You are on page 1of 10

Ageing Research Reviews 20 (2015) 1–10

Contents lists available at ScienceDirect

Ageing Research Reviews


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

Review

Cognitive frailty, a novel target for the prevention of elderly


dependency
Ruan Qingwei a,b , Yu Zhuowei a,b,c,∗ , Chen Ma a,b , Bao Zhijun b , Li Jin c , He Wei d
a
Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China
b
Research Center of Aging and Medicine, Shanghai Key Laboratory of Clinical Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University,
Shanghai 200040, China
c
Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
d
PET-CT Center, Department of Nuclear Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China

a r t i c l e i n f o a b s t r a c t

Article history: Frailty is a complex and heterogeneous clinical syndrome. Cognitive frailty has been considered as a sub-
Received 11 October 2014 type of frailty. In this study, we refine the definition of cognitive frailty based on existing reports about
Received in revised form frailty and the latest progress in cognition research. We obtain evidence from the literature regarding
12 December 2014
the role of pre-physical frailty in pathological aging. We propose that cognitive impairment of cogni-
Accepted 16 December 2014
tive frailty results from physical or pre-physical frailty and comprises two subtypes: the reversible and
Available online 30 December 2014
the potentially reversible. Reversible cognitive impairment is indicated by subjective cognitive decline
(SCD) and/or positive fluid and imaging biomarkers of amyloid-␤ accumulation and neurodegenera-
Keywords:
Cognitive frailty
tion. Potentially reversible cognitive impairment is MCI (CDR = 0.5). Based on the severity of cognitive
Physical frailty impairment, it is possible to determine the primary and secondary preventative measures for cognitive
Pre-physical frailty frailty. We further determine whether SCD is a component of pre-clinical AD or the early stage of other
Pre-MCI neurodegenerative diseases, which is required for guiding personal clinical intervention.
Subjective cognitive decline © 2014 Elsevier B.V. All rights reserved.
Pre-clinical AD

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
2. The rationale for cognitive frailty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2. Chronological order between physical frailty and cognitive impairment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.3. Support for pre-physical frailty as a cause of cognitive frailty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.4. Cognitive frailty and MCI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.5. Cognitive frailty and SCD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. The suggested definition of cognitive frailty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
4. The possible mechanisms of cognitive frailty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
5. The proposed tools to measure reversible cognitive frailty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
6. The prevention of cognitive frailty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7

1. Introduction

∗ Corresponding author at: Huadong Hospital, Shanghai Medical College, Fudan


Aging is the progressive and overall physiological decline of
University, 221 West Yan An Road, Shanghai 200040, China. Tel.: +86 21 62483180;
the reserves of an organism, which decreases the ability to gen-
fax: +86 21 62484981. erate adaptive responses and sustain homeostasis. Consequently,
E-mail address: qingweir@aliyun.com (Z. Yu). the body becomes more susceptible to stress, diseases and injuries.

http://dx.doi.org/10.1016/j.arr.2014.12.004
1568-1637/© 2014 Elsevier B.V. All rights reserved.
2 Q. Ruan et al. / Ageing Research Reviews 20 (2015) 1–10

The loss of essential body functions leads to age-related patholo- Shimada et al., 2013; Mhaoláin et al., 2012; Buchman et al., 2007,
gies, which ultimately lead to death (López-Otín et al., 2013; 2008). Comorbidities and physical frailty are often found in patients
Moskalev et al., 2014). Diseases accelerate aging, thereby dimin- with Alzheimer’s disease (AD) and are closely related to the het-
ishing the body’s adaptation via the relevant stress responses. erogeneity and clinical manifestations of this disease (Oosterveld
Multiple subclinical and age-related comorbidities in the elderly et al., 2014). Physical frailty, combined with cognitive impairment,
may exacerbate the decline in the physiological reserves of several is predictive of an increased risk of a poor prognosis. In other
systems, which would then result in a homeostatic imbalance or words, the phenotypic integration of cognitive impairment and
frailty (Clegg et al., 2013). Aging compromises the adaptive stress frailty clearly improves the predictive effectiveness of the latter
responses of an organ or the body to external stimuli, such as the regarding a poor prognosis in a 4-year follow-up of elderly subjects
ability to adapt in response to abrupt changes in health conditions. (in which the top five categories include death, disability, enter-
Frail elderly individuals display a greater apparent vulnerability to ing a nursing facility, hospitalization and a fall) (Rockwood, 2005;
minor diseases in that this population often has difficulty in retur- Avila-Funes et al., 2009; Cano et al., 2012; Kulmala et al., 2014).
ning to baseline equilibrium after mild stressors (Clegg et al., 2013; Cognitive frailty refers to the heterogeneous clinical syndrome
Ferrucci et al., 2002). Frailty is a pathological aging process that is that is found in elderly individuals, excluding those with AD and
reversible and occurs at an intermediate stage between age-related other dementias, that is characterized by concurrent physical frailty
diseases and a poor prognosis, such as disability or death (Moeley and potentially reversible cognitive impairment (Clinical dementia
et al., 2013; Clegg et al., 2014; Dorner et al., 2013; Tavassoli et al., rating score (CDR) = 0.5) (Kelaiditi et al., 2013). This implies that
2014). In other words, early intervention can improve the patient’s cognitive frailty is a form of pathological brain aging and a precur-
quality of life and reduce the adverse health consequences for the sor to neurodegenerative processes. Physical frailty and cognition
patient, thereby achieving the goal of healthy aging. are associated, however, the causal links between physical frailty
Based on different pathogeneses, frailty can be divided and cognitive impairment are not clear. The aim of the consensus
into physical frailty, cognitive frailty and psychosocial frailty group is to establish a conceptual framework for the study of cogni-
(Malmstrom and Morley, 2013; Panza et al., 2011). A broad con- tive impairment in individuals due to physical reasons (including a
sensus has been reached for the definition of physical frailty, psychological component resulting from physical impairment), but
which manifests as a reduction in body strength, stamina and not due to the presence of a concomitant neurological disease. The
physiological functions, which in turn, leads to an increased possi- significance of this definition lies in providing a novel target for sec-
bility of physical vulnerability, disability and death (Moeley et al., ondary prevention to stem elderly disability. However, developing
2013). Typically, physical frailty is suggested when at least three a definition requires a number of challenges to be addressed before
of the five following criteria are present: fatigue, decreased mus- the definition can be promoted clinically and used in future. In this
cle strength, low walking speed, reduced physical activities and article, we discuss the specific issues that are useful for clarifying
unintentional weight loss; pre-physical frailty is determined by the definition and research criteria for cognitive frailty in medical
the presence of one or two of the five criteria (Moeley et al., practice and clinical research.
2013). Cognitive frailty was proposed by Panza et al. (2006) when
these authors examined the risks of decreased cognitive functions 2.2. Chronological order between physical frailty and cognitive
modulated by vascular factors. Subsequent studies revealed that impairment
physical factors and cognition are crucial elements in predict-
ing risk of death (Panza et al., 2006, 2011; Pilotto et al., 2012). Because the underlying mechanisms for the physical impair-
To differentiate cognitive impairment not dementia (CIND) in ment due to cognitive decline are well known, the consensus group
the absence of physical frailty, a consensus on the definition of hopes to explain a cognitive decline caused by physical conditions
cognitive frailty was reached by the International Academy on or factors independent of neurodegenerative conditions. However,
Nutrition and Aging and the International Association of Gerontol- a consensus on the definition of cognitive frailty requires the simul-
ogy and Geriatrics in April, 2013 (Kelaiditi et al., 2013). Although taneous presence of both physical frailty and cognitive impairment
the emergence of cognitive frailty reflects a verifiable improve- (CDR = 0.5) (Kelaiditi et al., 2013). Thus, there may be confusion in
ment in preventing a poor prognosis of aging, many experts in designing clinical experiments. If physical frailty is present before
the field have provided beneficial suggestions for clarifying the cognitive impairment occurs, researchers of cognitive frailty should
definition of cognitive frailty (Woods et al., 2013; Buchman and recruit individuals with only physical frailty to carry out a lon-
Bennett, 2013; Dartigues and Amieva, 2014). Based on physical gitudinal study. Although a cross-sectional study design would
weakness and cognitive impairment, especially in light of the lat- be appropriate when both physical frailty and cognitive impair-
est consensus arising from studies of subjective cognitive decline ment (CDR = 0.5) are present, the subjects in the study may include
(SCD) and fluid and imaging biomarkers of amyloid-␤ accumula- individuals whose physical impairment may result from cognitive
tion and neurodegeneration, we herein refine the framework for decline (CDR = 0.5). About one-third of mild cognitive impairment
the definition and potential mechanisms of cognitive frailty, as well (MCI) patients experience obstacles in the instrumental activities
as relevant screening tools. Furthermore, we explore the signifi- of daily living (Morris, 2012). Moreover, these cognitively frail indi-
cance of our suggestions in preventing the progression of cognitive viduals can also be referred to as physically frail individuals with
frailty. MCI (CDR = 0.5).

2.3. Support for pre-physical frailty as a cause of cognitive frailty


2. The rationale for cognitive frailty
A growing body of research evidence supports connections
2.1. Overview between frailty, cognitive impairment and dementia. It has been
shown that cognitive frailty can be a prelude to degenerative neu-
The physical phenotype of frailty and cognitive impairment has rological diseases and that physical frailty can enhance the future
an obvious interrelationship. During the aging process, the inter- risk of MCI and dementia in cognitively normal populations, as
action of physical frailty and cognitive impairment causes a cycle well as accelerate the cognitive impairment of these individuals
of physical and cognitive functional decline and poor quality of (Boyle et al., 2010; Buchman et al., 2007; Auyeung et al., 2011).
life (Houles et al., 2012; Robertson et al., 2013; Han et al., 2014; Physical frailty can augment the risk of delirium and death in
Q. Ruan et al. / Ageing Research Reviews 20 (2015) 1–10 3

Fig. 1. The components of cognitive frailty and different trajectories of both physical and cognitive functions under specific conditions.

hospitalized elderly individuals (Eeles et al., 2012). Studies have 2.4. Cognitive frailty and MCI
revealed that cognitive impairment is secondary to a plethora of
illnesses, such as age-associated health problems, heart disease, A major controversial point regarding the definition of cognitive
hypertension, stroke, diabetes, chronic kidney disease, viral infec- frailty is reversible cognitive impairment (CDR = 0.5), which can
tions (e.g., HIV and hepatitis C) and substance use (Oosterveld be fairly confusing. It was proposed that a CDR value of 0.5 was
et al., 2014; Knopman and Roberts, 2010; Devlin et al., 2012; equivalent to the MCI stage (Hughes et al., 1982; Morris, 1993).
Hayden et al., 2010; Weiner and Seliger, 2014). In addition, an On one hand, this criterion makes it difficult to discriminate cog-
age-associated decline in health status is also a risk factor of AD nitive frailty from MCI, prodromal period AD and CIND; on the
and dementia (Song et al., 2011). The severity of physical frailty other hand, progressive neuronal cell loss during the MCI stage
can be evaluated by adopting a numerical score or studying a may surpass the physiological ability of the brain to compensate;
dynamic state over time (Puts et al., 2005a; Moeley et al., 2013; thus, irreversible cognitive damage might have already occurred
Tavassoli et al., 2014; Pilotto et al., 2012; de Vries et al., 2011, (Gomez-Isla et al., 1996; Aisen, 2008). Based on the neuropsychol-
2012; Bouillon et al., 2013; Ng et al., 2014). It was reported that ogical profile, MCI is divided into three subtypes: amnestic MCI
elderly subjects with physical frailty did indeed have a signifi- is considered to progress preferentially to AD; MCI with a slight
cantly higher degree and percentage of cognitive impairment than impairment of multiple cognitive domains may progress to either
those displayed by elderly individuals who were robust and had AD or vascular dementia or be part of the normal cognitive aging
pre-physical frailty. Additionally, the elderly subjects with physical process; and single-domain non-memory MCI may progress to
frailty combined with cognitive impairment exhibited even higher non-AD dementia (Portet et al., 2006). Some MCI patients have
rates of disability (Avila-Funes et al., 2009). A longitudinal study symptoms that are reversible and can recover to regain normal
with an average duration of five years revealed that mild physi- cognitive function. The cognitive functions of other patients may
cal impairment (according to a performance-based assessment) in even be stable and not change throughout the remainder of their
cognitively normal elderly adults might lead to cognitive decline, lives. However, more MCI patients exhibit an irreversible, pro-
which could later be clinically detected and was closely associ- gressive reduction in cognition (Golomb et al., 2004; Matthews
ated with the development of dementia due to AD (Wilkins et al., et al., 2008; Mitchell and Shiri-Feshki, 2009). In the early stage of
2013). The clinical manifestations of physical frailty also display an cognitive decline, cognitively impaired individuals often display a
apparent hierarchy. Weakness is the earliest emerging condition of functional loss in more complicated tasks (e.g., instrumental activi-
pre-physical frailty and, during this stage, the presence of slowness, ties of daily living), whereas the functional loss of simpler tasks (i.e.,
reduced activity/exhaustion and weight loss indicates a rapidly ris- activities of daily living) appears in the more severe stage of cogni-
ing risk of pre-physical frailty transforming into physical frailty tive impairment (Millán-Calenti et al., 2012; Njegovan et al., 2001;
(Xue et al., 2008) (Fig. 1). Frailty is a process of dynamic transforma- Barberger-Gateau et al., 1992). Studies have revealed that 34% of
tion. Based on clinical, functional, behavioral and biomarker-based MCI patients experience obstacles in performing the instrumental
differences, even pre-physical frailty is clearly different from nor- activities of daily living (e.g., managing the household economy),
mal aging and is part of a pathological aging process (Abellan van and this percentage is significantly higher than that (5%) of non-
Kan et al., 2008). Therefore, prompt intervention can achieve a bet- MCI populations (Morris, 2012). According to the definition, the
ter outcome, and pre-physical frailty has already become a target appearance of physical disabilities is clearly not a component of
for primary prevention (Abellan van Kan et al., 2008). Addition- physical frailty. Previous studies indicated that immunotherapy-
ally, a question has been raised as to whether pre-physical frailty based clinical trials involving humanized monoclonal antibodies
in patients should be incorporated as a physical factor contributing against amyloid ␤ peptide, bapineuzumab and solanezumab, failed.
to cognitive frailty (Dartigues and Amieva, 2014). We propose here, Moreover, in a 5-year comparative study, Ginkgo biloba extract and
that clinical pre-physical frailty should be adopted as a diagnostic a placebo were used as agents of intervention to examine a large
criterion of cognitive frailty. cohort of subjective cognitive impairment and/or MCI subjects
4 Q. Ruan et al. / Ageing Research Reviews 20 (2015) 1–10

(2854 individuals); the results revealed that Ginkgo biloba extract, 3. The suggested definition of cognitive frailty
a potent antioxidant, could not reduce the risk of progression of
impairment in AD patients. A lesson learned from these studies Based on the above rationale, we suggest the definition of cog-
is that the target (cognitive impairment in the study subjects) of nitive frailty to be a heterogeneous clinical syndrome of cognitive
secondary prevention should be a focus in pre-clinical AD or asymp- impairment (CDR ≤ 0.5) that develops in elderly individuals, is
tomatic AD (Doody et al., 2014; Vellas et al., 2012, 2013). At the caused by physical factors (e.g., physical frailty and pre-physical
asymptomatic stage, patients have produced in vivo signals (e.g., frailty) and is excluded from dementia resulting from AD or other
positive for certain biomarkers) that are indicative of their brain conditions. Cognitive frailty includes two subtypes, reversible and
diseases (Sperling et al., 2011, 2013; Caselli and Reiman, 2013). potential reversible cognitive frailty (Figs. 1 and 2). The cognitive
impairment of reversible cognitive frailty is SCD and/or positive
biomarkers resulting from physical factors. SCD and/or positive
2.5. Cognitive frailty and SCD biomarkers may occur at the late stage of pre-clinical AD or at
the pre-MCI stage due to other causes. Only SCD and/or posi-
Recently, the Subjective Cognitive Decline Initiative (SCD-I) tive biomarkers, unrelated to an acute event, clinical diagnosis of
Working Group formulated a basic conceptual framework for the neurodegenerative and mental conditions, are caused by physical
study of the common concepts of SCD, pre-MCI SCD and SCD factors and is cognitive impairment of reversible cognitive frailty
in pre-clinical AD (Jessen et al., 2014). SCD refers to a type of (Fig. 1). The cognitive impairment of potentially reversible cogni-
cognitive decline in which, although the subjects have altered tive frailty is MCI (CDR = 0.5), which had been discussed in detail by
subjective cognitive function, including subjective memory dys- the consensus group.
function or other types of deterioration in subjective cognitive
functions (e.g., executive function, attention, language and visu-
ospatial function), the individuals display a normal performance 4. The possible mechanisms of cognitive frailty
in cognitive testing (Fig. 1) (Jessen et al., 2014). SCD is char-
acterized by increasing compensatory cognitive efforts (normal The aging of organisms is caused by a series of complex and
cognitive performance range) and subtle cognitive decline in two interconnected events. Nutrition-sensing signals, such as growth
phases (Fig. 1). After the subtle cognitive decline, cognitive func- factors, insulin and insulin-like growth factor 1 (IGF 1), can acti-
tional reserve is not sufficient to maintain normal age-, sex-, and vate mammalian target of rapamycin receptor complex 1 and
education-adjusted performance due to increasing brain pathol- inhibit adenosine monophosphate (AMP)-activated protein kinase,
ogy and objective cognitive decline (MCI or prodromal AD) occurs which leads to elevated activities of p53 and p16 and, in turn,
(Fig. 1). Therefore, in the scope of cognitive impairment onward causes telomere shortening and DNA damage. As a consequence,
to the stage of dementia, SCD is a non-specific condition that mitochondrial biogenesis and function decline, which then ush-
not only may appear as the first symptom of pre-clinical AD (the ers in a series of events, including cell senescence, stem cell
asymptomatic at-risk stage of AD or pre-MCI stage of AD patients), depletion, alterations in intercellular signals, disorders of the regu-
but may also emerge in various types of pre-MCI patients, such latory systems, neuroendocrine dysfunction, inflammatory aging
as those with normal aging, certain personality traits (e.g., ner- and immune senescence. These events collectively culminate in
vousness or anxiety), an individual cultural background, non-AD the aging of the body (Fig. 2) (López-Otín et al., 2013; Sahin and
mental or neurological disorders, other diseases (e.g., cerebrovas- DePinho, 2013; Moskalev et al., 2014). Moreover, genetic and envi-
cular risk factors, stroke or former head injury), and drug-abuse ronmental elements, in conjunction with epigenetic mechanisms,
(Jessen et al., 2014). Longitudinal neuropsychological assessment lead to the accumulation of metabolic errors and molecular and
can indicate the time of symptomatic conversion to MCI. The util- cellular damages that are manifested as excessive exogenous and
ity of SCD is to pre-select subjects who may have an early cognitive endogenous genomic injury and inadequate DNA repair. Subse-
impairment before the MCI stage. The major advantages of enrich- quently, DNA exhibits a reduction in overall methylation but a
ment by SCD are its low cost, safety, convenience and a short localized methylation enhancement, altered histone modification
time in clinical application. However, it is noteworthy that some and chromatin remodeling, which compromise protein homeo-
subjects with reversible cognitive impairment probably do not stasis in various pathways and trigger autophagy, proteasome
have SCD. These individuals could be identified by biomarkers of degradation, protein refolding and protein aggregation (López-Otín
both amyloid ␤-accumulation and neurodegeneration or neuronal et al., 2013; Moskalev et al., 2014).
injury. Aging causes a state of increased vulnerability/susceptibility
Cognitive impairment (CDR = 0.5) in the present definition of and a gradual decrease in the physiological reserves of mul-
cognitive frailty, obviously ignores individuals with reversible cog- tiple organs, as well as pathologies associated with increased
nitive impairment, such as SCD and/or positive biomarkers of age, such as neuroendocrine dysfunction, systemic homeostatic
both amyloid ␤-accumulation and neurodegeneration or neuronal mechanisms (e.g., oxidative damage, inflammation and mitochon-
injury (CDR < 0.5). If MCI (CDR = 0.5) is a potentially reversible cog- drial activities), metabolic modifications, reduced physical activity,
nitive impairment, SCD and/or positive biomarkers are an early immune senescence and aberrant apoptosis and energy production
cognitive impairment that occur before MCI and have an evidently (Wang et al., 2010; Leng et al., 2004a,b; Walston et al., 2002, 2006,
reversible nature. As reversible cognitive damage is clearly optimal 2008; Ho et al., 2011; Akki et al., 2014; Ko et al., 2012; Puts et al.,
for serving as a secondary target for the prevention of cognitive 2005b; Lang et al., 2009) (Fig. 2). In the course of aging, hormesis
impairment, the level of the cognitive damage in cognitive frailty can only occur when there is a sufficient level of stress responses,
ought to include SCD and/or positive biomarkers which are lower which accelerate senescence and manifest as a collapse of stress-
than that with MCI. Because numerous causes may lead to SCD apart resistance, protein denaturation or aggregation, the accumulation
from AD, we adopt the appearance of pre-MCI SCD and/or posi- of DNA mutations, mitochondrial insufficiency, dysfunctional Ca2+
tive biomarkers, which have a relatively broad scope as diagnostic homeostasis, cellular senescence and apoptosis (Moskalev et al.,
criteria of cognitive frailty (Fig. 1) (Jessen et al., 2014). Obviously, 2014). When people age, the emergence of abnormalities in three
SCD with a clinical diagnosis of acute impairment, mental or neu- or more systems (e.g., pathophysiological modifications caused
rological diseases should be excluded as cognitive impairment of by cardiovascular and pulmonary diseases and diabetes) may
cognitive frailty. accelerate the depletion of physiological reserves and initiate
Q. Ruan et al. / Ageing Research Reviews 20 (2015) 1–10 5

Fig. 2. The cycle of physical frailty, cognitive frailty and cognitive impairment. The physical impairment caused cognitive frailty is outlined in red arrows, proposed cognitive
frailty components are outlined in red box with pink background and previous cognitive frailty components are outlined in blue dotted line.

frailty-related homeostatic failure. As a consequence, homeostasis (e.g., vitamin D deficiency), hormones (e.g., reduced testosterone,
becomes less likely to be sufficient for the maintenance and repair insulin resistance), lifestyle and mental health issues (Robertson
of the aging body, which in turn becomes vulnerable regarding et al., 2013; Houles et al., 2012; Kelaiditi et al., 2013). There are
stress responses, such that even minor stress events can lead to intricate connections among physical frailty, changes in cognitive
the above hormesis process and disproportionate changes in health function and the pathological alterations in dementia. A longitu-
(Clegg et al., 2013; Fried et al., 2001, 2009; Varadhan et al., 2008; dinal clinical-pathologic study of aging was performed to examine
Ferrucci et al., 2008; Kalyani et al., 2012; Chaves et al., 2008). the relationship between physical frailty in elderly subjects and
Genetic and environmental stressors may cause dysfunctions common postmortem age-related pathological signs in the brain,
in susceptible neurons of specific brain systems (e.g., medial tem- such as cerebral infarcts, the Lewy body pathway and the AD
poral lobe system and fronto-striatal system) and the decline of pathway; the results revealed that physical frailty was related to
structural (e.g., number of neurons and synapses) and functional the pathology of AD in older persons with and without demen-
reserves. Finally, heterogeneous brain aging or different neurode- tia (Buchman et al., 2008). Another longitudinal clinical-pathologic
generative disease occurs (Canevelli et al., 2014). Numerous studies study which had an average duration of 6.5 years and had no base-
have revealed that multiple risk factors that cause cognitive impair- line of dementia, revealed that physical frailty was associated with a
ment are also associated with the development and worsening higher risk of developing non-AD dementia but not AD (Gray et al.,
of physical frailty in older individuals (Robertson et al., 2013; 2013). Furthermore, a recent longitudinal study with an average
Houles et al., 2012; Kelaiditi et al., 2013). The risk factors include duration of 6 years revealed that physical frailty and altered cogni-
cardiovascular elements (e.g., diabetes, dyslipidemia, hyper- tive function were closely related and that brain pathology, which
tension, inflammation and hyperhomocysteinemia), nutrition encompasses AD pathology, macroinfarcts and nigral neuronal loss,
6 Q. Ruan et al. / Ageing Research Reviews 20 (2015) 1–10

displayed a significant correlation with the rate of change in both depression or anxiety), neurologic disease (apart from AD) (Jessen
physical frailty and cognition. It was therefore postulated that phys- et al., 2014).
ical frailty and changes in cognitive function may, in part, share a To identify individuals with reversible cognitive impairment
common pathological basis (Buchman et al., 2014). and in an absence of SCD, implementation of a personal secondary
According to our proposed definition, reversible cognitive intervention to the pre-MCI SCD caused by physical factors in
impairment of cognitive frailty results from pre-physical or phys- individuals with cognitive frailty, having one screening tool to dif-
ical frailty and may lead to an outcome of dementia originating ferentiate whether a patient with pre-MCI SCD is pre-clinical AD
from AD or other factors (Fig. 2). Thus far, direct evidence or another neurodegenerative disease in the subclinical stages is
for mechanistically studying cognitive frailty has been lacking, indispensable. Preclinical AD SCD research criteria, in combina-
but epidemiological results have suggested that heart disease, tion with biomarkers of preclinical AD, may be a useful screening
hypertension, stroke, diabetes, chronic kidney disease and an age- tool (Jessen et al., 2014; Sperling et al., 2013). A preliminary
associated decline in health status were recognized as AD, dementia questionnaire screening tool includes the following critical prop-
or cognitive decline risk factors (Okonkwo et al., 2010; Knopman erties: (1) a subjective decline in memory, rather than in the
and Roberts, 2010; Weiner and Seliger, 2014; Song et al., 2011; other domains of cognition; (2) an onset within the last five
Elwood et al., 2002). A prospective cohort study that had a dura- years; and (3) an age at onset of SCD ≥ 60 years. Biomarkers of
tion of 12 years and examined 750 community elderly subjects with preclinical AD include the following: (i) markers of amyloid ␤-
normal cognitive function revealed that physical frailty was closely accumulation, such as the level of amyloid ␤42 in cerebrospinal
associated with the risk of the occurrence of MCI (Boyle et al., 2010). fluid, and positron emission tomography (PET) amyloid imaging;
Likewise, another prospective cohort study which had a duration (ii) markers of neurodegeneration or neuronal injury, including the
of four years and examined 2737 elderly individuals with normal level of tau and phosphorylated tau protein in cerebrospinal fluid;
cognition, also showed that physical frailty was associated with a 18 F-fluorodeoxyglucose PET functional imaging or functional mag-

faster rate of cognitive decline (Auyeung et al., 2011). Until now, netic resonance imaging (MRI); and nerve degeneration or damage,
the mechanism of physical and/or pre-physical frailty that causes such as that revealed by the MRI-based detection of hippocampus
cognitive decline is unknown. An alternative two-hit vascular atrophy or cortical thinning. Based on various combinations of amy-
hypothesis of AD may be one of the mechanisms of cognitive decline loid ␤-aggregation, nerve degeneration and the subtle reduction of
by physical causes (Sagare et al., 2012; Zlokovic, 2011). Accord- cognitive function, pre-clinical AD can be divided into four stages
ing to the hypothesis, vascular risk factors and cerebrovascular (Sperling et al., 2013). In addition, homozygous ApoE ε4 is a useful
disorder lead to brain oligemia and hypoxia and/or blood-brain biomarker for late onset familial and sporadic AD patients. In the
barrier (BBB) dysfunction, which reduce amyloid ␤-peptide (A␤) absence of results in “classical” cognitive tests, we have to consider
vascular clearance across the BBB and increase A␤-production from the association of self-report measures with existing biomarkers.
A␤-precursor protein, casing A␤-accumulation and tau-related Because of the heterogeneous features of SCD in preclinical AD,
pathology in brain. the questionnaire preliminary screening tool does not compre-
hensively define the specific characteristics of SCD in preclinical
AD in individuals of cognitive frailty. According to current knowl-
5. The proposed tools to measure reversible cognitive edge, when all of the self-report measures, markers of amyloid
frailty ␤-accumulation and markers of neurodegeneration or neuronal
injury are positive, the subject may belong to stage 2 or 3 of the
Based on the mechanisms of frailty, including those of phe- proposed staging of preclinical AD by the US National Institute on
notypic, defect accumulation, the aggregation of multifunctional Aging (Sperling et al., 2013). When self-report measures and only
domains and multidimensions, various frailty-screening tech- markers of amyloid ␤-accumulation are positive, the subject is in
niques have been indicated (de Vries et al., 2011; Bouillon et al., stage 1 of the proposed staging of preclinical AD. Although amyloid
2013; Sternberg et al., 2011). Among these, Fried’s phenotypic ␤-accumulation is currently the first biomarker to indicate AD and
physiology-based screening is being preferentially used for frailty has a greater specificity compared with markers of neurodegen-
research (Fried et al., 2001), whereas scales for frailty resulting from eration or neuronal injury, it also may be positive in individuals
defect accumulation are more suitable for health management, who have dementia with Lewy bodies and/or cerebral amyloid
such as predicting whether an elderly individual requires hospital- angiopathy. When self-report measures and only markers of neu-
ization (Rockwood and Mitnitski, 2007). In addition, those clinical rodegeneration or neuronal injury are positive, the subject may
screening instruments for rapidly examining cognitive impairment have suspected non-Alzheimer pathology, including normal aging
in populations with physical frailty and cognitive frailty, such as and other age-related neurodegenerative diseases. The biomarker
the Simple “FRAIL” Questionnaire Screening Tool and the Rapid homozygous ApoE ε4 is helpful for the differentiation of diagnosis.
Cognitive Screen (RCS), can meet the daily examination require- When only self-report measures are positive, the subject is at stage
ments of clinicians in identifying subjects at risk (Moeley et al., 0 of the proposed staging of preclinical AD. Even if the individual
2013; Malmstrom and Morley, 2013). A broad consensus has been probably experiences early AD processes that are undetectable with
reached for the screening of pre-physical or physical frailty subjects current biomarkers, the subject should be considered as a normal
for cognitive frailty, whereas the screening of cognitive function individual after excluding psychiatric disorders. If only biomarkers
in reversible cognitive frailty is absent. We suggest the use of pre- are positive without self-report measures, the differentiation of the
MCI SCD research criteria with minor modifications, in combination diagnosis of preclinical AD from other neurodegenerative diseases
with suggested SCD features as a preliminary screening tool for is according to the proposed staging of preclinical AD by the US
cognitive function, including the following: (1) a self-experienced National Institute on Aging (Sperling et al., 2013).
persistent decline in cognitive capacity compared with a previously
normal status and unrelated to an acute event; (2) normal age-,
gender- and education-adjusted performance on standardized cog- 6. The prevention of cognitive frailty
nitive tests that are used to classify MCI or prodromal AD; and 1
and 2 must both be met. The exclusion criteria include the follow- Preventive intervention is required for elderly subjects in a pre-
ing: (1) MCI, prodromal AD or dementia and (2) conditions that physically frail state to reduce the risk of developing physical frailty
can be explained by a psychiatric disorder (reach the threshold of syndromes (Abellan van Kan et al., 2008; Lang et al., 2009). In
Q. Ruan et al. / Ageing Research Reviews 20 (2015) 1–10 7

addition, secondary interventions should be provided for elderly seem to improve muscle strength or bone density in healthy elderly
subjects with physical frailty syndromes that are in a reversible women (Friedlander et al., 2001). Moreover, low levels of 25(OH)
state to thereby avoid a reduction in physical functions (Abellan van D were strongly associated with the prevalence and incidence of
Kan et al., 2008). SCD is an effective target for cognitive impairment frailty, and moderately elevated levels of C-reactive protein (CRP)
intervention (Canevelli et al., 2013; Sperling et al., 2011). Elderly might play a role in the incidence of frailty (Puts et al., 2005b). How-
individuals with cognitive frailty comprise those with pre-physical ever, a series of studies indicated that low levels of 25(OH) D were
or physical frailty combined with pre-MCI SCD and/or positive fluid associated with the prevalence of frailty in both elderly women
and imaging biomarkers of amyloid-␤ accumulation and neurode- and men and that the level could predict a greater risk of frailty in
generation. In comparison with pre-physical and physical frailty elderly women, but not in elderly men (Ensrud et al., 2010, 2011).
syndromes, the prevention of cognitive frailty requires particu- Severe vitamin D deficiency was also reported to be associated with
lar treatments (Fig. 1). For the elderly subjects with reversible advanced-stage dementia in geriatric inpatients (Annweiler et al.,
cognitive frailty, primary preventive intervention includes the pro- 2011). Although there is no direct evidence supporting the idea
motion of physical activities, cognitive stimulation, exercise and a that a large-scale intervention with vitamin D can prevent or treat
healthy diet (e.g., a Mediterranean diet, the cessation of smoking, frailty, a meta-analysis demonstrated that the combined treatment
promoting emotional recovery, engaging in an active and socially with vitamin D and calcium, but not vitamin D alone, reduced mor-
integrated lifestyle, an ideal amount of daily sleep, the mainte- tality in the elderly subjects (Rejnmark et al., 2012). Sarcopenia is a
nance of a proper body weight and metabolic control (including common cause of frailty. Young plasma and GDF11 can rejuvenate
the control of dyslipidemia, diabetes and blood pressure)), as these aged organ and tissues of the aging mouse, increase physical per-
measures boost patients’ ability to fight the disease (Sternberg formance, such as cognitive function and muscular performance
et al., 2011; Desai et al., 2010). At this stage, secondary preven- (Villeda et al., 2014; Sinha et al., 2014). GDF11 or oxytocin can acti-
tions for cognitive imapirment such as preclinical AD0 and physical vate adult muscle stem cell, improving muscle regeneration and
frailty are suggested. For the elderly subjects with potential cogni- performance (Sinha et al., 2014; Elabd et al., 2014). These systemic
tive frailty, secondary prevention is required, which comprises a factors may potentially contribute to the treatment of cognitive
geriatric assessment determining the cause of cognitive frailty and frailty.
an evidence-based, medicinal, individualized multimodal interven-
tion (Fig. 1). Other measures, such as drug treatment for chronic
7. Conclusion
diseases, fall prevention, exercise and nutrition support, which tar-
get physical, nutritional, cognitive and psychological domains, may
Based on the progress of research involving studies of cognitive
delay the progression and secondary occurrence of cognitive frailty
impairment and frailty, we herein refine the definition of cognitive
related adverse outcomes (Sternberg et al., 2011; Desai et al., 2010;
frailty. Consequently, by referring to the level of cognitive impair-
Kelaiditi et al., 2013; Moeley et al., 2013). Lastly, for the elderly
ment caused by pre-physical or physical frailty, we may determine
subjects with irreversible cognitive impairment caused by physical
whether primary prevention or secondary prevention is indicated.
factors that may lead to adverse health-related outcomes, such as
This definition suggests that self-report measures with biomarkers
disability or death, a comprehensive geriatric assessment is always
be used to facilitate the diagnosis (e.g., different combinations of
required for intervention at this stage (Fig. 1). In other words, a
amyloid-␤ accumulation, neurodegeneration and subtle cognitive
multimodal intervention with the main goal of rehabilitation and
decline) and to further determine whether the SCD is a component
sustaining the quality of life is a suitable choice for these patients.
of pre-clinical SCD; this information can then be used for guiding
Many studies have shown that exercise can improve the mobil-
clinical intervention. However, this revised clinical screening crite-
ity and functional ability of frail elderly individuals (de Vries et al.,
rion of cognitive frailty still requires a large cohort of elderly to test
2012; Theou et al., 2011; Clegg et al., 2012). The positive influence
the predictive effectiveness of a poor prognosis. Moreover, more
of exercise on frailty is partially realized by reducing the systemic,
research is also required to determine the physical causes of SCD
chronic inflammation in the body (Robertson et al., 2013). Physical
from a physiological perspective.
activity can counteract sarcopenia, a major cause of physical frailty,
and cognitive decline (Landi et al., 2010). For example, aerobic
exercise and strength training for 12 weeks resulted in improved Conflict of interest statement
scores in functional capacity, cognition and quality of life (Langlois
et al., 2013). Protein nutritional supplements may act synergisti- None declared.
cally with resistance exercise in elderly persons to increase muscle
mass, reduce complications, improve grip strength and produce Acknowledgements
weight gain (Tieland et al., 2012a,b; Malafarina et al., 2013; Cawood
et al., 2012). However, one randomized controlled trial that inves- This work was supported by grants from the Shanghai Key Lab-
tigated the effects of exercise and nutritional supplementation in oratory of Clinical Geriatric Medicine Subject Construction (No.
very elderly frail individuals in a long-term care situation indi- 13dz2260700) and Shanghai Hospital Development Center Grant
cated that these treatments had no effect on muscle strength, gait (No. SHDC12014221).
speed, stair climbing or physical activity (Fiatarone et al., 1994).
Mouse experiments revealed that genetic and/or dietary interven-
tions that promote longevity are protective regarding aging-related References
physical frailty (Arum et al., 2014). In addition, several drugs have
Abellan van Kan, G., Rolland, Y., Bergman, H., Morley, J.E., Kritchevsky, S.B., Vellas,
been used in intervention studies of frailty, a 3-year follow-up B., 2008. The I.A.N.A Task Force on frailty assessment of older people in clinical
study in which an angiotensin-converting enzyme (ACE) inhibitor practice. J. Nutr. Health Aging 12 (1), 29–37.
Aisen, P.S., 2008. Treatment for MCI: is the evidence sufficient? Neurology 70 (22),
was used to treat the non-frail at a baseline level in women aged
2020–2021.
65–79 years old (N = 27,378); the results revealed that the inci- Akki, A., Yang, H., Gupta, A., Chacko, V.P., Yano, T., Leppo, M.K., Steenbergen, C.,
dence of frailty was similar in ACE inhibitor users and nonusers Walston, J., Weiss, R.G., 2014. Skeletal muscle ATP kinetics are impaired in frail
(Gray et al., 2009). It was also reported that testosterone improved mice. Age (Dordr.) 36 (1), 21–30.
Annweiler, C., Fantino, B., Le Gall, D., Schott, A.M., Berrut, G., Beauchet, O., 2011.
muscle strength but caused side effects in the cardiovascular and Severe vitamin D deficiency is associated with advanced-stage dementia in
respiratory systems (Basaria et al., 2010). In contrast, IGF 1 did not geriatric inpatients. J. Am. Geriatr. Soc. 59, 169–171.
8 Q. Ruan et al. / Ageing Research Reviews 20 (2015) 1–10

Arum, O., Rickman, D.J., Kopchick, J.J., Bartke, A., 2014. The slow-aging growth hor- community-dwelling elderly persons carried out by trained lay buddies: study
mone receptor/binding protein gene-disrupted (GHR-KO) mouse is protected protocol of a randomized controlled trial. BMC Public Health 27 (13), 1232.
from aging-resultant neuromusculoskeletal frailty. Age (Dordr.) 36 (1), 117–127. Eeles, E.M., White, S.V., O’Mahony, S.M., Bayer, A.J., Hubbard, R.E., 2012. The impact
Auyeung, T.W., Lee, J.S., Kwok, T., Woo, J., 2011. Physical frailty predicts future cog- of frailty and delirium on mortality in older inpatients. Age Ageing 41, 412–416.
nitive decline – a four-year prospective study in 2737 cognitively normal older Elabd, C., Cousin, W., Upadhyayula, P., Upadhyayula, P., Chen, R.Y., Chooljian, M.S.,
adults. J. Nutr. Health Aging 15 (8), 690–694. Li, J., Kung, S., Jiang, K.P., Conboy, I.M., 2014. Oxytocin is an age-specific circulat-
Avila-Funes, J.A., Amieva, H., Barberger-Gateau, P., Le Goff, M., Raoux, N., Ritchie, ing hormone that is necessary for muscle maintenance and regeneration. Nat.
K., Carrière, I., Tavernier, B., Tzourio, C., Gutiérrez-Robledo, L.M., Dartigues, J.F., Commun. 2014 (5), 4082.
2009. Cognitive impairment improves the predictive validity of the phenotype Elwood, P.C., Pickering, J., Bayer, A., Gallacher, J.E., 2002. Vascular disease and cog-
of frailty for adverse health outcomes: the three-city study. J. Am. Geriatr. Soc. nitive function in older men in the Caerphilly cohort. Age Ageing 31 (1), 43–48.
57 (3), 453–461. Ensrud, K.E., Ewing, S.K., Fredman, L., Hochberg, M.C., Cauley, J.A., Hillier, T.A., Cum-
Barberger-Gateau, P., Commenges, D., Gagnon, M., Letenneur, L., Sauvel, C., Dar- mings, S.R., Yaffe, K., Cawthon, P.M., Study of Osteoporotic Fractures Research
tigues, J.F., 1992. Instrumental activities of daily living as a screening tool for Group, 2010. Circulating 25-hydroxyvitamin D levels and frailty status in older
cognitive impairment and dementia in elderly community dwellers. J. Am. Geri- women. J. Clin. Endocrinol. Metab. 95 (12), 5266–5273.
atr. Soc. 40 (11), 1129–1134. Ensrud, K.E., Blackwell, T.L., Cauley, J.A., Cummings, S.R., Barrett-Connor, E., Dam,
Basaria, S., Coviello, A.D., Travison, T.G., Storer, T.W., Farwell, W.R., Jette, A.M., Eder, T.T., Hoffman, A.R., Shikany, J.M., Lane, N.E., Stefanick, M.L., Orwoll, E.S.,
R., Tennstedt, S., Ulloor, J., Zhang, A., Choong, K., Lakshman, K.M., Mazer, N.A., Cawthon, P.M., Osteoporotic Fractures in Men Study Group, 2011. Circulating
Miciek, R., Krasnoff, J., Elmi, A., Knapp, P.E., Brooks, B., Appleman, E., Aggarwal, 25-hydroxyvitamin D levels and frailty in older men: the osteoporotic fractures
S., Bhasin, G., Hede-Brierley, L., Bhatia, A., Collins, L., LeBrasseur, N., Fiore, L.D., in men study. J. Am. Geriatr. Soc. 59 (1), 101–106.
Bhasin, S., 2010. Adverse events associated with testosterone administration. N. Ferrucci, L., Cavazzini, C., Corsi, A., Bartali, B., Russo, C.R., Lauretani, F., Ferrucci,
Engl. J. Med. 363, 109–122. L., Cavazzini, C., Corsi, A.M., Bartali, B., Russo, C.R., Lauretani, F., Bandinelli, S.,
Bouillon, K., Kivimaki, M., Hamer, M., Sabia, S., Fransson, E.I., Singh-Manoux, A., Bandinelli, S., Guralnik, J.M., 2002. Biomarkers of frailty in older persons. J.
Gale, C.R., Batty, G.D., 2013. Measures of frailty in population-based studies: Endocrinol. Invest. 25 (10 Suppl.), 10–15.
an overview. BMC Geriatr. 21 (13), 64. Ferrucci, L., Giallauria, F., Schlessinger, D., 2008. Mapping the road to resilience:
Boyle, P.A., Buchman, A.S., Wilson, R.S., Leurgans, S.E., Bennett, D.A., 2010. Physical novel math for the study of frailty. Mech. Ageing Dev. 129 (11), 677–679.
frailty is associated with incident mild cognitive impairment in community- Fiatarone, M.A., O’Neill, E.F., Ryan, N.D., Clements, K.M., Solares, G.R., Nelson, M.E.,
based older persons. J. Am. Geriatr. Soc. 58, 248–255. Roberts, S.B., Kehayias, J.J., Lipsitz, L.A., Evans, W.J., 1994. Exercise training and
Buchman, A.S., Boyle, P.A., Wilson, R.S., Tang, Y., Bennett, D.A., 2007. Frailty is asso- nutritional supplementation for physical frailty in very elderly people. N. Engl.
ciated with incident Alzheimer’s disease and cognitive decline in the elderly. J. Med. 330, 1769–1775.
Psychosom. Med. 69, 483–489. Fried, L.P., Tangen, C.M., Walston, J., Newman, A.B., Hirsch, C., Gottdiener, J., Seeman,
Buchman, A.S., Schneider, J.A., Leurgans, S., Bennett, D.A., 2008. Physical frailty in T., Tracy, R., Kop, W.J., 2001. Frailty in older adults: evidence for a phenotype. J.
older persons is associated with Alzheimer disease pathology. Neurology 71 (7), Gerontol. A: Biol. Sci. Med. Sci. 56 (3), M146–M156.
499–504. Fried, L.P., Xue, Q.L., Cappola, A.R., Ferrucci, L., Chaves, P., Varadhan, R., Guralnik,
Buchman, A.S., Bennett, D.A., 2013. Cognitive frailty. J. Nutr. Health Aging 17 (9), J.M., Leng, S.X., Semba, R.D., Walston, J.D., Blaum, C.S., Bandeen-Roche, K., 2009.
738–739. Nonlinear multisystem physiological dysregulation associated with frailty in
Buchman, A.S., Yu, L., Wilson, R.S., Boyle, P.A., Schneider, J.A., Bennett, D.A., 2014. older women: implications for etiology and treatment. J. Gerontol. A: Biol. Sci.
Brain pathology contributes to simultaneous change in physical frailty and cog- Med. Sci. 64 (10), 1049–1057.
nition in old age. J. Gerontol. A: Biol. Sci. Med. Sci., pii: glu117. Friedlander, A.L., Butterfield, G.E., Moynihan, S., Grillo, J., Pollack, M., Holloway, L.,
Canevelli, M., Adali, N., Tainturier, C., 2013. Cognitive interventions targeting subjec- Friedman, L., Yesavage, J., Matthias, D., Lee, S., Marcus, R., Hoffman, A.R., 2001.
tive cognitive complaints. Am. J. Alzheimers Dis. Other Demen. 28 (6), 560–567. One year of insulin-like growth factor I treatment does not affect bone density,
Canevelli, M., Troili, F., Bruno, G., 2014. Reasoning about frailty in neurology: neu- body composition, or psychological measures in ostmenopausal women. J. Clin.
robiological correlates and clinical perspectives. J. Frailty Aging 3, 18–20. Endocrinol. Metab. 86, 1496–1503.
Cano, C., Samper-Ternent, R., Al Snih, S., Markides, K., Ottenbacher, K.J., 2012. Frailty Golomb, J., Kluger, A., Ferris, S.H., 2004. Mild cognitive impairment: historical devel-
and cognitive impairment as predictors of mortality in older Mexican Americans. opment and summary of research. Dialogues Clin. Neurosci. 6 (4), 351–367.
J. Nutr. Health Aging 16, 142–147. Gomez-Isla, T., Price, J.L., McKeel Jr., D.W., Morris, J.C., Growdon, J.H., Hyman, B.T.,
Caselli, R.J., Reiman, E.M., 2013. Characterizing the preclinical stages of Alzheimer’s 1996. Profound loss of layer II entorhinal cortex neurons occurs in very mild
disease and the prospect of presymptomatic intervention. J. Alzheimers Dis. 33 Alzheimer’s disease. J. Neurosci. 16, 4491–4500.
(Suppl. 1), S405–S416. Gray, S.L., LaCroix, A.Z., Aragaki, A.K., McDermott, M., Cochrane, B.B., Kooperberg,
Cawood, A.L., Elia, M., Stratton, R.J., 2012. Systematic review and meta-analysis of C.L., Murray, A.M., Rodriguez, B., Black, H., Woods, N.F., Women’s Health Initia-
the effects of high protein oral nutritional supplements. Ageing Res. Rev. 11, tive Observational Study, 2009. Angiotensin-converting enzyme inhibitor use
e278–e296. and incident frailty in women aged 65 and older: prospective findings from
Chaves, P.H., Varadhan, R., Lipsitz, L.A., Stein, P.K., Windham, B.G., Tian, J., Fleisher, the Women’s Health Initiative Observational Study. J. Am. Geriatr. Soc. 57 (2),
L.A., Guralnik, J.M., Fried, L.P., 2008. Physiological complexity underlying heart 297–303.
rate dynamics and frailty status in community-dwelling older women. J. Am. Gray, S.L., Anderson, M.L., Hubbard, R.A., LaCroix, A., Crane, P.K., McCormick, W.,
Geriatr. Soc. 56 (9), 1698–1703. Bowen, J.D., McCurry, S.M., Larson, E.B., 2013. Frailty and incident dementia. J.
Clegg, A., Barber, S., Young, J., Forster, A., Iliffe, S., 2012. Do home-based exercise Gerontol. A: Biol. Sci. Med. Sci. 68 (9), 1083–1090.
interventions improve outcomes for frail older people? Findings from a system- Han, E.S., Lee, Y., Kim, J., 2014. Association of cognitive impairment with frailty in
atic review. Rev. Clin. Gerontol. 22, 68–78. community-dwelling older adults. Int. Psychogeriatr. 26 (1), 155–163.
Clegg, A., Barber, S., Young, J., Iliffe, S., Forster, A., 2014. The home-based older peo- Hayden, K.M., Norton, M.C., Darcey, D., Ostbye, T., Zandi, P.P., Breitner, J.C., Welsh-
ple’s exercise (HOPE) trial: a pilot randomised controlled trial of a home-based Bohmer, K.A., Cache, County Study Investigators, 2010. Occupational exposure to
exercise intervention for older people with frailty. Age Ageing 43 (5), 687– pesticides increases the risk of incident AD: the Cache County study. Neurology
695. 74, 1524–1530.
Clegg, A., Young, J., Iliffe, S., Rikkert, M.O., Rockwood, K., 2013. Frailty in elderly Ho, Y.Y., Matteini, A.M., Beamer, B., Fried, L., Xue, Q.L., Arking, D.E., Chakravarti, A.,
people. Lancet 381 (9868), 752–762. Fallin, M.D., Walston, J., 2011. Exploring biologically relevant pathways in frailty.
Dartigues, J.F., Amieva, H., 2014. Cognitive frailty: rational and definition from an J. Gerontol. A: Biol. Sci. Med. Sci. 66 (9), 975–979.
(I.A.N.A./I.A.G.G.) international consensus group. J. Nutr. Health Aging 18 (1), 95. Houles, M., Canevelli, M., Abellan van kan, G., Ousset, P.J., Cesari, M., Vellas, B., 2012.
de Vries, N.M., Staal, J.B., van Ravensberg, C.D., Hobbelen, J.S., Olde Rikkert, M.G., Frailty and cognition. J. Frailty Aging 1 (2), 56–61.
Nijhuis-van der Sanden, M.W., 2011. Outcome instruments to measure frailty: Hughes, C.P., Berg, L., Danziger, W.L., Coben, L.A., Martin, R.L., 1982. A new clinical
a systematic review. Ageing Res. Rev. 10 (1), 104–114. scale for the staging of dementia. Br. J. Psychiatry 140, 566–572.
de Vries, N.M., van Ravensberg, C.D., Hobbelen, J.S., Olde Rikkert, M.G., Staal, Jessen, F., Amariglio, R.E., van Boxtel, M., Breteler, M., Ceccaldi, M., Chételat, G.,
J.B., Nijhuis-van der Sanden, M.W., 2012. Effects of physical exercise ther- Dubois, B., Dufouil, C., Ellis, K.A., van der Flier, W.M., Glodzik, L., van Harten,
apy on mobility, physical functioning, physical activity and quality of life in A.C., de Leon, M.J., McHugh, P., Mielke, M.M., Molinuevo, J.L., Mosconi, L., Osorio,
community-dwelling older adults with impaired mobility, physical disability R.S., Perrotin, A., Petersen, R.C., Rabin, L.A., Rami, L., Reisberg, B., Rentz, D.M.,
and/or multi-morbidity: a meta-analysis. Ageing Res. Rev. 11, 136–149. Sachdev, P.S., de la Sayette, V., Saykin, A.J., Scheltens, P., Shulman, M.B., Slavin,
Desai, A.K., Grossberg, G.T., Chibnall, J.T., 2010. Healthy brain aging: a road map. Clin. M.J., Sperling, R.A., Stewart, R., Uspenskaya, O., Vellas, B., Visser, P.J., Wagner,
Geriatr. Med. 26 (1), 1–16. M., Subjective Cognitive Decline Initiative (SCD-I) Working Group, 2014. A con-
Devlin, K.N., Gongvatana, A., Clark, U.S., Chasman, J.D., Westbrook, M.L., Tashima, ceptual framework for research on subjective cognitive decline in preclinical
K.T., Navia, B., Cohen, R.A., 2012. Neurocognitive effects of HIV, hepatitis C, and Alzheimer’s disease. Alzheimer’s Demen., pii: S1552-5260(14)00002-8.
substance use history. J. Int. Neuropsychol. Soc. 18 (1), 68–78. Kalyani, R.R., Varadhan, R., Weiss, C.O., Fried, L.P., Cappola, A.R., 2012. Frailty status
Doody, R.S., Thomas, R.G., Farlow, M., Iwatsubo, T., Vellas, B., Joffe, S., Kieburtz, K., and altered glucose–insulin dynamics. J. Gerontol. A: Biol. Sci. Med. Sci. 67 (12),
Raman, R., Sun, X., Aisen, P.S., Siemers, E., Liu-Seifert, H., Mohs, R., Alzheimer’s 1300–1306.
Disease Cooperative Study Steering Committee; Solanezumab Study Group, Kelaiditi, E., Cesari, M., Canevelli, M., van Kan, G.A., Ousset, P.J., Gillette-Guyonnet, S.,
2014. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. Ritz, P., Duveau, F., Soto, M.E., Provencher, V., Nourhashemi, F., Salvà, A., Robert,
N. Engl. J. Med. 370 (4), 311–321. P., Andrieu, S., Rolland, Y., Touchon, J., Fitten, J.L., Vellas, B., IANA/IAGG, 2014.
Dorner, T.E., Lackinger, C., Haider, S., Luger, E., Kapan, A., Luger, M., Schindler, Cognitive frailty: rational and definition from an (I.A.N.A./I.A.G.G.) international
K.E., 2013. Nutritional intervention and physical training in malnourished frail consensus group. J. Nutr. Health Aging 17, 726–734.
Q. Ruan et al. / Ageing Research Reviews 20 (2015) 1–10 9

Kulmala, J., Nykänen, I., Mänty, M., Hartikainen, S., 2014. Association between frailty Puts, M.T., Visser, M., Twisk, J.W., Deeg, D.J., Lips, P., 2005b. Endocrine and inflam-
and dementia: a population-based study. Gerontology 60 (1), 16–21. matory markers as predictors of frailty. Clin. Endocrinol. (Oxf.) 63 (4), 403–411.
Knopman, D.S., Roberts, R., 2010. Vascular risk factors: imaging and neuropathologic Rejnmark, L., Avenell, A., Masud, T., Anderson, F., Meyer, H.E., Sanders, K.M., Salo-
correlates. J. Alzheimers Dis. 20, 699–709. vaara, K., Cooper, C., Smith, H.E., Jacobs, E.T., Torgerson, D., Jackson, R.D., Manson,
Ko, F., Yu, Q., Xue, Q.L., Yao, W., Brayton, C., Yang, H., Fedarko, N., Walston, J., 2012. J.E., Brixen, K., Mosekilde, L., Robbins, J.A., Francis, R.M., Abrahamsen, B., 2012.
Inflammation and mortality in a frail mouse model. Age (Dordr.) 34 (3), 705–715. Vitamin D with calcium reduces mortality: patient level pooled analysis of
Landi, F., Abbatecola, A.M., Provinciali, M., Corsonello, A., Bustacchini, S., Manigrasso, 70,528 patients from eight major vitamin D trials. J. Clin. Endocrinol. Metab.
L., Cherubini, A., Bernabei, R., Lattanzio, F., 2010. Moving against frailty: does 97 (8), 2670–2681.
physical activity matter? Biogerontology 11, 537–545. Robertson, D.A., Savva, G.M., Kenny, R.A., 2013. Frailty and cognitive impairment – a
Lang, P.O., Michel, J.P., Zekry, D., 2009. Frailty syndrome: a transitional state in a review of the evidence and causal mechanisms. Ageing Res. Rev. 12 (4), 840–851.
dynamic process. Gerontology 55 (5), 539–549. Rockwood, K., 2005. Frailty and its definition: a worthy challenge. J. Am. Geriatr. Soc.
Langlois, F., Vu, T.T., Chasse, K., Dupuis, G., Kergoat, M.J., Bherer, L., 2013. Benefits of 53 (6), 1069–1070.
physical exercise training on cognition and quality of life in frail older adults. J. Rockwood, K., Mitnitski, A., 2007. Frailty in relation to the accumulation of deficits.
Gerontol. B: Psychol. Sci. Soc. Sci. 68, 400–404. J. Gerontol. A: Biol. Sci. Med. Sci. 62 (7), 722–727.
Leng, S.X., Yang, H., Walston, J.D., 2004a. Decreased cell proliferation and altered Sagare, A.P., Bell, R.D., Zlokovic, B.V., 2012. Neurovascular dysfunction and faulty
cytokine production in frail older adults. Aging Clin. Exp. Res. 16 (3), 249–252. amyloid ␤-peptide clearance in Alzheimer disease. Cold Spring Harb. Perspect.
Leng, S.X., Cappola, A.R., Andersen, R.E., Blackman, M.R., Koenig, K., Blair, M., Med. 2 (10), pii: a011452.
Walston, J.D., 2004b. Serum levels of insulin-like growth factor-I (IGF-I) and Sahin, E., DePinho, R.A., 2013. Axis of aging: telomeres, p53 and mitochondria. Nat.
dehydroepiandrosterone sulfate (DHEA-S), and their relationships with serum Rev. Mol. Cell Biol. 13, 397–404.
interleukin-6, in the geriatric syndrome of frailty. Aging Clin. Exp. Res. 16 (2), Shimada, H., Makizako, H., Doi, T., Yoshida, D., Tsutsumimoto, K., Anan, Y., Uemura,
153–157. K., Ito, T., Lee, S., Park, H., Suzuki, T., 2013. Combined prevalence of frailty and
López-Otín, C., Blasco, M.A., Partridge, L., Serrano, M., Kroemer, G., 2013. The hall- mild cognitive impairment in a population of elderly Japanese people. J. Am.
marks of aging. Cell 153 (6), 1194–1217. Med. Dir. Assoc. 14 (7), 518–524.
Malafarina, V., Uriz-Otano, F., Iniesta, R., Gil-Guerrero, L., 2013. Effectiveness of nutri- Sinha, M., Jang, Y.C., Oh, J., Khong, D., Wu, E.Y., Manohar, R., Miller, C., Regalado,
tional supplementation on muscle mass in treatment of sarcopenia in old age: S.G., Loffredo, F.S., Pancoast, J.R., Hirshman, M.F., Lebowitz, J., Shadrach, J.L.,
a systematic review. J. Am. Med. Dir. Assoc. 14, 10–17. Cerletti, M., Kim, M.J., Serwold, T., Goodyear, L.J., Rosner, B., Lee, R.T., Wagers,
Malmstrom, T.K., Morley, J.E., 2013. Frailty and cognition: linking two common A.J., 2014. Restoring systemic GDF11 levels reverses age-related dysfunction in
syndromes in older persons. J. Nutr. Health Aging 17 (9), 723–725. mouse skeletal muscle. Science 344 (6184), 649–652.
Matthews, F.E., Stephan, B.C., McKeith, I.G., Bond, J., Brayne, C., Medical Research Song, X., Mitnitski, A., Rockwood, K., 2011. Nontraditional risk factors combine to
Council Cognitive Function and Ageing Study, 2008. Two-year progression from predict Alzheimer disease and dementia. Neurology 77, 227–234.
mild cognitive impairment to dementia: to what extent do different definitions Sperling, R.A., Jack Jr., C.R., Aisen, P.S., 2011. Testing the right target and right drug
agree? J. Am. Geriatr. Soc. 56 (8), 1424–1433. at the right stage. Sci. Transl. Med. 3 (111), 111cm33.
Millán-Calenti, J.C., Tubío, J., Pita-Fernández, S., Rochette, S., Lorenzo, T., Maseda, A., Sperling, R.A., Karlawish, J., Johnson, K.A., 2013. Preclinical Alzheimer disease – the
2012. Cognitive impairment as predictor of functional dependence in an elderly challenges ahead. Nat. Rev. Neurol. 9 (1), 54–58.
sample. Arch. Gerontol. Geriatr. 54 (1), 197–201. Sternberg, S.A., Wershof Schwartz, A., Karunananthan, S., Karunananthan, S.,
Mhaoláin, A.M., Gallagher, D., Crosby, L., Ryan, D., Lacey, L., Coen, R.F., Coakley, D., Bergman, H., Mark Clarfield, A., 2011. The identification of frailty: a systematic
Walsh, J.B., Cunningham, C., Lawlor, B., 2012. Frailty and quality of life for people literature review. J. Am. Geriatr. Soc. 59 (11), 2129–2138.
with Alzheimer’s dementia and mild cognitive impairment. Am. J. Alzheimers Tavassoli, N., Guyonnet, S., Abellan Van Kan, G., Sourdet, S., Krams, T., Soto, M.E.,
Dis. Other Demen. 27 (1), 48–54. Subra, J., Chicoulaa, B., Ghisolfi, A., Balardy, L., Cestac, P., Rolland, Y., Andrieu, S.,
Mitchell, A.J., Shiri-Feshki, M., 2009. Rate of progression of mild cognitive impair- Nourhashemi, F., Oustric, S., Cesari, M., Vellas, B., Geriatric Frailty Clinic (G.F.C)
ment to dementia – meta-analysis of 41 robust inception cohort studies. Acta for Assessment of Frailty and Prevention of Disability Team, 2014. Description
Psychiatr. Scand. 119 (4), 252–265. of 1,108 older patients referred by their physician to the Geriatric Frailty Clinic
Moeley, J.E., Vellas, B., van kan, G.A., Anker, S.D., Bauer, J.M., Bernabei, R., Cesari, M., (G.F.C) for Assessment of Frailty and Prevention of Disability at the gerontopole.
Chumlea, W.C., Doehner, W., Evans, J., Fried, L.P., Guralnik, J.M., Katz, P.R., Malm- J. Nutr. Health Aging 18 (5), 457–464.
strom, T.K., McCarter, R.J., Gutierrez Robledo, L.M., Rockwood, K., von Haehling, Tieland, M., Dirks, M.L., van der Zwaluw, N., Verdijk, L.B., van de Rest, O., de Groot,
S., Vandewoude, M.F., Walston, J., 2013. Frailty consensus: a call to action. J. Am. L.C., van Loon, L.J., 2012a. Protein supplementation increases muscle mass gain
Med. Dir. Assoc. 14, 392–397. during prolonged resistance-type exercise training in frail elderly people: a
Morris, J.C., 1993. The clinical dementia rating (CDR): current version and scoring randomized, double-blind, placebo-controlled trial. J. Am. Med. Dir. Assoc. 13,
rules. Neurology 43 (11), 2412–2414. 713–719.
Morris, J.C., 2012. Revised criteria for mild cognitive impairment may compromise Tieland, M., van de Rest, O., Dirks, M.L., van der Zwaluw, N., Mensink, M., van Loon, L.J.,
the diagnosis of Alzheimer disease dementia. Arch. Neurol. 69 (6), 700–708. de Groot, L.C., 2012b. Protein supplementation improves physical performance
Moskalev, A.A., Aliper, A.M., Smit-McBride, Z., Buzdin, A., Zhavoronkov, A., 2014. in frail elderly people: a randomized, double-blind, placebo-controlled trial. J.
Genetics and epigenetics of aging and longevity. Cell Cycle 13 (7), 1063–1077. Am. Med. Dir. Assoc. 13 (720), e726.
Ng, T.P., Feng, L., Nyunt, M.S., 2014. Frailty in older persons: multisystem risk factors Theou, O., Stathokostas, L., Roland, K.P., Jakobi, J.M., Patterson, C., Vandervoort, A.A.,
and the frailty risk index (FRI). J. Am. Med. Dir. Assoc. 15 (9), 635–642. Jones, G.R., 2011. The effectiveness of exercise interventions for the management
Njegovan, V., Hing, M.M., Mitchell, S.L., Molnar, F.J., 2001. The hierarchy of functional of frailty: a systematic review. J. Aging Res. 2011, 569194.
loss associated with cognitive decline in older persons. J. Gerontol. A: Biol. Sci. Varadhan, R., Seplaki, C.L., Xue, Q.L., Bandeen-Roche, K., Fried, L.P., 2008.
Med. Sci. 56 (10), M638–M643. Stimulus–response paradigm for characterizing the loss of resilience in homeo-
Okonkwo, O.C., Cohen, R.A., Gunstad, J., Tremont, G., Alosco, M.L., Poppas, A., 2010. static regulation associated with frailty. Mech. Ageing Dev. 129 (11), 666–670.
Longitudinal trajectories of cognitive decline among older adults with cardio- Vellas, B., Coley, N., Ousset, P.J., Berrut, G., Dartigues, J.F., Dubois, B., Grandjean, H.,
vascular disease. Cerebrovasc. Dis. 30 (4), 362–373. Pasquier, F., Piette, F., Robert, P., Touchon, J., Garnier, P., Mathiex-Fortunet, H.,
Oosterveld, S.M., Kessels, R.P., Hamel, R., Ramakers, I.H., Aalten, P., Verhey, F.R., Andrieu, S., GuidAge Study Group, 2012. Long-term use of standardised Ginkgo
Sistermans, N., Smits, L.L., Pijnenburg, Y.A., van der Flier, W.M., Olde Rikkert, biloba extract for the prevention of Alzheimer’s disease (GuidAge): a randomised
M.G., Melis, R.J., 2014. The influence of co-morbidity and frailty on the clinical placebo-controlled trial. Lancet Neurol. 11 (10), 851–859.
manifestation of patients with Alzheimer’s disease. J. Alzheimers Dis. 42 (2), Vellas, B., Carrillo, M.C., Sampaio, C., Brashear, H.R., Siemers, E., Hampel, H., Schnei-
501–509. der, L.S., Weiner, M., Doody, R., Khachaturian, Z., Cedarbaum, J., Grundman, M.,
Panza, F., D’Introno, A., Colacicco, A.M., Capurso, C., Parigi, A.D., Capurso, S.A., Caselli, Broich, K., Giacobini, E., Dubois, B., Sperling, R., Wilcock, G.K., Fox, N., Scheltens,
R.J., Pilotto, A., Scafato, E., Capurso, A., Solfrizzi, V., 2006. Cognitive frailty: pre- P., Touchon, J., Hendrix, S., Andrieu, S., Aisen, P., EU/US/CTAD Task Force Mem-
dementia syndrome and vascular risk factors. Neurobiol. Aging 27 (7), 933– bers, 2013. Designing drug trials for Alzheimer’s disease: what we have learned
940. from the release of the phase III antibody trials: a report from the EU/US/CTAD
Panza, F., Solfrizzi, V., Frisardi, V., Maggi, S., Sancarlo, D., Adante, F., D’Onofrio, Task Force. Alzheimers Dement. 9 (4), 438–444.
G., Seripa, D., Pilotto, A., 2011. Different models of frailty in predementia and Villeda, S.A., Plambeck, K.E., Middeldorp, J., Castellano, J.M., Mosher, K.I., Luo, J.,
dementia syndromes. J. Nutr. Health Aging 15 (8), 711–719. Smith, L.K., Bieri, G., Lin, K., Berdnik, D., Wabl, R., Udeochu, J., Wheatley, E.G.,
Pilotto, A., Rengo, F., Marchionni, N., Sancarlo, D., Fontana, A., Panza, F., Ferrucci, L., Zou, B., Simmons, D.A., Xie, X.S., Longo, F.M., Wyss-Coray, T., 2014. Young blood
Study Group FIRI-SIGG, 2012. Comparing the prognostic accuracy for all-cause reverses age-related impairments in cognitive function and synaptic plasticity
mortality of frailty instruments: a multicentre 1-year follow-up in hospitalized in mice. Nat. Med. 20 (6), 659–663.
older patients. PLoS ONE 7 (1), e29090. Walston, J., McBurnie, M.A., Newman, A., Tracy, R.P., Kop, W.J., Hirsch, C.H., Gottdi-
Portet, F., Ousset, P.J., Visser, P.J., Frisoni, G.B., Nobili, F., Scheltens, P., Vellas, B., ener, J., Fried, L.P., Cardiovascular Health Study, 2002. Frailty and activation of
Touchon, J., MCI Working Group of the European Consortium on Alzheimer’s the inflammation and coagulation systems with and without clinical comorbidi-
Disease (EADC), 2006. Mild cognitive impairment (MCI) in medical practice: a ties: results from the Cardiovascular Health Study. Arch. Intern. Med. 162 (20),
critical review of the concept and new diagnostic procedure. Report of the MCI 2333–2341.
Working Group of the European Consortium on Alzheimer’s Disease. J. Neurol. Walston, J., Hadley, E.C., Ferrucci, L., Guralnik, J.M., Newman, A.B., Studenski, S.A.,
Neurosurg. Psychiatry 77 (6), 714–718. Ershler, W.B., Harris, T., Fried, L.P., 2006. Research agenda for frailty in older
Puts, M.T., Lips, P., Deeg, D.J., 2005a. Static and dynamic measures of frailty pre- adults: toward a better understanding of physiology and etiology: summary
dicted decline in performance-based and self-reported physical functioning. J. from the American Geriatrics Society/National Institute on Aging Research Con-
Clin. Epidemiol. 58 (11), 1188–1198. ference on Frailty in Older Adults. J. Am. Geriatr. Soc. 54, 991–1001.
10 Q. Ruan et al. / Ageing Research Reviews 20 (2015) 1–10

Walston, J., Fedarko, N., Yang, H., Leng, S., Beamer, B., Espinoza, S., Lipton, Wilkins, C.H., Roe, C.M., Morris, J.C., Galvin, J.E., 2013. Mild physical impairment
A., Zheng, H., Becker, K., 2008. The physical and biological characterization predicts future diagnosis of dementia of the Alzheimer’s type. J. Am. Geriatr.
of a frail mouse model. J. Gerontol. A: Biol. Sci. Med. Sci. 63 (4), 391– Soc. 61 (7), 1055–1059.
398. Woods, A.J., Cohen, R.A., Pahor, M., 2013. Cognitive frailty: frontiers and challenges.
Wang, G.C., Kao, W.H., Murakami, P., Xue, Q.L., Chiou, R.B., Detrick, B., J. Nutr. Health Aging 17 (9), 741–743.
McDyer, J.F., Semba, R.D., Casolaro, V., Walston, J.D., Fried, L.P., 2010. Xue, Q.L., Bandeen-Roche, K., Varadhan, R., Zhou, J., Fried, L.P., 2008. Initial man-
Cytomegalovirus infection and the risk of mortality and frailty in older women: ifestations of frailty criteria and the development of frailty phenotype in the
a prospective observational cohort study. Am. J. Epidemiol. 171 (10), 1144– Women’s Health and Aging Study II. J. Gerontol. A: Biol. Sci. Med. Sci. 63 (9),
1152. 984–990.
Weiner, D.E., Seliger, S.L., 2014. Cognitive and physical function in chronic kidney Zlokovic, B.V., 2011. Neurovascular pathways to neurodegeneration in Alzheimer’s
disease. Curr. Opin. Nephrol. Hypertens. 23 (3), 291–297. disease and other disorders. Nat. Rev. Neurosci. 12 (12), 723–738.

You might also like