onder, G., Fried, L.P., 2019. Frailty:
could talk: myokines as a biomarker of frailty. Exp. Gerontol. 127, 110715 https:// implications for clinical practice and public health. Lancet 394, 1365–1375. https://
doi.org/10.1016/j.exger.2019.110715. doi.org/10.1016/S0140-6736(19)31786-6.
Compté, N., Zouaoui Boudjeltia, K., Vanhaeverbeek, M., De Breucker, S., Tassignon, J., Hou, P., Xue, H.-P., Mao, X.-E., Li, Y.-N., Wu, L.-F., Liu, Y.-B., 2018. Inflammation
Trelcat, A., Pepersack, T., Goriely, S., 2013. Frailty in old age is associated with markers are associated with frailty in elderly patients with coronary heart disease.
decreased interleukin-12/23 production in response to toll-like receptor ligation. Aging 10, 2636–2645. https://doi.org/10.18632/aging.101575.
PLoS One 8, e65325. https://doi.org/10.1371/journal.pone.0065325. Howlett, S.E., Rockwood, M.R.H., Mitnitski, A., Rockwood, K., 2014. Standard
Coto Montes, A., Boga, J.A., Bermejo Millo, C., Rubio González, A., Potes Ochoa, Y., Vega laboratory tests to identify older adults at increased risk of death. BMC Med. 12, 171.
Naredo, I., Martínez Reig, M., Romero Rizos, L., Sánchez Jurado, P.M., Solano, J.J., https://doi.org/10.1186/s12916-014-0171-9.
Abizanda, P., Caballero, B., 2017. Potential early biomarkers of sarcopenia among Hubbard, R.E., O’Mahony, M.S., Savva, G.M., Calver, B.L., Woodhouse, K.W., 2009.
independent older adults. Maturitas 104, 117–122. https://doi.org/10.1016/j. Inflammation and frailty measures in older people. J. Cell. Mol. Med. 13,
maturitas.2017.08.009. 3103–3109. https://doi.org/10.1111/j.1582-4934.2009.00733.x.
Cruz-Jentoft, A.J., Bahat, G.G., Bauer, J.J., Boirie, Y., Bruyère, O., Cederholm, T., Inglés, M., Gambini, J., Carnicero, J.A., García-García, F.J., Rodríguez-Mañas, L., Olaso-
Cooper, C., Landi, F., Rolland, Y., Sayer, A.A., Schneider, S.M., Sieber, C.C., González, G., Dromant, M., Borrás, C., Viña, J., 2014. Oxidative stress is related to
Topinkova, E., Vandewoude, M., Visser, M., Zamboni, M., Writing Group for the frailty, not to age or sex, in a geriatric population: lipid and protein oxidation as
European Working Group on Sarcopenia in Older People 2 (EWGSOP2), and the E.G. biomarkers of frailty. J. Am. Geriatr. Soc. 62, 1324–1328. https://doi.org/10.1111/
for E, Bautmans, I., Baeyens, J.P., Cesari, M., Cherubini, A., Kanis, J., Maggio, M., jgs.12876.
Martin, F., Michel, J.P., Pitkala, K., Reginster, J.Y., Rizzoli, R., Sánchez- Jiang, S., Kang, L., Zhu, M., Zhang, X., Wang, C., Cai, J., Liu, X., 2020. A potential urinary
Rodríguez, D., Schols, J., 2019. Sarcopenia: revised European consensus on biomarker to determine frailty status among older adults. Arch. Gerontol. Geriatr.
definition and diagnosis. Age Ageing 48, 601. https://doi.org/10.1093/ageing/ 88, 104038 https://doi.org/10.1016/j.archger.2020.104038.
afz046. Justice, J.N., Ferrucci, L., Newman, A.B., Aroda, V.R., Bahnson, J.L., Divers, J.,
Curcio, F., Ferro, G., Basile, C., Liguori, I., Parrella, P., Pirozzi, F., Della-Morte, D., Espeland, M.A., Marcovina, S., Pollak, M.N., Kritchevsky, S.B., Barzilai, N.,
Gargiulo, G., Testa, G., Tocchetti, C.G., Bonaduce, D., Abete, P., 2016. Biomarkers in Kuchel, G.A., 2018. A framework for selection of blood-based biomarkers for
sarcopenia: a multifactorial approach. Exp. Gerontol. 85, 1–8. https://doi.org/ geroscience-guided clinical trials: report from the TAME Biomarkers Workgroup.
10.1016/j.exger.2016.09.007. GeroScience 40, 419–436. https://doi.org/10.1007/s11357-018-0042-y.
Darvin, K., Randolph, A., Ovalles, S., Halade, D., Breeding, L., Richardson, A., Kalyani, R.R., Varadhan, R., Weiss, C.O., Fried, L.P., Cappola, A.R., 2012. Frailty status
Espinoza, S.E., 2014. Plasma protein biomarkers of the geriatric syndrome of frailty. and altered dynamics of circulating energy metabolism hormones after oral glucose
J. Gerontol. Biol. Sci. Med. Sci. 69, 182–186. https://doi.org/10.1093/gerona/ in older women. J. Nutr. Health Aging 16, 679–686. https://doi.org/10.1007/
glt183. s12603-012-0369-5.
De Souza, R.J., Mente, A., Maroleanu, A., Cozma, A.I., Ha, V., Kishibe, T., Uleryk, E., Kelley, G.A., Kelley, K.S., Roberts, S., Haskell, W., 2011. Efficacy of aerobic exercise and
Budylowski, P., Schünemann, H., Beyene, J., Anand, S.S., 2015. Intake of saturated a prudent diet for improving selected lipids and lipoproteins in adults: a meta-
and trans unsaturated fatty acids and risk of all cause mortality, cardiovascular analysis of randomized controlled trials. BMC Med. 9, 74. https://doi.org/10.1186/
disease, and type 2 diabetes: systematic review and meta-analysis of observational 1741-7015-9-74.
studies. Br. Med. J. 351, h3978. https://doi.org/10.1136/bmj.h3978.

11
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

Kelley, G.A., Kelley, K.S., Tran, Z.V., 2005. Exercise, lipids, and lipoproteins in older Ma, L., Niu, H., Sha, G., Zhang, Y., Liu, P., Li, Y., 2019. Serum SIRT1 is associated with
adults: a meta-analysis. Prev. Cardiol. 8, 206–214. https://doi.org/10.1111/j.0197- frailty and adipokines in older adults. J. Nutr. Health Aging 23, 246–250. https://
3118.2005.03769.x. doi.org/10.1007/s12603-018-1149-7.
Kim, K.S., Park, K.S., Kim, M.J., Kim, S.K., Cho, Y.W., Park, S.W., 2014. Type 2 diabetes Mahmoudi, R., Jaisson, S., Badr, S., Jaidi, Y., Bertholon, L.-A., Novella, J.-L., Gillery, P.,
is associated with low muscle mass in older adults. Geriatr. Gerontol. Int. 14, 2019. Post-translational modification-derived products are associated with frailty
115–121. https://doi.org/10.1111/ggi.12189. status in elderly subjects. Clin. Chem. Lab. Med. 57, 1153–1161. https://doi.org/
Kiss, R., Farkas, N., Jancso, G., Kovacs, K., Lenard, L., 2019. Determination of frail state 10.1515/cclm-2018-1322.
and association of frailty with inflammatory markers among cardiac surgery patients Marcos-Pérez, D., Sánchez-Flores, M., Maseda, A., Lorenzo-López, L., Millán-Calenti, J.C.,
in a Central European patient population. Clin. Hemorheol. Microcirc. 76, 341–350. Gostner, J.M., Fuchs, D., Pásaro, E., Laffon, B., Valdiglesias, V., 2018. Frailty in older
https://doi.org/10.3233/CH-190681. adults is associated with plasma concentrations of inflammatory mediators but not
Kochlik, B., Stuetz, W., Pérès, K., Féart, C., Tegner, J., Rodriguez-Mañas, L., Grune, T., with lymphocyte subpopulations. Front. Immunol. 9, 1056. https://doi.org/
Weber, D., 2019. Associations of plasma 3-methylhistidine with frailty status in 10.3389/fimmu.2018.01056.
french cohorts of the FRAILOMIC initiative. J. Clin. Med. 8, 1010. https://doi.org/ Marcos-Pérez, D., Sánchez-Flores, M., Maseda, A., Lorenzo-López, L., Millán-Calenti, J.C.,
10.3390/jcm8071010. Pásaro, E., Laffon, B., Valdiglesias, V., 2019. Serum cortisol but not oxidative stress
Komici, K., Gnemmi, I., Bencivenga, L., Vitale, D.F., Rengo, G., Di Stefano, A., biomarkers are related to frailty: results of a cross-sectional study in Spanish older
Eleuteri, E., 2020. Impact of galectin-3 circulating levels on frailty in elderly patients adults. J. Toxicol. Environ. Heal. Part A 1–11. https://doi.org/10.1080/
with systolic heart failure. J. Clin. Med. 9, 2229. https://doi.org/10.3390/ 15287394.2019.1654639.
jcm9072229. Margiotta, E., Miragoli, F., Callegari, M.L., Vettoretti, S., Caldiroli, L., Meneghini, M.,
Krams, T., Cesari, M., Guyonnet, S., Abellan Van Kan, G., Cantet, C., Vellas, B., Zanoni, F., Messa, P., 2020. Gut microbiota composition and frailty in elderly
Rolland, Y., 2016. Is the 25-hydroxy-vitamin d serum concentration a good marker patients with Chronic Kidney Disease. PLoS One 15, e0228530. https://doi.org/
of frailty? J. Nutr. Health Aging 20, 1034–1039. https://doi.org/10.1007/s12603- 10.1371/journal.pone.0228530.
016-0714-1. Marzetti, E., Calvani, R., Cesari, M., Tosato, M., Cherubini, A., Di Bari, M., Pahor, M.,
Kuo, Y.-H., Wang, T.-F., Liu, L.-K., Lee, W.-J., Peng, L.-N., Chen, L.-K., 2019. Savera, G., Collamati, A., D’Angelo, E., Bernabei, R., Landi, F., 2016.
Epidemiology of sarcopenia and factors associated with it among community- Operationalization of the physical frailty & sarcopenia syndrome: rationale and
dwelling older adults in Taiwan. Am. J. Med. Sci. 357, 124–133. https://doi.org/ clinical implementation. Transl. Med. UniSa. 13, 29–32.
10.1016/j.amjms.2018.11.008. Marzetti, E., Calvani, R., Lorenzi, M., Marini, F., D’Angelo, E., Martone, A.M., Celi, M.,
Landi, F., Calvani, R., Cesari, M., Tosato, M., Maria Martone, A., Ortolani, E., Savera, G., Tosato, M., Bernabei, R., Landi, F., 2014a. Serum levels of C-terminal agrin fragment
Salini, S., N Sisto, A., Picca, A., Marzetti, E., Martone, A.M., Ortolani, E., Savera, G., (CAF) are associated with sarcopenia in older hip fractured patients. Exp. Gerontol.
Salini, S., Sisto, A., Picca, A., Marzetti, E., 2018. Sarcopenia: an overview on current 60, 79–82. https://doi.org/10.1016/j.exger.2014.10.003.
definitions, diagnosis and treatment. Curr. Protein Pept. Sci. 19, 633–638. https:// Marzetti, E., Guerra, F., Calvani, R., Marini, F., Biancolillo, A., Gervasoni, J.,
doi.org/10.2174/1389203718666170607113459. Primiano, A., Coelho-Júnior, H.J., Landi, F., Bernabei, R., Bucci, C., Picca, A., 2020.
Landi, F., Calvani, R., Cesari, M., Tosato, M., Martone, A.M., Bernabei, R., Onder, G., Circulating mitochondrial-derived vesicles, inflammatory biomarkers and amino
Marzetti, E., 2015. Sarcopenia as the biological substrate of physical frailty. Clin. acids in older adults with physical frailty and sarcopenia: a preliminary BIOSPHERE
Geriatr. Med. 31, 367–374. https://doi.org/10.1016/j.cger.2015.04.005. multi-marker study using sequential and orthogonalized covariance selection –
Landi, F., Calvani, R., Lorenzi, M., Martone, A.M., Tosato, M., Drey, M., D’Angelo, E., linear discrimi. Front. Cell Dev. Biol. 8, 56441. https://doi.org/10.3389/
Capoluongo, E., Russo, A., Bernabei, R., Onder, G., Marzetti, E., 2016. Serum levels fcell.2020.564417.
of C-terminal agrin fragment (CAF) are associated with sarcopenia in older Marzetti, E., Lorenzi, M., Antocicco, M., Bonassi, S., Celi, M., Mastropaolo, S.,
multimorbid community-dwellers: results from the ilSIRENTE study. Exp. Gerontol. Settanni, S., Valdiglesias, V., Landi, F., Bernabei, R., Onder, G., 2014b. Shorter
79, 31–36. https://doi.org/10.1016/j.exger.2016.03.012. telomeres in peripheral blood mononuclear cells from older persons with sarcopenia:
Leng, S., Chaves, P., Koenig, K., Walston, J., 2002. Serum interleukin-6 and hemoglobin results from an exploratory study. Front. Aging Neurosci. 6, 233. https://doi.org/
as physiological correlates in the geriatric syndrome of frailty: a pilot study. J. Am. 10.3389/fnagi.2014.00233.
Geriatr. Soc. 50, 1268–1271. https://doi.org/10.1046/j.1532-5415.2002.50315.x. Marzetti, E., Picca, A., Marini, F., Biancolillo, A., Coelho-Junior, H.J., Gervasoni, J.,
Leng, S.X., Tian, X., Matteini, A., Li, H., Hughes, J., Jain, A., Walston, J.D., Fedarko, N.S., Bossola, M., Cesari, M., Onder, G., Landi, F., Bernabei, R., Calvani, R., 2019.
2011. IL-6-independent association of elevated serum neopterin levels with Inflammatory signatures in older persons with physical frailty and sarcopenia: the
prevalent frailty in community-dwelling older adults. Age Ageing 40, 475–481. frailty “cytokinome” at its core. Exp. Gerontol. 122, 129–138. https://doi.org/
https://doi.org/10.1093/ageing/afr047. 10.1016/j.exger.2019.04.019.
Leng, S.X., Xue, Q.-L., Tian, J., Huang, Y., Yeh, S.-H., Fried, L.P., 2009. Associations of Michaud, M., Balardy, L., Moulis, G., Gaudin, C., Peyrot, C., Vellas, B., Cesari, M.,
neutrophil and monocyte counts with frailty in community-dwelling disabled older Nourhashemi, F., 2013. Proinflammatory cytokines, aging, and age-related diseases.
women: results from the Women’s Health and Aging Studies I. Exp. Gerontol. 44, J. Am. Med. Dir. Assoc. 14, 877–882. https://doi.org/10.1016/j.jamda.2013.05.009.
511–516. https://doi.org/10.1016/j.exger.2009.05.005. Milot, E., Morissette-Thomas, V., Li, Q., Fried, L.P., Ferrucci, L., Cohen, A.A., 2014.
Leng, S.X., Xue, Q.-L., Tian, J., Walston, J.D., Fried, L.P., 2007. Inflammation and frailty Trajectories of physiological dysregulation predicts mortality and health outcomes in
in older women. J. Am. Geriatr. Soc. 55, 864–871. https://doi.org/10.1111/j.1532- a consistent manner across three populations. Mech. Ageing Dev. 141–142, 56–63.
5415.2007.01186.x. https://doi.org/10.1016/j.mad.2014.10.001.
Liang, Y.-D., Liu, Q., Du, M.-H., Liu, Z., Yao, S.-M., Zheng, P.-P., Wan, Y.-H., Sun, N., Mitnitski, A.B., Mogilner, A.J., Rockwood, K., 2001. Accumulation of deficits as a proxy
Li, Y.-Y., Liu, J.-P., Luo, Y., Cai, J.-P., Yang, J.-F., Wang, H., 2020. Urinary 8-oxo-7,8- measure of aging. Sci. World J. 323–336. https://doi.org/10.1100/tsw.2001.58.
dihydroguanosine as a potential biomarker of frailty for elderly patients with Mochizuki, T., Yano, K., Ikari, K., Okazaki, K., 2019. Sarcopenia-associated factors in
cardiovascular disease. Free Radic. Biol. Med. 152, 248–254. https://doi.org/ Japanese patients with rheumatoid arthritis: a cross-sectional study. Geriatr.
10.1016/j.freeradbiomed.2020.03.011. Gerontol. Int. 19, 907–912. https://doi.org/10.1111/ggi.13747.
Lim, J.P., Leung, B.P., Ding, Y.Y., Tay, L., Ismail, N.H., Yeo, A., Yew, S., Chong, M.S., Mohamad, M., Ebeid, S., Shawky Khater, M., Alsadany, M., 2018. Interferon- gamma-
2015. Monocyte chemoattractant protein-1: a proinflammatory cytokine elevated in inducible guanosine triphosphate cyclohydrolase 1 (GTP-CH1) pathway is associated
sarcopenic obesity. Clin. Interv. Aging 10, 605–609. https://doi.org/10.2147/CIA. with frailty in Egyptian elderly. Rep. Biochem. Mol. Biol. 7, 52–58.
S78901. Montaruli, A., Castelli, L., Mulè, A., Scurati, R., Esposito, F., Galasso, L., Roveda, E.,
Lin, C.-H., Liao, C.-C., Huang, C.-H., Tung, Y.-T., Chang, H.-C., Hsu, M.-C., Huang, C.-C., 2021. Biological rhythm and chronotype: new perspectives in health. Biomolecules
2017. Proteomics analysis to identify and characterize the biomarkers and physical 11, 487. https://doi.org/10.3390/biom11040487.
activities of non-frail and frail older adults. Int. J. Med. Sci. 14, 231–239. https:// Morley, J.E., Vellas, B., Abellan van Kan, G., Anker, S.D., Bauer, J.M., Bernabei, R.,
doi.org/10.7150/ijms.17627. Cesari, M., Chumlea, W.C., Doehner, W., Evans, J., Fried, L.P., Guralnik, J.M.,
Liu, C.K., Lyass, A., Larson, M.G., Massaro, J.M., Wang, N., D’Agostino, R.B.S., Katz, P.R., Malmstrom, T.K., McCarter, R.J., Gutierrez Robledo, L.M., Rockwood, K.,
Benjamin, E.J., Murabito, J.M., 2016. Biomarkers of oxidative stress are associated von Haehling, S., Vandewoude, M.F., Walston, J., 2013. Frailty consensus: a call to
with frailty: the Framingham Offspring Study. Age 38, 1. https://doi.org/10.1007/ action. J. Am. Med. Dir. Assoc 392–397. https://doi.org/10.1016/j.
s11357-015-9864-z. jamda.2013.03.022.
Looman, W.M., Fabbricotti, I.N., Blom, J.W., Jansen, A.P.D., Lutomski, J.E., Munguia, L., Solis, V., Meaney, E., Ramirez-Sanchez, I., Villarreal, F., Perez-Duran, J.,
Metzelthin, S.F., Huijsman, R., TOPICS-MDS Research Consortium, 2018. The frail Rubio-Gayosso, I., Ortiz, A., Ceballos, G., Najera, N., 2018. Association of physical
older person does not exist: development of frailty profiles with latent class analysis. performance tests with frailty indicators and oxidative stress markers in a sample of
BMC Geriatr. 18, 84. https://doi.org/10.1186/s12877-018-0776-5. a community-dwelling elderly population. Biomed. Res. 29, 3344–3350. https://doi.
López-Otín, C., Blasco, M.A., Partridge, L., Serrano, M., Kroemer, G., 2013. The org/10.4066/biomedicalresearch.29-18-806.
hallmarks of aging. Cell 153, 1194–1217. https://doi.org/10.1016/j. Musarò, A., McCullagh, K., Paul, A., Houghton, L., Dobrowolny, G., Molinaro, M.,
cell.2013.05.039. Barton, E.R., L Sweeney, H., Rosenthal, N., 2001. Localized Igf-1 transgene
Lu, Y., Karagounis, L.G., Ng, T.P., Carre, C., Narang, V., Wong, G., Tan, C.T.Y., Nyunt, M. expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat.
S.Z., Gao, Q., Abel, B., Poidinger, M., Fulop, T., Bosco, N., Larbi, A., 2020. Systemic Genet. 27, 195–200. https://doi.org/10.1038/84839.
and metabolic signature of sarcopenia in community-dwelling older adults. Musarò, A., McCullagh, K.J.A., Naya, F.J., Olson, E.N., Rosenthal, N., 1999. IGF-1
J. Gerontol. - Ser. Biol. Sci. Med. Sci. 75, 309–317. https://doi.org/10.1093/gerona/ induces skeletal myocyte hypertrophy through calcineurin in association with
glz001. GATA-2 and NF-ATc1. Nature 400, 581–585. https://doi.org/10.1038/23060.
Lustosa, L.P., Batista, P.P., Pereira, D.S., Pereira, L.S.M., Scianni, A., Ribeiro-Samora, G. Muscedere, J., Kim, P.M., Afilalo, J., Balion, C., Baracos, V.E., Bowdish, D., Cesari, M.,
A., 2017. Comparison between parameters of muscle performance and inflammatory Erusalimsky, J.D., Fülöp, T., Heckman, G., Howlett, S.E., Khadaroo, R.G.,
biomarkers of non-sarcopenic and sarcopenic elderly women. Clin. Interv. Aging 12, Kirkland, J.L., Rodriguez Mañas, L., Marzetti, E., Paré, G., Raina, P., Rockwood, K.,
1183–1191. https://doi.org/10.2147/CIA.S139579. Sinclair, A., Skappak, C., Verschoor, C., Walter, S., 2019. Proceedings of the

12
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

Canadian frailty network workshop: identifying biomarkers of frailty to support German population: results from the ESTHER cohort study. Gerontology 61,
frailty risk assessment, diagnosis and prognosis. Toronto, January 15, 2018. 407–415. https://doi.org/10.1159/000380881.
J. Frailty Aging 8, 106–116. https://doi.org/10.14283/jfa.2019.12. Schwingshackl, L., Morze, J., Hoffmann, G., 2020. Mediterranean diet and health status:
Namioka, N., Hanyu, H., Hirose, D., Hatanaka, H., Sato, T., Shimizu, S., 2017. Oxidative active ingredients and pharmacological mechanisms. Br. J. Pharmacol. 177,
stress and inflammation are associated with physical frailty in patients with 1241–1257. https://doi.org/10.1111/bph.14778.
Alzheimer’s disease. Geriatr. Gerontol. Int. 17, 913–918. https://doi.org/10.1111/ Seeman, T.E., McEwen, B.S., Rowe, J.W., Singer, B.H., 2001. Allostatic load as a marker
ggi.12804. of cumulative biological risk: MacArthur studies of successful aging. Proc. Natl.
Nani, A., Murtaza, B., Sayed Khan, A., Khan, N.A., Hichami, A., 2021. Antioxidant and Acad. Sci. USA. 98, 4770–4775. doi:10.1073/pnas.081072698.
anti-inflammatory potential of polyphenols contained in mediterranean diet in Semmarath, W., Seesen, M., Yodkeeree, S., Sapbamrer, R., Ayood, P., Malasao, R.,
obesity: molecular mechanisms. Molecules 26, 985. https://doi.org/10.3390/ Siviroj, P., Limtrakul Dejkriengkraikul, P., 2019. The association between frailty
molecules26040985. indicators and blood-based biomarkers in early-old community dwellers of Thailand.
Nascimento, C.M.C., Zazzetta, M.S., Gomes, G.A.O., Orlandi, F.S., Gramani-Say, K., Int. J. Environ. Res. Public Health 16, 3457. https://doi.org/10.3390/
Vasilceac, F.A., Gratao, A.C.M., Pavarini, S.C.I., Cominetti, M.R., 2018. Higher levels ijerph16183457.
of tumor necrosis factor beta are associated with frailty in socially vulnerable Seok, W.P., Goodpaster, B.H., Strotmeyer, E.S., Kuller, L.H., Broudeau, R., Kammerer, C.,
community-dwelling older adults. BMC Geriatr. 18, 268. https://doi.org/10.1186/ De Rekeneire, N., Harris, T.B., Schwartz, A.V., Tylavsky, F.A., Yong-Wook, C.,
s12877-018-0961-6. Newman, A.B., 2007. Accelerated loss of skeletal muscle strength in older adults
National Institutes of Health, 2014. Study Quality Assessment Tools. 〈https://www.nh with type 2 diabetes: the health, aging, and body composition study. Diabetes Care
lbi.nih.gov/health-topics/study-quality-assessment-tools〉 (accessed 28 May 2021). 30, 1507–1512. https://doi.org/10.2337/dc06-2537.
Page, M.J., McKenzie, J.E., Bossuyt, P.M., Boutron, I., Hoffmann, T.C., Mulrow, C.D., Serviddio, G., Romano, A.D., Greco, A., Rollo, T., Bellanti, F., Altomare, E.,
Shamseer, L., Tetzlaff, J.M., Akl, E.A., Brennan, S.E., Chou, R., Glanville, J., Vendemiale, G., 2009. Frailty syndrome is associated with altered circulating redox
Grimshaw, J.M., Hróbjartsson, A., Lalu, M.M., Li, T., Loder, E.W., Mayo-Wilson, E., balance and increased markers of oxidative stress. Int. J. Immunopathol. Pharmacol.
McDonald, S., McGuinness, L.A., Stewart, L.A., Thomas, J., Tricco, A.C., Welch, V.A., 22, 819–827. https://doi.org/10.1177/039463200902200328.
Whiting, P., Moher, D., 2021. The PRISMA 2020 statement: an updated guideline for Sierra, F., 2016. The emergence of geroscience as an interdisciplinary approach to the
reporting systematic reviews. PLoS Med 18, e1003583. https://doi.org/10.1371/ enhancement of health span and life span. Cold Spring Harb. Perspect. Med. 6,
journal.pmed.1003583. a025163. https://doi.org/10.1101/cshperspect.a025163.
Parsons, T.J., Papachristou, E., Atkins, J.L., Papacosta, O., Ash, S., Lennon, L.T., Stolz, E., Mayerl, H., Freidl, W., 2019. Fluctuations in frailty among older adults. Age
Whincup, P.H., Ramsay, S.E., Wannamethee, S.G., 2019. Physical frailty in older Ageing 48, 547–552. https://doi.org/10.1093/ageing/afz040.
men: prospective associations with diet quality and patterns. Age Ageing 48, Strehl, C., Ehlers, L., Gaber, T., Buttgereit, F., 2019. Glucocorticoids-all-rounders tackling
355–360. https://doi.org/10.1093/ageing/afy216. the versatile players of the immune system. Front. Immunol. 10, 1744. https://doi.
Pedersen, B.K., Febbraio, M., 2006. Exercise and interleukin-6 action. Expert Rev. org/10.3389/fimmu.2019.01744.
Endocrinol. Metab. 1, 319–321. https://doi.org/10.1586/17446651.1.3.319. Su, L., Hao, Q.-K., Liu, S., Dong, B.-R., 2017. Monocytes related inflammatory biomarkers
Picca, A., Beli, R., Calvani, R., Coelho-Júnior, H.J., Landi, F., Bernabei, R., Bucci, C., are associated with frailty syndrome. Int. J. Gerontol. 11, 225–229. https://doi.org/
Guerra, F., Marzetti, E., 2020. Older adults with physical frailty and sarcopenia show 10.1016/j.ijge.2017.08.004.
increased levels of circulating small extracellular vesicles with a specific Szanton, S.L., Allen, J.K., Seplaki, C.L., Bandeen-Roche, K., Fried, L.P., 2009. Allostatic
mitochondrial signature. Cells 9, 973. https://doi.org/10.3390/cells9040973. load and frailty in the women’s health and aging studies. Biol. Res. Nurs. 10,
Picca, A., Coelho-Junior, H.J., Cesari, M., Marini, F., Miccheli, A., Gervasoni, J., 248–256. https://doi.org/10.1177/1099800408323452.
Bossola, M., Landi, F., Bernabei, R., Marzetti, E., Calvani, R., 2019a. The Tajar, A., Lee, D.M., Pye, S.R., O’Connell, M.D.L., Ravindrarajah, R., Gielen, E.,
metabolomics side of frailty: toward personalized medicine for the aged. Exp. Boonen, S., Vanderschueren, D., Pendleton, N., Finn, J.D., Bartfai, G., Casanueva, F.
Gerontol. 126, 110692 https://doi.org/10.1016/j.exger.2019.110692. F., Forti, G., Giwercman, A., Han, T.S., Huhtaniemi, I.T., Kula, K., Lean, M.E.J.,
Picca, A., Ponziani, F.R., Calvani, R., Marini, F., Biancolillo, A., Coelho-Junior, H.J., Punab, M., Wu, F.C.W., O’Neill, T.W., 2013. The association of frailty with serum 25-
Gervasoni, J., Primiano, A., Putignani, L., Del Chierico, F., Reddel, S., Gasbarrini, A., hydroxyvitamin D and parathyroid hormone levels in older European men. Age
Landi, F., Bernabei, R., Marzetti, E., 2019b. Gut microbial, inflammatory and Ageing 42, 352–359. https://doi.org/10.1093/ageing/afs162.
metabolic signatures in older people with physical frailty and sarcopenia: results Tay, L., Leung, B.P., Wee, S., Tay, K.S., Ali, N., Chan, M., Lim, W.S., 2018. Association of
from the BIOSPHERE study. Nutrients 12, 65. https://doi.org/10.3390/ nutrition and immune-endocrine dysfunction with muscle mass and performance in
nu12010065. cognitively impaired older adults. Arch. Gerontol. Geriatr. 75, 20–27. https://doi.
Powers, S.K., Lynch, G.S., Murphy, K.T., Reid, M.B., Zijdewind, I., 2016. Disease-induced org/10.1016/j.archger.2017.11.008.
skeletal muscle atrophy and fatigue. Med. Sci. Sports Exerc. 48, 2307–2319. https:// Ter Borg, S., de Groot, L.C.P.G.M., Mijnarends, D.M., de Vries, J.H.M., Verlaan, S.,
doi.org/10.1249/MSS.0000000000000975. Meijboom, S., Luiking, Y.C., Schols, J.M.G.A., 2016. Differences in nutrient intake
Qu, T., Walston, J.D., Yang, H., Fedarko, N.S., Xue, Q.-L., Beamer, B.A., Ferrucci, L., and biochemical nutrient status between sarcopenic and nonsarcopenic older adults-
Rose, N.R., Leng, S.X., 2009a. Upregulated ex vivo expression of stress-responsive results from the maastricht sarcopenia study. J. Am. Med. Dir. Assoc. 17, 393–401.
inflammatory pathway genes by LPS-challenged CD14(+) monocytes in frail older https://doi.org/10.1016/j.jamda.2015.12.015.
adults. Mech. Ageing Dev. 130, 161–166. https://doi.org/10.1016/j. Theou, O., Brothers, T.D., Mitnitski, A., Rockwood, K., 2013. Operationalization of frailty
mad.2008.10.005. using eight commonly used scales and comparison of their ability to predict all-cause
Qu, T., Yang, H., Walston, J.D., Fedarko, N.S., Leng, S.X., 2009b. Upregulated monocytic mortality. J. Am. Geriatr. Soc. 61, 1537–1551. https://doi.org/10.1111/jgs.12420.
expression of CXC chemokine ligand 10 (CXCL-10) and its relationship with serum Tjia, J., Rothman, M.R., Kiely, D.K., Shaffer, M.L., Holmes, H.M., Sachs, G.A., Mitchell, S.
interleukin-6 levels in the syndrome of frailty. Cytokine 46, 319–324. https://doi. L., 2010. Daily medication use in nursing home residents with advanced dementia.
org/10.1016/j.cyto.2009.02.015. J. Am. Geriatr. Soc. 58, 880–888. https://doi.org/10.1111/j.1532-
Rockwood, K., Song, X., MacKnight, C., Bergman, H., Hogan, D.B., McDowell, I., 5415.2010.02819.x.
Mitnitski, A., 2005. A global clinical measure of fitness and frailty in elderly people. Tosti, V., Bertozzi, B., Fontana, L., 2018. Health benefits of the mediterranean diet:
Can. Med. Assoc. J. 173, 489–495. https://doi.org/10.1503/cmaj.050051. metabolic and molecular mechanisms. J. Gerontol. - Ser. Biol. Sci. Med. Sci. 73,
Rodríguez-Mañas, L., Angulo, J., Carnicero, J.A., El Assar, M., García-García, F.J., 318–326. https://doi.org/10.1093/gerona/glx227.
Sinclair, A.J., 2021. Dual effects of insulin resistance on mortality and function in Tuttle, C.S.L., Thang, L.A.N., Maier, A.B., 2020. Markers of inflammation and their
non-diabetic older adults: findings from the Toledo Study of Healthy Aging. 2021. association with muscle strength and mass: a systematic review and meta-analysis.
doi: 10.1007/s11357–021-00384–4. Ageing Res. Rev. 64, 101185 https://doi.org/10.1016/j.arr.2020.101185.
Rong, Y.-D., Bian, A.-L., Hu, H.-Y., Ma, Y., Zhou, X.-Z., 2018. Study on relationship Valdiglesias, V., Marcos-Pérez, D., Lorenzi, M., Onder, G., Gostner, J.M., Strasser, B.,
between elderly sarcopenia and inflammatory cytokine IL-6, anti-inflammatory Fuchs, D., Bonassi, S., 2018. Immunological alterations in frail older adults: a cross
cytokine IL-10. BMC Geriatr. 18, 308. https://doi.org/10.1186/s12877-018-1007-9. sectional study. Exp. Gerontol. 112, 119–126. https://doi.org/10.1016/j.
Ruangsuriya, J., Siviroj, P., Ayood, P., Ongprasert, K., Kaewtunjai, N., exger.2018.09.010.
Srichairatanakool, S., Tuntiwechapikul, W., Chittrakul, J., 2019. Biomarker: trolox Valentini, A., Cianfarani, M.A., Tarantino, U., Di Daniele, N., Bertoli, A., 2019.
equivalent antioxidant capacity and telomere length of Thai elderly people with Osteoprotegerin as a biomarker of geriatric frailty syndrome. Aging 11, 4900–4909.
frailty. J. Heal. Sci. 9, 144–150. https://doi.org/10.17532/JHSCI.2019.737. https://doi.org/10.18632/aging.102083.
Rusanova, I., Diaz-Casado, M.E., Fernández-Ortiz, M., Aranda-Martínez, P., Guerra- Varadhan, R., Yao, W., Matteini, A., Beamer, B.A., Xue, Q.L., Yang, H., Manwani, B.,
Librero, A., García-García, F.J., Escames, G., Mañas, L., Acuña-Castroviejo, D., 2018. Reiner, A., Jenny, N., Parekh, N., Daniele Fallin, M., Newman, A., Bandeen-
Analysis of plasma microRNAs as predictors and biomarkers of aging and frailty in Roche, K., Tracy, R., Ferrucci, L., Walston, J., 2014. Simple biologically informed
humans. Oxid. Med. Cell. Longev. 2018, 7671850. https://doi.org/10.1155/2018/ infammatory index of two serum cytokines predicts 10 year all-cause mortality in
7671850. older adults. J. Gerontol. - Ser. Biol. Sci. Med. Sci. 69 A, 165–173. https://doi.org/
Saeki, C., Kanai, T., Nakano, M., Oikawa, T., Torisu, Y., Saruta, M., Tsubota, A., 2020. 10.1093/gerona/glt023.
Low serum branched-chain amino acid and insulin-like growth factor-1 levels are Vettoretti, S., Caldiroli, L., Armelloni, S., Ferrari, C., Cesari, M., Messa, P., 2019.
associated with sarcopenia and slow gait speed in patients with liver cirrhosis. Sarcopenia is associated with malnutrition but not with systemic inflammation in
J. Clin. Med. 9, 3239. https://doi.org/10.3390/jcm9103239. older persons with advanced CKD. Nutrients 11, 1378. https://doi.org/10.3390/
Sánchez-Castellano, C., Martín-Aragón, S., Bermejo-Bescós, P., Vaquero-Pinto, N., Miret- nu11061378.
Corchado, C., Merello de Miguel, A., Cruz-Jentoft, A.J., 2020. Biomarkers of Walston, J., McBurnie, M.A., Newman, A., Tracy, R.P., Kop, W.J., Hirsch, C.H.,
sarcopenia in very old patients with hip fracture. J. Cachex. Sarcopenia Muscle 11, Gottdiener, J., Fried, L.P., 2002. Frailty and activation of the inflammation and
478–486. https://doi.org/10.1002/jcsm.12508. coagulation systems with and without clinical comorbidities: results from the
Saum, K.-U., Dieffenbach, A.K., Jansen, E.H.J.M., Schottker, B., Holleczek, B., Hauer, K., Cardiovascular Health Study. Arch. Intern. Med. 162, 2333–2341. https://doi.org/
Brenner, H., 2015. Association between oxidative stress and frailty in an elderly 10.1001/archinte.162.20.2333.

13
A. Picca et al. Ageing Research Reviews 73 (2022) 101530

Wilcox, P.G., Wakai, Y., Walley, K.R., Cooper, D.J., Road, J., 1994. Tumor necrosis factor in elderly people with diabetes-a cross-sectional study. BMC Geriatr. 20, 200.
α decreases in vivo diaphragm contractility in dogs. Am. J. Respir. Crit. Care Med. https://doi.org/10.1186/s12877-020-01601-z.
150, 1368–1373. https://doi.org/10.1164/ajrccm.150.5.7952566. Yang, Y., Hao, Q., Flaherty, J.H., Cao, L., Zhou, J., Su, L., Shen, Y., Dong, B., 2018.
Wu, I.-C., Shiesh, S.-C., Kuo, P.-H., Lin, X.-Z., 2009. High oxidative stress is correlated Comparison of procalcitonin, a potentially new inflammatory biomarker of frailty, to
with frailty in elderly chinese. J. Am. Geriatr. Soc. 57, 1666–1671. https://doi.org/ interleukin-6 and C-reactive protein among older Chinese hospitalized patients.
10.1111/j.1532-5415.2009.02392.x. Aging Clin. Exp. Res. 30, 1459–1464. https://doi.org/10.1007/s40520-018-0964-3.
Yalcin, A., Silay, K., Balik, A.R., Avcioğlu, G., Aydin, A.S., 2018. The relationship Yeap, B.B., Paul Chubb, S.A., Lopez, D., Ho, K.K.Y., Hankey, G.J., Flicker, L., 2013.
between plasma interleukin-15 levels and sarcopenia in outpatient older people. Associations of insulin-like growth factor-I and its binding proteins and testosterone
Aging Clin. Exp. Res. 30, 783–790. https://doi.org/10.1007/s40520-017-0848-y. with frailty in older men. Clin. Endocrinol. 78, 752–759. https://doi.org/10.1111/
Yanagita, I., Fujihara, Y., Iwaya, C., Kitajima, Y., Tajima, M., Honda, M., Teruya, Y., cen.12052.
Asakawa, H., Ito, T., Eda, T., Yamaguchi, N., Kayashima, Y., Yoshimoto, M., Yoo, J.-I., Ha, Y.-C., Choi, H., Kim, K.-H., Lee, Y.-K., Koo, K.-H., Park, K.-S., 2018.
Harada, M., Yoshimoto, S., Aida, E., Yanase, T., Nawata, H., Muta, K., 2020. Low Malnutrition and chronic inflammation as risk factors for sarcopenia in elderly
serum albumin, aspartate aminotransferase, and body mass are risk factors for frailty patients with hip fracture. Asia Pac. J. Clin. Nutr. 27, 527–532. https://doi.org/
10.6133/apjcn.082017.02.

14

You might also like