Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jun 18.
Published in final edited form as: J Am Med Dir Assoc. 2011 Mar 4;12(4):249–256. doi: 10.1016/j.jamda.2011.01.003

Sarcopenia: An Undiagnosed Condition in Older Adults. Current Consensus Definition: Prevalence, Etiology, and Consequences

International Working Group on Sarcopenia1
PMCID: PMC3377163  NIHMSID: NIHMS379523  PMID: 21527165

Abstract

Sarcopenia, the age associated loss of skeletal muscle mass and function, has considerable societal consequences for the development of frailty, disability and health care planning. A group of geriatricians and scientists from academia and industry met in Rome, Italy on November 18, 2009 to arrive at a consensus definition of sarcopenia. The current consensus definition was approved unanimously by the meeting participants and is as follows: Sarcopenia is defined as the age-associated loss of skeletal muscle mass and function. The causes of sarcopenia are multi-factorial and can include disuse, altered endocrine function, chronic diseases, inflammation, insulin resistance, and nutritional deficiencies. While cachexia may be a component of sarcopenia, the two conditions are not the same. The diagnosis of sarcopenia should be considered in all older patients who present with observed declines in physical function, strength, or overall health. Sarcopenia should specifically be considered in patients who are bedridden, cannot independently rise from a chair, or who have a measured gait speed less that 1.0 m·s−1. Patients who meet these criteria should further undergo body composition assessment using dual energy x-ray absorptiometry (DXA) with sarcopenia being defined using currently validated definitions. A diagnosis of sarcopenia is consistent with a gait speed of less than 1 m·s−1 and an objectively measured low muscle mass (eg: appendicular mass relative to ht2 that is ≤ 7.23 kg/ m2 in men ≤ 5.67 kg/ m2 in men). Sarcopenia is a highly prevalent condition in older persons that leads to disability, hospitalization and death.

Keywords: muscle, aging, body composition, function, disability


“The sixth age shifts
Into the lean and slipper’d pantaloon
With spectacles on nose and pouch on side,
His youthful hose well sav’d, a world to wide
For his shrunk shank”

Shakespeare, As You Like It, Act II, Scene VII, lines 157–161

A reduction in lean body mass and an increase in fat mass is one of the most striking and consistent changes associated with advancing age. Skeletal muscle (1) and bone mass are the principal (if not exclusive) components of lean body mass to decline with age. These changes in body composition appear to occur throughout life and have important functional and metabolic consequences. The term sarcopenia (From the Greek: sarx for flesh, penia for loss) was first used by Irwin Rosenberg (2). It was originally described by Evans and Campbell (3) and further defined (4) as age related loss of muscle mass. This loss of muscle results in decreased strength, metabolic rate, aerobic capacity and thus, functional capacity. Subsequently, a number of authors have defined sarcopenia more specifically as a subgroup of older persons with muscle mass depletion, usually defined as being two standard deviations below the mean muscle mass of younger persons (usually age 35 years) (5). Since 1994 when 4 articles on sarcopenia were published, there has been an exponential increase in the number of publications reaching 140 in 2006 (6). This has been mirrored by an increase in citations to articles on sarcopenia going from 0 in 1996 to 2221 in 2006. Over this time sarcopenia has become recognized as an important geriatric condition and a key precursor to the development of frailty (7, 8). Much like osteopenia (bone density) predicts risk of a bone fracture, sarcopenia is a powerful predictor of late-life disability. The purpose of this article is to define sarcopenia, provide guidelines for assessment and briefly describe its prevalence, etiology, and consequences. Sarcopenia has “come of age” and should be recognized as a preventable and treatable condition among geriatric patients.

In 1931, Critchley noted that muscle loss occurs with aging and is most noticeable in intrinsic hand and foot muscles (9). Sarcopenia very likely begins in early adulthood (10) with atrophy and loss of type II muscle fibers (11, 12) and continues throughout life as a result of complex interaction of environmental and genetic causes. The direct effect of sarcopenia, on strength is illustrated by the dramatic age-associated decline in the world weight lifting records. These records decline by 30% in men and over 50% in women between the ages of 30 to 60 years. (13). Longitudinal studies have shown a clear decline in muscle mass, strength and power beginning at approximately 35 years of age (14). Strength and power decline to a greater extent than does muscle mass (15). In addition to sarcopenia, intramusculater lipid, termed myosteatosis (16), increases with age and increasing body fatness. Janssen et al. (17) estimated that sarcopenia results in an excess cost to the health care system of the United States of $18.4 billion a year (year 2001), due to associated disability.

Current Consensus Definition

A meeting was convened on November 18, 2009 in Rome, Italy with the express purpose of arriving at a consensus definition of sarcopenia. Because there has been no true consensus of the appropriate criteria for when an individual may be said to be sarcopenic, recognition of this treatable condition has been lacking. The following definition was the current consensus of the group of scientists and geriatricians that were present at the meeting. In addition, this definition was reviewed by a number of researchers in the area of skeletal muscle and aging. All participants are listed in the appendix:

“Sarcopenia is the age-associated loss of skeletal muscle mass and function. Sarcopenia is a complex syndrome that is associated with muscle mass loss alone or in conjunction with increased fat mass. The causes of sarcopenia are multi-factorial and can include disuse, changing endocrine function, chronic diseases, inflammation, insulin resistance, and nutritional deficiencies. While cachexia may be a component of sarcopenia, the two conditions are not the same.”

There was unanimous agreement that the presence of sarcopenia should be evaluated in older patients who have clinically observed declines in physical functioning, strength, or health status (Table 1). Clinicians should also consider sarcopenia in patients who present with difficulties in performing activitites of daily living, have a history of recurrent falls, have documented recent weight loss, have recently been hospitalized, or have chronic conditions associated with muscle loss (eg: Type II diabetes, chronic heart failure, chronic obstructive pulmonary disease, chronic kidney disease, rheumatoid arthritis, and malignancies).

Table 1.

Initial Patient Presentation to Evaluate for Sarcopenia

  • Noted decline in function, strength, “health” status

  • Self-reported mobility-related difficulty

  • History of recurrent falls

  • Recent unintentional weight loss (> 5%)

  • Post-hospitalization

  • Other chronic conditions (eg: Type II diabetes, CHF*, COPD**, CKD***, RA****, and Cancer)

*

CHF = chronic heart failure;

**

COPD = chronic obstructive pulmonary disease;

***

CKD = chronic kidney disease;

****

RA = rheumatoid arthritis

There was consensus on the panel that sarcopenia could be effectively targeted by assessing physical functioning in at risk patients (Table 2). Sarcopenia should be considered in patients who are bedridden, non-ambulatory, or who cannot rise from a chair unassisted. In addition, for patients who are ambulatory and can arise from a chair, gait speed should be assessed across a 4 meter course. Patients with a measured gait speed less than 1.0 m·s−1 should be referred for body composition assessment using whole body dual energy x-ray absorptiometry (DXA).

Table 2.

Targeting Sarcopenia

  • Assess patient for reduced physical functioning (or weakness).

  • Consider Sarcopenia in patients who are non-ambulatory or who cannot rise from a chair unassisted.

  • Assess habitual gait speed over a 4 meter course.

  • Patients with a habitual gait speed < 1.0 m/sec. should be considered for quantitative measurement of body composition by DXA.

The diagnosis of sarcopenia should be based on having a low whole body or appendicular fat free mass in combination with poor physical functioning. Current methods index appendicular fat free mass to height squared or whole body fat free mass to height squared. In patients with poor functional capacity, most easily identified using gait speed of than 1 m·s−1, sarcopenia can be diagnosed when the lean mass is less than 20%tile of values for healthy young adults. Currently objective cutpoints can be made for sarcopenia in men at an appendicular fat lean mass/ ht2 (aLM/Ht2) of ≤ 7.23 kg/ m2 and in women at ≤ 5.67 kg/ m2 (18).

Muscle Aging

Over the age span from 20 to 80 years of age, there is approximately a 30% reduction in muscle mass and a decline in cross-sectional area of about 20% (14). This is due to a decline in both muscle fiber size and number (10). There is no consensus on whether there is a selective loss of specific muscle fiber types. Early cross-sectional studies demonstrated a shift in muscle fiber composition with a higher type I/type II fiber ratio with advancing age (11). Larsson et al (12) suggested a preferential loss of type II fibers with advancing age, potentially starting in early adulthood. Type II fibers demonstrate selective atrophy (with a preservation of Type I fiber area) with age (19, 20). This is due to a reduction in high intensity activities that recruit these fibers, while type I fibers are used for most activities of daily living and during submaximal exercise (e.g. walking). An increase in hybrid type I and II fibers (21, 22) with advancing age has been described. Within the muscle, there is a decrease in non-contractile area along with a decrease in cross-bridging between the fibers. Single fiber intrinsic force is decreased. There is a decline in the number of T-tubule dihydropyridine receptors and an increase in uncoupled Ryanodine receptors. Twitch contraction time is slower and maximum shortening speed is lower.

Ultrasound studies have demonstrated the importance in tendon changes in altering muscle power with aging (21, 2325). With aging there is a decrease in tendon stiffness which, coupled with the shortening of muscle fascicles, results in smaller pennation angles (26, 27) and a decrease in specific force (i.e., fascicle force/physiological cross-sectional area). This may be one cause of decreasing strength with advancing age. In general, aging is associated with a greater decline in lower body than upper body and extensor compared to flexor strength (28). Overall, there is a much greater decline in strength than muscle mass with the decline in isometric knee extensor strength being between 55 to 76% (22, 29). These changes may be a cause of the decline in gait velocity that occurs with aging.

Etiology and biochemical basis for Sarcopenia

Sarcopenia is a universal phenomenon with a complex, multi-factorial etiology. Many of the potential causes vary by the age of the individual and are summarized in Table 3. The major factors considered to be involved include genetic heritability (3032), nutritional status (protein intake, energy intake, and vitamin D status) (3338), physical activity (3942), hormonal changes (declines in serum testosterone and growth hormone) (43, 44), insulin resistance (4547), atheroscelorosis (4850) and changes in circulating pro-inflammatory cytokines (51).

Table 3.

Sarcopenia Etiology by Age

Age Potential causes Effects
20–40 Decreased physical activity, decreased type II muscle fiber size and amount, maintenance of type I fibers (10) Maintenance of VO2max with exercise training, sprinting capacity is reduced
40–60 Loss of motor units accelerates (19). Decreased physical activity, increased body fatness (89), decreased androgens Decreased aerobic and sprinting capacity even with rigorous exercise, increased body fatness, insulin resistance 204, decreased muscle protein synthesis (90)
60–70 Decreased physical activity, reduced androgen and growth factor levels (44, 91), menopause, increased total body and visceral fat (92), chronic disease, impaired appetite regulation Inflammation (increased cytokine levels) (93), insulin resistance and type 2 diabetes (94), nutritional deficiencies (protein, vitamin D, and other micronutrients) (38), reduced muscle protein synthesis (90)
70+ Further reduction in physical activity, bouts of enforced inactivity due to illness, hospitalization depression, increased body fatness Fear of falling, low functional capacity (95), mild cognitive impairment, inflammation and increased muscle protein breakdown (96, 97)

At the molecular level, Sarcopenia results from a disproportionate decrease in skeletal muscle protein synthesis and/or an increase skeletal muscle protein breakdown. Anabolic hormones and muscular activity drive the system through activation of the phosphatidyl inositol3 kinase/serine-threonine kinase AKT system (52). This system stimulates muscle protein synthesis through the activation of the mammalian target of rapamycin (mTOR) and SGKI and inhibits atrophy by physophorylating the forkhead protein FOXO. Phosphorylated FOXO is inactivated thus reduces expression of the E3 ligase, Atrogin I and subsequently preventing protein degradation by the ubiquitin-proteasome system (53). A greater expression of MuRF-1 and atrogin-1 expression has been observed in aged rodent muscle compared to young along with a 90% higher level of ubiquitin conjugates. Increased availability of amino acids, particularly, branched chain amino acids stimulate mTOR (54). Elevated levels of angiotensin II inhibit phosphorylation of FOXO and stimulate capsase 3, which cleaves actomysin, allowing the actin and myosin to be degraded by the ubiquitin-proteasome system. This may explain the association of angiotensin converting enzyme inhibitors with increase muscle mass (55). Glucocorticoids inhibit AKT activity (55). Cytokines stimulate MURF 1 (muscle Ring finger), which, like atrogin, activates the ubiquitin-proteasome system (56). Myostatin D inhibits cell cycling through SMAD3 and MyoD, thus inhibiting the production of satellite cells (52) while testosterone increases satellite cell production by stimulating β-catenin. Cytokines cause DNA fragmentation and apoptosis by stimulating NFkB to produce Capsase 8. While little data comparing human skeletal muscle expression of factors affecting the expression of the ubiquitin-proteasome system, a recent study (57) examined baseline characteristics of young (23 ± 2y) and old (85 ± 1y) women as well as their response to a bout of resistance exercise. At baseline, the older women expressed FOXO3A and MuRF-1 genes at higher levels than did the young women. In response to a bout of resistance exercise all of the women demonstrated a substantial increase in the expression of MuRF-1, however the older women also showed a greater expression of atrogin-1, perhaps indicating a greater muscle proteolytic response to exercise.

Epidemiology

The measurement of muscle mass in humans is difficult with most of the available methods requiring assumptions that may not always be valid and with variable degrees of accuracy and difficulty. The most direct measurement currently available is urinary creatinine measured over 24-hour periods (58). Other, more indirect measures, include anthropometry (59), bioelectrical impedance, dual-energy x-ray absorptiometry (60), imaging techniques (e.g., computed tomography and magnetic resonance imaging ), ultrasound, total body potassium and neutron activation (6163). Most indirect measures of fat free mass assume, incorrectly, that skeletal muscle remains a constant component of 60% of fat free mass (64). Thus, some authors use only appendicular skeletal mass and correct this for height. Recent studies have demonstrated that not only is muscle mass reduced with advancing age, but the quality of muscle may also change. Increased skeletal muscle lipid, assessed by computerized tomography, is increased with advancing age and increased total body fatness (65). The Health and Body Composition Study, a longitudinal study of more than 3,000 older (age 70 – 79 y at baseline) has demonstrated the strong association of muscle mass with strength as well as the changing quality of skeletal muscle in late life (66). This and other studies show that sarcopenia is associated with reductions in strength, however the relationship between muscle mass and force production deteriorates with advancing age (65, 6769).

Several studies have quantified sarcopenia by indexing fat-free mass or appendicular fat-free mass by divided by height squared, fat mass or total mass. Using an index of aLM/Ht2 in the New Mexico Study, the prevalence of sarcopenia (i.e. aLM/Ht2 2 SD below a young reference group) was originally determined to be over 50% in persons older than 80 years (5). Subsequent studies in this population using more direct estimates found a prevalence of 12% for persons 60 to 70 years of age and nearly 30% for persons over 80 years (5, 70). Janssen et al. (71) using an index of lean/total mass and bioelectric impedance data from NH ANES III, found the prevalence of sarcopenia (−2SD) in persons 60 years of age and older was 7% to 10%. Women were more likely to be sarcopenic than were men in this study, but based on different indices, others have reported the opposite (7276). Table 4 compares a number of different studies on the prevalence of sarcopenia. A common finding of all of these approaches is that sarcopenia, defined as reduced fat free mass, is highly prevalent in older people and that it increases with advancing age.

Table 4.

Indices and prevalence of Sarcopenia.

Citation Method Sarcopenia Index Reference population Gender N Age (years) Prevalence
Baumgartner et al. 1998(5) Anthropometrics Appendicular lean mass/ht2
m ≤ 7.26 kg/m2
f ≤ 5.45 k/m2
Rosetta study (98) (m/f 18–40 yrs) m/f 883 61–70
71–80
≥80
13%
24%
50%
Melton et al. 2000 (76) DXA Appendicular lean mass/ht2
m ≤ 7.26 kg/m2
f ≤ 5.45 k/m2
Rosetta study (98) (m/f 18–40 yrs) m
f
100
99
≥70 28%
52%
Morley et al. 2001 (70) DXA Appendicular lean mass/ht2
m ≤ 7.26 kg/m2
f ≤ 5.45 k/m2
Rosetta study (98) (ref.) (m/f 18–40 yrs) m/f 199 <70
≥80
12%
30%
Janssen et al, 2002 (71) Bioelectrical impedance Ratio of muscle mass/total body mass
m≤31.5%
f ≤22.1%
NHANES III m
f
2,224
2,278
≥60
≥60
7%
10%
Tanko et al, 2002 (75) DXA Appendicular lean mass/ht2
f ≤ 5.4 k/m2
Rosetta study (98) (m/f 18–40 yrs) f 67 ≥70 12%
Ianuzzi-Sacich et al, 2002 (74) DXA Appendicular lean mass/ht2
m ≤ 7.26 kg/m2
f ≤ 5.45k/m2
Rosetta study (98) (m/f 18–40 yrs) m
f
142
195
≥65 27%
23%
Gillette-Guyonnet et al, 2003 (73) Appendicular lean mass/ht2
f ≤ 5.45 k/m2
Rosetta study (98) (m/f 18–40 yrs) f 1,321 ≥75 10%
Newman et al, 2003 (18) DXA Appendicular lean mass/ht2
m≤ 7.23 kg/m2
f ≤ 5.67 kg/m2
Health Aging and Body Composition baseline cohort m
f
1,435
1,549
70–79 20%
20%
Castillo et al, 2004 (72) Bioelectrical Impedance Fat free mass
m ≤ 47.9 kg
f ≤ 34.7 kg
(99)(m/f 25–44) m
f
694
1,006
70–75
≥85
4% 3%
16%
13%
Jansson et al, 2004 (100) Bioelectrical Impedance Total muscle mass/ht2
m ≤ 8.50 kg/m2
f ≤ 5.75 kg/m2
NHANES III m
f
2,223
2,276
≥60 11%
9%
Jansson et al, 2004(100) Bioelectrical Impedance Total lean mass/ht2
m ≤ 8.50kg/m2
f ≤ 5.75 kg/m2
Cardiovascular Health Study M
f
2,196
2,840
≥65 17%
11%
Schaap et al, 2006(101) DXA Longitudinal follow-up LASA study >3% loss of appendicular lean mass LASA study m
f
328 15%*

longitudinal analysis with sarcopenia defined as a loss of appendicular muscle mass of >3% in three years

DXA = dual x-ray absorptiometry; f = female, m = male

Sarcopenia and Disability

Sarcopenia is correlated with functional decline and disability (5, 16, 71, 76, 77). Findings are often stronger in men than in women, depending on the indexing method used. Sarcopenia has also been associated with increased mortality (78), although (79) weakness has been demonstrated to be a more powerful predictor of mortality in elderly people than muscle mass. In the longitudinal Rancho Bernardo study, sarcopenia was shown to be predictive of falls (72). Janssen (80) examined 5,036 men and women over 65 enrolled in the Cardiovascular Health Study. He reported that the likelihood of disability was 79% greater for those with “severe” sarcopenia (< SD below that of a 30-yr old person, based on bioelectric impedance and using lean mass/ height squared but not significantly different for those with “moderate” sarcopenia compared with those with normal muscle mass. During 8-year follow-up, only those with severe sarcopenia were more likely to develop physical disability. Sarcopenia has also been found to predict nosocomial infection during hospitalization (81). The term sarcopenic obesity was first used by Heber et al. (82) in 1996 and describes persons with reduced body mass out of proportion to their adipose mass. Sarcopenic obesity is associated with disability, gait problems and falls to a greater extent than persons with “proportionate” sarcopenia (83). In an 8 year longitudinal study, Baumgartner et al. (83), found that “obese sarcopenia” was a better predictor of physical disabilities, abnormalities in gait or balance and falls in the past year than either sarcopenia or obesity alone. This observation has been confirmed in the Framingham and NHANES (National Health and Nutrition Examination Survey) studies demonstrating that elderly people with high body fat and low muscle mass had the highest rate of disabilities (84). These data point to the fact that the development of disability and impaired mobility in older people is a complex etiology. Muscle mass is an important, but not the only, predictor of muscle strength or physical function. Fat has several adverse effects on muscle function. Higher body fatness and older age have been associated with greater intramuscular lipid and reduced muscle quality, defined as reduced strength/cross sectional area (66, 85). It is also possible that higher body fatness decreases the capacity to generate power (force × speed) and muscle power is more closely related to functional capacity that muscle strength (86). Several indices of sarcopenia that account for muscle and fat mass have been examined in relationship to function. These studies illustrate the complexity in defining sarcopenia in relationship to fat mass (18). When fat mass is considered the role of lean mass per se is apparently small. One study of older women compared BMI to lean mass/total mass and aLM/Ht2 and found only the former two indices were associated with ADL difficulties (87).

As work continues to define the relationship of both lean mass and fat mass together as they relate to disability, refinements in defining sarcopenia are likely to develop. Currently, to classify an individual as sarcopenic, the index of aLM/Ht2 has had the most support, particularly in men. Better reference values and perhaps sex-specific metrics may prove to provide more precise prediction of future disability in older adults.

Conclusion

Sarcopenia represents a major cause of disability and increased health costs in older persons. It is very common but like most geriatric syndromes, seldom recognized by physicians. Identification of sarcopenic patients at greatest risk can be performed using an easy to perform assessment of mobility, such as gait speed and commonly obtained measures of body composition (88). DXA instruments are used to assess bone density for the identification of those at greatest risk for the development of osteoporosis. Advances in instrumentation and software allow for an accurate and precise measure of fat free mass in elderly people. Similarly, elderly people should be screened for low muscle mass and poor functional capacity. These individuals have a very high risk of loss of independence and premature death. A number of promising treatments for sarcopenia are currently under investigation including physical activity, nutritional therapies, androgen therapy, and other behavioral and pharmacological strategies. However, until professional organizations, Centers for Medicare and Medicaid Services and the Food and Drug Administration recognize sarcopenia as a treatable geriatric condition its identification, treatment and the continued development of potential “anti-sarcopenia” agents will be limited.

We propose here in this manuscript a consensus definition of sarcopenia. This definition defines a population of patients that should be considered for evaluation of sarcopenia, a set of guidelines to target patients who may be sarcopenic for further evaluation, and an objective definition of sarcopenia. The use of the current consensus definition for when an individual can be said to be sarcopenic should provide the criteria for who should be considered for treatment of this condition. Although presently limited, available treatments for sarcopenia include interventions to promote healthy eating and increased physical activity.

Acknowledgments

Author Contributions:

  • Roger A. Fielding, Ph.D. (Co-Chair): co-chaired meeting, drafted manuscript, reviewed and edited manuscript

  • Bruno Vellas M.D. (Co-Chair): co-chaired meeting, reviewed and edited manuscript

  • William J. Evans, Ph.D.: drafted manuscript, reviewed and edited manuscript

  • Shalender Bhasin, M.D. : reviewed and edited manuscript

  • John E. Morley, M.D.: drafted manuscript, reviewed and edited manuscript

  • Anne B. Newman, M.D., M.P.H.,: reviewed and edited manuscript

  • Gabor Abellan van Kan: reviewed and edited manuscript

  • Sandrine Andrieu: reviewed and edited manuscript

  • Juergen Bauer: reviewed and edited manuscript

  • Denis Breuille: reviewed and edited manuscript

  • Tommy Cederholm: reviewed and edited manuscript

  • Julie Chandler: reviewed and edited manuscript

  • Capucine De Meynard: reviewed and edited manuscript

  • Lorenzo Donini: reviewed and edited manuscript

  • Tamara Harris: reviewed and edited manuscript

  • Aimo Kannt: reviewed and edited manuscript

  • Florence Keime Guibert: reviewed and edited manuscript

  • Graziano Onder: reviewed and edited manuscript Dimitris Papanicolaou: reviewed and edited manuscript

  • Yves Rolland: reviewed and edited manuscript

  • Daniel Rooks: reviewed and edited manuscript

  • Cornel Seiber: reviewed and edited manuscript

  • Elisabeth Souhami: reviewed and edited manuscript

  • Sjors Verlaan: reviewed and edited manuscript

  • Mauro Zamboni: reviewed and edited manuscript

  • Not in attendance:

  • Alan Sinclair: reviewed and edited manuscript

  • Heike Bischoff: reviewed and edited manuscript

Sources of support: Partial support for this meeting in the form of travel costs were provided by GlaxoSmithKline and Abbott, Chiesi, Danone, Merck, Nestlé, Novartis, Sanofi Aventis

Working Group on Sarcopenia:

Roger A. Fielding, Ph.D. (Co-Chair)1, Bruno Vellas M.D. (Co-Chair)2, William J. Evans, Ph.D.3, Shalender Bhasin, M.D.4, John E. Morley, M.D.5,*, Anne B. Newman6,*, M.D., M.P.H., Gabor Abellan7, Sandrine Andrieu8, Juergen Bauer9, Denis Breuille10, Tommy Cederholm11, Julie Chandler12, Capucine De Meynard13, Lorenzo Donini14, Tamara Hams15, Aimo Kannt16, Florence Keime Guibert17, Graziano Onder18, Dimitris Papanicolaou19, Yves Rolland20, Daniel Rooks21, CornelSieber22, Elisabeth Souhami23, Sjors Verlaan24, Mauro Zamboni25

Footnotes

*

Not in attendance: John Morley

1

Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, USA, roger.fielding@tufts.edu;

2

Gerontopole, Inserm U558 CHU Toulouse, France, vellas.b@chu-toulouse.fr;

3

GlaxoSmithKline, Research Triangle Park, USA, william.j.evans@gsk.com;

4

Boston University School of Medicine Boston, USA, Shalender.Bhasin@bmc.org;

5

University Medical School, St Louis, USA, morley@slu.edu;

6

University of Pittsburgh, Pittsburgh, USA. newmana@edc.pitt.edu;

7

Hôpital La Grave – Casselardit, Toulouse, France, abellan-van-kan-g@chu-toulouse.fr;

8

Inserm U 558, Faculté de médecine, Toulouse, France, sandrieu@cict.fr;

9

University of Erlangen-Nuremberg, Nuremberg, Germany, juergen.bauer@klinikum-nuernberg.de;

10

Centre de Recherche Nestlé, Lausanne, Switzerland, denis.breuille@rdls.nestle.com;

11

Uppsala University, Uppsala, Sweden, Tommy.cederholm@pubcare.uu.se;

12

Merck Research Laboratories, Rah way, USA, julie_chandler@merck.com;

13

Laboratory Chiesi, Courbevoie, France, c.demeynard@chiesifi-ance.com;

14

Università degli Studi Roma « La Sapienza », Roma, Italy, lm.donini@fastwebnet.it;

15

National Institut on Aging, Bethesda, USA, hairis99@nia.nih.gov;

16

Sanofi-Aventis Deutschland GmbH, Frankfurt am Main Germany, Aimo.Kannt@sanofi-aventis.com;

17

Institut de Recherches Internationales SERVIER, Courbevoie, France, Florence.keime-guibert@fr.netgrs.com;

18

Universita Cattalica del Sacre Cuore, Roma, Italy, Graziano_onder@rm.unicatt.it;

19

Merck Research Laboratories Rahway, USA, dimitris_papanicolaou@merck.com;

20

Hôspital La Grave – Casselardit, Toulouse, France, rolland.y@chu-toulouse.fr;

21

Novartis Institutes, Cambridge, USA, Daniel.rooks@novartis.com;

22

Klinikum Nurnberg Nord, Nurnberg, Germany, cornel.sieber@klinikum-nuemberg.de;

23

Sanofi-Avends R&D, Croix de Berny, France, Elisabeth.Souhami@sanofi-aventis.com;

24

Danone Research-Centre for Specialised Nutrition, Schiphol Airport, The Netherlands, sjors.verlaan@danone.com;

25

Universita di Verona, Verona, Italy, mauro.zamboni@univr.it

Financial disclosures: Partial support for this meeting in the form of travel costs were provided by GlaxoSmithKline and Abbott, Chiesi, Danone, Merck, Nestlé, Novartis, Sanofi Aventis

References

  • 1.Tzankoff SP, Norris AH. Longitudinal changes in basal metabolic rate in man. J Appl Physiol. 1978;33:536–9. doi: 10.1152/jappl.1978.45.4.536. [DOI] [PubMed] [Google Scholar]
  • 2.Rosenberg IH. Sarcopenia: origins and clinical relevance. J Nutr. 1997 May;127(5 Suppl):990S–lS. doi: 10.1093/jn/127.5.990S. [DOI] [PubMed] [Google Scholar]
  • 3.Evans WJ, Campbell WW. Sarcopenia and age–related changes in body composition and functional capacity. J Nutr. 1993;123:465–8. doi: 10.1093/jn/123.suppl_2.465. [DOI] [PubMed] [Google Scholar]
  • 4.Evans W. What is sarcopenia? J Gerontol. 1995;50A(special issue):5–8. [Google Scholar]
  • 5.Baumgartner RN, Koehler KM, Gallagher D, et al. Epidemiology of sarcopenia among the elderly in New Mexico. Am J Epidemiol. 1998;147(8):755–63. doi: 10.1093/oxfordjournals.aje.a009520. [DOI] [PubMed] [Google Scholar]
  • 6.http://apps.isiknowledge.com/WoS/CIW.cgi
  • 7.Morley JE. Anorexia, sarcopenia, and aging. Nutrition. 2001;17(7–8):660–3. doi: 10.1016/s0899-9007(01)00574-3. [DOI] [PubMed] [Google Scholar]
  • 8.Morley JE, Kim MJ, Haren MT, et al. Frailty and the aging male. Aging Male. 2005 Sep-Dec;8(3–4):135–40. doi: 10.1080/13685530500277232. [DOI] [PubMed] [Google Scholar]
  • 9.Critchley M. The neurology of old age. Lancet. 1931;1:1221–30. [Google Scholar]
  • 10.Lexell J, Henriksson–Larsen K, Wimblod B, Sjostrom M. Distribution of different fiber types in human skeletal muscles: Effects of aging studied in whole muscle cross sections. Muscle Nerve. 1983;6:588–95. doi: 10.1002/mus.880060809. [DOI] [PubMed] [Google Scholar]
  • 11.Larsson L. Morphological and functional characteristics of the aging skeletal muscle in man. Acta Physiol Scand Suppl. 1978;457(Suppl):1–36. [PubMed] [Google Scholar]
  • 12.Larsson L. Histochemical characteristics of human skeletal muscle during aging. Acat Physiol Scand. 1983;117:469–71. doi: 10.1111/j.1748-1716.1983.tb00024.x. [DOI] [PubMed] [Google Scholar]
  • 13.www.mastersweightlifting.org/records.htm
  • 14.Frontera WR, Hughes VA, Fielding RA, et al. Aging of skeletal muscle: a 12–yr longitudinal study. Journal of Applied Physiology. 2000;88(4):1321–6. doi: 10.1152/jappl.2000.88.4.1321. [DOI] [PubMed] [Google Scholar]
  • 15.Ferrucci L, Guralnik JM, Buchner D, et al. Departures from linearity in the relationship between measures of muscular strength and physical performance of the lower extremities: the Women’s Health and Aging Study. J Gerontol A Biol Sci Med Sci. 1997 Sep;52(5):M275–85. doi: 10.1093/gerona/52a.5.m275. [DOI] [PubMed] [Google Scholar]
  • 16.Delmonico MJ, Harris TB, Lee JS, et al. Alternative definitions of sarcopenia, lower extremity performance, and functional impairment with aging in older men and women. J Am Geriatr Soc. 2007 May;55(5):769–74. doi: 10.1111/j.1532-5415.2007.01140.x. [DOI] [PubMed] [Google Scholar]
  • 17.Janssen I, Shepard DS, Katzmarzyk PT, Roubenoff R. The healthcare costs of sarcopenia in the United States. J Am Geriatr Soc. 2004 Jan;52(1):80–5. doi: 10.1111/j.1532-5415.2004.52014.x. [DOI] [PubMed] [Google Scholar]
  • 18.Newman AB, Kupelian V, Visser M, et al. Sarcopenia: alternative definitions and associations with lower extremity function. J Am Geriatr Soc. 2003 Nov;51(11):1602–9. doi: 10.1046/j.1532-5415.2003.51534.x. [DOI] [PubMed] [Google Scholar]
  • 19.Lexell J, Taylor CC, Sjostrom M. What is the cause of the ageing atrophy? Total number, size and proportion of different fiber types studied in whole vastus lateralis muscle from 15– to 83–year–old men. J Neurol Sci. 1988 Apr;84(2–3):275–94. doi: 10.1016/0022-510x(88)90132-3. [DOI] [PubMed] [Google Scholar]
  • 20.Porter MM, Vandervoort AA, Lexell J. Aging of human muscle: structure, function and adaptability. Scand J Med Sci Sports. 1995 Jun;5(3):129–42. doi: 10.1111/j.1600-0838.1995.tb00026.x. [DOI] [PubMed] [Google Scholar]
  • 21.Reeves ND, Narici MV, Maganaris CN. Myotendinous plasticity to ageing and resistance exercise in humans. Exp Physiol. 2006 May;91(3):483–98. doi: 10.1113/expphysiol.2005.032896. [DOI] [PubMed] [Google Scholar]
  • 22.Doherty TJ. Invited review: Aging and sarcopenia. J Appl Physiol. 2003 Oct;95(4):1717–27. doi: 10.1152/japplphysiol.00347.2003. [DOI] [PubMed] [Google Scholar]
  • 23.Narici MV, Maganaris CN. Adaptability of elderly human muscles and tendons to increased loading. J Anat. 2006 Apr;208(4):433–43. doi: 10.1111/j.1469-7580.2006.00548.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Onambele GL, Narici MV, Maganaris CN. Calf muscle–tendon properties and postural balance in old age. J Appl Physiol. 2006 Jun;100(6):2048–56. doi: 10.1152/japplphysiol.01442.2005. [DOI] [PubMed] [Google Scholar]
  • 25.Sipila S, Suominen H. Quantitative ultrasonography of muscle: detection of adaptations to training in elderly women. Arch Phys Med Rehabil. 1996 Nov;77(11):1173–8. doi: 10.1016/s0003-9993(96)90143-4. [DOI] [PubMed] [Google Scholar]
  • 26.Kubo K, Kanehisa H, Azuma K, et al. Muscle architectural characteristics in young and elderly men and women. Int J Sports Med. 2003 Feb;24(2):125–30. doi: 10.1055/s-2003-38204. [DOI] [PubMed] [Google Scholar]
  • 27.Mian OS, Thorn JM, Ardigo LP, et al. Gastrocnemius muscle–tendon behaviour during walking in young and older adults. Acta Physiol (Oxf) 2007 Jan;189(1 ):57–65. doi: 10.1111/j.1748-1716.2006.01634.x. [DOI] [PubMed] [Google Scholar]
  • 28.Newman AB, Lee JS, Visser M, et al. Weight change and the conservation of lean mass in old age: the Health, Aging and Body Composition Study. Am J Clin Nutr. 2005 Oct;82(4):872–8. doi: 10.1093/ajcn/82.4.872. quiz 915–6. [DOI] [PubMed] [Google Scholar]
  • 29.Rolland YM, Perry HM, 3rd, Patrick P, et al. Loss of appendicular muscle mass and loss of muscle strength in young postmenopausal women. J Gerontol A Biol Sci Med Sci. 2007 Mar;62(3):330–5. doi: 10.1093/gerona/62.3.330. [DOI] [PubMed] [Google Scholar]
  • 30.Welle S, Brooks Al, Delehanty JM, et al. Gene expression profile of aging in human muscle. Physiol Genomics. 2003 Jul 7;14(2):149–59. doi: 10.1152/physiolgenomics.00049.2003. [DOI] [PubMed] [Google Scholar]
  • 31.Carey KA, Farnfield MM, Tarquinio SD, et al. Impaired expression of Notch signaling genes in aged human skeletal muscle. J Gerontol A Biol Sci Med Sci. 2007 Jan;62(1):9–17. doi: 10.1093/gerona/62.1.9. [DOI] [PubMed] [Google Scholar]
  • 32.Schrager MA, Roth SM, Ferrell RE, et al. Insulin-like growth factor-2 genotype, fat-free mass, and muscle performance across the adult life span. J Appl Physiol. 2004 Dec;97(6):2176–83. doi: 10.1152/japplphysiol.00985.2003. [DOI] [PubMed] [Google Scholar]
  • 33.Chapman IM, Macintosh CG, Morley JE, et al. The anorexia of ageing. Biogerontology. 2002;3(1–2):67–71. doi: 10.1023/a:1015211530695. [DOI] [PubMed] [Google Scholar]
  • 34.Volpi E, Sheffield-Moore M, Rasmussen BB, et al. Basal muscle amino acid kinetics and protein synthesis in healthy young and older men. JAMA. 2001;286( 10):1206–12. doi: 10.1001/jama.286.10.1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Katsanos CS, Kobayashi H, Sheffield-Moore M, et al. Aging is associated with diminished accretion of muscle proteins after the ingestion of a small bolus of essential amino acids. Am J Clin Nutr. 2005 Nov;82(5):1065–73. doi: 10.1093/ajcn/82.5.1065. [DOI] [PubMed] [Google Scholar]
  • 36.Campbell WW, Crim MC, Dallal GE, et al. Increased protein requirements in elderly people: new data and retrospective reassessments. Am J Clin Nutr. 1994 Oct;60(4):501–9. doi: 10.1093/ajcn/60.4.501. [DOI] [PubMed] [Google Scholar]
  • 37.Campbell WW, Trappe TA, Wolfe RR, et al. The recommended dietary allowance for protein may not be adequate for older people to maintain skeletal muscle. J Gerontol A Biol Sci Med Sci. 2001 Jun;56(6):M373–80. doi: 10.1093/gerona/56.6.m373. [DOI] [PubMed] [Google Scholar]
  • 38.Visser M, Deeg DJ, Lips P. Low vitamin D and high parathyroid hormone levels as determinants of loss of muscle strength and muscle mass (sarcopenia): the Longitudinal Aging Study Amsterdam. J Clin Endocrinol Metab. 2003 Dec;88(12):5766–72. doi: 10.1210/jc.2003-030604. [DOI] [PubMed] [Google Scholar]
  • 39.Kuh D, Bassey EJ, Butterworth S, et al. Grip strength, postural control, and functional leg power in a representative cohort of British men and women: associations with physical activity, health status, and socioeconomic conditions. Journals of Gerontology Series A-Biological Sciences & Medical Sciences. 2005;60(2):224–31. doi: 10.1093/gerona/60.2.224. [DOI] [PubMed] [Google Scholar]
  • 40.Hughes VA, Roubenoff R, Wood M, et al. Anthropometric assessment of 10-y changes in body composition in the elderly. Am J Clin Nutr. 2004;80(2):475–82. doi: 10.1093/ajcn/80.2.475. [DOI] [PubMed] [Google Scholar]
  • 41.Hughes VA, Frontera WR, Roubenoff R, et al. Longitudinal changes in body composition in older men and women: role of body weight change and physical activity. Am J Clin Nutr. 2002;76(2):473–81. doi: 10.1093/ajcn/76.2.473. [DOI] [PubMed] [Google Scholar]
  • 42.Kortebein P, Ferrando A, Lombeida J, et al. Effect of 10 days of bed rest on skeletal muscle in healthy older adults. Jama. 2007 Apr 25;297(16):1772–4. doi: 10.1001/jama.297.16.1772-b. [DOI] [PubMed] [Google Scholar]
  • 43.Baumgartner RN, Waters DL, Gallagher D, et al. Predictors of skeletal muscle mass in elderly men and women. Mech Ageing Dev. 1999;107(2):123–36. doi: 10.1016/s0047-6374(98)00130-4. [DOI] [PubMed] [Google Scholar]
  • 44.Morley JE. Hormones and the aging process. J Am Geriatr Soc. 2003 Jul;51(7 Suppl):S333–7. doi: 10.1046/j.1365-2389.2003.51344.x. [DOI] [PubMed] [Google Scholar]
  • 45.Guillet C, Boirie Y. Insulin resistance: a contributing factor to age-related muscle mass loss? Diabetes Metab. 2005 Dec;31(Spec No 2):5S20–5S6. doi: 10.1016/s1262-3636(05)73648-x. [DOI] [PubMed] [Google Scholar]
  • 46.Park SW, Goodpaster BH, Lee JS, et al. Excessive Loss of Skeletal Muscle Mass in Older Adults with Type 2 Diabetes. Diabetes Care. 2009 Jun 23; doi: 10.2337/dc09-0264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Park SW, Goodpaster BH, Strotmeyer ES, et al. Decreased muscle strength and quality in older adults with type 2 diabetes: the health, aging, and body composition study. Diabetes. 2006 Jun;55(6):1813–8. doi: 10.2337/db05-1183. [DOI] [PubMed] [Google Scholar]
  • 48.McDermott MM, Greenland P, Liu K, et al. Leg symptoms in peripheral arterial disease: associated clinical characteristics and functional impairment. JAMA. 2001 Oct 3;286(13):1599–606. doi: 10.1001/jama.286.13.1599. [DOI] [PubMed] [Google Scholar]
  • 49.McDermott MM, Guralnik JM, Albay M, et al. Impairments of muscles and nerves associated with peripheral arterial disease and their relationship with lower extremity functioning: the InCHIANTI Study. J Am Geriatr Soc. 2004 Mar;52(3):405–10. doi: 10.1111/j.1532-5415.2004.52113.x. [DOI] [PubMed] [Google Scholar]
  • 50.McDermott MM, Guralnik JM, Ferrucci L, et al. Physical activity, walking exercise, and calf skeletal muscle characteristics in patients with peripheral arterial disease. J Vasc Surg. 2007 Jul;46(l):87–93. doi: 10.1016/j.jvs.2007.02.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ferrucci L, Penninx BW, Volpato S, et al. Change in muscle strength explains accelerated decline of physical function in older women with high interleukin-6 serum levels. J Am Geriatr Soc. 2002 Dec;50(12):1947–54. doi: 10.1046/j.1532-5415.2002.50605.x. [DOI] [PubMed] [Google Scholar]
  • 52.Kandarian SC, Jackman RW. Intracellular signaling during skeletal muscle atrophy. Muscle Nerve. 2006 Feb;33(2):155–65. doi: 10.1002/mus.20442. [DOI] [PubMed] [Google Scholar]
  • 53.Clavel S, Coldefy AS, Kurkdjian E, et al. Atrophy-related ubiquitin ligases, atrogin-1 and MuRFl are up-regulated in aged rat Tibialis Anterior muscle. Mech Ageing Dev. 2006 Oct;127(10):794–801. doi: 10.1016/j.mad.2006.07.005. [DOI] [PubMed] [Google Scholar]
  • 54.Stipanuk MH. Leucine and protein synthesis: mTOR and beyond. Nutr Rev. 2007 Mar;65(3):122–9. doi: 10.1111/j.1753-4887.2007.tb00289.x. [DOI] [PubMed] [Google Scholar]
  • 55.Carter CS, Onder G, Kritchevsky SB, et al. Angiotensin-converting enzyme inhibition intervention in elderly persons: effects on body composition and physical performance. J Gerontol A Biol Sci Med Sci. 2005 Nov;60(11):1437–46. doi: 10.1093/gerona/60.11.1437. [DOI] [PubMed] [Google Scholar]
  • 56.Lecker SH, Jagoe RT, Gilbert A, et al. Multiple types of skeletal muscle atrophy involve a common program of changes in gene expression. Faseb J. 2004;18(1 ):39–51. doi: 10.1096/fj.03-0610com. [DOI] [PubMed] [Google Scholar]
  • 57.Raue U, Slivka D, Jemiolo B, et al. Proteolytic gene expression differs at rest and after resistance exercise between young and old women. J Gerontol A Biol Sci Med Sci. 2007 Dec;62(12):1407–12. doi: 10.1093/gerona/62.12.1407. [DOI] [PubMed] [Google Scholar]
  • 58.Heymsfield SB, Arteaga C, McManus C, et al. Measurement of muscle mass in humans: validity of the 24-hour urinary creatinine method. Am J Clin Nutr. 1983;37:478–94. doi: 10.1093/ajcn/37.3.478. [DOI] [PubMed] [Google Scholar]
  • 59.Heymsfield SB, McManus C, Smith J, et al. Anthropometric measurement of muscle mass: revised equations for calculating bone-free arm muscle area. Am J Clin Nutr. 1982;36:680–90. doi: 10.1093/ajcn/36.4.680. [DOI] [PubMed] [Google Scholar]
  • 60.Heymsfield SB, Smit R, Aulet M. Appendicular skeletal muscle mass: measurement by dual photon absorptiometry. Am J Clin Nutr. 1990;52:214–8. doi: 10.1093/ajcn/52.2.214. [DOI] [PubMed] [Google Scholar]
  • 61.Cohn SH, Ellis KJ, Wallach S. In vivo neutron activation analysis: clinical potential in body composition studies. Amer J Med. 1974;57:683–6. doi: 10.1016/0002-9343(74)90841-9. [DOI] [PubMed] [Google Scholar]
  • 62.Cohn SH, Vartsky D, Yasumura S, et al. Indexes of body cell mass: nitrogen versus potassium. Am J Physiol. 1983;244:E305–E10. doi: 10.1152/ajpendo.1983.244.3.E305. [DOI] [PubMed] [Google Scholar]
  • 63.Reeves ND, Maganaris CN, Narici MV. Ultrasonographic assessment of human skeletal muscle size. Eur J Appl Physiol. 2004 Jan;91(1):1 16–8. doi: 10.1007/s00421-003-0961-9. [DOI] [PubMed] [Google Scholar]
  • 64.Heymsfield SB, Waki M. Body composition in humans:Advancesin tne development of multicompartment chemical models. Nutrition reviews. 1991;49:91–108. doi: 10.1111/j.1753-4887.1991.tb02997.x. [DOI] [PubMed] [Google Scholar]
  • 65.Goodpaster BH, Carlson CL, Visser M, et al. Attenuation of skeletal muscle and strength in the elderly: The Health ABC Study. Journal of Applied Physiology. 2001;90(6):2157–65. doi: 10.1152/jappl.2001.90.6.2157. [DOI] [PubMed] [Google Scholar]
  • 66.Goodpaster BH, Park SW, Harris TB, et al. The loss of skeletal muscle strength, mass, and quality in older adults: the health, aging and body composition study. J Gerontol A Biol Sci Med Sci. 2006 Oct;61( 10):1059–64. doi: 10.1093/gerona/61.10.1059. [DOI] [PubMed] [Google Scholar]
  • 67.Ojanen T, Rauhala T, Hakkinen K. Strength and power profiles of the lower and upper extremities in master throwers at different ages. J Strength Cond Res. 2007 Feb;21(1):216–22. doi: 10.1519/00124278-200702000-00039. [DOI] [PubMed] [Google Scholar]
  • 68.Bruce SA, Newton D, Woledge RC. Effect of age on voluntary force and cross-sectional area of human adductor possicis muscle. Q J Exp Physiology. 1989;74:359–62. doi: 10.1113/expphysiol.1989.sp003278. [DOI] [PubMed] [Google Scholar]
  • 69.Vandervoort AA, McComas AJ. Contractile changes in opposing muscles of the human ankle joint with aging. J Appl Physiol. 1986 Jul;61(1):361–7. doi: 10.1152/jappl.1986.61.1.361. [DOI] [PubMed] [Google Scholar]
  • 70.Morley JE, Baumgartner RN, Roubenoff R, et al. Sarcopenia. J Lab Clin Med. 2001 Apr;137(4):231–43. doi: 10.1067/mlc.2001.113504. [DOI] [PubMed] [Google Scholar]
  • 71.Janssen I, Heymsfield SB, Ross R. Low relative skeletal muscle mass (sarcopenia) in older persons is associated with functional impairment and physical disability. J Am Geriatr Soc. 2002 May;50(5):889–96. doi: 10.1046/j.1532-5415.2002.50216.x. [DOI] [PubMed] [Google Scholar]
  • 72.Castillo EM, Goodman-Gruen D, Kritz-Silverstein D, et al. Sarcopenia in elderly men and women: the Rancho Bernardo study. Am J Prev Med. 2003 Oct;25(3):226–31. doi: 10.1016/s0749-3797(03)00197-1. [DOI] [PubMed] [Google Scholar]
  • 73.Gillette-Guyonnet S, Nourhashemi F, Andrieu S, et al. Body composition in French women 75+ years of age: the EPIDOS study. Mech Ageing Dev. 2003 Mar;124(3):311–6. doi: 10.1016/s0047-6374(02)00198-7. [DOI] [PubMed] [Google Scholar]
  • 74.Iannuzzi-Sucich M, Prestwood KM, Kenny AM. Prevalence of sarcopenia and predictors of skeletal muscle mass in healthy, older men and women. J Gerontol A Biol Sci Med Sci. 2002 Dec;57(12):M772–7. doi: 10.1093/gerona/57.12.m772. [DOI] [PubMed] [Google Scholar]
  • 75.Tanko LB, Movsesyan L, Mouritzen U, et al. Appendicular lean tissue mass and the prevalence of sarcopenia among healthy women. Metabolism. 2002 Jan;51(1):69–74. doi: 10.1053/meta.2002.28960. [DOI] [PubMed] [Google Scholar]
  • 76.Melton LJ, 3rd, Khosla S, Crowson CS, et al. Epidemiology of sarcopenia. Journal of the American Geriatrics Society. 2000;48(6):625–30. [PubMed] [Google Scholar]
  • 77.Lauretani F, Russo CR, Bandinelli S, et al. Age-associated changes in skeletal muscles and their effect on mobility: an operational diagnosis of sarcopenia. J Appl Physiol. 2003 Nov;95(5):1851–60. doi: 10.1152/japplphysiol.00246.2003. [DOI] [PubMed] [Google Scholar]
  • 78.Metter EJ, Talbot LA, Schrager M, Conwit R. Skeletal muscle strength as a predictor of all-cause mortality in healthy men. J Gerontol A Biol Sci Med Sci. 2002 Oct;57(10):B359–65. doi: 10.1093/gerona/57.10.b359. [DOI] [PubMed] [Google Scholar]
  • 79.Newman AB, Kupelian V, Visser M, et al. Strength, but not muscle mass, is associated with mortality in the health, aging and body composition study cohort. J Gerontol A Biol Sci Med Sci. 2006 Jan;61(1):72–7. doi: 10.1093/gerona/61.1.72. [DOI] [PubMed] [Google Scholar]
  • 80.Janssen I. Influence of sarcopenia on the development of physical disability: the Cardiovascular Health Study. J Am Geriatr Soc. 2006 Jan;54(1):56–62. doi: 10.1111/j.1532-5415.2005.00540.x. [DOI] [PubMed] [Google Scholar]
  • 81.Cosqueric G, Sebag A, Ducolombier C, et al. Sarcopenia is predictive of nosocomial infection in care of the elderly. Br J Nutr. 2006 Nov;96(5):895–901. doi: 10.1017/bjn20061943. [DOI] [PubMed] [Google Scholar]
  • 82.Heber D, Ingles S, Ashley JM, et al. Clinical detection of sarcopenic obesity by bioelectrical impedance analysis. Am J Clin Nutr. 1996 Sep;64(3 Suppl):472S–7S. doi: 10.1093/ajcn/64.3.472S. [DOI] [PubMed] [Google Scholar]
  • 83.Baumgartner RN, Wayne SJ, Waters DL, et al. Sarcopenic obesity predicts instrumental activities of daily living disability in the elderly. Obes Res. 2004 Dec;12(12):1995–2004. doi: 10.1038/oby.2004.250. [DOI] [PubMed] [Google Scholar]
  • 84.Davison KK, Ford ES, Cogswell ME, et al. Percentage of body fat and body mass index are associated with mobility limitations in people aged 70 and older from NHANES III. J Am Geriatr Soc. 2002 Nov;50(11):1802–9. doi: 10.1046/j.1532-5415.2002.50508.x. [DOI] [PubMed] [Google Scholar]
  • 85.Goodpaster BH. Intramuscular lipid content is increased in obesity and decreased by weight loss. American Journal of Physiology. 1999;277(6 Pt 1):E1130–41. [Google Scholar]
  • 86.Bassey EJ, Fiatarone MA, O’Neill EF, et al. Leg extensor power and functional performance in very old men and women. Clin Sci. 1992;82:321–7. doi: 10.1042/cs0820321. [DOI] [PubMed] [Google Scholar]
  • 87.Zoico E, Di Francesco V, Guralnik JM, et al. Physical disability and muscular strength in relation to obesity and different body composition indexes in a sample of healthy elderly women. Int J Obes Relat Metab Disord. 2004 Feb;28(2):234–41. doi: 10.1038/sj.ijo.0802552. [DOI] [PubMed] [Google Scholar]
  • 88.Guralnik JM, Ferrucci L, Simonsick EM, et al. Lower-extremity function in persons over the age of 70 years as a predictor of sebsequent disability. N Engl J Med. 1995;332:556–61. doi: 10.1056/NEJM199503023320902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Visser M, Harris TB, Langlois J, et al. Body fat and skeletal muscle mass in relation to physical disability in very old men and women of the Framingham Heart Study. J Gerontol A Biol Sci Med Sci. 1998;53(3):M214–21. doi: 10.1093/gerona/53a.3.m214. [DOI] [PubMed] [Google Scholar]
  • 90.Short KR, Nair KS. The effect of age on protein metabolism. Curr Opin Clin Nutr Metab Care. 2000 Jan;3(1):39–44. doi: 10.1097/00075197-200001000-00007. [DOI] [PubMed] [Google Scholar]
  • 91.Vermeulen A, Goemaere S, Kaufman JM. Testosterone, body composition and aging. Journal of Endocrinological Investigation. 1999;22(5 Suppl):110–6. [PubMed] [Google Scholar]
  • 92.Schrager MA, Metter EJ, Simonsick E, et al. Sarcopenic obesity and inflammation in the InCHIANTI study. J Appl Physiol. 2007 Mar;102(3):919–25. doi: 10.1152/japplphysiol.00627.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Di Iorio A, Cherubini A, Volpato S, et al. Markers of inflammation, Vitamin E and peripheral nervous system function The InCHIANTI study. Neurobiol Aging. 2005 Aug 19; doi: 10.1016/j.neurobiolaging.2005.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Evans WJ, Farrell PA. The aging pancreas: The effects of aging on insulin secretion and action. In: Jefferson JS, Cherrington AD, editors. The Handbook of Physiology. Oxford: Oxford University Press; 2001. pp. 969–99. [Google Scholar]
  • 95.Guralnik JM, Ferrucci L, Pieper CF, et al. Lower extremity function and subsequent disability: consistency across studies, predictive models, and value of gait speed alone compared with the short physical performance battery. J Gerontol A Biol Sci Med Sci. 2000 Apr;55(4):M221–31. doi: 10.1093/gerona/55.4.m221. [DOI] [PubMed] [Google Scholar]
  • 96.Cesari M, Kritchevsky SB, Baumgartner RN, et al. Sarcopenia, obesity, and inflammation--results from the Trial of Angiotensin Converting Enzyme Inhibition and Novel Cardiovascular Risk Factors study. Am J Clin Nutr. 2005 Aug;82(2):428–34. doi: 10.1093/ajcn.82.2.428. [DOI] [PubMed] [Google Scholar]
  • 97.Cesari M, Penninx BW, Pahor M, et al. Inflammatory markers and physical performance in older persons: the InCHIANTI study. J Gerontol A Biol Sci Med Sci. 2004 Mar;59(3):242–8. doi: 10.1093/gerona/59.3.m242. [DOI] [PubMed] [Google Scholar]
  • 98.Gallagher D, Visser M, De Meersman RE, et al. Appendicular skeletal muscle mass: effects of age, gender, and ethnicity. Journal of Applied Physiology. 1997;83(1):229–39. doi: 10.1152/jappl.1997.83.1.229. [DOI] [PubMed] [Google Scholar]
  • 99.Pichard C, Kyle UG, Bracco D, et al. Reference values of fat-free and fat masses by bioelectrical impedance analysis in 3393 healthy subjects. Nutrition. 2000;16:245–54. doi: 10.1016/s0899-9007(00)00256-2. [DOI] [PubMed] [Google Scholar]
  • 100.Janssen I, Baumgartner RN, Ross R, et al. Skeletal muscle outpoints associated with elevated physical disability risk in older men and women. Am J Epidemiol. 2004 Feb 15;159(4):413–21. doi: 10.1093/aje/kwh058. [DOI] [PubMed] [Google Scholar]
  • 101.Schaap LA, Pluijm SM, Deeg DJ, Visser M. Inflammatory markers and loss of muscle mass (sarcopenia) and strength. Am J Med. 2006 Jun;119(6):526, e9–17. doi: 10.1016/j.amjmed.2005.10.049. [DOI] [PubMed] [Google Scholar]

RESOURCES