You are on page 1of 16

Downloaded from http://perspectivesinmedicine.cshlp.

org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

Effects of Exercise and Aging on Skeletal Muscle

Giovanna Distefano1 and Bret H. Goodpaster1,2


1
Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida 32804
2
Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827
Correspondence: bret.goodpaster@flhosp.org

A substantial loss of muscle mass and strength (sarcopenia), a decreased regenerative capac-
ity, and a compromised physical performance are hallmarks of aging skeletal muscle. These
changes are typically accompanied by impaired muscle metabolism, including mitochon-
drial dysfunction and insulin resistance. A challenge in the field of muscle aging is to dis-
sociate the effects of chronological aging per se on muscle characteristics from the secondary
influence of lifestyle and disease processes. Remarkably, physical activity and exercise are
well-established countermeasures against muscle aging, and have been shown to attenuate
age-related decreases in muscle mass, strength, and regenerative capacity, and slow or
prevent impairments in muscle metabolism. We posit that exercise and physical activity
can influence many of the changes in muscle during aging, and thus should be emphasized
as part of a lifestyle essential to healthy aging.
www.perspectivesinmedicine.org

keletal muscle aging is characterized by a changes that likely further compromise muscle
S number of structural and functional changes
that are associated with increased physical lim-
function (Gonzalez-Freire et al. 2014; Men-
donca et al. 2016). Additionally, age-related al-
itations and risk for disease. The progressive loss terations in muscle metabolism, including insu-
of muscle mass and function, a condition or lin sensitivity and mitochondrial capacity, have
process referred to as sarcopenia (Rosenberg been extensively investigated (Tonkonogi et al.
1997; Cruz-Jentoft et al. 2010), has long been 2003; Amati et al. 2009; Consitt et al. 2013; Por-
recognized as being among the most remarkable ter et al. 2015; Distefano et al. 2016). Despite the
and deleterious of these changes. Studies per- significant advances in the field of muscle aging,
formed in the 1980s and 1990s identified fun- many questions remain, especially concerning
damentally important structural changes in the the roles of both mass and function, along
aging muscle (Lexell et al. 1983, 1986, 1988; with the underlying mechanisms of sarcopenia
Forsberg et al. 1991; Overend et al. 1992; Lexell and the bioenergetics of the aged muscle.
1997), as well as a diminished regenerative ca- A challenge in the study of skeletal muscle
pacity and satellite cell potential (Snow 1977; aging is to decipher whether the deterioration
Schultz and Lipton 1982). These adaptations of muscle function is attributed to age per se, or
are accompanied by neurological and vascular rather is a consequence of lifestyle and disease.

Editors: Juleen R. Zierath, Michael J. Joyner, and John A. Hawley


Additional Perspectives on The Biology of Exercise available at www.perspectivesinmedicine.org
Copyright # 2018 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a029785
Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785

1
Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

G. Distefano and B.H. Goodpaster

As proposed by Busse (1969), a combination of balance between energy intake and energy ex-
primary and secondary aging occurs (Busse penditure have been shown to exacerbate these
1969). Primary aging corresponds to the inevi- changes. In the following sections, we discuss
table changes in cellular structure and function the robust influence of obesity and physical ac-
that happens independent of lifestyle, environ- tivity on the age-related muscle changes. The
mental influences, or disease. Changes involving distinct and combined effects of primary aging,
interactions of primary aging with environmen- obesity, and reduced physical activity on skeletal
tal influences and disease define secondary ag- muscle, although not completely understood,
ing. While considerable efforts have been made are presented in Table 1. While aging is associ-
to identify potential interventions that prevent ated with decreases in muscle mass, strength,
or diminish primary aging, physical activity and and regenerative capacity, its effect on insulin
exercise are feasible and well-established coun- sensitivity and mitochondrial capacity is highly
termeasures against secondary aging (Booth influenced by obesity and physical activity. In-
et al. 2011). Exercise improves cardiorespiratory creased body fat likely potentiates the loss of
fitness in older men and women (Pruchnic et al. muscle mass and strength, and is associated
2004) and decreases the likelihood of several with insulin sensitivity, mitochondrial dysfunc-
deadly chronic diseases (Kyu et al. 2016). Exer- tion, and impaired regenerative capacity. Con-
cise also increases myofiber size (Bamman et al. versely, physical activity can improve all of these
2003), whole muscle mass (Harridge et al. 1999), muscle characteristics (Fig. 1). While resistance
muscle quality (Da Boit et al. 2016), improves exercise is generally used to increase muscle
functional abilities (Da Boit et al. 2016), and mass and strength, and aerobic exercise is
attenuates age-related decreases in muscle known to improve insulin sensitivity and mito-
strength (Goodpaster et al. 2008) and increases chondrial capacity, additional studies are need-
www.perspectivesinmedicine.org

in fat infiltration (Goodpaster et al. 2008). Fur- ed to better understand the optimal type and
thermore, exercise can prevent age-associated amount of physical activity required to improve
muscle insulin resistance (Amati et al. 2009) muscle health.
and diminished mitochondrial capacity (Safdar
et al. 2010).
The purpose of this review is to (1) describe Muscle Mass, Structure, and Strength
the effect of aging on several skeletal muscle char- A decline in skeletal muscle mass (atrophy) be-
acteristics, (2) discuss the influence of primary gins during the third or fourth decade of life,
and secondary aging on these processes, and (3) and approximately 10% of muscle can be lost by
examine the preventive and therapeutic effects of the age of 50 years (Lexell et al. 1988). The rate
physical activity and exercise on skeletal muscle
aging. We will focus on the effects of aging on
muscle morphology, mass, strength, insulin sen- Table 1. Skeletal muscle characteristics and how they
sitivity, mitochondrial capacity, and regenera- are affected by aging, obesity, and physical activity
tive potential. Additionally, we will discuss the Physical
beneficial effects of an active lifestyle to prevent Muscle characteristic Aging Obesity activity
or counteract age-related muscular changes.
Muscle mass   
Muscle strength   
SKELETAL MUSCLE CHANGES WITH Insulin sensitivity $  
CHRONOLOGICAL AGING AND THE Mitochondrial capacity
INFLUENCE OF LIFESTYLE Content $  
Function $  
Skeletal muscle has a remarkable capacity to Turnover ? ? ?
adapt to the demands imposed on it, a process Regenerative capacity   
known as muscle plasticity. While chronological  Decrease, increase, $ no change, $ no change or
aging can promote changes in muscle, an im- small effect, ? not well established.

2 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785


www.perspectivesinmedicine.org

Impaired regeneration Better regeneration

Muscle injury Muscle injury


Aging

Poor nutrition Good nutrition


↑ Fibrosis formation Sedentary lifestyle Active lifestyle
↓ Fibrosis formation

A. Unhealthy B. Healthy

↑ Intermuscular adipose tissue ↓ Intermuscular adipose tissue

Impaired
Better
contractile properties
contractile properties

Muscle mass
Muscle strength
Physical performance
Glucose Glucose
↑ Inflammation
Mobility limitations
Increased fall risk
Insulin resistant Insulin sensitive
Functional limitations
Institutionalization
Mitochondrial dysfunction Mortality Healthy mitochondria

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785


Figure 1. Sedentary lifestyle contributes to an “unhealthy aging.” (A) In this scenario, impairments on muscle regeneration
are exacerbated because of a reduction in satellite cell content and a decreased myogenic potential and increased fibrosis.
Unhealthy aging is also characterized by an augmented accumulation of intermuscular adipocytes. Furthermore, obesity is
known to exacerbate the low-grade inflammation that occurs with aging. These lifestyle factors may also impair myofiber
contractile function. Poor nutrition and physical inactivity are more strongly associated with insulin resistance and
mitochondrial dysfunction than chronological age, although they may exacerbate or accelerate loss of muscle mass,
strength, and functional performance and energy metabolism. Conversely, exercise can profoundly improve muscle me-
tabolism, strength, and function, which can attenuate or prevent some of the negative age-associated changes and will
translate to a “healthy” aging. (B) In this scenario, exercise reduces or delays the declines in muscle regeneration, increases
satellite cells and enhances their activation, improves myogenic potential, and reduces fibrosis formation. Exercise can also
reduce the age-associated accumulation of intermuscular fat and alter intramyocellular lipids (IMCLs). Exercise improves
muscle cell and tissue contractile function. Exercise can robustly improve impaired muscle metabolism, such as insulin
resistance and mitochondrial dysfunction, all of which are likely linked to enhanced neuromuscular activation and vascular
Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

function. The combination of good nutrition and an active lifestyle can minimize the declines in muscle mass, strength, and
physical performance that are observed in older people, all of which likely prevent or delay mobility limitations, disability,
Skeletal Muscle Aging and Exercise

3
institutionalization, and morbidity, translating into healthy aging. This illustration used elements from Servier Medical Art,
www.servier.com/Powerpoint-image-bank.
Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

G. Distefano and B.H. Goodpaster

of muscle loss then accelerates so that by the Interestingly, an improved single fiber contrac-
seventh and eighth decades of life about tile function was reported for oldest-old subjects
0.7% – 0.8% of lower limb muscles is reduced (87 –90 yr old) when compared to young sub-
per year in both men and women (Koster et al. jects, suggesting a compensatory mechanism
2011). The reduction in whole muscle mass is to the decrements in whole muscle function
mainly explained by the reduced number of my- (Grosicki et al. 2016). Impairments of muscle
ofibers, and to a lesser degree by a decrease in strength are likely due not only to decreases in
myofiber area (Lexell et al. 1988). During mus- muscle lean mass, but also a combination of
cle atrophy, signaling pathways that regulate factors that may include a decline in voluntary
muscle size are altered. Among the potential neural drive (Clark and Taylor 2011), impaired
factors underlying the age-related muscle atro- neuromuscular control such as lower motoneu-
phy, decreased mitochondrial capacity and in- ron firing rates (Connelly et al. 1999) and nerve
creased oxidative stress (Fulle et al. 2004; Mar- conduction velocities (Metter et al. 1998), in-
zetti et al. 2013), impaired satellite cell function creases in noncontractile adipose tissue (Good-
(Alway et al. 2014), as well as increased inflam- paster et al. 2008), and excitation – contraction
mation (Schaap et al. 2009) have received con- uncoupling (Payne and Delbono 2004; Delbono
siderable attention. The specific mechanisms 2011).
involved in the age-related loss of muscle mass The aging muscle undergoes several mor-
are the scope of several contemporary investiga- phological changes, which in turn may affect
tions (Peake et al. 2010; Bonaldo and Sandri muscle strength and physical performance.
2013; Sandri et al. 2013; Rudrappa et al. 2016), These muscular changes are likely linked to
yet remain incompletely understood, especially age-related changes in the central and peripheral
in humans. nervous systems, including a gradual loss of mo-
www.perspectivesinmedicine.org

Concomitant with muscle atrophy, muscle toneurons and degeneration of neuromuscular


strength declines with aging, and together they junctions (Gonzalez-Freire et al. 2014). With
define sarcopenia (Cruz-Jentoft et al. 2010). aging, denervation of fibers belonging to a single
Muscle strength significantly decreases after motor unit (usually fast) occurs (Lexell et al.
50 – 60 years of age (Murray et al. 1980; Lindle 1983). This denervation is followed by immedi-
et al. 1997; Lynch et al. 1999). The rates of decline ate reinnervation by the remaining motoneu-
are approximately 2% – 4% per year (Bassey and rons (usually slow). The reinnervation of muscle
Harries 1993; Frontera et al. 2000a; Goodpaster fibers by a different type of motoneuron results
et al. 2006; Delmonico et al. 2009), and are great- in fiber type conversion and fiber type grouping
er in lower limbs when compared to upper limbs (Lexell et al. 1986). With advancing aging, mo-
(Landers et al. 2001; Amaral et al. 2014). Inter- toneurons lose their regenerative capacity and
estingly, the loss of muscle strength is about some muscle fibers remain denervated resulting
three times greater than the rates of muscle at- in their ultimate death. Atrophy of type II my-
rophy (Goodpaster et al. 2006; Koster et al. ofibers (Lexell et al. 1988; Lexell and Taylor 1991;
2011). Therefore, the specific strength (i.e., Joseph et al. 2012), followed by an increased ac-
strength per unit of muscle) significantly de- cumulation of noncontractile components, in-
creases with aging, suggesting a decline in mus- cluding adipose and connective tissues, is also
cle quality (Goodpaster et al. 2006; Koster et al. observed (Alnaqeeb et al. 1984; Kent-Braun et al.
2011). The processes responsible for the loss of 2000). Furthermore, older subjects have impair-
muscle strength have not been elucidated. Some ments in the vascular system, including a com-
studies have shown an age-related deterioration promised central arterial compliance (Tanaka
in single fiber contractile function (Larsson et et al. 2000), endothelial wall function (Groen
al. 1997; Frontera et al. 2000b). One study of et al. 2014), and a reduced muscle capillary den-
young and old subjects matched for physical sity (Groen et al. 2014). These vascular changes
activity levels, however, did not observe these have the potential to further compromise skel-
impairments with aging (Trappe et al. 2003). etal muscle function by affecting the delivery of

4 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

Skeletal Muscle Aging and Exercise

oxygen, hormones, growth factors, nutrients, ments in muscle function have not been eluci-
and amino acids. dated. Resistance exercise acutely increases
No interventions likely completely prevent muscle protein synthesis out to 72 h (Miller et
the age-associated loss of muscle mass and al. 2005), and although older subjects have a
strength. However, secondary modifiable fac- reduced rate of muscle protein synthesis com-
tors have shown to play a key role in modulating pared to younger individuals, their proportion-
these changes. In this context, sarcopenia can al response to exercise is similar (Schulte and
present with increased body fat (obesity), a con- Yarasheski 2001). Exercise can also prevent the
dition known as sarcopenic obesity. The in- age-associated intermuscular adipose tissue in-
creased adiposity in older subjects negatively filtration (Goodpaster et al. 2008) and improve
impacts muscle function independent of the neural and vascular function of older subjects
loss of muscle mass (Koster et al. 2011). Long- (Nishimune et al. 2014; Messi et al. 2016;
term exposure to obesity has also been related Verdijk et al. 2016).
with poor handgrip strength later in life (Sten- The degree of improvements in muscle mass
holm et al. 2008). In addition, obesity-related and strength in response to resistance exercise
conditions such as inflammatory and endocrine have shown to vary according to the subject age
diseases, including type 2 diabetes, can poten- (Trappe et al. 2001; Raue et al. 2009). While
tiate the declines in muscle mass and strength similar increases in muscle mass were observed
(Park et al. 2007). Chronic low-grade inflam- in young and 74-yr-old women after resistance
mation is generally observed with aging, can exercise (Trappe et al. 2001), no improvements
be amplified by obesity (Schrager et al. 2007), in muscle mass was observed in octogenarian
and is associated with the age-related decreases women after training (Raue et al. 2009). How-
in muscle mass and strength (Schaap et al. ever, other reports of improvements in muscle
www.perspectivesinmedicine.org

2009). The mechanisms by which inflammation mass and strength observed after resistance
contributes to sarcopenia are not completely exercise in very old individuals (.85 yr old)
understood (Peake et al. 2010). suggests that skeletal muscle may partially re-
Physical inactivity is a key secondary factor tains the capacity to adapt to the mechanical
affecting muscle aging. Physical inactivity in- load (Fiatarone et al. 1990; Harridge et al.
duced by controlled bed rest induces several 1999). Improvements in single muscle fiber
harmful muscular adaptations, including re- contractile properties have also been observed
ductions in muscle volume and power, that in older individuals after both aerobic (Harber
are more severe in older than younger subjects et al. 2009) and resistance (Trappe et al. 2000)
(Pisot et al. 2016). These impairments were not exercise training, but these improvements have
completely restored in older individuals after a shown to be diminished in very older subjects
14-day recovery period that included nutrition- (Slivka et al. 2008; Raue et al. 2009). Despite the
al support and exercise (Pisot et al. 2016). Con- generally consistent response of muscle to exer-
versely, an active lifestyle has been reported to cise, the degree of response varies considerably.
attenuate sarcopenia and prevent body fat accu- While some studies have reported similar im-
mulation and inflammation (Safdar et al. 2010). provements among sexes (Tracy et al. 1999;
Resistance exercise promotes muscle hypertro- Leenders et al. 2013), others have reported
phy and improves strength and physical perfor- greater absolute and relative exercise-related im-
mance. This includes significant improvements provements in muscle mass and strength in men
or prevention of declines in myofiber (Bamman than in women (Ivey et al. 2000; Bamman et al.
et al. 2003) and whole muscle (Harridge et al. 2003; Da Boit et al. 2016). Although specific
1999) size, strength (Tracy et al. 1999; Bamman mechanisms have been proposed to explain
et al. 2003), muscle quality (Da Boit et al. the diminished response to exercise in aging
2016), and physical performance (Fiatarone (Mera et al. 2016), there are likely multiple fac-
et al. 1990; Da Boit et al. 2016). The underlying tors at play, which deserve more thorough sys-
mechanisms for exercise-induced improve- tematic investigation.

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785 5


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

G. Distefano and B.H. Goodpaster

Muscle Metabolism development of insulin resistance (Amati et al.


2011).
Insulin Sensitivity
Chronic exercise in older men and women
In addition to their essential role on mobility maintains high insulin sensitivity (Amati et al.
and physical performance, skeletal muscles play 2009; Amati et al. 2012) similar to that of young
a crucial role in whole-body metabolism. They endurance-trained athletes (Dube et al. 2016).
modulate blood glucose levels by insulin-medi- Together, these findings strongly support the
ated glucose uptake, and when this is impaired, idea that age per se is not the cause of skeletal
insulin resistance can lead to type 2 diabetes. muscle metabolic impairments, and that chron-
While there is a higher prevalence of type 2 ic exercise is a primary determinant of insulin
diabetes in older adults (Wild et al. 2004), the sensitivity. Indeed, one bout of exercise is suffi-
effects of aging on insulin resistance are less cient to induce acute improvements in muscle
clear. A number of studies have reported a de- glucose uptake and insulin sensitivity (Heath
cline in insulin sensitivity with aging (Rowe et et al. 1983). These improvements are still pres-
al. 1983; Fink et al. 1986). The causes of insulin ent up to 1– 2 days after the bout of exercise
resistance are complex and not fully under- (Mikines et al. 1988; Cartee et al. 1989; Naga-
stood. Among the factors that have shown to sawa et al. 1991). Importantly, the capacity of
play a role are impaired mitochondrial function exercise to improve insulin sensitivity and glu-
(Petersen et al. 2003), increased oxidative stress cose uptake is maintained at old age (Dube et al.
(Anderson et al. 2009), increased inflammation 2008; Bienso et al. 2015). Additionally, a 5-year
(Shoelson et al. 2006), and lipotoxicity (Amati longitudinal study performed in older subjects
2012). This of course raises questions about the (70 –79 yr old) showed that maintaining mod-
primary versus secondary effects of aging on est physical activity through walking dimin-
www.perspectivesinmedicine.org

insulin resistance. Indeed, studies indicate ished the odds of developing or worsening met-
that increasing age per se is not a major deter- abolic syndrome, including insulin sensitivity
minant of insulin sensitivity (Lalia et al. 2016), (Peterson et al. 2010). Although the specific
but rather obesity, body fat distribution, and mechanisms by which exercise improves insulin
physical inactivity much more profoundly in- sensitivity are not fully understood, it is clear
fluence insulin sensitivity (Lanza et al. 2008; that many of the myocellular factors implicated
Amati et al. 2009, 2012; Karakelides et al. in aging and insulin resistance are also affected
2010). Both young and older obese subjects by exercise.
have lower insulin sensitivity when compared
with lean individuals, independent of age
Mitochondrial Capacity
(Karakelides et al. 2010). Additionally, no dif-
ference in insulin sensitivity is observed be- Mitochondria are essential organelles for prop-
tween highly trained young and adults (Lanza er cellular function and play a key role in skeletal
et al. 2008). These findings support the concept muscle bioenergetics. The effect of aging on
that changes in insulin sensitivity with chrono- skeletal muscle mitochondria has been exten-
logical aging are likely secondary to changes in sively investigated for several decades, but the
body fat and physical activity. In support of results are contradictory. A substantial number
this, the variation in body fat distribution of animal and human studies have reported de-
among older subjects is associated with their creases in mitochondrial content with chrono-
inflammatory profile (Koster et al. 2010). Ad- logical aging, expressed by a reduced number,
ditionally, although high levels of intramyocel- density, or size of mitochondria (Orlander et al.
lular lipid (IMCL) content are observed in both 1978; Conley et al. 2000; Crane et al. 2010), and
athletes and in insulin resistance subjects with decreased mitochondrial DNA and protein ex-
type 2 diabetes (Goodpaster et al. 2001), spe- pression (Rooyackers et al. 1996; Short et al.
cific lipids in muscle (i.e., diacylglycerols and 2005; Lanza et al. 2008). Functional declines
ceramides), likely play a significant role in the have also been reported including reductions

6 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

Skeletal Muscle Aging and Exercise

in ATP production (Drew et al. 2003; Short et al. mitochondrial fusion and fission (Bori et al.
2005; Mansouri et al. 2006; Lanza et al. 2008), 2012; Konopka et al. 2014), others have de-
mitochondrial respiration (Trounce et al. 1989; scribed both an increased fission (Iqbal et al.
Kerner et al. 2001; Tonkonogi et al. 2003; Ku- 2013), or increased fusion (Leduc-Gaudet
maran et al. 2005), mitochondrial enzymatic et al. 2015) in older muscles.
activities (Trounce et al. 1989; Boffoli et al. The contradictory associations between ag-
1994; Proctor et al. 1995; Rooyackers et al. ing and mitochondria could be because of dif-
1996; Houmard et al. 1998; Tonkonogi et ferences in study methodology. Several diverse
al. 2003; Lanza et al. 2008; Crane et al. 2010), measures can be used to assess mitochondrial
and increased reactive oxygen species (ROS) content and function (Lanza and Nair 2010;
production (Mansouri et al. 2006; Chabi et al. Hepple 2014), and it is possible that aging
2008). Additionally, in vivo human studies have does not affect all mitochondrial characteristics
shown reduced maximal ATP flux with aging in equally. Additionally, studies performed in iso-
the gastrocnemius (McCully et al. 1993), vastus lated mitochondria, a method that does not
lateralis (Conley et al. 2000; Larsen et al. 2012), preserve the complex structural arrangement
and soleus (Petersen et al. 2003) muscles. of mitochondria, have been reported to exagger-
Despite a number of studies describing age- ate functional age-related impairments when
related changes in mitochondrial capacity, sev- compared to permeabilized myofibers (Picard
eral animal and human studies have reported no et al. 2010). In addition to study methodology,
age-related trends related to mitochondrial con- factors associated with secondary aging such as
tent (Rasmussen et al. 2003; Gouspillou et al. body fat and physical activity levels influence
2014), enzymatic activity (Brierley et al. 1996; skeletal muscle mitochondrial capacity and the
Chabi et al. 2008), ATP synthesis (Barrientos expression of mitochondrial fission and fusion
www.perspectivesinmedicine.org

et al. 1996; Rasmussen et al. 2003), mitochon- proteins (Hutter et al. 2007; Distefano et al.
drial respiration (Barrientos et al. 1996; Kerner 2016). Obese subjects have displayed reduced
et al. 2001; Hutter et al. 2007; Chabi et al. 2008; capacity for lipid oxidation, and lowered activ-
Gouspillou et al. 2014; Distefano et al. 2016), ity of mitochondrial enzymes (Kim et al. 2000;
activity of electron transport chain complexes Thyfault et al. 2004). Similarly, decreased phys-
(Barrientos et al. 1996; Brierley et al. 1996; Ras- ical activity can adversely affect mitochondrial
mussen et al. 2003), and ROS production (Drew capacity (Booth and Holloszy 1977; Ringholm
et al. 2003; Tonkonogi et al. 2003; Gouspillou et al. 2011).
et al. 2014). Furthermore, in vivo studies have The ability of exercise to increase mito-
failed to show changes in maximal ATP flux with chondrial content and function is well docu-
aging (Chretien et al. 1998; Kent-Braun and Ng mented (Holloszy et al. 1970; Dohm et al.
2000; Lanza et al. 2005, 2007). 1973). Exercise training, including endurance
In addition to mitochondrial content and and resistance exercise, stimulates mitochondri-
function, recent studies have highlighted the al biogenesis through increases in the peroxi-
importance of mitochondrial morphology and some proliferator-activated receptor g coactiva-
turnover for proper mitochondrial function tor 1a (PGC-1a) (Baar et al. 2002; Geng et al.
(Detmer and Chan 2007), Briefly, mitochondria 2010). Furthermore, recent studies have sug-
integrity relies on the efficiency of quality con- gested that exercise can improve function/effi-
trol processes, and their morphology are regu- ciency of mitochondria through remodeling of
lated by continuous fusion, fission, and mito- the mitochondrial network (fusion, fission, and
phagy (Ono et al. 2001; Twig et al. 2008a,b). autophagy) (Cartoni et al. 2005; Ding et al.
Limited animal and human studies to date 2010; Perry et al. 2010; Smuder et al. 2011).
have examined mitochondrial quality control Several studies have shown that mitochondrial
processes in muscle aging, but the results are function is not affected by chronological aging,
inconsistent and need to be further investigated. but rather by decreases in physical activity that
While some have found no age-related trends in normally occurs with aging (Barrientos et al.

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785 7


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

G. Distefano and B.H. Goodpaster

1996; Brierley et al. 1996). No differences in mi- Romanello et al. 2010), but the exact role of
tochondrial content and respiration was ob- these processes in regulating muscle mass still
served between young and old subjects matched remains poorly understood in humans. Recent
for physical activity, both engaged in moderate- evidence has also shown that lower mitochon-
to vigorous-intensity exercise training (Gous- drial capacity and efficiency is associated with
pillou et al. 2014). Studies that included a group reduced physical performance in older adults
of older trained subjects have been performed in (Coen et al. 2013). Similarly, high-functioning
an attempt to investigate whether maintenance elderly individuals have been shown to main-
of physical activity levels during aging can pre- tain muscle mass and mitochondrial capacity,
vent decreases in mitochondrial capacity. Mito- whereas low-functioning elderly individuals
chondrial content, biogenesis, electron transfer show decreased muscle mass and mitochondrial
chain function, and antioxidant capacity is function in comparison to young individuals
preserved in skeletal muscle of active older in- (Joseph et al. 2012). These results suggest a po-
dividuals (Safdar et al. 2010). Similarly, muscle tential role for mitochondria in sarcopenia and
biopsies from well-trained seniors who exer- physical function in aging.
cised regularly in the previous 30 years showed
that lifelong physical exercise delays age-associ-
Muscle Regenerative Capacity
ated skeletal muscle declines (Zampieri et al.
2014). These well-trained seniors presented bet- Skeletal muscle has an amazing capacity for
ter mitochondria organization, including pre- regeneration that relies on resident stem cells,
served fiber morphology and ultrastructure of also identified as satellite cells. In response to
intracellular organelles involved in calcium han- muscle injury or stress, quiescent satellite cells
dling and ATP production, and lowered expres- are activated, proliferate, and differentiate into
www.perspectivesinmedicine.org

sion of genes related to autophagy and ROS in a myogenic lineage to endure regeneration or
comparison with health-matched sedentary se- muscle growth (Yin et al. 2013). Some activated
niors. Likewise, age-related declines in oxidative satellite cells self-renew and return to quies-
capacity (Proctor et al. 1995), mitochondrial cence to maintain the satellite cell pool (Yin
ATP production (Lanza et al. 2008), and citrate et al. 2013). Impairments in any phase of the
synthase activity (Lanza et al. 2008) have been satellite cell cycle may result in a deficient
observed in sedentary subjects, but not in en- muscle regeneration that can lead to detriments
durance-trained subjects. in muscle contractile function. Aged skeletal
Recent evidence has also suggested that mi- muscle has previously been shown to possess
tochondria may play a key role in sarcopenia. An a diminished (Joanisse et al. 2016) or delayed
imbalance between mitochondrial fusion and regeneration (Shavlakadze et al. 2010), and an
fission and an impaired mitochondrial turnover increased potential for fibrosis formation after
resulting from insufficient biogenesis and/or injury (Brack et al. 2007). A reduced number
defective autophagic removal of dysfunctional and impaired function of satellite cells is also
mitochondria are all factors that may be in- observed (Brack et al. 2005; Chakkalakal et al.
volved in the loss of muscle mass during aging. 2012; Sousa-Victor et al. 2014). Additionally,
Specifically, mitochondrial dysfunction and in- aged muscle has attenuated regrowth following
creased ROS production stimulates catabolic atrophy-inducing events when compared to
signaling pathways and muscle atrophy by acti- younger muscles (Pisot et al. 2016), but the
vating the two major proteolytic systems: the specific mechanisms responsible for the loss
ubiquitin proteasome and the autophagy lyso- of growth capacity are unknown. Due to their
some (Sandri et al. 2004; Tong et al. 2009). Sev- essential role in muscle regeneration and
eral in vitro and animal studies have provided growth, several studies have investigated wheth-
information on the molecular pathways in- er an impaired satellite cell potential would lead
volved in these processes (Sandri et al. 2004; to sarcopenia. Although some have supported
Mammucari et al. 2007; Masiero et al. 2009; this hypothesis (Verdijk et al. 2007), strong re-

8 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

Skeletal Muscle Aging and Exercise

cent evidence has failed to support these asso- shown to possess a similar density of satellite
ciations (Fry et al. 2015). cells in type I and II myofibers despite a decrease
Recent discoveries have revealed potential in myofiber distribution and cross-sectional
molecular and cellular mechanisms responsible area (Mackey et al. 2014). Additionally, im-
for the age-related impairments in muscle re- proved muscle regeneration in trained mice
generation. Satellite cell optimal function relies has been observed with a concomitant improve-
on the appropriate support from the systemic ment in vascularization and inflammatory re-
environment (circulation), local microenviron- sponse (Joanisse et al. 2016).
ment (niche), as well as their intrinsic capacity
(Conboy et al. 2005; Yin et al. 2013; Sousa-Vic-
tor et al. 2014). Heterochronic parabiosis stud- CONCLUDING REMARKS AND FUTURE
ies, in which young and old animals join the PERSPECTIVES
same circulatory system, highlight the influence The isolated impact of aging on skeletal muscle
of the systemic environment on satellite cell is difficult to disentangle from the many other
function demonstrating an improved regenera- factors that change concurrently with aging,
tive capacity of old satellite cells when exposed including decreased moderate- to vigorous-in-
to a young environment (Conboy et al. 2005). tensity physical activity and increased sedentary
Proper stem cell function has also been shown behavior, as well as augmented adiposity. On
to depend on growth factors, trophic factors, one hand, no single intervention can complete-
and cytokines from the surrounding myofiber ly prevent the age-related loss of muscle mass,
microenvironment (Jasper and Kennedy 2012). strength, and regenerative capacity. On the oth-
Recently, it has been shown that, in addition to er, exercise and physical activity can signifi-
extrinsic factors, intrinsic satellite cell changes
www.perspectivesinmedicine.org

cantly attenuate, or in some cases prevent, these


are also responsible for the age-related defective declines in muscle metabolism and function.
regenerative capacity (Sousa-Victor et al. 2014). Skeletal muscle partially retains its plasticity
These alterations were present in satellite cells in response to exercise with aging, providing
from very old animals, and were not rejuvenated compelling evidence that many of the negative
by a youthful environment (Sousa-Victor et al. age-associated changes in muscle function and
2014). metabolism are caused by lifestyle changes
In addition to chronological aging, other secondary to aging, most notably physical inac-
secondary factors have been shown to influence tivity. The fields of muscle aging and exercise
muscle regenerative capacity. An impaired mus- physiology have synergized to provide impor-
cle regeneration after injury is observed in obese tant insights into primary effects of aging on
mice when compared to normal weight controls muscle, and which age-associated changes can
(Nguyen et al. 2011; Fu et al. 2016), including a be attenuated or prevented by exercise. We need
decreased expression of myogenic genes, num- to better understand, however, which specific
ber of newly formed regenerated fibers, and sat- responses to exercise differ in older subjects,
ellite cell pool (Fu et al. 2016). The increased the underlying mechanism by which aging
inflammation that occurs with obesity has also may affect acute and chronic exercise responses,
been shown to contribute to these impairments and the variation in the individual response to
(Brown et al. 2015). Physical activity has been exercise. Moreover, the field of muscle aging
shown to positively affect the regenerative ca- needs to link the myocellular responses to exer-
pacity of older muscles. Both resistance and en- cise to many of its health benefits.
durance exercise training ranging from few
weeks to months have shown to increase the
number of satellite cells in old animals and sub- REFERENCES
jects (Roth et al. 2001; Verdijk et al. 2009;
Alnaqeeb MA, Al Zaid NS, Goldspink G. 1984. Connective
Leenders et al. 2013; Joanisse et al. 2016). Im- tissue changes and physical properties of developing and
portantly, lifelong endurance runners have been ageing skeletal muscle. J Anat 139: 677 –689.

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785 9


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

G. Distefano and B.H. Goodpaster

Alway SE, Myers MJ, Mohamed JS. 2014. Regulation of sat- Booth FW, Laye MJ, Roberts MD. 2011. Lifetime sedentary
ellite cell function in sarcopenia. Front Aging Neurosci 6: living accelerates some aspects of secondary aging. J Appl
246. Physiol (1985) 111: 1497– 1504.
Amaral JF, Alvim FC, Castro EA, Doimo LA, Silva MV, Novo Bori Z, Zhao Z, Koltai E, Fatouros IG, Jamurtas AZ, Dour-
Junior JM. 2014. Influence of aging on isometric muscle oudos II, Terzis G, Chatzinikolaou A, Sovatzidis A, Dra-
strength, fat-free mass and electromyographic signal ganidis D, et al. 2012. The effects of aging, physical train-
power of the upper and lower limbs in women. Braz J ing, and a single bout of exercise on mitochondrial
Phys Ther 18: 183– 190. protein expression in human skeletal muscle. Exp Geron-
Amati F. 2012. Revisiting the diacylglycerol-induced insulin tol 47: 417– 424.
resistance hypothesis. Obes Rev 13: 40–50. Brack AS, Bildsoe H, Hughes SM. 2005. Evidence that sat-
Amati F, Dube JJ, Coen PM, Stefanovic-Racic M, Toledo FG, ellite cell decrement contributes to preferential decline in
Goodpaster BH. 2009. Physical inactivity and obesity nuclear number from large fibres during murine age-
underlie the insulin resistance of aging. Diabetes Care related muscle atrophy. J Cell Sci 118: 4813–4821.
32: 1547–1549. Brack AS, Conboy MJ, Roy S, Lee M, Kuo CJ, Keller C,
Amati F, Dube JJ, Alvarez-Carnero E, Edreira MM, Cho- Rando TA. 2007. Increased Wnt signaling during aging
mentowski P, Coen PM, Switzer GE, Bickel PE, Stefa- alters muscle stem cell fate and increases fibrosis. Science
novic-Racic M, Toledo FG, et al. 2011. Skeletal muscle 317: 807–810.
triglycerides, diacylglycerols, and ceramides in insulin Brierley EJ, Johnson MA, James OF, Turnbull DM. 1996.
resistance: Another paradox in endurance-trained ath- Effects of physical activity and age on mitochondrial
letes? Diabetes 60: 2588– 2597. function. QJM 89: 251–258.
Amati F, Pennant M, Azuma K, Dube JJ, Toledo FG, Rossi Brown LA, Lee DE, Patton JF, Perry RAJr, Brown JL, Baum JI,
AP, Kelley DE, Goodpaster BH. 2012. Lower thigh sub- Smith-Blair N, Greene NP, Washington TA. 2015. Diet-
cutaneous and higher visceral abdominal adipose tissue induced obesity alters anabolic signalling in mice at the
content both contribute to insulin resistance. Obesity 20: onset of skeletal muscle regeneration. Acta Physiol 215:
1115– 1117. 46– 57.
Anderson EJ, Lustig ME, Boyle KE, Woodlief TL, Kane DA, Busse EW. 1969. Theories of aging. In Behavior and adapta-
Lin CT, Price JWIII, Kang L, Rabinovitch PS, Szeto HH, tion in late life (ed. Busse EW, Pfeiffer E). Little, Brown,
et al. 2009. Mitochondrial H2O2 emission and cellular Boston, MA.
www.perspectivesinmedicine.org

redox state link excess fat intake to insulin resistance in Cartee GD, Young DA, Sleeper MD, Zierath J, Wallberg-
both rodents and humans. J Clin Invest 119: 573 –581. Henriksson H, Holloszy JO. 1989. Prolonged increase
Baar K, Wende AR, Jones TE, Marison M, Nolte LA, Chen in insulin-stimulated glucose transport in muscle after
M, Kelly DP, Holloszy JO. 2002. Adaptations of skeletal exercise. Am J Physiol 256: E494– E499.
muscle to exercise: Rapid increase in the transcriptional Cartoni R, Leger B, Hock MB, Praz M, Crettenand A, Pich S,
coactivator PGC-1. FASEB J 16: 1879–1886. Ziltener JL, Luthi F, Deriaz O, Zorzano A, et al. 2005.
Bamman MM, Hill VJ, Adams GR, Haddad F, Wetzstein CJ, Mitofusins 1/2 and ERRa expression are increased in
Gower BA, Ahmed A, Hunter GR. 2003. Gender differ- human skeletal muscle after physical exercise. J Physiol
ences in resistance-training-induced myofiber hypertro- 567: 349–358.
phy among older adults. J Gerontol A Biol Sci Med Sci 58: Chabi B, Ljubicic V, Menzies KJ, Huang JH, Saleem A, Hood
108–116. DA. 2008. Mitochondrial function and apoptotic sus-
Barrientos A, Casademont J, Rotig A, Miro O, Urbano-Mar- ceptibility in aging skeletal muscle. Aging Cell 7: 2 –12.
quez A, Rustin P, Cardellach F. 1996. Absence of relation- Chakkalakal JV, Jones KM, Basson MA, Brack AS. 2012. The
ship between the level of electron transport chain activ- aged niche disrupts muscle stem cell quiescence. Nature
ities and aging in human skeletal muscle. Biochem 490: 355–360.
Biophys Res Commun 229: 536–539. Chretien D, Gallego J, Barrientos A, Casademont J, Cardel-
Bassey EJ, Harries UJ. 1993. Normal values for handgrip lach F, Munnich A, Rotig A, Rustin P. 1998. Biochemical
strength in 920 men and women aged over 65 years, parameters for the diagnosis of mitochondrial respirato-
and longitudinal changes over 4 years in 620 survivors. ry chain deficiency in humans, and their lack of age-
Clin Sci (Lond) 84: 331–337. related changes. Biochem J 329: 249–254.
Bienso RS, Olesen J, Gliemann L, Schmidt JF, Matzen MS, Clark BC, Taylor JL. 2011. Age-related changes in motor
Wojtaszewski JF, Hellsten Y, Pilegaard H. 2015. Effects of cortical properties and voluntary activation of skeletal
exercise training on regulation of skeletal muscle glucose muscle. Curr Aging Sci 4: 192–199.
metabolism in elderly men. J Gerontol A Biol Sci Med Sci
Coen PM, Jubrias SA, Distefano G, Amati F, Mackey DC,
70: 866 –872. Glynn NW, Manini TM, Wohlgemuth SE, Leeuwenburgh
Boffoli D, Scacco SC, Vergari R, Solarino G, Santacroce G, C, Cummings SR, et al. 2013. Skeletal muscle mitochon-
Papa S. 1994. Decline with age of the respiratory chain drial energetics are associated with maximal aerobic ca-
activity in human skeletal muscle. Biochim Biophys Acta pacity and walking speed in older adults. J Gerontol A Biol
1226: 73–82. Sci Med Sci 68: 447–455.
Bonaldo P, Sandri M. 2013. Cellular and molecular mecha- Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman
nisms of muscle atrophy. Dis Model Mech 6: 25–39. IL, Rando TA. 2005. Rejuvenation of aged progenitor cells
Booth FW, Holloszy JO. 1977. Cytochrome c turnover in rat by exposure to a young systemic environment. Nature
skeletal muscles. J Biol Chem 252: 416– 419. 433: 760–764.

10 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

Skeletal Muscle Aging and Exercise

Conley KE, Jubrias SA, Esselman PC. 2000. Oxidative ca- acteristics and substrate energetics in lifelong endurance
pacity and ageing in human muscle. J Physiol 526: 203 – athletes. Med Sci Sports Exerc 48: 472– 480.
210. Fiatarone MA, Marks EC, Ryan ND, Meredith CN, Lipsitz
Connelly DM, Rice CL, Roos MR, Vandervoort AA. 1999. LA, Evans WJ. 1990. High-intensity strength training in
Motor unit firing rates and contractile properties in ti- nonagenarians. Effects on skeletal muscle. JAMA 263:
bialis anterior of young and old men. J Appl Physiol 3029–3034.
(1985) 87: 843– 852. Fink RI, Wallace P, Olefsky JM. 1986. Effects of aging on
Consitt LA, Van Meter J, Newton CA, Collier DN, Dar MS, glucose-mediated glucose disposal and glucose trans-
Wojtaszewski JF, Treebak JT, Tanner CJ, Houmard JA. port. J Clin Invest 77: 2034–2041.
2013. Impairments in site-specific AS160 phosphoryla- Forsberg AM, Nilsson E, Werneman J, Bergstrom J, Hult-
tion and effects of exercise training. Diabetes 62: 3437– man E. 1991. Muscle composition in relation to age and
3447. sex. Clin Sci 81: 249–256.
Crane JD, Devries MC, Safdar A, Hamadeh MJ, Tarnopolsky Frontera WR, Hughes VA, Fielding RA, Fiatarone MA, Evans
MA. 2010. The effect of aging on human skeletal muscle WJ, Roubenoff R. 2000a. Aging of skeletal muscle: A 12-
mitochondrial and intramyocellular lipid ultrastructure.
yr longitudinal study. J Appl Physiol (1985) 88: 1321–
J Gerontol A Biol Sci Med Sci 65: 119–128.
1326.
Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm
Frontera WR, Suh D, Krivickas LS, Hughes VA, Goldstein R,
T, Landi F, Martin FC, Michel JP, Rolland Y, Schneider
Roubenoff R. 2000b. Skeletal muscle fiber quality in older
SM, et al. 2010. Sarcopenia: European consensus on def-
men and women. Am J Physiol Cell Physiol 279: C611–
inition and diagnosis: Report of the European Working
C618.
Group on Sarcopenia in Older People. Age Ageing 39:
412–423. Fry CS, Lee JD, Mula J, Kirby TJ, Jackson JR, Liu F, Yang L,
Da Boit M, Sibson R, Meakin JR, Aspden RM, Thies F, Mendias CL, Dupont-Versteegden EE, McCarthy JJ, et al.
Mangoni AA, Gray SR. 2016. Sex differences in the re- 2015. Inducible depletion of satellite cells in adult, sed-
sponse to resistance exercise training in older people. entary mice impairs muscle regenerative capacity with-
Physiol Rep 4: e12834. out affecting sarcopenia. Nat Med 21: 76– 80.
Delbono O. 2011. Expression and regulation of excitation- Fu X, Zhu M, Zhang S, Foretz M, Viollet B, Du M. 2016.
contraction coupling proteins in aging skeletal muscle. Obesity impairs skeletal muscle regeneration through in-
Curr Aging Sci 4: 248 –259. hibition of AMPK. Diabetes 65: 188–200.
www.perspectivesinmedicine.org

Delmonico MJ, Harris TB, Visser M, Park SW, Conroy MB, Fulle S, Protasi F, Di Tano G, Pietrangelo T, Beltramin A,
Velasquez-Mieyer P, Boudreau R, Manini TM, Nevitt M, Boncompagni S, Vecchiet L, Fano G. 2004. The contri-
Newman AB, et al. 2009. Longitudinal study of muscle bution of reactive oxygen species to sarcopenia and mus-
strength, quality, and adipose tissue infiltration. Am J cle ageing. Exp Gerontol 39: 17–24.
Clin Nutr 90: 1579– 1585. Geng T, Li P, Okutsu M, Yin X, Kwek J, Zhang M, Yan Z.
Detmer SA, Chan DC. 2007. Functions and dysfunctions of 2010. PGC-1a plays a functional role in exercise-induced
mitochondrial dynamics. Nat Rev Mol Cell Biol 8: 870 – mitochondrial biogenesis and angiogenesis but not fiber-
879. type transformation in mouse skeletal muscle. Am J Phys-
Ding H, Jiang N, Liu H, Liu X, Liu D, Zhao F, Wen L, Liu S, Ji iol Cell Physiol 298: C572–C579.
LL, Zhang Y. 2010. Response of mitochondrial fusion and Gonzalez-Freire M, de Cabo R, Studenski SA, Ferrucci L.
fission protein gene expression to exercise in rat skeletal 2014. The neuromuscular junction: Aging at the cross-
muscle. Biochim Biophys Acta 1800: 250– 256. road between nerves and muscle. Front Aging Neurosci 6:
Distefano G, Standley RA, Dube JJ, Carnero EA, Ritov VB, 208.
Stefanovic-Racic M, Toledo FG, Piva SR, Goodpaster BH, Goodpaster BH, He J, Watkins S, Kelley DE. 2001. Skeletal
Coen PM. 2016. Chronological age does not influence muscle lipid content and insulin resistance: Evidence for
ex-vivo mitochondrial respiration and quality control a paradox in endurance-trained athletes. J Clin Endocri-
in skeletal muscle. J Gerontol A Biol Sci Med Sci doi: nol Metab 86: 5755–5761.
10.1093/gerona/glw102. Goodpaster BH, Park SW, Harris TB, Kritchevsky SB, Nevitt
Dohm GL, Huston RL, Askew EW, Fleshood HL. 1973. Ef- M, Schwartz AV, Simonsick EM, Tylavsky FA, Visser M,
fects of exercise, training, and diet on muscle citric acid Newman AB. 2006. The loss of skeletal muscle strength,
cycle enzyme activity. Can J Biochem 51: 849– 854. mass, and quality in older adults: The health, aging and
Drew B, Phaneuf S, Dirks A, Selman C, Gredilla R, Lezza A, body composition study. J Gerontol A Biol Sci Med Sci 61:
Barja G, Leeuwenburgh C. 2003. Effects of aging and 1059–1064.
caloric restriction on mitochondrial energy production Goodpaster BH, Chomentowski P, Ward BK, Rossi A, Glynn
in gastrocnemius muscle and heart. Am J Physiol Regul NW, Delmonico MJ, Kritchevsky SB, Pahor M, Newman
Integr Comp Physiol 284: R474– R480. AB. 2008. Effects of physical activity on strength and
Dube JJ, Amati F, Stefanovic-Racic M, Toledo FG, Sauers SE, skeletal muscle fat infiltration in older adults: A random-
Goodpaster BH. 2008. Exercise-induced alterations in ized controlled trial. J Appl Physiol (1985) 105: 1498–
intramyocellular lipids and insulin resistance: The ath- 1503.
lete’s paradox revisited. Am J Physiol Endocrinol Metab Gouspillou G, Sgarioto N, Kapchinsky S, Purves-Smith F,
294: E882–E888. Norris B, Pion CH, Barbat-Artigas S, Lemieux F, Taivas-
Dube JJ, Broskey NT, Despines AA, Stefanovic-Racic M, salo T, Morais JA, et al. 2014. Increased sensitivity to
Toledo FG, Goodpaster BH, Amati F. 2016. Muscle char- mitochondrial permeability transition and myonuclear

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785 11


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

G. Distefano and B.H. Goodpaster

translocation of endonuclease G in atrophied muscle of Karakelides H, Irving BA, Short KR, O’Brien P, Nair KS.
physically active older humans. FASEB J 28: 1621– 1633. 2010. Age, obesity, and sex effects on insulin sensitivity
Groen BB, Hamer HM, Snijders T, van Kranenburg J, Frijns and skeletal muscle mitochondrial function. Diabetes 59:
D, Vink H, van Loon LJ. 2014. Skeletal muscle capillary 89– 97.
density and microvascular function are compromised Kent-Braun JA, Ng AV. 2000. Skeletal muscle oxidative ca-
with aging and type 2 diabetes. J Appl Physiol (1985) pacity in young and older women and men. J Appl Physiol
116: 998– 1005. (1985) 89: 1072– 1078.
Grosicki GJ, Standley R, Murach KA, Raue U, Minchev K, Kent-Braun JA, Ng AV, Young K. 2000. Skeletal muscle con-
Coen P, Newman AB, Cummings S, Harris T, Kritchevsky tractile and noncontractile components in young and
S, et al. 2016. Improved single muscle fiber quality in the older women and men. J Appl Physiol (1985) 88: 662–
oldest-old. J Appl Physiol (1985) 121: 878 –884. 668.
Harber MP, Konopka AR, Douglass MD, Minchev K, Ka- Kerner J, Turkaly PJ, Minkler PE, Hoppel CL. 2001. Aging
minsky LA, Trappe TA, Trappe S. 2009. Aerobic exercise skeletal muscle mitochondria in the rat: Decreased un-
training improves whole muscle and single myofiber size coupling protein-3 content. Am J Physiol Endocrinol
Metab 281: E1054– E1062.
and function in older women. Am J Physiol Regul Integr
Comp Physiol 297: R1452– R1459. Kim JY, Hickner RC, Cortright RL, Dohm GL, Houmard JA.
2000. Lipid oxidation is reduced in obese human skeletal
Harridge SD, Kryger A, Stensgaard A. 1999. Knee extensor
muscle. Am J Physiol Endocrinol Metab 279: E1039–
strength, activation, and size in very elderly people fol- E1044.
lowing strength training. Muscle Nerve 22: 831 –839.
Konopka AR, Suer MK, Wolff CA, Harber MP. 2014. Mark-
Heath GW, Gavin JRIII, Hinderliter JM, Hagberg JM, ers of human skeletal muscle mitochondrial biogenesis
Bloomfield SA, Holloszy JO. 1983. Effects of exercise and quality control: Effects of age and aerobic exercise
and lack of exercise on glucose tolerance and insulin sen- training. J Gerontol A Biol Sci Med Sci 69: 371– 378.
sitivity. J Appl Physiol Respir Environ Exerc Physiol 55:
Koster A, Stenholm S, Alley DE, Kim LJ, Simonsick EM,
512–517.
Kanaya AM, Visser M, Houston DK, Nicklas BJ, Tylavsky
Hepple RT. 2014. Mitochondrial involvement and impact in FA, et al. 2010. Body fat distribution and inflammation
aging skeletal muscle. Front Aging Neurosci 6: 211. among obese older adults with and without metabolic
Holloszy JO, Oscai LB, Don IJ, Mole PA. 1970. Mitochon- syndrome. Obesity 18: 2354– 2361.
www.perspectivesinmedicine.org

drial citric acid cycle and related enzymes: Adaptive re- Koster A, Ding J, Stenholm S, Caserotti P, Houston DK,
sponse to exercise. Biochem Biophys Res Commun 40: Nicklas BJ, You T, Lee JS, Visser M, Newman AB, et al.
1368– 1373. 2011. Does the amount of fat mass predict age-related
Houmard JA, Weidner ML, Gavigan KE, Tyndall GL, Hickey loss of lean mass, muscle strength, and muscle quality in
MS, Alshami A. 1998. Fiber type and citrate synthase older adults? J Gerontol A Biol Sci Med Sci 66: 888 –895.
activity in the human gastrocnemius and vastus lateralis Kumaran S, Panneerselvam KS, Shila S, Sivarajan K, Pan-
with aging. J Appl Physiol (1985) 85: 1337– 1341. neerselvam C. 2005. Age-associated deficit of mitochon-
Hutter E, Skovbro M, Lener B, Prats C, Rabol R, Dela F, drial oxidative phosphorylation in skeletal muscle: Role
Jansen-Durr P. 2007. Oxidative stress and mitochondrial of carnitine and lipoic acid. Mol Cell Biochem 280: 83–89.
impairment can be separated from lipofuscin accumula- Kyu HH, Bachman VF, Alexander LT, Mumford JE, Afshin A,
tion in aged human skeletal muscle. Aging Cell 6: 245 – Estep K, Veerman JL, Delwiche K, Iannarone ML, Moyer
256. ML, et al. 2016. Physical activity and risk of breast cancer,
colon cancer, diabetes, ischemic heart disease, and ische-
Iqbal S, Ostojic O, Singh K, Joseph AM, Hood DA. 2013.
mic stroke events: Systematic review and dose-response
Expression of mitochondrial fission and fusion regulato- meta-analysis for the Global Burden of Disease Study
ry proteins in skeletal muscle during chronic use and 2013. BMJ 354: i3857.
disuse. Muscle Nerve 48: 963–970.
Lalia AZ, Dasari S, Johnson ML, Robinson MM, Konopka
Ivey FM, Roth SM, Ferrell RE, Tracy BL, Lemmer JT, Hurlbut AR, Distelmaier K, Port JD, Glavin MT, Esponda RR, Nair
DE, Martel GF, Siegel EL, Fozard JL, Jeffrey Metter E, et al. KS, et al. 2016. Predictors of whole-body insulin sensi-
2000. Effects of age, gender, and myostatin genotype on tivity across ages and adiposity in adult humans. J Clin
the hypertrophic response to heavy resistance strength Endocrinol Metab 101: 626– 634.
training. J Gerontol A Biol Sci Med Sci 55: M641–M648. Landers KA, Hunter GR, Wetzstein CJ, Bamman MM,
Jasper H, Kennedy BK. 2012. Niche science: The aging stem Weinsier RL. 2001. The interrelationship among muscle
cell. Cell Cycle 11: 2959–2960. mass, strength, and the ability to perform physical tasks
Joanisse S, Nederveen JP, Baker JM, Snijders T, Iacono C, of daily living in younger and older women. J Gerontol A
Parise G. 2016. Exercise conditioning in old mice im- Biol Sci Med Sci 56: B443– B448.
proves skeletal muscle regeneration. FASEB J 30: 3256– Lanza IR, Nair KS. 2010. Mitochondrial metabolic function
3268. assessed in vivo and in vitro. Curr Opin Clin Nutr Metab
Joseph AM, Adhihetty PJ, Buford TW, Wohlgemuth SE, Lees Care 13: 511– 517.
HA, Nguyen LM, Aranda JM, Sandesara BD, Pahor M, Lanza IR, Befroy DE, Kent-Braun JA. 2005. Age-related
Manini TM, et al. 2012. The impact of aging on mito- changes in ATP-producing pathways in human skeletal
chondrial function and biogenesis pathways in skeletal muscle in vivo. J Appl Physiol (1985) 99: 1736–1744.
muscle of sedentary high- and low-functioning elderly Lanza IR, Larsen RG, Kent-Braun JA. 2007. Effects of old age
individuals. Aging Cell 11: 801– 809. on human skeletal muscle energetics during fatiguing

12 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

Skeletal Muscle Aging and Exercise

contractions with and without blood flow. J Physiol 583: peroxide release and oxidative damage in mouse hind-
1093– 1105. limb skeletal muscle during aging. Mech Ageing Dev 127:
Lanza IR, Short DK, Short KR, Raghavakaimal S, Basu R, 298 –306.
Joyner MJ, McConnell JP, Nair KS. 2008. Endurance ex- Marzetti E, Calvani R, Cesari M, Buford TW, Lorenzi M,
ercise as a countermeasure for aging. Diabetes 57: 2933– Behnke BJ, Leeuwenburgh C. 2013. Mitochondrial dys-
2942. function and sarcopenia of aging: From signaling path-
Larsen RG, Callahan DM, Foulis SA, Kent-Braun JA. 2012. ways to clinical trials. Int J Biochem Cell Biol 45: 2288–
Age-related changes in oxidative capacity differ between 2301.
locomotory muscles and are associated with physical ac- Masiero E, Agatea L, Mammucari C, Blaauw B, Loro E,
tivity behavior. Appl Physiol Nutr Metab 37: 88–99. Komatsu M, Metzger D, Reggiani C, Schiaffino S, Sandri
Larsson L, Li X, Frontera WR. 1997. Effects of aging on M. 2009. Autophagy is required to maintain muscle
shortening velocity and myosin isoform composition in mass. Cell Metab 10: 507– 515.
single human skeletal muscle cells. Am J Physiol 272: McCully KK, Fielding RA, Evans WJ, Leigh JSJr, Posner JD.
C638– C649. 1993. Relationships between in vivo and in vitro measure-
Leduc-Gaudet JP, Picard M, St-Jean Pelletier F, Sgarioto N, ments of metabolism in young and old human calf mus-
Auger MJ, Vallee J, Robitaille R, St-Pierre DH, Gouspillou cles. J Appl Physiol (1985) 75: 813– 819.
G. 2015. Mitochondrial morphology is altered in atro- Mendonca GV, Pezarat-Correia P, Vaz JR, Silva L, Heffernan
phied skeletal muscle of aged mice. Oncotarget 6: 17923– KS. 2016. Impact of aging on endurance and neuromus-
17937. cular physical performance: The role of vascular senes-
Leenders M, Verdijk LB, van der Hoeven L, van Kranenburg cence. Sports Med doi: 10.1007/s40279-016-0596-8.
J, Nilwik R, van Loon LJ. 2013. Elderly men and women Mera P, Laue K, Ferron M, Confavreux C, Wei J, Galan-Diez
benefit equally from prolonged resistance-type exercise M, Lacampagne A, Mitchell SJ, Mattison JA, Chen Y, et al.
training. J Gerontol A Biol Sci Med Sci 68: 769 –779. 2016. Osteocalcin signaling in myofibers is necessary and
Lexell J. 1997. Evidence for nervous system degeneration sufficient for optimum adaptation to exercise. Cell Metab
with advancing age. J Nutr 127: 1011S– 1013S. 23: 1078–1092.
Lexell J, Taylor CC. 1991. Variability in muscle fibre areas in Messi ML, Li T, Wang ZM, Marsh AP, Nicklas B, Delbono O.
whole human quadriceps muscle: Effects of increasing 2016. Resistance training enhances skeletal muscle inner-
age. J Anat 174: 239– 249. vation without modifying the number of satellite cells or
www.perspectivesinmedicine.org

Lexell J, Henriksson-Larsen K, Winblad B, Sjostrom M. their myofiber association in obese older adults. J Ger-
1983. Distribution of different fiber types in human skel- ontol A Biol Sci Med Sci 71: 1273– 1280.
etal muscles: Effects of aging studied in whole muscle Metter EJ, Conwit R, Metter B, Pacheco T, Tobin J. 1998. The
cross sections. Muscle Nerve 6: 588– 595. relationship of peripheral motor nerve conduction veloc-
Lexell J, Downham D, Sjostrom M. 1986. Distribution of ity to age-associated loss of grip strength. Aging 10: 471–
different fibre types in human skeletal muscles. Fibre 478.
type arrangement in m. vastus lateralis from three groups Mikines KJ, Sonne B, Farrell PA, Tronier B, Galbo H. 1988.
of healthy men between 15 and 83 years. J Neurol Sci 72: Effect of physical exercise on sensitivity and responsive-
211–222. ness to insulin in humans. Am J Physiol 254: E248–E259.
Lexell J, Taylor CC, Sjostrom M. 1988. What is the cause of Miller BF, Olesen JL, Hansen M, Dossing S, Crameri RM,
the ageing atrophy? Total number, size and proportion of Welling RJ, Langberg H, Flyvbjerg A, Kjaer M, Babraj JA,
different fiber types studied in whole vastus lateralis mus-
et al. 2005. Coordinated collagen and muscle protein
cle from 15- to 83-year-old men. J Neurol Sci 84: 275 –
synthesis in human patella tendon and quadriceps mus-
294.
cle after exercise. J Physiol 567: 1021– 1033.
Lindle RS, Metter EJ, Lynch NA, Fleg JL, Fozard JL, Tobin J,
Murray MP, Gardner GM, Mollinger LA, Sepic SB. 1980.
Roy TA, Hurley BF. 1997. Age and gender comparisons of
Strength of isometric and isokinetic contractions: Knee
muscle strength in 654 women and men aged 20– 93 yr. J
muscles of men aged 20 to 86. Phys Ther 60: 412– 419.
Appl Physiol (1985) 83: 1581– 1587.
Nagasawa J, Sato Y, Ishiko T. 1991. Time course of in vivo
Lynch NA, Metter EJ, Lindle RS, Fozard JL, Tobin JD, Roy
insulin sensitivity after a single bout of exercise in rats. Int
TA, Fleg JL, Hurley BF. 1999. Muscle quality. I: Age-asso-
ciated differences between arm and leg muscle groups. J Sports Med 12: 399– 402.
J Appl Physiol (1985) 86: 188– 194. Nguyen MH, Cheng M, Koh TJ. 2011. Impaired muscle
Mackey AL, Karlsen A, Couppe C, Mikkelsen UR, Nielsen regeneration in ob/ob and db/db mice. ScientificWorld
RH, Magnusson SP, Kjaer M. 2014. Differential satellite Journal 11: 1525– 1535.
cell density of type I and II fibres with lifelong endurance Nishimune H, Stanford JA, Mori Y. 2014. Role of exercise in
running in old men. Acta Physiol (Oxf ) 210: 612– 627. maintaining the integrity of the neuromuscular junction.
Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Muscle Nerve 49: 315–324.
Del Piccolo P, Burden SJ, Di Lisi R, Sandri C, Zhao J, et al. Ono T, Isobe K, Nakada K, Hayashi JI. 2001. Human cells are
2007. FoxO3 controls autophagy in skeletal muscle in protected from mitochondrial dysfunction by comple-
vivo. Cell Metab 6: 458 –471. mentation of DNA products in fused mitochondria.
Mansouri A, Muller FL, Liu Y, Ng R, Faulkner J, Hamilton Nat Genet 28: 272 –275.
M, Richardson A, Huang TT, Epstein CJ, Van Remmen H. Orlander J, Kiessling KH, Larsson L, Karlsson J, Aniansson
2006. Alterations in mitochondrial function, hydrogen A. 1978. Skeletal muscle metabolism and ultrastructure

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785 13


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

G. Distefano and B.H. Goodpaster

in relation to age in sedentary men. Acta Physiol Scand Raue U, Slivka D, Minchev K, Trappe S. 2009. Improvements
104: 249– 261. in whole muscle and myocellular function are limited
Overend TJ, Cunningham DA, Paterson DH, Lefcoe MS. with high-intensity resistance training in octogenarian
1992. Thigh composition in young and elderly men de- women. J Appl Physiol (1985) 106: 1611– 1617.
termined by computed tomography. Clin Physiol 12: Ringholm S, Bienso RS, Kiilerich K, Guadalupe-Grau A,
629–640. Aachmann-Andersen NJ, Saltin B, Plomgaard P, Lundby
Park SW, Goodpaster BH, Strotmeyer ES, Kuller LH, Brou- C, Wojtaszewski JF, Calbet JA, et al. 2011. Bed rest reduces
deau R, Kammerer C, de Rekeneire N, Harris TB, metabolic protein content and abolishes exercise-in-
Schwartz AV, Tylavsky FA, et al. 2007. Accelerated loss of duced mRNA responses in human skeletal muscle. Am
skeletal muscle strength in older adults with type 2 dia- J Physiol Endocrinol Metab 301: E649– E658.
betes: The health, aging, and body composition study. Romanello V, Guadagnin E, Gomes L, Roder I, Sandri C,
Diabetes Care 30: 1507– 1512. Petersen Y, Milan G, Masiero E, Del Piccolo P, Foretz M, et
Payne AM, Delbono O. 2004. Neurogenesis of excitation- al. 2010. Mitochondrial fission and remodelling contrib-
contraction uncoupling in aging skeletal muscle. Exerc utes to muscle atrophy. EMBO J 29: 1774– 1785.
Sport Sci Rev 32: 36–40.
Rooyackers OE, Adey DB, Ades PA, Nair KS. 1996. Effect of
Peake J, Della Gatta P, Cameron-Smith D. 2010. Aging and age on in vivo rates of mitochondrial protein synthesis in
its effects on inflammation in skeletal muscle at rest and human skeletal muscle. Proc Natl Acad Sci 93: 15364–
following exercise-induced muscle injury. Am J Physiol 15369.
Regul Integr Comp Physiol 298: R1485– R1495.
Rosenberg IH. 1997. Sarcopenia: Origins and clinical rele-
Perry CG, Lally J, Holloway GP, Heigenhauser GJ, Bonen A, vance. J Nutr 127: 990S– 991S.
Spriet LL. 2010. Repeated transient mRNA bursts precede
increases in transcriptional and mitochondrial proteins Roth SM, Martel GF, Ivey FM, Lemmer JT, Tracy BL, Metter
during training in human skeletal muscle. J Physiol 588: EJ, Hurley BF, Rogers MA. 2001. Skeletal muscle satellite
4795– 4810. cell characteristics in young and older men and women
Petersen KF, Befroy D, Dufour S, Dziura J, Ariyan C, Roth- after heavy resistance strength training. J Gerontol A Biol
man DL, DiPietro L, Cline GW, Shulman GI. 2003. Mi- Sci Med Sci 56: B240–B247.
tochondrial dysfunction in the elderly: Possible role in Rowe JW, Minaker KL, Pallotta JA, Flier JS. 1983. Charac-
insulin resistance. Science 300: 1140– 1142. terization of the insulin resistance of aging. J Clin Invest
71: 1581–1587.
www.perspectivesinmedicine.org

Peterson MJ, Morey MC, Giuliani C, Pieper CF, Evenson KR,


Mercer V, Visser M, Brach JS, Kritchevsky SB, Goodpaster Rudrappa SS, Wilkinson DJ, Greenhaff PL, Smith K, Idris I,
BH, et al. 2010. Walking in old age and development of Atherton PJ. 2016. Human skeletal muscle disuse atro-
metabolic syndrome: The health, aging, and body com- phy: Effects on muscle protein synthesis, breakdown, and
position study. Metab Syndr Relat Disord 8: 317 –322. insulin resistance—A qualitative review. Front Physiol 7:
Picard M, Ritchie D, Wright KJ, Romestaing C, Thomas 361.
MM, Rowan SL, Taivassalo T, Hepple RT. 2010. Mito- Safdar A, Hamadeh MJ, Kaczor JJ, Raha S, Debeer J, Tarno-
chondrial functional impairment with aging is exagger- polsky MA. 2010. Aberrant mitochondrial homeostasis
ated in isolated mitochondria compared to permeabi- in the skeletal muscle of sedentary older adults. PLoS
lized myofibers. Aging Cell 9: 1032– 1046. ONE 5: e10778.
Pisot R, Marusic U, Biolo G, Mazzucco S, Lazzer S, Grassi B, Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard
Reggiani C, Toniolo L, di Prampero PE, Passaro A, et al. A, Walsh K, Schiaffino S, Lecker SH, Goldberg AL. 2004.
2016. Greater loss in muscle mass and function but small- Foxo transcription factors induce the atrophy-related
er metabolic alterations in older compared with younger ubiquitin ligase atrogin-1 and cause skeletal muscle atro-
men following 2 wk of bed rest and recovery. J Appl Phys- phy. Cell 117: 399– 412.
iol (1985) 120: 922– 929.
Sandri M, Barberi L, Bijlsma AY, Blaauw B, Dyar KA, Milan
Porter C, Hurren NM, Cotter MV, Bhattarai N, Reidy PT,
G, Mammucari C, Meskers CG, Pallafacchina G, Paoli A,
Dillon EL, Durham WJ, Tuvdendorj D, Sheffield-Moore
et al. 2013. Signalling pathways regulating muscle mass in
M, Volpi E, et al. 2015. Mitochondrial respiratory capac-
ageing skeletal muscle: The role of the IGF1-Akt-mTOR-
ity and coupling control decline with age in human skel-
etal muscle. Am J Physiol Endocrinol Metab 309: E224– FoxO pathway. Biogerontology 14: 303– 323.
E232. Schaap LA, Pluijm SM, Deeg DJ, Harris TB, Kritchevsky SB,
Proctor DN, Sinning WE, Walro JM, Sieck GC, Lemon PW. Newman AB, Colbert LH, Pahor M, Rubin SM, Tylavsky
1995. Oxidative capacity of human muscle fiber types: FA, et al. 2009. Higher inflammatory marker levels in
Effects of age and training status. J Appl Physiol (1985) older persons: Associations with 5-year change in muscle
78: 2033–2038. mass and muscle strength. J Gerontol A Biol Sci Med Sci
64: 1183–1189.
Pruchnic R, Katsiaras A, He J, Kelley DE, Winters C, Good-
paster BH. 2004. Exercise training increases intramyocel- Schrager MA, Metter EJ, Simonsick E, Ble A, Bandinelli S,
lular lipid and oxidative capacity in older adults. Am J Lauretani F, Ferrucci L. 2007. Sarcopenic obesity and
Physiol Endocrinol Metab 287: E857–E862. inflammation in the InCHIANTI study. J Appl Physiol
Rasmussen UF, Krustrup P, Kjaer M, Rasmussen HN. 2003. (1985) 102: 919–925.
Experimental evidence against the mitochondrial theory Schulte JN, Yarasheski KE. 2001. Effects of resistance train-
of aging. A study of isolated human skeletal muscle mi- ing on the rate of muscle protein synthesis in frail elderly
tochondria. Exp Gerontol 38: 877– 886. people. Int J Sport Nutr Exerc Metab 11: S111–S118.

14 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

Skeletal Muscle Aging and Exercise

Schultz E, Lipton BH. 1982. Skeletal muscle satellite cells: oxidative power but unchanged antioxidative capacity in
Changes in proliferation potential as a function of age. skeletal muscle from aged humans. Pflugers Arch 446:
Mech Ageing Dev 20: 377 –383. 261 –269.
Shavlakadze T, McGeachie J, Grounds MD. 2010. Delayed Tracy BL, Ivey FM, Hurlbut D, Martel GF, Lemmer JT, Siegel
but excellent myogenic stem cell response of regenerating EL, Metter EJ, Fozard JL, Fleg JL, Hurley BF. 1999. Muscle
geriatric skeletal muscles in mice. Biogerontology 11: quality. II. Effects Of strength training in 65- to 75-yr-old
363–376. men and women. J Appl Physiol (1985) 86: 195– 201.
Shoelson SE, Lee J, Goldfine AB. 2006. Inflammation and Trappe S, Godard M, Gallagher P, Carroll C, Rowden G,
insulin resistance. J Clin Invest 116: 1793– 1801. Porter D. 2001. Resistance training improves single mus-
Short KR, Bigelow ML, Kahl J, Singh R, Coenen-Schimke J, cle fiber contractile function in older women. Am J Phys-
Raghavakaimal S, Nair KS. 2005. Decline in skeletal mus- iol Cell Physiol 281: C398–C406.
cle mitochondrial function with aging in humans. Proc Trappe S, Gallagher P, Harber M, Carrithers J, Fluckey J,
Natl Acad Sci 102: 5618– 5623. Trappe T. 2003. Single muscle fibre contractile properties
Slivka D, Raue U, Hollon C, Minchev K, Trappe S. 2008. in young and old men and women. J Physiol 552: 47–58.
Single muscle fiber adaptations to resistance training in Trounce I, Byrne E, Marzuki S. 1989. Decline in skeletal
old (.80 yr) men: Evidence for limited skeletal muscle muscle mitochondrial respiratory chain function: Possi-
plasticity. Am J Physiol Regul Integr Comp Physiol 295: ble factor in ageing. Lancet 1: 637 –639.
R273–R280. Twig G, Elorza A, Molina AJ, Mohamed H, Wikstrom JD,
Smuder AJ, Kavazis AN, Min K, Powers SK. 2011. Exercise Walzer G, Stiles L, Haigh SE, Katz S, Las G, et al. 2008a.
protects against doxorubicin-induced markers of au- Fission and selective fusion govern mitochondrial segre-
tophagy signaling in skeletal muscle. J Appl Physiol gation and elimination by autophagy. EMBO J 27: 433–
(1985) 111: 1190–1198. 446.
Snow MH. 1977. The effects of aging on satellite cells in Twig G, Hyde B, Shirihai OS. 2008b. Mitochondrial fusion,
skeletal muscles of mice and rats. Cell Tissue Res 185: fission and autophagy as a quality control axis: The bio-
399–408. energetic view. Biochim Biophys Acta 1777: 1092– 1097.
Sousa-Victor P, Gutarra S, Garcia-Prat L, Rodriguez-Ubreva Verdijk LB, Koopman R, Schaart G, Meijer K, Savelberg HH,
J, Ortet L, Ruiz-Bonilla V, Jardi M, Ballestar E, Gonzalez van Loon LJ. 2007. Satellite cell content is specifically
S, Serrano AL, et al. 2014. Geriatric muscle stem cells reduced in type II skeletal muscle fibers in the elderly.
www.perspectivesinmedicine.org

switch reversible quiescence into senescence. Nature Am J Physiol Endocrinol Metab 292: E151–E157.
506: 316– 321. Verdijk LB, Gleeson BG, Jonkers RA, Meijer K, Savelberg
Stenholm S, Harris TB, Rantanen T, Visser M, Kritchevsky HH, Dendale P, van Loon LJ. 2009. Skeletal muscle hy-
SB, Ferrucci L. 2008. Sarcopenic obesity: Definition, pertrophy following resistance training is accompanied
cause and consequences. Curr Opin Clin Nutr Metab by a fiber type-specific increase in satellite cell content in
Care 11: 693 –700. elderly men. J Gerontol A Biol Sci Med Sci 64: 332– 339.
Tanaka H, Dinenno FA, Monahan KD, Clevenger CM, De- Verdijk LB, Snijders T, Holloway TM, van Kranenburg J, van
Souza CA, Seals DR. 2000. Aging, habitual exercise, and Loon LJ. 2016. Resistance training increases skeletal mus-
dynamic arterial compliance. Circulation 102: 1270– cle capillarization in healthy older men. Med Sci Sports
1275. Exerc 48: 2157–2164.
Thyfault JP, Kraus RM, Hickner RC, Howell AW, Wolfe RR, Wild S, Roglic G, Green A, Sicree R, King H. 2004. Global
Dohm GL. 2004. Impaired plasma fatty acid oxidation in prevalence of diabetes: Estimates for the year 2000 and
extremely obese women. Am J Physiol Endocrinol Metab projections for 2030. Diabetes Care 27: 1047– 1053.
287: E1076– E1081. Yin H, Price F, Rudnicki MA. 2013. Satellite cells and the
Tong JF, Yan X, Zhu MJ, Du M. 2009. AMP-activated protein muscle stem cell niche. Physiol Rev 93: 23–67.
kinase enhances the expression of muscle-specific ubiq- Zampieri S, Pietrangelo L, Loefler S, Fruhmann H, Voge-
uitin ligases despite its activation of IGF-1/Akt signaling lauer M, Burggraf S, Pond A, Grim-Stieger M, Cvecka J,
in C2C12 myotubes. J Cell Biochem 108: 458 –468. Sedliak M, et al. 2014. Lifelong physical exercise delays
Tonkonogi M, Fernstrom M, Walsh B, Ji LL, Rooyackers O, age-associated skeletal muscle decline. J Gerontol A Biol
Hammarqvist F, Wernerman J, Sahlin K. 2003. Reduced Sci Med Sci 70: 163–173.

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029785 15


Downloaded from http://perspectivesinmedicine.cshlp.org/ on July 3, 2023 - Published by Cold Spring Harbor Laboratory Press

Effects of Exercise and Aging on Skeletal Muscle


Giovanna Distefano and Bret H. Goodpaster

Cold Spring Harb Perspect Med 2018; doi: 10.1101/cshperspect.a029785 originally published online April
21, 2017

Subject Collection The Biology of Exercise

Exosomes as Mediators of the Systemic Effects of Exercise and Aging on Skeletal Muscle
Adaptations to Endurance Exercise Giovanna Distefano and Bret H. Goodpaster
Adeel Safdar and Mark A. Tarnopolsky
Molecular Basis of Exercise-Induced Skeletal Muscle-Adipose Tissue Cross Talk
Muscle Mitochondrial Biogenesis: Historical Kristin I. Stanford and Laurie J. Goodyear
Advances, Current Knowledge, and Future
Challenges
Christopher G.R. Perry and John A. Hawley
Exercise Metabolism: Fuels for the Fire Performance Fatigability: Mechanisms and Task
Mark Hargreaves and Lawrence L. Spriet Specificity
Sandra K. Hunter
Health Benefits of Exercise Adaptations to Endurance and Strength Training
Gregory N. Ruegsegger and Frank W. Booth David C. Hughes, Stian Ellefsen and Keith Baar
Molecular Regulation of Exercise-Induced Muscle The Bioenergetics of Exercise
Fiber Hypertrophy P. Darrell Neufer
Marcas M. Bamman, Brandon M. Roberts and
Gregory R. Adams
Physiological Redundancy and the Integrative Effects of Exercise on Vascular Function,
Responses to Exercise Structure, and Health in Humans
Michael J. Joyner and Jerome A. Dempsey Daniel J. Green and Kurt J. Smith
On the Run for Hippocampal Plasticity Control of Muscle Metabolism by the Mediator
C'iana Cooper, Hyo Youl Moon and Henriette van Complex
Praag Leonela Amoasii, Eric N. Olson and Rhonda
Bassel-Duby
Molecular Basis for Exercise-Induced Fatigue: Theoretical and Biological Evaluation of the Link
The Importance of Strictly Controlled Cellular Ca between Low Exercise Capacity and Disease Risk
2+ Handling Lauren Gerard Koch and Steven L. Britton
Arthur J. Cheng, Nicolas Place and Håkan
Westerblad

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright © 2018 Cold Spring Harbor Laboratory Press; all rights reserved

You might also like