You are on page 1of 16

Ageing Research Reviews 67 (2021) 101305

Contents lists available at ScienceDirect

Ageing Research Reviews


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

Review

The emerging role of the sympathetic nervous system in skeletal muscle


motor innervation and sarcopenia
Osvaldo Delbono a, b, c, d, *, Anna Carolina Zaia Rodrigues a, d, Henry Jacob Bonilla a,
Maria Laura Messi a
a
Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
b
Department of Internal Medicine, The Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC, USA
c
Department of Internal Medicine, The Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
d
Department of Internal Medicine, The Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA

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

Keywords: Examining neural etiologic factors’role in the decline of neuromuscular function with aging is essential to our
Skeletal muscle understanding of the mechanisms underlying sarcopenia, the age-dependent decline in muscle mass, force and
Sarcopenia power. Innervation of the skeletal muscle by both motor and sympathetic axons has been established, igniting
Motoneuron
interest in determining how the sympathetic nervous system (SNS) affect skeletal muscle composition and
Autonomic nervous system
Sympathetic nervous system
function throughout the lifetime. Selective expression of the heart and neural crest derivative 2 gene in peripheral
Neuromuscular junction SNs increases muscle mass and force regulating skeletal muscle sympathetic and motor innervation; improving
Denervation acetylcholine receptor stability and NMJ transmission; preventing inflammation and myofibrillar protein
Aging degradation; increasing autophagy; and probably enhancing protein synthesis. Elucidating the role of central SNs
will help to define the coordinated response of the visceral and neuromuscular system to physiological and
pathological challenges across ages.
This review discusses the following questions: (1) Does the SNS regulate skeletal muscle motor innervation?
(2) Does the SNS regulate presynaptic and postsynaptic neuromuscular junction (NMJ) structure and function?
(3) Does sympathetic neuron (SN) regulation of NMJ transmission decline with aging? (4) Does maintenance of
SNs attenuate aging sarcopenia? and (5) Do central SN group relays influence sympathetic and motor muscle
innervation?

Introduction and gait speed falls below 0.8 m/s, a clinical diagnosis of sarcopenia is
reached (Studenski et al., 2011). Although, the average population un­
Age-dependent decline in muscle mass, force and power, termed dergoing sarcopenia varies according to the nutritional status and
sarcopenia (Rosenberg, 1989), impairs mobility to increasing the risk of physical activity, more than 50 % of people over 80 undergo sarcopenia
falls, institutionalization, comorbidity, and premature death (Larsson (von Haehling et al., 2010), and all others exhibit some degree of muscle
et al., 2019; Tieland et al., 2018). In addition to impairing quality of life atrophy and loss of muscle function (Larsson et al., 2019). Those with
in the adult population, sarcopenia is also a major risk factor for many sarcopenia subjects are not necessarily frail; sarcopenia is about twice as
age-associated diseases (Chin et al., 2013; DeAndrade et al., 2018; Landi common as frailty (von Haehling et al., 2010).
et al., 2012; Maeda and Akagi, 2016; Nishiguchi et al., 2016; Srikanthan Physiological and pathological studies show that innervation and
et al., 2010; Tournadre et al., 2019; Upadhya et al., 2015). Sedentary systemic factors exert a strong influence on skeletal muscle mass,
people lose 0.5 percent of lean muscle mass every year between age 25 composition and function (Delbono, 2003; Larsson et al., 2019). Sar­
and 60; the rate doubles to about 1 percent after 60, and doubles again copenia is associated with: (a) excitation-contraction uncoupling (Del­
each decade thereafter (Lynch et al., 1999). Once muscle mass in older bono, 2011a; Delbono et al., 1995; Wang et al., 2002), which diminishes
adults falls below two standard deviations of the mean of young controls muscle specific force (force normalized to cross-sectional area)

* Corresponding author at: Wake Forest School of Medicine, Medical Center Boulevard, Department of Internal Medicine, Gerontology and Geriatric Medicine,
Winston-Salem, NC 27157, USA.
E-mail address: odelbono@wakehealth.edu (O. Delbono).

https://doi.org/10.1016/j.arr.2021.101305
Received 2 December 2020; Received in revised form 6 January 2021; Accepted 15 February 2021
Available online 18 February 2021
1568-1637/© 2021 Elsevier B.V. All rights reserved.
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

(Gonzalez et al., 2000a, b; Wang et al., 2000); (b) decreased number of denervation remains unknown (Delbono, 2011a). Several groups have
muscle fibers as well as fiber atrophy (Dutta, 1997; Lexell, 1995); (c) reported skeletal muscle denervation and reinnervation and motor unit
changes in fiber type (Frontera et al., 2000; Larsson et al., 1991); (d) remodeling or loss in aging rodents or humans (Doherty et al., 1993;
diminished muscle power related to decreased maximal isometric force Einsiedel and Luff, 1992; Hashizume et al., 1988; Johnson et al., 1995;
and slower sliding of actin on myosin (Brooks and Faulkner, 1994; Hook Kanda and Hashizume, 1989, 1992; Larsson and Ansved, 1995; Larsson
et al., 1999); (e) impaired recovery after eccentric contraction (Faulkner et al., 1991, 2019; Larsson and Edstrom, 1986; Zhang et al., 1996).
et al., 1993); and (f) deposit of intra- and extra-myocellular lipids with Motor-unit remodeling leads to changes in fiber-type composition (Pette
consequent changes in skeletal muscle quality (Choi et al., 2012). Motor and Staron, 2001; Schiaffino and Reggiani, 1994, 1996).
and/or sympathetic muscle denervation trigger and/or exacerbate some During development, muscle fiber phenotype is determined by in­
of these processes. teractions among subpopulations of ventral spinal cord motor neurons
Explanations for these age-dependent changes in muscle properties that activate contraction at rates ranging from 10 (slow fibers) to 100
include: alterations in myofiber calcium homeostasis (Delbono, 2011b; (fast-fatigue resistant) to 150 Hz (fast-fatigue sensitive) (Buller, 1960a,
Jimenez-Moreno et al., 2008; Tang et al., 2011), decreased muscle b; Greensmith and Vrbova, 1996). During aging, motor-unit remodeling
loading associated with sedentary lifestyle, (Landi et al., 2014; Tseng appears to involve denervation of fast muscle fibers and their reinner­
et al., 1995) oxidative damage (Jackson, 2016; Jang et al., 2010; vation by axonal sprouting from slow fibers (Frey et al., 2000; Kadhir­
Leeuwenburgh et al., 1998; Vasilaki et al., 2017; Weindruch, 1995), esan et al., 1996; Larsson, 1995; Lexell, 1995). When denervation
impaired mitochondrial quality control (Picca et al., 2019), exceeds reinnervation, that population of muscle fibers becomes atro­
age-dependent decrease in insulin-like growth factor-1 (IGF-1) expres­ phic and non-functional. Although denervation contributes to skeletal
sion or tissue sensitivity (Owino et al., 2001; Renganathan et al., 1997; muscle atrophy and functional impairment with aging (Larsson and
Schiaffino et al., 2013), hormonal and metabolic alterations (Kamwa Ansved, 1995), its time course, and prevalence in human and animal
et al., 2020; Vitale et al., 2016), impaired autophagy (Masiero et al., models of aging remain undetermined. Combining contraction force
2009; Sakuma et al., 2016; White et al., 2016; Wokke et al., 1990), recordings with muscle immunostaining for the neural cell-adhesion
accumulation of senescence cells (Tchkonia et al., 2013; Zhu et al., molecule (NCAM) protein, a marker of fiber denervation and reinner­
2014), inflammation (Ferrucci et al., 1999, 2002; Tchkonia et al., 2013), vation in rat studies showed that the old have significantly more
decline in satellite cell proliferation (Garcia-Prat et al., 2016; Hwang denervated fibers than the young (Andersson et al., 1993; Gosztonyi
and Brack, 2018), and loss of neural control in skeletal muscle (Delbono, et al., 2001). Moreover, the area of denervated fibers, detected by pos­
2003). Unfortunately, some of these mechanisms are technically diffi­ itive staining with NCAM antibodies, accounts for a significant fraction
cult to test in the whole organism, particularly in mammalian species. of the decline in specific force (Urbancheck et al., 2001). Human skeletal
Pharmacological interventions produce numerous side effects, have muscle studies confirm NCAM’s usefulness as a denervation/ reinner­
yielded inconsistent results, and their rationales have been challenged vation marker (Biral et al., 2008; Messi et al., 2016; Sonjak et al., 2019).
(Fry et al., 2015; Jackson, 2016; Larsson et al., 2019; Ristow, 2014; We reported that muscle denervation in aging mice is more extensive
Sakellariou et al., 2017; Wang and Hekimi, 2015). Still, the interaction than previous functional and structural assays predicted. Two sodium
between nerve and skeletal muscle is clearly crucial for both tissues to channel isoforms are expressed in skeletal muscle, the TTX-sensitive
survive and function adequately throughout life (Delbono, 2003; Payne Nav1.4 or SkM1 and the TTX-resistant Nav1.5 or SkM2 (Kallen et al.,
et al., 2006b). Does muscle atrophy and weakness result from primary 1990; Trimmer et al., 1989). The SkM2 mRNA level is highest in early
neural or muscular etiologic factors or a combination? Does development, when TTX-insensitive channels predominate, but declines
age-dependent maintenance of sympathetic neuron transcription fac­ rapidly with age as SkM1 mRNA increases; SkM2 mRNA is not detect­
tors, particularly the heart and neural crest derivative 2 (Hand2) (Hen­ able in normally innervated adult skeletal muscle but increases greater
dershot et al., 2008; Rodrigues et al., 2020; Stanzel et al., 2016), than 100-fold after denervation; rat cardiac muscle has abundant SkM2
attenuate motor denervation? Examining neural etiologic factors’ role in mRNA but no detectable SkM1 message (Kallen et al., 1990). To assess
the decline of neuromuscular function with aging is essential to our under­ the extent of denervation in aging rodents, we combined electrophysi­
standing of the mechanisms underlying sarcopenia. ological and immunohistochemical assays to detect the expression of
This review focuses on how aging influences skeletal muscle sym­ TTX-resistant Nav1.5 in flexor digitorum brevis muscles from
pathetic and motor innervation and how their alteration mediates sar­ young-adult and senescent mice. Sodium current density measured with
copenia pathogenesis. It discusses basic aspects of muscle innervation, the macropatch cell-attached technique did not show significant dif­
even at a young age, that are incompletely understood. This review is ferences between muscle fibers from young and old mice. The TTX
organized to consider the following questions: (1) Does the sympathetic dose-response curve, using the whole-cell voltage-clamp technique,
nervous system (SNS) regulate skeletal muscle motor innervation? (2) showed three populations of fibers in senescent mice: one similar to fi­
Does the SNS regulate presynaptic and postsynaptic neuromuscular bers from young mice (TTX sensitive); another similar to fibers from
junction (NMJ) structure and function? (3) Does sympathetic neuron experimentally denervated muscle (TTX resistant); and an intermediate
regulation of NMJ transmission decline with aging? (4) Does mainte­ group. Partially and fully denervated fibers made up approximately 50
nance of sympathetic neurons attenuate aging sarcopenia? and (5) Do percent of the total tested, which aligns with the percentage of fibers
central sympathetic neuron group relays influence sympathetic and positive for the Nav1.5 channel, as shown by specific immunostaining
motor muscle innervation? (Wang et al., 2005) Discrepant results related to the time course, loca­
tion, and extent of denervation in rodents (Hendrickse et al., 2018) can
1. Does the SNS regulate skeletal muscle motor innervation? be accounted for by the age of the animals and the denervation and
Relationship between sarcopenia and the dual muscle immunofluorescence techniques used. These studies revealed that new
innervation technical approaches are needed to accurately assess muscle denervation,
which, although subtle, can be devastating over time (Delbono, 2003).
Sarcopenia is characterized by heterogeneous muscle fiber size and
myosin heavy-chain composition (Schiaffino, 2018; Schiaffino and Increased frequency of hybrid fiber types and quantification of myosin
Reggiani, 1994), selective myofiber atrophy, myofiber grouping, and heavy chain (MHC) composition in aging skeletal muscle
progressive NMJ degeneration, leading to complete or partial synapse
denervation, and Schwann cell depletion (Delbono, 2003; Li and Since innervation regulates MHC expression (Schiaffino and
Thompson, 2011; Wang et al., 2005). Despite its relevance to the onset Reggiani, 1996), muscle morphometric analysis of MHC predominance
and outcome of sarcopenia, the etiology of age-related skeletal muscle is critical to understanding the myofiber/motoneuron relationship in

2
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

health and disease (Lefeuvre et al., 1996). Determining fibers’ shapes, reorientation of costameres (rib-like structures formed by dystrophin
sizes, and arrangements elucidates muscle tissue morphology and and β-dystroglycan) (Bezakova and Lomo, 2001), proliferation of triadic
overall health (Gropp, 2017), while immunohistochemistry (IHC) using membranes (Salvatori et al., 1988), reduction of voltage-gated calcium
MHC antibodies is a widely accepted approach to examine skeletal channels, and alterations to the sarcoplasmic reticulum Ca2+ release
muscle fiber type composition (Bloemberg and Quadrilatero, 2012). channel. (Delbono, 1992; Delbono et al., 1997; Delbono and Stefani,
However useful for compositional studies of larger portions of the body, 1993; Wang et al., 2000). Costameric proteins transmit lateral forces and
computed tomography (CT) scan, magnetic resonance imaging (MRI), provide structural integrity in contracting mechanically loaded muscle
and dual-energy X-ray absorptiometry cannot visualize the microscopic fibers (Straub and Campbell, 1997). Along with muscle activity, muscle
structural features of skeletal muscle. Performed with the skill needed to agrin at a concentration two orders of magnitude lower than the effec­
correctly interpret and generate reproducible results, IHC offers the best tive concentration of neural agrin, regulates the organization of cyto­
combination of molecular and microscopic morphological information skeletal proteins in skeletal muscle fibers (Bezakova and Lomo, 2001).
for the analysis of skeletal muscle. These molecular changes in aging muscle should be explored (Taetzsch
Both semiautomatic and automatic approaches have been developed and Valdez, 2018). Compared to young and middle-aged mice, aging
to aid IHC analysis (Bergmeister et al., 2016; Briguet et al., 2004; Lau C57BL/6 mice show an increase in agrin, calcitonin gene-related pep­
et al., 2018; Mayeuf-Louchart et al., 2018; Miazaki et al., 2015; Pertl tide, growth associated protein 43, fibroblast growth factor binding
et al., 2013; Smith and Barton, 2014; Wen et al., 2018); but aging protein 1, and transforming growth factor-β1at the NMJs of the tibialis
skeletal muscle poses difficulties that young, healthy muscle tissue does anterior, soleus, extensor digitorum longus, and gracilis muscles as ev­
not. Failing autophagy leads to intracellular protein deposits (Jokl and idence of increased development, plasticity, and maintenance of NMJ
Blanco, 2016). Aging skeletal muscle usually displays some degree of signaling with aging (Blasco et al., 2020).
atrophy; more fibers express more than one MHC (hybrid); interstices The studies reported above strongly associate neural alterations with
are infiltrated with fat cells, lipofuscin, and fibrous tissue, and angulated the onset and gradual decline of aging skeletal muscle function. In­
small myofibers disperse heterogeneously throughout the tissue (Dis­ terventions focused on motor neurons, their axons, and associated non-
tefano and Goodpaster, 2018; Hepple, 2018; Purves-Smith et al., 2014). neuronal cells, and the NMJ may slow or even reverse age-related im­
These characteristics can affect the efficacy of muscle analysis programs pairments of skeletal muscle.
that rely on tissue homogeneity.
We designed a reliable, user-friendly, and accessible approach to Dual innervation of the skeletal muscle
quantifying basic muscle morphometric parameters that does not
require computer experts or statisticians (Bonilla et al., 2020). It em­ Unlike the skeletal motor system, which innervates striated muscles,
phasizes image thresholding and editing capability to properly quantify the autonomic nervous system (ANS) innervates the smooth muscle
hybrid fibers and to identify muscle fibers, regardless of their degree of organs and tissues (Powley, 2008). Although skeletal muscle sympa­
atrophy, respectively (Bonilla et al., 2020). thetic muscle innervation was described over a century ago (Boeke,
1909); myofiber sympathetic innervation remained unexplored, prob­
Plasticity at the aging neuromuscular junction ably due to technical limitations. Sympathetic axons travel through the
peripheral nervous system together with motor and sensory axons and
NMJ formation involves a number of resident molecules, such as low- then surrounding blood vessels (Eichmann and Brunet, 2014; Wang
density lipoprotein (LDL)-related receptor protein 4 (LRP4) (Messéant et al., 2020a). to reach the muscle fiber (Griffin and Thompson, 2008).
et al., 2017; Weatherbee et al., 2006), agrin (differentiation) (Ruegg and Sympathectomy and sympathetic neuron-labeling experiments using
Bixby, 1998), Dok-7 (synaptogenesis) (Okada et al., 2006), and the monosynaptic retrograde tracer β-subunit of cholera toxin (CTβ)
muscle-specific kinase (MuSK) (synapse maintenance) (Koneczny et al., helped to elucidate the innervation of mouse hindlimb muscles by par­
2014), and others. MuSK participates in three signaling pathways: avertebral sympathetic ganglia and ventral horn motoneurons (Card and
Agrin-LRP4, Wnt, and MuSK-BMP, via distinct domains. Although ge­ Enquist, 2012; Kerman et al., 2003; Rodrigues et al., 2018). For a long
netic studies have elucidated the role these proteins play in NMJ time, sympathetic innervation of skeletal muscle was thought to be
structure and function, how they contribute to its age-associated func­ restricted to the blood vessels until it was observed in the intra- and
tional decline remains ill-defined (Fish and Fallon, 2020). extrafusal fibers of several muscles (Barker and Saito, 1981). This
Age-associated structural and functional alterations of the neuromus­ finding sparked researchers’ interest in the role of postganglionic sym­
cular junction such as instability of neuromuscular junction have been pathetic neurons and their neurotransmitter(s) in myofiber composition,
reported (Balice-Gordon, 1997; Khan et al., 2016; Payne and Delbono, trophism, and function. Even at birth, sympathetic innervation and
2004; Payne et al., 2006a; Rodrigues et al., 2018; Valdez et al., 2010). interaction with NMJs in hindlimb, diaphragm, and levator auris longus
The events leading to denervation in aging mammals are less understood muscles is extensive and, in the extensor digitorum longus muscle, in­
than the formation and activity-dependent editing of neuromuscular creases until adulthood (Straka et al., 2018).
synaptic connections (Personius and Balice-Gordon, 2000). Activity has The ANS coordinates homeostasis through sympathetic and para­
been shown to play a crucial role (Personius and Balice-Gordon, 2000), sympathetic outflows and the enteric nervous system (Robertson et al.,
prompting the interesting hypothesis that a similar mechanism affects 2012). The SNS regulates the function of many tissues, and its genetic,
senescent mammals. While exercise and calorie restriction attenuate autoimmune, or degenerative alteration results in a variety of clinical
NMJ spatial distortion in aging mice and humans (Messi et al., 2016; disorders affecting the brain, spinal cord, and/or nerve structure and
Valdez et al., 2010), the levels required are highly variable and difficult function. While it also plays a role in skeletal muscle regeneration after
to sustain. Such interventions must be personalized and closely injury (Beitzel et al., 2007) and the ability to perform short intensive
monitored. tasks like the Wingate test (Le Panse et al., 2007), its deterioration with
Nerve terminal remodeling during the period of target absence does age results in incapacitating symptoms (Delbono, 2003; Gonzalez-Freire
not adequately explain the subsequent changes to the NMJ. Mechanisms et al., 2014; Lipsitz and Novak, 2008). Its functional impairment and the
for remodeling the synapse include failure of the regenerating muscle resulting muscle weakness are manifestations of Alzheimer’s disease
fiber to contact the old basal lamina and nerve terminal, growth of the (Jensen-Daham et al., 2015) and such synucleinopathies as Parkinson’s
nerve terminal and its glia toward the regenerating fiber, and remod­ disease, dementia with Lewy bodies, multiple system atrophy, and pure
eling of the initial contact as the motor nerve terminal becomes varicose autonomic failure (Askmark et al., 2001; Cano-Jaimez et al., 2010;
(Li and Thompson, 2011). Femminella et al., 2014; Janig, 2012; Kandinov et al., 2011; Low and
In skeletal muscles from aging rodents, motor denervation causes Bennarroch, 2008; Mu et al., 2013; Munver and Volfson, 2006; Okamoto

3
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

and Biaggioni, 2012; Ooi et al., 1997; Orimo et al., 2007; Tachi et al., acetylcholine receptor stability is extensive. It regulates NMJ trans­
1995). Muscle weakness associated with autonomic dysfunction is also mission, maintains optimal Gαi2 expression, and prevents any increase
obvious in adrenal insufficiency, Lambert-Eaton myasthenic syndrome, in histone deacetylase 4 (Hdac4), myogenin, the muscle RING-finger
and chronic fatigue syndrome (Beitzel et al., 2007; Lindquist and Stan­ protein-1 MuRF1 (a.k.a. TRIM63), and muscle-specific microRNA-206
gel, 2011; O’Suilleabhain et al., 1998). Aging produces several changes (miR-206). The SNS also prevents elevations in MuRF1 levels, muscle
in the ANS that may impair adaptations to common physiologic stressors atrophy, and maintains postsynaptic membrane acetylcholine receptor
and increase the risk of developing diseases that further harm autonomic (AChR) levels (Rodrigues et al., 2018), as depicted in Fig. 1.
function. After sympathectomy, levels of the neurotransmitter noradrenaline
We used a microsurgical approach to muscle sympathetic denerva­ (NA) decreased 80 percent or more in soleus and EDL muscles, while
tion to define the role of the SNS in regulating NMJ stability and func­ plasma catecholamines were not affected (Silveira et al., 2014), which
tion in adult mice (Khan et al., 2016; Rodrigues et al., 2019). A decrease indicates that the surgical removal of lumbar paravertebral sympathetic
in synaptic vesicle release and quantum content indicates that motor ganglia did not prevent the adrenal gland medulla from releasing
axon function is impaired. Next, to determine how sympathetic and adrenaline into the bloodstream. Since the β2-AR (adrenergic receptor)
motor axons in the spinal nerve communicate under normal and path­ is a heterotrimeric guanine nucleotide-binding protein (G pro­
ological conditions, we examined whether sympathectomy alters mye­ tein)-coupled receptor, and muscle NA levels and receptor activity
lination, myelin thickness, and/or neurofilament (NF) phosphorylation decrease with sympathectomy (Silveira et al., 2014), we examined
in large axons like those in motor neurons. We found increased variation whether sympathectomy led to decreased Gαi2 activation, a mechanism
in myelin thickness, probably a result of Schwann cell reprogramming postulated to activate myogenin (Mei et al., 2011). Minetti et al. reported
(Jessen and Mirsky, 2016). Myelinating Schwann cells modulate NF that Gαi2 promotes skeletal muscle hypertrophy, myoblast differentia­
phosphorylation, axonal caliber, and slow axonal transport (de Waegh tion, and muscle regeneration, indicating that receptors that act through
et al., 1992). Although our analysis at the sciatic-tibio-peroneal nerve Gα(i2) might be targets to prevent skeletal muscle wasting (Minetti
showed no downregulation of genes encoding axonal myelination, we et al., 2014).
found a reactive increase in neurotrophin gene transcription. NFs are We found Gαi2 significantly decreased in hindlimb mouse muscles
abundant in motor and other neurons with large-diameter axons (Ohara after sympathectomy, and that restoring it using a human Gαi2 (Q205 L)
et al., 1993; Sakaguchi et al., 1993). The mechanism underlying cross­ mutant, subcloned into an adeno-associated viral vector, prevented
talk between sympathetic and motor axons remains unclear. The inter­ decreases in membrane AChR levels and increases in MuRF1, Hdac4,
action of both axons with dephosphorylated NFs, revealed by myogenin, and β2-AR expression. These results indicate that increasing
transmission electron microscopy, may interfere with microtubules’ muscle levels of Gαi2 prevents skeletal muscle motor denervation (Rodrigues
transport of enzymes and peptide precursors (Purves et al., 2011). et al., 2018).
The influence of the SNS on muscle motor innervation and

Fig. 1. Model of SNS regulation of the NMJ. The SNS regulates NMJ motor axon vesicle release and postsynaptic AChR stability through the Gαi2-Hdac4-myogenin-
MuRF1-miR-206 pathway. (Rodrigues et al., 2018) AChR: acetylcholine receptor; DRG: dorsal root ganglion; Hdac4: histone deacetylase-4; IML: intermediolateral
column; NA: noradrenaline.

4
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

The SNS regulates skeletal muscle motor innervation constitutively activated form of Gαi2 (Moxham and Malbon, 1996) using
an adeno-associated virus, AAV-Gαi2(Q205 L), prevented both. Myoge­
To address whether the SNS regulates skeletal muscle motor inner­ nin induces muscle and serum miR-206, which suppresses Hdac4 and
vation, we studied the impact of regional and systemic sympathetic promotes reinnervation (Moresi et al., 2010). SNS denervation of blood
ablation on muscle motor innervation using transcriptome analysis, vessels may affect NMJ molecular composition and function, but specific
retrograde tracing of the sympathetic outflow to the skeletal muscle, and Gαi2(Q205 L) expression in myofibers (Minetti et al., 2014) is consistent
electrophysiological, imaging, and protein analysis of NMJ trans­ with a direct sympathetic effect on NMJs. Also, muscle-flow recordings
mission. We and others also used state-of-the-art microsurgical tech­ have shown that epinephrine’s effect on neuromuscular transmission is
niques in several wild-type and transgenic mice, including C57BL6, independent of concomitant vascular changes produced by the cate­
enzyme E3 ubiquitin-protein ligase TRIM63/muscle specific RING finger cholamine (Bowman and Raper, 1966).
1 (MuRF1KO), and dopamine beta-hydroxylase (DBH)-tomato and Thy-
1 mice (Khan et al., 2016; Rodrigues et al., 2019). 2. Does the SNS regulate presynaptic and postsynaptic NMJ
After SNS ablation, we found elevated levels of MuRF1 transcript and structure and function? Sympathomimetics can be used to
protein. MuRF1 is involved in muscle trophism and maintenance answer this question
(Moriscot et al., 2010). It is expressed in both type-I and type-II fibers,
preferentially the latter; highly enriched in the postterminal; and in­ To explore sympathetic neuron-motoneuron cross-talk and its influ­
teracts with AChR in endocytic structures (Rudolf et al., 2013). The ence on NMJ transmission, we tested the effects of the sympathetic
procedure evoked an increase in E3 ligase that led to an overall increase postganglionic neurotransmitter, noradrenaline, and sympathomimetics
in AChR but a decrease in sarcolemmal AChR, indicating that sympa­ on such motoneuron-terminal targets as TRPV1 and P/Q- and N-type
thetic neurons influence AChR partition between the sarcolemmal and voltage-activated Ca2+ channels and the resulting Ca2+ fluxes and syn­
subsarcolemmal compartments. Moreover, sympathectomized aptic vesicle release dynamics (Khuzakhmetova and Bukharaeva, 2020;
MuRF1KO mice showed AChR partitioned much as it is in innervated Kuba, 1970; Rodrigues et al., 2018; Tsentsevitsky et al., 2019). The
young mice (Rodrigues et al., 2018). These results support the conclu­ impact of SNS ablation and sympathomimetics on the amplitude of the
sion that MuRF1 mediates SNS regulation of the fraction of receptors compound muscle action potential has also been reported (Khan et al.,
located at the postterminal NMJ sarcolemma. 2016).
Severing the sciatic nerve is considered a model of motor muscle Catecholamines act on specific receptors located on the cell surface
denervation since flaccid paralysis and massive atrophy ensue (Bodine of target organs. These receptors, expressed throughout the body, are
et al., 2001; Carlson et al., 2002; Machado et al., 2019). Rapidly, classed in two groups. The α-subtype includes the Gq-coupled receptors
following nerve transection, MuRF1 and F-Box Protein 32 (Fbxo32, a.k.a. α1A, α1B, and α1D and Gi-coupled receptors α2A, α2B, and α2C. The
atrogin-1) increase (Bodine et al., 2001), while at day 14, MuRF1, but not β-subtype comprises the Gs-coupled β1 and Gs/Gi-coupled β2 and β3
Fbxo32, was significantly increased after motor denervation in both receptors. β1 and α2B are particularly important in understanding the
soleus and tibialis anterior muscles as determined by immunoblot mechanisms by which the SNS regulates NMJ transmission (Rodrigues
(Moriscot et al., 2010). Note that treatment with the β2-AR agonist et al., 2019; Wang et al., 2020a), and β2 directly regulates skeletal
clenbuterol prevents denervation-induced increases in Fbxo32 levels muscle mass and metabolism (Lynch and Ryall, 2008).
(Goncalves et al., 2012). These results indicate that maintaining sympa­ Noradrenaline (NA), the main neurotransmitter of ganglionic sym­
thetic activity or stimulating β2-AR prevents Fbxo32 levels from rising and pathetic neurons, modulates neuromuscular transmission (Kuba, 1970).
that muscle sympathetic ablation mimics, to some extent, muscle motor Regional surgical removal or systemic chemical ablation of mouse hin­
denervation and may account for changes in muscle gene expression previ­ dlimb SNS, causes a dramatic decrease in muscle NA (Silveira et al.,
ously attributed primarily to motoneuron ablation. 2014). and the release of synaptic vesicles (Rodrigues et al., 2018).
Whether MuRF1 mediates muscle atrophy is controversial. Trans­ Administration of sympathomimetic agents prevents the effects of
genic muscle-specific overexpression of MuRF1 does not cause muscle chemical sympathectomy and reestablishes the compound muscle action
atrophy (Hirner et al., 2008), but the same group demonstrated that potential disrupted by SNS ablation, demonstrating that SNS innerva­
MuRF1 KO dramatically prevents denervation-induced atrophy in tion controls NMJ homoeostasis (Khan et al., 2016). Their use in vivo
mouse TA muscle (Moriscot et al., 2010). More recently, that induction and ex vivo helped us to define the acute and chronic effects of the SNS
of genes associated with the NMJ is blunted in MuRF1 KO mice has been on muscle function, and investigating how postganglionic sympathetic
reported (Furlow et al., 2013). Thus, we cannot rule out MuRF1 axons and their neurotransmitter(s) affect myofiber composition and
participation in denervation-induced muscle atrophy. function clarified the process of aging sarcopenia. β2-AR agonists in­
The increase in MuRF1 after sympathectomy suggests that the SNS crease protein synthesis in denervated muscle and after injury (Gon­
inhibits its expression under physiological conditions. We ruled out the calves et al., 2012), by suppressing proteolysis (Navegantes et al., 2009).
participation of several alternative pathways (Bonaldo and Sandri, While the postsynaptic mechanism is well-established, these findings
2013), and a potential effect of β2-AR (Rodrigues et al., 2018). We also reveal a presynaptic mechanism based on crosstalk between the motor and
found an increase in protein kinase regulatory subunit-I alpha (PKA RIα) sympathetic nervous systems at the NMJ.
but a decrease in the PKA(RIIα)/PKA(RIα) ratio, which we recently The discovery prompted investigation of NMJ presynaptic vesicle
proposed as a marker of the failure of postsynaptic AChR recycling and release in response to acute ex-vivo or chronic, repetitive in-vivo
membrane insertion (Xu et al., 2017). An alternative mechanism for the administration of β-AR agonists. We focused on their effects on extra­
elevation in MuRF1 levels after SNS ablation could be a decrease in cellular Ca2+ concentration, TRPV1, and P/Q- and N-type voltage-
muscle autophagy. However, although lysosome degradation of pro­ activated Ca2+ channels in young adult mice to determine how the
teasomes has been demonstrated in rat liver (Cuervo et al., 1995) its role SNS influences NMJ transmission (Rodrigues et al., 2019).
in the skeletal muscle is not known. If sympathomimetic agents enhance NMJ transmission and reme­
The elevated Hdac4 concentration at the NMJ further supports the diate muscle wasting and strength in aged rodents (Carter et al., 1991;
concept that sympathetic ablation impairs motor axon function (Cohen Ryall and Lynch, 2008), why do endogenously secreted catecholamines
et al., 2007; Moresi et al., 2010). MuRF1 regulation by the canonical fail to maintain muscle mass and force in the absence of significant
Gαi2-myogenin cascade has been reported (Minetti et al., 2014), but changes in β2-AR levels? Recent studies support a novel mechanism by
whether SNS dysfunction is involved is unknown. We found that sym­ which motor and sympathetic neurons interact at the NMJ presynapse
pathectomy downregulates Gαi2, which has been shown to increase (Khan et al., 2016; Rodrigues et al., 2018, 2019; Wang et al., 2020a).
Hdac4 (Minetti et al., 2014), and infecting the TA muscle with the Impaired neuronal crosstalk can explain, at least partially, progressive

5
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

atrophy and weakness with aging (Rodrigues et al., 2019). 3. Does sympathetic neuron regulation of NMJ transmission
We examined whether the SNS regulates motor neuron axon function decline with aging?
and neuromuscular transmission in situ—without any exogenous
intervention, including delivery of catecholamines or sympathomimetic To address this question, we analyzed sciatic-peroneal nerve
agents. Using endogenous neurotransmitters to regulate motoneuron immunolabeling co-registration and NMJ transmission recordings in a
synaptic vesicle release allowed fine-tuning of the acute stress response novel mouse ex-vivo preparation. The sympathetic-peroneal nerve/
under physiological conditions. Our comprehensive approach included lumbricalis muscle preparation (SPNL) maintains the anatomical
nerve immunolabeling co-registration and electrophysiological re­ complexity of the relationship between the sympathetic and motor
cordings in a novel mouse ex-vivo preparation that preserves the nervous systems. This approach unveils the molecular substrate that
complexity of the interaction between motoneuron and SNS at the NMJ accounts for the influence of endogenous sympathetic neurons on
and muscle innervation in the living mouse. Defining the mechanisms by motoneuron-muscle transmission in young mice and older rodents,
which the SNS regulates neuromuscular properties may have broad health providing a unique opportunity to examine whether sympathetic neuron
implications, particularly if we can learn how to counteract sarcopenia, regulation of NMJ transmission declines with age (Wang et al., 2020a).
which impairs gait and mobility in older adults and people suffering from Compared to young mice, geriatric mice show no significant change
neurodegenerative diseases (Wang et al., 2020a). in NMJ-transmission frequency but higher MEPP amplitude and lower
Fig. 2 shows how sympathetic axon depolarization leads to N-type EPP amplitude and quantal content (Wang et al., 2020a). MEPP fre­
Ca2+ channel activation, Ca2+ influx, and NA release. This neurotrans­ quency and amplitude vary according to the muscle examined. In
mitter then acts on sympathetic axon α2B- and β1-AR to inhibit N-type age-matched CBF-1 mice, MEPP frequency decreased in EDL, soleus, and
Ca2+ channels mainly through Gi and possibly exclusively via G(o)/G(i) EDC but not gluteus maximus or diaphragm muscles, while MEPP
and activate them through Gαs. N-type VGCC Ca2+ influx levels may amplitude increased in all but diaphragm (Banker et al., 1983).
result from the balance between yohimbine-sensitive α2-AR autor­ In aging lumbricalis NMJs, sympathetic neuron stimulation does not
egulation and β1-AR activation. Other rapid pertussis toxin (PTX)- increase MEPP frequency. Our nerve image co-registration analysis in­
insensitive G-protein pathways may involve NA-independent activating dicates that the strong decline in motoneuron β1-AR expression with
mechanisms (G#) (Mirotznik et al., 2000). Propranolol, a β1/β2-AR aging is the factor limiting its synaptic vesicle release. In contrast to the
blocker, and atenolol, a β1-AR blocker, prevent β1-AR activation and reported increase in β-AR in the cerebral cortex, hypothalamus (Meitzen
diminish NA release at the NMJ (Wang et al., 2020b). et al., 2013), and hippocampus with aging (Santulli and Iaccarino,
Since NA has no effect on miniature end-plate potential (MEPP) 2013), our data show a significant decline in β1-AR in
frequency, end-plate potential (EPP) amplitude, and quantal content in choline-acetyltransferase and β1- and α2B-AR in tyrosine hydroxylase
the presence of propranolol (Rodrigues et al., 2019), we proposed the immunoreactive axons. We proposed that the age-dependent decline in
motoneuron as a novel target of the SNS. We also proposed that Ca2+ β1-AR expression in sympathetic neurons contributes to diminished NA
influx through TRPV1 channels and P/Q-type Ca2+ channels (Cav2.1) release (Wang et al., 2020a).
determines the axoplasmic Ca2+ concentration at rest and during motor For functional recordings, we used the SPNL technique to determine
neuron depolarization, respectively. These data indicate that by acting on whether sympathetic neuron regulation of motoneurons synaptic vesicle
β1-AR, NA modulates TRPV1 and P/Q-type VGCC and, therefore, moto­ release mediated by β1- and α2B-AR is diminished in geriatric mice.
neuron Ca2+ concentration and the likelihood of ACh synaptic vesicle release However, this approach has some limitations. First, SPNL cannot assess
(Fig. 2). the influence of sympathetic relays from the brain and brainstem nuclei

Fig. 2. Downregulation of preterminal adrenergic receptors and its impact on motoneuron synaptic vesicle release with aging. AP: axon potential. Red arrows
indicate decreased function or protein expression with aging. Dashed lines indicate proposed pathways. Adapted from (Rodrigues et al., 2019; Wang et al., 2020a).
AP: action potential; EPP: end-plate potential; NA: noradrenaline; QC: quantal content; tSC: terminal Schwann cell; VGCC: voltage-gated calcium channel.

6
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

on the intermediolateral column of the spinal cord. Second, it cannot 4. Does maintenance of sympathetic neurons attenuate aging
assess the systemic influence of the humoral milieu on the structures sarcopenia?
involved in neuromuscular transmission. Third, repetitive stimulation
might exhaust catecholamine and/or acetylcholine neuron pools. Future The transcription factor Hand2 points to an intervention that will maintain
experiments in anaesthetized, live mice can overcome these limitations. sympathetic neurons and attenuate sarcopenia with aging
SNS activation may influence the activity of peripheral targets other
than skeletal muscle to modify neuromuscular transmission and muscle The process by which sympathetic neurons are maintained during
innervation. Optogenetic studies targeting specific areas of the SNS development has been amply investigated (Rohrer, 2011). However, the
should address this question (see below). factors that sustain its integrity into old age are unknown. The differ­
Older adults younger than 75 show modest effects of aging on basal entiation of neural crest cells into NA sympathetic neurons is complex
SNS activity (Ng et al., 1994). Above 75, concentrations of basal plasma and requires a network of transcription factors. One of them, Hand2,
NA and NA responses to a stressor were higher than did either younger plays a prominent role in adult sympathetic neuron maintenance of NA
subjects (Barnes et al., 1982; Esler et al., 1995; Hoeldtke and Cilmi, genes and cell survival as demonstrated by axotomy (Pellegrino et al.,
1985; Mazzeo et al., 1997; Palmer et al., 1978; Veith et al., 1986). 2011) and in conditional Hand2 KO sympathetic neurons (Stanzel et al.,
Exaggerated increased basal plasma NA suggests advanced age has 2016). The Hand2 gene encodes a protein in the basic helix-loop-helix
specific effects on the sympathoneural and sympathoadrenomedullary family of transcription factors. Eliminating it in adult sympathetic
components of the SNS (Pascualya et al., 1999), Both increased release neurons reduces expression of the genes involved in synapse develop­
and decreased clearance would account for high NA levels with ment and function (Stanzel et al., 2016). Moreover, in vitro knockdown
advanced age (Pascualya et al., 1999). SNS hyperactivity with aging is of Hand2 in differentiated, postmitotic neurons leads to a selective loss
associated with ARs desensitization, downregulation, and changes in of TH and DBH with no effect on generic neuronal genes such as
subtypes expression patterns (Insel, 1993). Cell-signaling molecules, neuron-specific class III beta-tubulin (Tuj1), embryonic lethal, abnormal
such as GPRC kinases, are responsible for β-AR phosphorylation, vision-like RNA-binding protein level 3 (ELAVL3 or HuC) and superior
desensitization, and downregulation (Urasawa et al., 1991), which may cervical ganglia (Schmidt et al., 2009). Conditional Hand2 knockout in
contribute to the dysfunction of the target (Pfleger et al., 2019); for Hand2-DBHCre mouse embryos caused a selective reduction of TH and a
example, the age-dependent impairment of NMJ transmission in massive reduction of proliferating, DBH-expressing immature neurons
response to sympathetic stimulation. β1-AR is among the most studied (Schmidt et al., 2009). Despite the detailed examination of Hand2’s
AR subtypes, probably due to its role in excitation-contraction coupling function in neurogenesis, and noradrenergic differentiation in embry­
in normal and diseased hearts. Chronic ischemic heart failure, common onic, postnatal, and adult mice (Rohrer, 2011), analysis of its pivotal
in older adults, is associated with decreased β1-AR expression after a role in sympathetic neuron genotype and phenotype in aged mammals is
period of increased expression that also increases cAMP and missing and necessary for its potential to attenuate or reverse
protein-kinase A. Phosphorylated GATA4 then mediates the upregula­ sarcopenia.
tion of miRNA let7, which directly targets ADRB1 mRNA 3’UTR to
repress β1-AR (Du et al., 2017). Whether this mechanism operates in Hand2 expression declines throughout the mouse’s life, but inducing it even
distal axons is unknown. Overall, regulation of β-AR expression in the at advanced ages preserves the number and size of paravertebral
nervous system and especially the nerve is unclear. sympathetic ganglia neurons
Our data on the age-dependent decline in α2-AR levels are consistent
with their diminished responsiveness in prejunctional noradrenergic Hand2 plays a prominent role in adult sympathetic neurons’ adren­
nerve terminals reported in the cardiovascular system (Docherty, 2002). ergic differentiation and maintenance (Doxakis et al., 2008; Tsarovina
However, the mechanism leading to the age-dependent decline in their et al., 2010). Fig. 3 shows the convergence of a hierarchical network
expression in the spinal nerve remains unknown. comprised of the bone morphogenetic proteins (BMPs), paired-like ho­
In summary, the age-dependent decline in sympathetic axon β1 and meobox 2b (Phox2b), achaete-scute family BHLH transcription factor 1
α2B and motoneuron β1-AR accounts for the decline in EPP amplitude (Ascl1), and the insulinoma-associated-1 (Insm1) that regulates its
and synaptic vesicle quantum content (Fig. 2). expression. GATA2 and its downstream GATA3 transcription factors are
also required for the survival of embryonic and adult sympathetic neu­
The pharmacological vs the physiological approach to restor SNS influence rons (Tsarovina et al., 2010), but their role in old age has not been
on NMJ transmission explored.
Adult nerve injury suppresses Hand2, which directly regulates DBH
Maintaining or recovering sympathetic muscle innervation
throughout life is undoubtedly preferable to chronic use of AR phar­
macological agents. Drug administration cannot mimic the complex,
concerted tissue response to endogenous sympathetic neurotransmit­
ters, which depends on the AR subtype’s repertoire and location and the
intricate relationship between central and peripheral nervous system
relays. Loss of receptor selectivity, tolerance, and tachyphylaxis can also
trigger adverse effects. Moreover, pharmaceutical compounds reach
stable levels in blood, precluding any adaptation to physiological or
pathological challenges. Thus, we cannot prevent or reverse age-related
decline in skeletal muscle mass and force until we develop interventions
that target the molecular mechanisms governing NMJ instability and muscle
denervation. Fig. 3. The transcription factor Hand2 regulates noradrenergic differentiation
and sympathetic neuron maintenance. Relationship among the bone morpho­
genetic protein (BMP)-induced transcription factors paired-like homeobox 2b
(Phox2b), Achaete-Scute Family BHLH Transcription Factor 1 (Ascl1),
Insulinoma-associated 1 (Insm1), and Hand2 (black arrows). Adapted from
(Rohrer, 2011). Ascl1: Achaete-scute homolog 1; BMP: bone morphogenetic
protein; Hand2: heart and neural crest derivative 2; Insm1: insulinoma asso­
ciated 1; Phox2b: paired-like homeobox 2b.

7
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

expression (Rychlik et al., 2005) and, most important, the genes con­ Sympathetic denervation of various target organs
trolling synaptic and neurotransmission functions in adult sympathetic
neurons (Stanzel et al., 2016). Based on the reported role of the tran­ In old mice, we found that blood vessels were still surrounded by
scription factors supporting developing and adult autonomic neurons sympathetic axons, but their projections toward myofibers and NMJs
(Stanzel et al., 2016), we examined whether inducing HAND2 expres­ were fewer, depriving the muscle of the dense sympathetic network
sion exclusively in sympathetic neurons would attenuate (a) motor observed in young mammals (Rodrigues et al., 2018, 2020; Straka et al.,
denervation, (b) impaired NMJ transmission, and (c) loss of muscle mass 2018). The skeletal muscle does not seem to be the only organ pro­
and function in old mice. We delivered a viral vector carrying Hand2 gressively losing sympathetic innervation density and complexity, since
expression or an empty vector (EV) exclusively to sympathetic neurons. blood vessels and the heart have been reported to exhibit marked
Since Hand2 expression is restricted to the peripheral SNS (Hendershot deprivation of sympathetic axons with aging (Brown et al., 2019; Li
et al., 2008), we ruled out any influence of central sympathetic nuclei on et al., 2003; Rengo et al., 2016). The pulmonary nervous system consists
our measures. of afferent and efferent nerve fibers that help to maintain organ ho­
Neurons from 22-month-old EV-treated mice were not immunore­ meostasis (Brandenberger and Mühlfeld, 2017). In the lower airways,
active to Hand2 antibody probably because they had less than 20 aging is associated with a reduction in noradrenergic innervation of the
percent of the transcript recorded at postnatal day-5 (Rodrigues et al., bronchial tree (Ricci et al., 1997) and a decrease in vasoactive intestinal
2020). Sympathetic neuron hypermethylation may account, at least peptide concentration and nerve fiber density (Geppetti et al., 1988).
partially, for this age-dependent decrease in Hand2 gene transcription, Evidence for a decrease in sympathetic control of intestinal function in
since CpG islands, but not C combined with any other nucleotide (CH or aged rodents has been reported (Baker et al., 1991), and the timing
CHH) exhibit hypermethylation in paravertebral sympathetic ganglia aligns with the onset and rate of myenteric cell loss (Cowen et al., 2000;
neuron with aging (Rodrigues et al., 2020). Epigenetic silencing of El-Salhy et al., 1999; Phillips and Powley, 2001, 2007). Noradrenergic
Hand2 has been reported in other territories in older adults, for instance networks are abundant in young adult rats, but in aged rats, noradren­
in the development of endometrial cancer, which occurs in older women ergic innervation is less dense, and splenic NA levels are significantly
(Jones et al., 2013). Hand2 undergoes age-associated DNA hyper­ lower. A relationship between diminished noradrenergic innervation
methylation in epithelial tissues (Maegawa et al., 2010). The mouse and reduced immune responsiveness in aging mammals has been pro­
intestine, a tissue densely innervated by the SNS, shows a high rate of posed (Felten et al., 1987).
Hand2 hypermethylation as a function of age, suggesting that Hand2 can The mechanism by which organs lose sympathetic axon terminals is
be hypermethylated in other cells as well (Maegawa et al., 2010). incompletely understood. During development, target tissues secrete
Sustained Hand2 expression in sympathetic neurons in old mice trophic factors to regulate axon branching at sympathetic nerve termi­
preserves ganglia sympathetic neurons size to a significant extent, which nals, an essential step in forming functional connections. Compart­
indicates that Hand2 ameliorates the loss of ganglia neurons with aging mentalized neuronal cultures revealed that the autocrine Wnt5a-
(Stanzel et al., 2016). Conditional deletion of Hand2 reveals its critical (retinoic acid-related orphan receptor [ROR]) signaling loop directs
functions in neurogenesis during development (Hendershot et al., 2008) sympathetic axon branching during target innervation (Ryu et al.,
and adulthood (Stanzel et al., 2016). Preserving the number and size of 2013). Although Wnt5a induced β-catenin nuclear translocation in
sympathetic neurons in Hand2-treated mice extends and enriches the vascular smooth muscle cells (VSMCs) from young and old mice, CREB
complexity of sympathetic muscle innervation with aging. phosphorylation and Wnt1-inducible signaling pathway protein-1
The increase in phosphorylated PKA RI and mechanistic target of (WISP-1) upregulation did not occur in old VSMCs. Wnt5a-mediated
rapamycin complex 1 (mTORC1) with Hand2 increases phosphorylation survival was lost with aging due to impaired CREB activation and
of the cAMP-response element binding protein CREB modulator- CREM, WISP-1 regulation. Better understanding of the effect of age on Wnt
which translocates to the nucleus. Both CREB and CREM, bind a specific signaling may identify targets to promote VSMC survival in elderly pa­
cis element - cAMP-response element (CRE) - in the promoter region of tients (Brown et al., 2019). Due to the relevance of skeletal muscle
skeletal muscle target genes (Zheng et al., 2002). In the Hand2-treated sympathetic innervation, we tested whether maintaining postganglionic
mice, upregulation of Crtc3, a member of the CREB regulated tran­ sympathetic neurons by Hand2 expression prevents or attenuates NMJ
scription co-activator gene family, is consistent with activation of transmission and muscle innervation.
cAMP-responsive genes. CREM, a top upstream regulator of genes
controlled by sympathetic neurons expressing Hand2 in old age. CREM Hand2 expression enhances NMJ transmission and muscle innervation
targets include the TH and dHand, two genes essential for SN function
and maintenance (Rauen et al., 2013). In summary, low levels of PKA RI Sympathetic neuron Hand2 enhances spontaneous and evoked NMJ
phosphorylation (Xu et al., 2017), together with genomic hyper­ transmission and prevents myofiber motor and sympathetic denerva­
methylation may account for the age-dependent decrease in Hand2 gene tion. It also preserves the complexity of muscle sympathetic innervation
transcription. and its proximity to the NMJ. Fragmentation and simplification of the
postterminals are hallmarks of age-dependent deterioration of skeletal
Hand2 increases skeletal muscle mass and force and whole-body strength muscle innervation (Li and Thompson, 2011), while sustained Hand2
but not spontaneous mobility or endurance expression by sympathetic neurons preserves their molecular integrity
and morphology. Hand2-treated mice had fewer small postterminal
The increase in muscle mass in Hand2- compared to EV-treated mice fragments than the EV group and showed an approximately five-fold
was associated with enhanced muscle strength, which we attribute to decrease when the number of fragments was normalized to the muscle
the increase in cross-sectional area (CSA) of type-II fibers in extensor area. The wider range of postterminal fragment sizes in the Hand2 group
digitorum longus (EDL) muscle, and type-I and type-II fibers in soleus indicates that, to a significant degree, muscle sympathetic innervation
muscle (Rodrigues et al., 2020). ameliorates fragmentation.
In the EV- compared to the Hand2-treated mice, electrical stimula­ Hand2 significantly preserves skeletal muscle sympathetic innerva­
tion of distal peroneal nerves resulted in unsustained and weaker lum­ tion, as shown by (a) higher TH levels in nerve and muscle, detected by
bricalis muscle peak force. However, this effect was more pronounced in immunoblot; (b) longer muscle sympathetic axon path; and the shorter
response to muscle activation through the nerve. These data indicate distance between sympathetic neurons and NMJs, confirmed by
that in addition to muscle sympathetic innervation, aging impairs motor immunohistochemistry. NMJ pre- and postterminal co-localization and
innervation and NMJ transmission, and Hand2 expression in sympa­ quantification of neurofilament (NF) path length per muscle area shows
thetic neurons rescues them significantly (Rodrigues et al., 2020). that the preservation of sympathetic innervation enhances myofiber

8
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

motor innervation. The mechanism underlying preservation of muscle skeletal muscle (Pigna et al., 2018), show that it preserves skeletal
sympathetic and motor innervation is uncertain; however, low protein muscle structure following long-term denervation, suggestging that its
phosphatase-2 (PP2A), and protein phosphatase-1 (PP1) levels and more role in muscle denervation is more complex than previously thought. It
NF-heavy chain (NFH-), NF-medium chain (NFM-), and NF-light chain might involve oxidative stress, the ubiquitin-proteasome system, and
(NFL) subunit phosphorylation indicate that NF phosphorylation plays a autophagy (Pigna et al., 2018).
role in stabilizing muscle motor innervation in Hand2-treated mice. This Hand2 also downregulates myogenin and myoD, both associated
conclusion is further supported by confocal and electron microscopy with denervation in gastrocnemius and tibialis anterior muscles. Myo­
studies on muscle sympathetic ablation, which show compromised NF genin and myoD genes are regulated by electrical activity (Eftimie et al.,
phosphorylation and disorganized axon NF and microtubules (Rodri­ 1991; Tang and Goldman, 2006). Downregulating these genes reduces
gues et al., 2020, 2019). MuRF1 and atrogin-1. MuRF1 is expressed in both type-I and type-II
Additionally, Hand2 upregulates the genes encoding critical com­ fibers, but predominantly the latter; it is highly enriched in the post­
ponents of NMJ and myofiber innervation, including Nrcam, a neural terminal; interacts with AChR in endocytic structures (Rudolf et al.,
cell-adhesion molecule, its product, NCAM, which is a marker of muscle 2013) and participates in muscle trophism and maintenance (Moriscot
denervation/reinnervation (Messi et al., 2016), and Nrxn1, a presyn­ et al., 2010). Consistently, Hand2-induced preservation of muscle
aptic membrane cell-adhesion molecule that primarily binds to neuro­ sympathetic innervation downregulates both MuRF1 and atrogin and
ligins, forming a complex required for efficient neurotransmission prevents AChR endocytosis.
(Hadley et al., 2006). Hand2 also upregulates the In old mice, the PKA RIα subunit is largely removed from the NMJ,
microtubule-associated protein-2 (MAP2) gene, which may stabilize the while PKA RIIα and RIIβ are enriched there (Xu et al., 2017). Induced
microtubules against depolimerization, synapse differentiation inducing sustained levels of Hand2 in sympathetic neurons, like other in­
1 like (Sindig1l), which plays a role in the shaping of synaptic specificity terventions to counter muscle denervation (Xu et al., 2017), upregulates
(Cadwell et al., 2016), and the GDNF receptor Gfra1, expressed at PKA RI in the TA muscle, and downregulates PKA RII and the PKA
myelinated nerves and NMJ. Both GDNF and Gfra1 play a crucial role in RII/PKA RI ratio in both GA and TA muscles. These results support the
muscle innervation and GDNF signaling (Hase et al., 1999; Henderson hypothesis that muscle sympathetic innervation prevents activation of the
et al., 1994; Rodrigues et al., 2020). These results indicate that Hand2 signaling cascade reported in skeletal muscle denervation (Moresi et al.,
expression enhances NMJ transmission and muscle innervation in the old. 2010), by maintaining Gαi2 expression.

G protein-coupled receptor (GPCR) signaling plays a pivotal role in Mechanisms by which Hand2 prevents age-related decline in muscle mass
sympathetic neuron-mediated myofiber maintenance and force. Role of Akt, atrogins, inflammation, mTORC1, and autophagy
(Fig. 4)
The GPCR constitute the largest family of membrane receptors. It is
responsible for cell signaling upon binding to hormones, neurotrans­ The protein kinase B (a.k.a. Akt) promotes myotube hypertrophy by
mitters, or ions. Notably, many pharmacological agents target these stimulating protein synthesis through mTOR activity while inhibiting
receptors, stressing their involvement in pathophysiological conditions the forkhead box FoxO to reduce MuRF1 abundance, preventing
and therapeutics (Rosenbaum et al., 2009). In contrast to stimulating Gs, myofibrillar protein turnover (Rommel et al., 2001). Hand2 treatment
the inhibitory subunit Gαi2 isoform blocks adenylate cyclase activation increases muscle Akt phosphorylation, a critical step in the activation of
and cAMP production. Physiological or pathological circumstances that the noncanonical β-AR pathway, which leads to FoXO1 phosphorylation
decrease levels of sympathetic neurotransmitter or Gαi2 expression and exclusion from the nucleus and downregulation of MuRF1 and
prevents the myogenin- and MuRF1-mediated pro-cachectic action of atrogin/MAFbx (Rodrigues et al., 2020). This mechanism, together with
tumor necrosis factor alpha (TNF-α) and myofiber atrophy (Minetti downregulation of the Hdac4-myogenin-MyoD-atrogin/MuRF1
et al., 2014). Minetti and coworkers showed that Gαi2 induces muscle signaling cascade (see above), reduces atrogin expression to prevent
hypertrophy by promoting mTOR and protein synthesis in an muscle atrophy (Ryall et al., 2008).
Akt-independent mechanism, while inhibiting atrophy in response to Activation of the transcription factor nuclear factor kappa B (NF-κB)
TNF-α. Ablation of muscle sympathetic innervation decreases muscle NA is a fulcrum in the processes that mediate muscle atrophy. The signifi­
(Silveira et al., 2014), and Gαi2 protein levels leading to muscle atrophy cance of NF-κB as a key regulator of muscle atrophy has been empha­
(Rodrigues et al., 2018). The model of muscle sympathetic ablation sized by several in vivo studies, which have demonstrated that NF-κB-
shows downregulation of Gαi2, which activates the targeted therapies can abrogate muscle atrophy (Thoma and Lightfoot,
Hdac4-myogenin-MyoD cascade to raise MuRF1 levels and increase 2018). NFκB signaling plays a role in age-related skeletal muscle atrophy
AChR destabilization (Rodrigues et al., 2019). Note that both experi­ probably by activating the senescence-associated secretory phenotype
mental sympathetic ablation and aging can cause vasculature denerva­ (Salminen et al., 2009). Moreover, Hand2 decreased the inhibitor IκB
tion, which might influence the deterioration of muscle structure and and NFκB phosphorylation (Rodrigues et al., 2020). These results sup­
function. However, muscle flow recordings have shown that the effect of port our RNA-sequencing analysis showing that Hand2-induced toll-like
epinephrine on neuromuscular transmission is independent of receptor 5 (Tlr5) downregulation decreases IκB activation, leading to
concomitant vascular changes produced by the catecholamine (Bowman nuclear exclusion of NFκB and preventing activation of the canonical
and Raper, 1966). These data indicate that catecholamines trigger intra­ pro-inflammatory pathway. Consistently, downregulation of
myocellular signaling independently of their vasoactive function. TNF-related-weak-inducer-of-apoptosis (TWEAK) and Toll-like recep­
tor-1 (TLR-1) and interleukin (IL) 16, which play roles in the innate
Sympathetic neuron Hand2 prevents Gαi2 downregulation with aging immune system and the cell cycle, respectively, contribute to SNS
regulation of inflammation and immune-system homeostasis (Révész
We found that Hand2 expression in sympathetic neurons prevents et al., 2014). Cytokines downregulate Hand2 in cultured sympathetic
the decrease in Gαi2 levels with aging and activation of the signaling neurons, while Hand2 overexpression rescues the noradrenergic
cascade that has been associated with muscle motor denervation phenotype. These observations support a third mechanism for decreased
(Minetti et al., 2014). It also lowers Hdac4 concentration to improve Hand2 transcription with aging, in addition to reduced levels of PKA RI
motor-axon function (Cohen et al., 2007; Moresi et al., 2010). Hdac4 (Xu et al., 2017) and Hand2 genomic DNA hypermethylation.
mediates skeletal muscle response to denervation, suggesting its inhi­ Rapamycin-induced mammalian target of rapamycin complex 1
bition as a target for treating neurogenic muscle atrophy. However, (mTORC1) inhibition prevents age-related muscle loss, while its chronic
studies in mutant mice where HDAC4 is specifically deleted in the activation advances muscle damage and loss (Tang et al., 2019),

9
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

Fig. 4. Schematic representation of Hand2-induced expression in sympathetic neurons and its impact on skeletal muscle sympathetic and motor innervation in old
mice. Selective expression of Hand2 in the mouse peripheral sympathetic system from middle- through old-age increases muscle mass and force by (a) regulating
skeletal muscle sympathetic and motor innervation; (b) improving AChR stability and NMJ transmission; (c) preventing inflammation and myofibrillar protein
degradation; (d) increasing autophagy; and (e) probably enhancing protein synthesis. (Rodrigues et al., 2020). AChR: acetylcholine receptor; CREB: cAMP response
element-binding protein; CREM: cAMP Responsive Element Modulator; FoXO1: Forkhead box protein O1; IκB: inhibitor of nuclear factor kappa B; IML: inter­
mediolateral column; LC-I/LC-II: microtubule-associated protein light chain; mTOR: the mechanistic target of rapamycin; NA: noradrenaline; NFκB: nuclear factor
kappa-light-chain-enhancer of activated B cells; NFH: neurofilament heavy chain; NFL: neurofilament light polypeptide; NFM: neurofilament medium chain; Nrcam:
Neuronal cell adhesion molecule; Nrxn1: neurexin-1-alpha; p62: 62 kDa protein; PKA: protein kinase A; TH: tyrosine hydroxylase; TSC2: tuberous sclerosis complex
2; TLR5: Toll-like receptor 5.

inhibiting constitutive and starvation-induced autophagy in skeletal autophagy (Laplante and Sabatini, 2012).
muscle (Castets et al., 2013). We found in both gastrocnemius and Does increased macroautophagy improve skeletal muscle mass and/
tibialis anterior muscles, that Hand2 treatment increases mTORC1 or function in old mice? One common feature of all age-related changes
phosphorylation at serine 2448, fine-tuning its activity at the “repressor at the tissue level is the accumulation of damage and harmful modifi­
domain (Chiang and Abraham, 2005; Figueiredo et al., 2017). Deleting cations in DNA, proteins, lipids, and cellular organelles; autophagic ef­
this site strongly activates mTORC1 (Sekulic et al., 2000). Increased ficiency declines over time, and intracellular waste products amass
phosphorylation at the repressor domain prevents Unc-51-like kinase (Salminen et al., 2009). Robust genetic evidence across species supports
(ULK) and autophagy-related-13 (ATG13) phosphorylation, activating the inhibition of autophagy in aging (Carnio et al.; Rubinsztein et al.,

10
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

2011). Analysis of muscle autophagy showed lower p62 and higher motor innervation remains unknown. Elucidating the functional role of
ATG-7 levels and LC3-I lipidation in the Hand2 group than the EV group. brainstem sympathetic neuron projections to preganglionic noradrenergic
Atg7 ablation induces NMJ degeneration and precocious aging (Carnio neurons will help to define the coordinated response of the visceral and
et al., 2014), which suggests that high ATG-7 levels are required to neuromuscular system to physiological and pathological challenges.
improve muscle autophagy. Autophagy selectively degrades the
LC3-binding adaptor p62 protein (sequestosome-1 [SQSTM1]) (Ichi­ 6. Conclusions and future directions
mura et al., 2008), and since a drop in p62 is associated with activation
of the autophagic process, it serves as a readout of autophagic flux, The SNS regulates several primary synaptic and non-synaptic events.
together with LC3-I conversion to LC3-II (Bjorkoy et al., 2005; Sei­ Inducing Hand2 expression in sympathetic neurons from middle-aged
benhener et al., 2004). Non-significant changes in lysosome-associated mice improves their NMJ transmission, myofiber innervation, muscle
membrane protein 2 (Lamp2) and heat-shock cognate protein of 70 mass, and function maintenance as they age. Despite recent progress in
kDa (HSC70) levels indicate that macroautophagy, but not our understanding of the process by which the SNS regulates skeletal
chaperone-mediated autophagy, is activated by sustained Hand2 muscle structure and function with aging, several questions remain.
expression in sympathetic neurons with aging. Note that direct stimu­ Hand2 is critical for sympathetic neurons maintenance in adult mice,
lation of sympathetic neurons activates muscle postsynaptic β2-AR, but its expression is regulated by transcription factors, such as BMPs,
cAMP production, and import of the transcriptional co-activator ASCL1, Phox2b, and Insm1 (Rohrer, 2011), and their expression levels
peroxisome proliferator-activated receptor γ-coactivator 1α and function have not been examined in aging organisms. Also, despite
(PPARGC1A) into the myonucleus (Khan et al., 2016), which increases the beneficial impact of Hand2 induced expression on muscle innerva­
autophagy (Vainshtein et al., 2015). tion, we do not know whether Hand2 expression induced for a longer
In summary, preventing decline in peripheral sympathetic neuron time, and up to very old age (geriatric) will still ameliorate sarcopenia.
Hand2 expression with aging attenuates sympathetic and motor muscle Treated middle-aged mice should be examined not only 6-months, but
denervation and aging sarcopenia (Rodrigues et al., 2020). 10–12 months after Hand2- or EV-treatment.
Whether mechanisms other than hypermethylation influence Hand2
5. Do central sympathetic neuron relays influence sympathetic expression in aging sympathetic neurons remains unknown. Although
and motor muscle innervation? some of these mechanisms are discussed above, the potential role of the
recently identified enhancer RNAs (eRNAs) should be investigated.
The studies reviewed above indicate that activation of peripheral eRNAs are a novel class of short, noncoding RNAs transcribed from
sympathetic neurons increase NMJ transmission. However, we must still active enhancer regions in the genome. They regulate downstream genes
define the specific neuronal groups in the peripheral and central nervous promoting DNA looping/interactions, regulating chromatin accessi­
system that account for SNS influence on NMJ transmission; for bility, and interfering in transcription, which is clinically relevant (Li
example, whether or how postganglionic sympathetic neurons regulate et al., 2014, 2016; Mora et al., 2016).
synaptic vesicle release in vivo or central autonomic relays regulate NMJ We do not know whether the age-dependent decline in central
transmission. Previous studies have associated the pontine locus autonomic nucleus composition and/or function accounts for skeletal
coeruleus (LC) (a.k.a. A6 nucleus) with human mobility, motility, and motor denervation and sarcopenia. A decrease in LC neuron density is
skeletal muscle physiology (Buchman et al., 2012; Del Tredici and associated with impaired mobility in older adults, which indicates that
Braak, 2013; Dobbins and Feldman, 1994; Jacobs et al., 1991; Robinson, central sympathetic relays may play a critical role in physical activity
1978; Xiang et al., 2014). A series of experiments in our laboratory (Buchman et al., 2012). Our research shows that postganglionic, but not
focused on lumbar postganglionic sympathetic neurons, and the LC, a LC neurons regulate NMJ transmission. Whether other brainstem
subdivision of which projects to preganglionic neurons in the spinal cord noradrenergic neuron groups more densely projected to the IML influ­
intermediolateral (IML) column to address this issue. Combining opto­ ence mammalians’ motility is an open question for future research on
genetics (a technique that involves the use of light to control neuron the mechanisms of aging sarcopenia.
activity that have been genetically modified to express light-sensitive Recent findings can be translated to human sarcopenia. Non-
ion channels) with NMJ transmission recordings provided a unique pathogenic viral vectors with long-lasting expression, recently tested
opportunity to determine the precise role of postganglionic sympathetic in rodents, could be used for safe, efficient gene delivery in humans in
neurons and LC neurons in NMJ transmission in adult living mice the near future (Rodrigues et al., 2020). Nanomaterials or lipid micelles
(Rodrigues et al., 2020). are alternative approaches to deliver Hand2 to ganglionic sympathetic
We crossed Ai32(RCL-ChR2(H134R/EYFP)) (Madisen et al., 2012) neurons.
and TH-Cre (Savitt et al., 2005) to create a mouse model that would
express channelrodhopsin-2 (ChR2) in the central and peripheral Declaration of Competing Interest
noradrenergic neurons. We concluded that in vitro and in living mice
sympathetic axon optostimulation, mediated by β1-ARs, enhances NMJ The authors declare that they have no conflicts of interest.
transmission and LC does not, despite its projections to the IML
(Rodrigues et al., 2020), perhaps because they do not establish en passant Acknowledgments
synapse with preganglionic sympathetic neurons. Confocal z-stack pro­
jections of sections through the spinal cord showed that the injected The National Institutes of Health grants R01AG057013 and
AAV2/rh8-PRSx8-EGFP-W viral vector appears at both the dorsal horn R01AG057013-02S1, and the Wake Forest Claude D. Pepper Older
and the IML (Bruinstroop et al., 2012). Also, we stimulated LC at fre­ Americans Independence Center (P30-AG21332) supported this work
quencies below and above 5 Hz with no obvious response, which in­ and the original research published by the Delbono laboratory.
dicates that by innervating the muscle spindle (Radovanovic et al.,
2015), LC modulates posture (Carter et al., 2010), but not NMJ References
transmission.
Andersson, A.M., Olsen, M., Zhernosekov, D., Gaardsvoll, H., Krog, L., Linnemann, D.,
Detailed analysis of central neuroanatomical circuitries show that
Bock, E., 1993. Age-related changes in expression of the neural cell adhesion
several brainstem noradrenergic regions project to the IML, including molecule in skeletal muscle: a comparative study of newborn, adult and aged rats.
the noradrenergic bulbospinal A1 and A2 areas (Rodovalho et al., 2020) Biochem. J. 290 (Pt 3), 641–648.
and pontine A5, A6, and A7 (Bruinstroop et al., 2012). The functional
relevance of these CNS projections for NMJ transmission and muscle

11
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

Askmark, H., Eeg-Olofsson, K., Johansson, A., Nilsson, P., Olsson, Y., Aquilonius, S., Carlson, B.M., Borisov, A.B., Dedkov, E.I., Khalyfa, A., Kostrominova, T.Y.,
2001. Parkinsonism and neck extensor myopathy: A new syndrome or coincidental Macpherson, P.C., Wang, E., Faulkner, J.A., 2002. Effects of long-term denervation
findings? Arch. Neurol. 58, 232–237. on skeletal muscle in old rats. J. Gerontol. A Biol. Sci. Med. Sci. 57, B366–374.
Baker, D.M., Watson, S.P., Santer, R.M., 1991. Evidence for a decrease in sympathetic Carnio, S., LoVerso, F., Baraibar, Martin A., Longa, E., Khan, Muzamil M., Maffei, M.,
control of intestinal function in the aged rat. Neurobiol. Aging 12, 363–365. Reischl, M., Canepari, M., Loefler, S., Kern, H., Blaauw, B., Friguet, B., Bottinelli, R.,
Balice-Gordon, R.J., 1997. Age-related changes in neuromuscular innervation. Muscle Rudolf, R., Sandri, M., 2014. Autophagy impairment in muscle induces
Nerve S5, S83–S87. neuromuscular junction degeneration and precocious aging. Cell Rep. 8 (5),
Banker, B.Q., Kelly, S.S., Robbins, N., 1983. Neuromuscular transmission and correlative 1509–1521.
morphology in young and old mice. J. Physiol. 339, 355–377. Carter, W.J., Dang, A.Q., Faas, F.H., Lynch, M.E., 1991. Effects of clenbuterol on skeletal
Barker, D., Saito, M., 1981. Autonomic innervation of receptors and muscle fibres in cat muscle mass, body composition, and recovery from surgical stress in senescent rats.
skeletal muscle. Proc. R. Soc. Lond. B Biol. Sci. 212, 317–332. Metabolism 40, 855–860.
Barnes, R.F., Raskind, M., Gumbretch, G., Halter, J.B., 1982. The effects of age on the Carter, M.E., Yizhar, O., Chikahisa, S., Nguyen, H., Adamantidis, A., Nishino, S.,
plasma catecholamine response to mental stress in man. J. Clin. Endocrinol. Metab. Deisseroth, K., de Lecea, L., 2010. Tuning arousal with optogenetic modulation of
54, 64–69. locus coeruleus neurons. Nat. Neurosci. 13, 1526–1533.
Beitzel, F., Sillence, M.N., Lynch, G.S., 2007. Beta-Adrenoceptor signaling in Castets, P., Lin, S., Rion, N., Fulvio, S., Romanino, K., Guridi, M., 2013. Sustained
regenerating skeletal muscle after beta-agonist administration. Am. J. Physiol. activation of mTORC1 in skeletal muscle inhibits constitutive and starvation-induced
Endocrinol. Metab. 293, E932–940. autophagy and causes a severe, late-onset myopathy. Cell Metab. 17, 731–744.
Bergmeister, K.D., Gröger, M., Aman, M., Willensdorfer, A., Manzano-Szalai, K., Chiang, G.G., Abraham, R.T., 2005. Phosphorylation of mammalian target of rapamycin
Salminger, S., Aszmann, O.C., 2016. Automated muscle fiber type population (mTOR) at Ser-2448 is mediated by p70S6 kinase. J. Biol. Chem. 280, 25485–25490.
analysis with ImageJ of whole rat muscles using rapid myosin heavy chain Chin, S.O., Rhee, S.Y., Chon, S., Hwang, Y.-C., Jeong, I.-K., Oh, S., Ahn, K.J., Chung, H.Y.,
immunohistochemistry. Muscle Nerve 54, 292–299. Woo, J.-T., Kim, S.-W., Kim, J.-W., Kim, Y.S., Ahn, H.-Y., 2013. Sarcopenia is
Bezakova, G., Lomo, T., 2001. Muscle activity and muscle agrin regulate the organization independently associated with cardiovascular disease in older Korean adults: the
of cytoskeletal proteins and attached acetylcholine receptor (AchR) aggregates in korea national health and nutrition examination survey (KNHANES) from 2009.
skeletal muscle fibers. J. Cell Biol. 153, 1453–1463. PLoS One 8, e60119.
Biral, D., Kern, H., Adami, N., Boncompagni, S., Protasi, F., Carraro, U., 2008. Atrophy- Choi, S.J., Shively, C.A., Register, T.C., Stehle, J., High, K., Ip, E., Feng, X.,
resistant fibers in permanent peripheral denervation of human skeletal muscle. Kritchevsky, S.B., Nicklas, B.J., Delbono, O., 2012. Force generation capacity of
Neurol. Res. 30, 137–144. single muscle fibers of vastus lateralis declines with age and correlates with physical
Bjorkoy, G., Lamark, T., Brech, A., Outzen, H., Perander, M., Overvatn, A., Stenmark, H., function in african green vervet monkeys. J. Gerontol. Biol. Sci. 68 (3), 258–267.
Johansen, T., 2005. p62/SQSTM1 forms protein aggregates degraded by autophagy Cohen, T.J., Waddell, D.S., Barrientos, T., Lu, Z., Feng, G., Cox, G.A., Bodine, S.C., Yao, T.
and has a protective effect on huntingtin-induced cell death. J. Cell Biol. 171, P., 2007. The histone deacetylase HDAC4 connects neural activity to muscle
603–614. transcriptional reprogramming. J. Biol. Chem. 282, 33752–33759.
Blasco, A., Gras, S., Mòdol-Caballero, G., Tarabal, O., Casanovas, A., Piedrafita, L., Cowen, T., Johnson, R., Soubeyre, V., Santer, R.M., 2000. Restricted diet rescues rat
Barranco, A., Das, T., Pereira, S.L., Navarro, X., Rueda, R., Esquerda, J.E., enteric motor neurones from age related cell death. Gut 47, 653–660.
Calderó, J., 2020. Motoneuron deafferentation and gliosis occur in association with Cuervo, A.M., Palmer, A., Rivett, A.J., Knecht, E., 1995. Degradation of proteasomes by
neuromuscular regressive changes during ageing in mice. J. Cachexia Sarcopenia lysosomes in rat liver. Eur. J. Biochem. 227, 792–800.
Muscle 6, 1628–1660. de Waegh, S.M., Lee, V.M., Brady, S.T., 1992. Local modulation of neurofilament
Bloemberg, D., Quadrilatero, J., 2012. Rapid determination of myosin heavy chain phosphorylation, axonal caliber, and slow axonal transport by myelinating Schwann
expression in rat, mouse, and human skeletal muscle using multicolor cells. Cell 68, 451–463.
immunofluorescence analysis. PLoS One 7, e35273. DeAndrade, J., Pedersen, M., Garcia, L., Nau, P., 2018. Sarcopenia is a risk factor for
Bodine, S.C., Latres, E., Baumhueter, S., Lai, V.K., Nunez, L., Clarke, B.A., complications and an independent predictor of hospital length of stay in trauma
Poueymirou, W.T., Panaro, F.J., Na, E., Dharmarajan, K., Pan, Z.Q., Valenzuela, D. patients. J. Surg. Res. 221, 161–166.
M., DeChiara, T.M., Stitt, T.N., Yancopoulos, G.D., Glass, D.J., 2001. Identification of Del Tredici, K., Braak, H., 2013. Dysfunction of the locus coeruleus-norepinephrine
ubiquitin ligases required for skeletal muscle atrophy. Science 294, 1704–1708. system and related circuitry in Parkinson’s disease-related dementia. J. Neurol.
Boeke, J., 1909. Ueber eine aus marklosen Fasern hervorgehende zweite Art von Neurosurg. Psychiatry 84, 774–783.
hypolemmalen Nervenendplatten bei den quergestreiften Muskelfasern der Delbono, O., 1992. Calcium current activation and charge movement in denervated
Vertebraten. Anat. Anz. 35, 481–484. mammalian skeletal muscle fibres. J. Physiol. 451, 187–203.
Bonaldo, P., Sandri, M., 2013. Cellular and molecular mechanisms of muscle atrophy. Delbono, O., 2003. Neural control of aging skeletal muscle. Aging Cell 2, 21–29.
Dis. Model. Mech. 6, 25–39. Delbono, O., 2011a. Excitation-contraction coupling regulation in aging skeletal muscle.
Bonilla, H.J., Messi, M.L., Sadieva, K.A., Hamilton, C.A., Buchman, A.S., Delbono, O., In: Lynch, G.S. (Ed.), Sarcopenia-Age-Related Muscle Wasting and Weakness.
2020. Semiautomatic morphometric analysis of skeletal muscle obtained by needle Mechanisms and Treatment. Springer, Victoria, pp. 113–134.
biopsy in older adults. GeroScience 6, 1431–1443. Delbono, O., 2011b. Expression and regulation of excitation-contraction coupling
Bowman, W.C., Raper, C., 1966. Effects of sympathomimetic amines on neuromuscular proteins in aging skeletal muscle. Curr. Aging Sci. 4 (3), 248–259.
transmission. Br. J. Pharmacol. Chemother. 27, 313–331. Delbono, O., Stefani, E., 1993. Calcium current inactivation in denervated rat skeletal
Brandenberger, C., Mühlfeld, C., 2017. Mechanisms of lung aging. Cell Tissue Res. 367, muscle fibres. J. Physiol. 460, 173–183.
469–480. Delbono, O., O’Rourke, K.S., Ettinger, W.H., 1995. Excitation-calcium release uncoupling
Briguet, A., Courdier-Fruh, I., Foster, M., Meier, T., Magyar, J.P., 2004. Histological in aged single human skeletal muscle fibers. J. Membr. Biol. 148, 211–222.
parameters for the quantitative assessment of muscular dystrophy in the mdx-mouse. Delbono, O., Renganathan, M., Messi, M.L., 1997. Regulation of mouse skeletal muscle L-
Neuromuscul. Disord. 14, 675–682. type Ca2+ channel by activation of the insulin-like growth factor-1 receptor.
Brooks, S.V., Faulkner, J.A., 1994. Skeletal muscle weakness in old age: underlying J. Neurosci. 17, 6918–6928.
mechanisms. Med. Sci. Sports Exerc. 26, 432–439. Distefano, G., Goodpaster, B.H., 2018. Effects of exercise and aging on skeletal muscle.
Brown, B.A., Connolly, G.M., Mill, C.E.J., Williams, H., Angelini, G.D., Johnson, J.L., Cold Spring Harb. Perspect. Med. 8 (3), a029785.
George, S.J., 2019. Aging differentially modulates the Wnt pro-survival signalling Dobbins, E.G., Feldman, J.L., 1994. Brainstem network controlling descending drive to
pathways in vascular smooth muscle cells. Aging Cell 18, e12844. phrenic motoneurons in rat. J. Comp. Neurol. 347, 64–86.
Bruinstroop, E., Cano, G., Vanderhorst, V.G.J.M., Cavalcante, J.C., Wirth, J., Sena- Docherty, J.R., 2002. Age-related changes in adrenergic neuroeffector transmission.
Esteves, M., Saper, C.B., 2012. Spinal projections of the A5, A6 (locus coeruleus), and Auton. Neurosci. 96, 8–12.
A7 noradrenergic cell groups in rats. J. Comp. Neurol. 520, 1985–2001. Doherty, T.J., Vandervoort, A.A., Taylor, A.W., Brown, W.F., 1993. Effects of motor unit
Buchman, A.S., Nag, S., Shulman, J.M., Lim, A.S., VanderHorst, V.G., Leurgans, S.E., losses on strength in older men and women. J. Appl. Physiol. 74, 868–874.
Schneider, J.A., Bennett, D.A., 2012. Locus coeruleus neuron density and Doxakis, E., Howard, L., Rohrer, H., Davies, A.M., 2008. HAND transcription factors are
parkinsonism in older adults without Parkinson’s disease. Mov. Disord. 27, required for neonatal sympathetic neuron survival. EMBO Rep. 9, 1041–1047.
1625–1631. Du, Y., Zhang, M., Zhao, W., Shu, Y., Gao, M., Zhuang, Y., Yang, T., Mu, W., Li, T., Li, X.,
Buller, A.J., Eccles, J.C., Eccles, R.M., 1960a. Differentiation of fast and slow muscles in Sun, F., Pan, Z., Lu, Y., 2017. Let-7a regulates expression of β1-adrenoceptors and
the cat hind limb. J. Physiol. 150, 399–416. forms a negative feedback circuit with the β1-adrenoceptor signaling pathway in
Buller, A.J., Eccles, J.C., Eccles, R.M., 1960b. Interactions between motoneurones and chronic ischemic heart failure. Oncotarget 8, 8752–8764.
muscles in respect of the characteristic speeds of their responses. J. Physiol. 150, Dutta, C., 1997. Significance of sarcopenia in the elderly. J. Nutr. 127, 992S–993S.
417–439. Eftimie, R., Brenner, H.R., Buonanno, A., 1991. Myogenin and MyoD join a family of
Cadwell, C.R., Palasantza, A., Jiang, X., Berens, P., Deng, Q., Yilmaz, M., Reimer, J., skeletal muscle genes regulated by electrical activity. Proc. Natl. Acad. Sci. U. S. A.
Shen, S., Bethge, M., Tolias, K.F., Sandberg, R., Tolias, A.S., 2016. 88, 1349–1353.
Electrophysiological, transcriptomic and morphologic profiling of single neurons Eichmann, A., Brunet, I., 2014. Arterial innervation in development and disease. Sci.
using Patch-seq. Nat. Biotechnol. 34, 199–203. Transl. Med. 6, 252.
Cano-Jaimez, M., Pérez-Sánchez, F., Milán, M., Buendía, P., Ambrosio, S., Fariñas, I., Einsiedel, L.J., Luff, A.R., 1992. Effect of partial denervation on motor units in the ageing
2010. Vulnerability of peripheral catecholaminergic neurons to MPTP is not rat medial gastrocnemius. J. Neurol. Sci. 112, 178–184.
regulated by α-synuclein. Neurobiol. Dis. 38, 92–103. El-Salhy, M., Sandström, O., Holmlund, F., 1999. Age-induced changes in the enteric
Card, J.P., Enquist, L.W., 2012. Use and visualization of neuroanatomical viral nervous system in the mouse. Mech. Ageing Dev. 107, 93–103.
transneuronal tracers. In: Badoer, E. (Ed.), Visualization Techniques: From Esler, M.D., Thompson, J.M., Kaye, D.M., Turner, A.G., Jennings, G.L., Cox, H.S.,
Immunohistochemistry to Magnetic Resonance Imaging. Humana Press, Totowa, NJ, Lambert, G.W., Seals, D.R., 1995. Effects of aging on the responsiveness of the
pp. 225–268. human cardiac sympathetic nerves to stressors. Circulation 91, 351–358.

12
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

Faulkner, J.A., Brooks, S.V., Opiteck, J.A., 1993. Injury to skeletal muscle fibers during Hepple, R.T., 2018. When motor unit expansion in ageing muscle fails, atrophy ensues.
contractions: conditions of occurrence and prevention. Phys. Ther. 73, 911–921. J. Physiol. 596, 1545–1546.
Felten, S.Y., Bellinger, D.L., Collier, T.J., Coleman, P.D., Felten, D.L., 1987. Decreased Hirner, S., Krohne, C., Schuster, A., Hoffmann, S., Witt, S., Erber, R., Sticht, C., Gasch, A.,
sympathetic innervation of spleen in aged fischer 344 rats. Neurobiol. Aging 8, Labeit, S., Labeit, D., 2008. MuRF1-dependent regulation of systemic carbohydrate
159–165. metabolism as revealed from transgenic mouse studies. J. Mol. Biol. 379, 666–677.
Femminella, G.D., Rengo, G., Komici, K., Iacotucci, P., Petraglia, L., Pagano, G., de Hoeldtke, R.D., Cilmi, K.M., 1985. Effects of aging on catecholamine metabolism. J. Clin.
Lucia, C., Canonico, V., Bonaduce, D., Leosco, D., Ferrara, N., 2014. Autonomic Endocrinol. Metab. 60, 479–484.
dysfunction in Alzheimer’s disease: tools for assessment and review of the literature. Hook, P., Li, X., Sleep, J., Hughes, S., Larsson, L., 1999. In vitro motility speed of slow
J. Alzheimers Dis. 42, 369–377. myosin extracted from single soleus fibres from young and old rats. J. Physiol. 520,
Ferrucci, L., Harris, T.B., Guralnik, J.M., Tracy, R.P., Corti, M.C., Cohen, H.J., 463–471.
Penninx, B., Pahor, M., Wallace, R., Havlik, R.J., 1999. Serum IL-6 level and the Hwang, A.B., Brack, A.S., 2018. Chapter Ten - muscle stem cells and aging. In:
development of disability in older persons. J. Am. Geriatr. Soc. 47, 639–646. Sassoon, D. (Ed.), Current Topics in Developmental Biology. Academic Press,
Ferrucci, L., Penninx, B.W., Volpato, S., Harris, T.B., Bandeen-Roche, K., Balfour, J., pp. 299–322.
Leveille, S.G., Fried, L.P., Md, J.M., 2002. Change in muscle strength explains Ichimura, Y., Kominami, E., Tanaka, K., Komatsu, M., 2008. Selective turnover of p62/
accelerated decline of physical function in older women with high interleukin-6 A170/SQSTM1 by autophagy. Autophagy 4, 1063–1066.
serum levels. J. Am. Geriatr. Soc. 50, 1947–1954. Insel, P.A., 1993. Adrenergic receptors, G proteins, and cell regulation: implications for
Figueiredo, V.C., Markworth, J.F., Cameron-Smith, D., 2017. Considerations on mTOR aging research. Exp. Gerontol. 28, 341–348.
regulation at serine 2448: implications for muscle metabolism studies. Cell. Mol. Life Jackson, M.J., 2016. Reactive oxygen species in sarcopenia: Should we focus on excess
Sci. 74, 2537–2545. oxidative damage or defective redox signaling? Mol. Aspects Med. 50, 33–40.
Fish, L.A., Fallon, J.R., 2020. Multiple MuSK signaling pathways and the aging Jacobs, B.L., Abercrombie, E.D., Fornal, C.A., Levine, E.S., Morilak, D.A., Stafford, I.L.,
neuromuscular junction. Neurosci. Lett. 731, 135014. 1991. Single-unit and physiological analyses of brain norepinephrine function in
Frey, D., Schneider, C., Xu, L., Borg, J., Spooren, W., Caroni, P., 2000. Early and selective behaving animals. Prog. Brain Res. 88, 159–165.
loss of neuromuscular synapse subtypes with low sprouting competence in Jang, Y.C., Lustgarten, M.S., Liu, Y., Muller, F.L., Bhattacharya, A., Liang, H., Salmon, A.
motoneuron diseases. J. Neurosci. 20, 2534–2542. B., Brooks, S.V., Larkin, L., Hayworth, C.R., Richardson, A., Van Remmen, H., 2010.
Frontera, W.R., Suh, D., Krivickas, L.S., Hughes, V.A., Goldstein, R., Roubenoff, R., 2000. Increased superoxide in vivo accelerates age-associated muscle atrophy through
Skeletal muscle fiber quality in older men and women. Am. J. Physiol. Cell Physiol. mitochondrial dysfunction and neuromuscular junction degeneration. FASEB J. 24,
279, C611–618. 1376–1390.
Fry, C.S., Lee, J.D., Mula, J., Kirby, T.J., Jackson, J.R., Liu, F., Yang, L., Mendias, C.L., Janig, W., 2012. Complex Regional Pain Syndrome, 3rd ed. Academic, Amsterdam.
Dupont-Versteegden, E.E., McCarthy, J.J., Peterson, C.A., 2015. Inducible depletion Jensen-Daham, C., Waldemar, G., Jensen, T.S., Malmqvist, L., Moeller, M.M.,
of satellite cells in adult, sedentary mice impairs muscle regenerative capacity Andersen, B.B., Høgh, P., Ballegaard, M., 2015. Autonomic dysfunction in patients
without affecting sarcopenia. Nat. Med. 21, 76–80. with mild to moderate Alzheimer’s disease. J. Alzheimer Dis. 47, 681–689.
Furlow, J.D., Watson, M.L., Waddell, D.S., Neff, E.S., Baehr, L.M., Ross, A.P., Bodine, S. Jessen, K.R., Mirsky, R., 2016. The repair Schwann cell and its function in regenerating
C., 2013. Altered gene expression patterns in muscle ring finger 1 null mice during nerves. J. Physiol. 594, 3521–3531.
denervation- and dexamethasone-induced muscle atrophy. Physiol. Genomics 45, Jimenez-Moreno, R., Wang, Z.M., Guerring, R.C., Delbono, O., 2008. Sarcoplasmic
1168–1185. reticulum Ca2+ release declines in muscle fibers from aging mice. Biophys. J. 94 (8),
Garcia-Prat, L., Martinez-Vicente, M., Perdiguero, E., Ortet, L., Rodriguez-Ubreva, J., 3178–3188.
Rebollo, E., Ruiz-Bonilla, V., Gutarra, S., Ballestar, E., Serrano, A.L., Sandri, M., Johnson, H., Mossberg, K., Arvidsson, U., Piehl, F., Hokfelt, T., Ulfhake, B., 1995.
Munoz-Canoves, P., 2016. Autophagy maintains stemness by preventing senescence. Increase in alpha-CGRP and GAP-43 in aged motoneurons: a study of peptides,
Nature 529, 37–42. growth factors, and ChAT mRNA in the lumbar spinal cord of senescent rats with
Geppetti, P., De Rossi, M., Mione, M., Renzi, D., Amenta, F., 1988. Age-related changes in symptoms of hindlimb incapacities. J. Comp. Neurol. 359, 69–89.
vasoactive intestinal polypeptide levels and distribution in the rat lung. J. Neural Jokl, E.J., Blanco, G., 2016. Disrupted autophagy undermines skeletal muscle adaptation
Transm. 74, 1–10. and integrity. Mamm. Genome 27, 525–537.
Goncalves, D.A., Silveira, W.A., Lira, E.C., Graca, F.A., Paula-Gomes, S., Zanon, N.M., Jones, A., Teschendorff, A.E., Li, Q., Hayward, J.D., Kannan, A., Mould, T., West, J.,
Kettelhut, I.C., Navegantes, L.C., 2012. Clenbuterol suppresses proteasomal and Zikan, M., Cibula, D., Fiegl, H., Lee, S.-H., Wik, E., Hadwin, R., Arora, R., Lemech, C.,
lysosomal proteolysis and atrophy-related genes in denervated rat soleus muscles Turunen, H., Pakarinen, P., Jacobs, I.J., Salvesen, H.B., Bagchi, M.K., Bagchi, I.C.,
independently of Akt. Am. J. Physiol. Endocrinol. Metab. 302, E123–133. Widschwendter, M., 2013. Role of DNA methylation and epigenetic silencing of
Gonzalez, A., Kirsch, W.G., Shirokova, N., Pizarro, G., Brum, G., Pessah, I.N., Stern, M.D., HAND2 in endometrial Cancer development. PLoS Med. 10, e1001551.
Cheng, H., Rios, E., 2000a. Involvement of multiple intracellular release channels in Kadhiresan, V.A., Hassett, C.A., Faulkner, J.A., 1996. Properties of single motor units in
calcium sparks of skeletal muscle. Proc. Natl. Acad. Sci. U. S. A. 97, 4380–4385. medial gastrocnemius muscles of adult and old rats. J. Physiol. 493, 543–552.
Gonzalez, E., Messi, M.L., Delbono, O., 2000b. The specific force of single intact extensor Kallen, R.G., Sheng, Z.H., Yang, J., Chen, L.Q., Rogart, R.B., Barchi, R.L., 1990. Primary
digitorum longus and soleus mouse muscle fibers declines with aging. J. Membr. structure and expression of a sodium channel characteristic of denervated and
Biol. 178, 175–183. immature rat skeletal muscle. Neuron 4, 233–242.
Gonzalez-Freire, M., de Cabo, R., Studenski, S., Ferrucci, L., 2014. The Neuromuscular Kamwa, V., Welch, C., Hassan-Smith, Z.K., 2020. The endocrinology of sarcopenia and
Junction (NMJ): aging at the crossroad between nerves and muscle. Front. Aging frailty. Minerva Endocrinol. Online ahead of print.
Neurosci. 6, 208. Kanda, K., Hashizume, K., 1989. Changes in properties of the medial gastrocnemius
Gosztonyi, G., Naschold, U., Grozdanovic, Z., Stoltenburg-Didinger, G., Gossrau, R., motor units in aging rats. J. Neurophysiol. 737–746.
2001. Expression of Leu-19 (CD56, N-CAM) and nitric oxide synthase (NOS) I in Kanda, K., Hashizume, K., 1992. Factors causing difference in force output among motor
denervated and reinnervated human skeletal muscle. Microsc. Res. Tech. 55, units in the rat medial gastrocnemius muscle. J. Physiol. 448, 677–695.
187–197. Kandinov, B., Korczyn, A.D., Rabinowitz, R., Nefussy, B., Drory, V.E., 2011. Autonomic
Greensmith, L., Vrbova, G., 1996. Motoneurone survival: a functional approach. Trends impairment in a transgenic mouse model of amyotrophic lateral sclerosis. Auton.
Neurosci. 19, 450–455. Neurosci. 159, 84–89.
Griffin, J.W., Thompson, W.J., 2008. Biology and pathology of nonmyelinating Schwann Kerman, I.A., Enquist, L.W., Watson, S.J., Yates, B.J., 2003. Brainstem substrates of
cells. Glia 56, 1518–1531. sympatho-motor circuitry identified using trans-synaptic tracing with pseudorabies
Gropp, K.E., 2017. Skeletal muscle toolbox. Toxicol. Pathol. 45, 939–942. virus recombinants. J. Neurosci. 23, 4657–4666.
Hadley, D., Murphy, T., Valladares, O., Hannenhalli, S., Ungar, L., Kim, J., Bucan, M., Khan, M.M., Lustrino, D., Silveira, W.A., Wild, F., Straka, T., Issop, Y., O’Connor, E.,
2006. Patterns of sequence conservation in presynaptic neural genes. Genome Biol. Cox, D., Reischl, M., Marquardt, T., Labeit, D., Labeit, S., Benoit, E., Molgó, J.,
7, R105. Lochmüller, H., Witzemann, V., Kettelhut, I.C., Navegantes, L.C.C., Pozzan, T.,
Hase, A., Suzuki, H., Arahata, K., Akazawa, C., 1999. Expression of human GFR alpha-1 Rudolf, R., 2016. Sympathetic innervation controls homeostasis of neuromuscular
(GDNF receptor) at the neuromuscular junction and myelinated nerves. Neurosci. junctions in health and disease. Proc. Natl. Acad. Sci. 113, 746–750.
Lett. 269, 55–57. Khuzakhmetova, V., Bukharaeva, E., 2020. Adrenaline facilitates synaptic transmission
Hashizume, K., Kanda, K., Burke, R., 1988. Medial gastrocnemius motor nucleus in the by synchronizing release of acetylcholine quanta from motor nerve endings. Cell.
rat: age-related changes in the number and size of motoneurons. J. Comp. Neurol. Mol. Neurobiol. Online ahead of print.
269, 425–430. Koneczny, I., Cossins, J., Vincent, A., 2014. The role of muscle-specific tyrosine kinase
Hendershot, T.J., Liu, H., Clouthier, D.E., Shepherd, I.T., Coppola, E., Studer, M., (MuSK) and mystery of MuSK myasthenia gravis. J. Anat. 224, 29–35.
Firulli, A.B., Pittman, D.L., Howard, M.J., 2008. Conditional deletion of Hand2 Kuba, K., 1970. Effects of catecholamines on the neuromuscular junction in the rat
reveals critical functions in neurogenesis and cell type-specific gene expression for diaphragm. J. Physiol. 211, 551–570.
development of neural crest-derived noradrenergic sympathetic ganglion neurons. Landi, F., Liperoti, R., Russo, A., Giovannini, S., Tosato, M., Capoluongo, E., Bernabei, R.,
Dev. Biol. 319, 179–191. Onder, G., 2012. Sarcopenia as a risk factor for falls in elderly individuals: results
Henderson, C.E., Phillips, H.S., Pollock, R.A., Davies, A.M., Lemeulle, C., Armanini, M., from the ilSIRENTE study. Clin. Nutr. 31, 652–658.
Simmons, L., Moffet, B., Vandlen, R.A., Simpson, L., Koliatsos, V.E., Rosenthal, A., Landi, F., Marzetti, E., Martone, A.M., Bernabei, R., Onder, G., 2014. Exercise as a
et al., 1994. GDNF: a potent survival factor for motoneurons present in peripheral remedy for sarcopenia. Curr. Opin. Clin. Nutr. Metab. Care 17 (1), 25–31.
nerve and muscle. Science 266, 1062–1064. Laplante, M., Sabatini, D.M., 2012. mTOR signaling in growth control and disease. Cell
Hendrickse, P., Galinska, M., Hodson-Tole, E., Degens, H., 2018. An evaluation of 149, 274–293.
common markers of muscle denervation in denervated young-adult and old rat Larsson, L., 1995. Motor units: remodeling in aged animals. J. Gerontol. Ser. A Biol. Sci.
gastrocnemius muscle. Exp. Gerontol. 106, 159–164. Med. Sci. 50, 91–95.

13
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

Larsson, L., Ansved, T., 1995. Effects of ageing on the motor unit. Prog. Neurobiol. 45, Minetti, G.C., Feige, J.N., Bombard, F., Heier, A., Morvan, F., Nürnberg, B., Leiss, V.,
397–458. Birnbaumer, L., Glass, D.J., Fornaro, M., 2014. Gαi2 signaling is required for skeletal
Larsson, L., Edstrom, L., 1986. Effects of age on enzyme-histochemical fibre spectra and muscle growth, regeneration, and satellite cell proliferation and differentiation. Mol.
contractile properties of fast- and slow-twitch skeletal muscles in the rat. J. Neurol. Cell. Biol. 34, 619–630.
Sci. 76, 69–89. Mirotznik, R.R., Zheng, X., Stanley, E.F., 2000. G-protein types involved in calcium
Larsson, L., Ansved, T., Edstrom, L., Gorza, L., Schiaffino, S., 1991. Effects of age on channel inhibition at a presynaptic nerve terminal. J. Neurosci. 20, 7614–7621.
physiological, immunohistochemical and biochemical properties of fast-twitch single Mora, A., Sandve, G.K., Gabrielsen, O.S., Eskeland, R., 2016. In the loop: promoter-
motor units in the rat. J. Physiol. 443, 257–275. enhancer interactions and bioinformatics. Brief Bioinform 17, 980–995.
Larsson, L., Degens, H., Li, M., Salviati, L., Lee, Yi., Thompson, W., Kirkland, J.L., Moresi, V., Williams, A.H., Meadows, E., Flynn, J.M., Potthoff, M.J., McAnally, J.,
Sandri, M., 2019. Sarcopenia: aging-related loss of muscle mass and function. Shelton, J.M., Backs, J., Klein, W.H., Richardson, J.A., Bassel-Duby, R., Olson, E.N.,
Physiol. Rev. 99, 427–511. 2010. Myogenin and class II HDACs control neurogenic muscle atrophy by inducing
Lau, Y.S., Xu, L., Gao, Y., Han, R., 2018. Automated muscle histopathology analysis using E3 ubiquitin ligases. Cell 143, 35–45.
CellProfiler. Skelet. Muscle 8 (1), 32. Moriscot, A.S., Baptista, I.L., Bogomolovas, J., Witt, C., Hirner, S., Granzier, H.,
Le Panse, B., Arlettaz, A., Portier, H., Lecoq, A.M., De Ceaurriz, J., Collomp, K., 2007. Labeit, S., 2010. MuRF1 is a muscle fiber-type II associated factor and together with
Effects of acute salbutamol intake during supramaximal exercise in women. Br. J. MuRF2 regulates type-II fiber trophicity and maintenance. J. Struct. Biol. 170,
Sports Med. 41, 430–434. 344–353.
Leeuwenburgh, C., Hansen, P., Shaish, A., Holloszy, J.O., Heinecke, J.W., 1998. Markers Moxham, C.M., Malbon, C.C., 1996. Insulin action impaired by deficiency of the G-
of protein oxidation by hydroxyl radical and reactive nitrogen species in tissues of protein subunit Gi[alpha]2. Nature 379, 840–844.
aging rats. Am. J. Physiol. 274, R453–461. Mu, L., Sobotka, S., Chen, J., Su, H., Sanders, I., Adler, C.H., Shill, H.A., Caviness, J.N.,
Lefeuvre, B., Crossin, F., Fontaine-Perus, J., Bandman, E., Gardahaut, M.F., 1996. Samanta, J.E., Beach, T.G., Arizona Parkinson’s Disease, C, 2013. Alpha-synuclein
Innervation regulates myosin heavy chain isoform expression in developing skeletal pathology and axonal degeneration of the peripheral motor nerves innervating
muscle fibers. Mech. Dev. 58, 115–127. pharyngeal muscles in Parkinson disease. J. Neuropathol. Exp. Neurol. 72, 119–129.
Lexell, J., 1995. Human aging, muscle mass, and fiber type composition. J. Gerontol. Ser. Munver, R., Volfson, I.A., 2006. Adrenal insufficiency: diagnosis and management. Curr.
A Biol. Sci. Med. Sci. 50, 11–16. Urol. Rep. 7, 80–85.
Li, Y., Thompson, W.J., 2011. Nerve terminal growth remodels neuromuscular synapses Navegantes, L.C., Baviera, A.M., Kettelhut, I.C., 2009. The inhibitory role of sympathetic
in mice following regeneration of the postsynaptic muscle fiber. J. Neurosci. 31, nervous system in the Ca2+-dependent proteolysis of skeletal muscle. Braz. J. Med.
13191–13203. Biol. Res. 42, 21–28.
Li, S.T., Holmes, C., Kopin, I.J., Goldstein, D.S., 2003. Aging-related changes in cardiac Ng, A.V., Callister, R., Johnson, D.G., Seals, D.R., 1994. Sympathetic neural reactivity to
sympathetic function in humans, assessed by 6-18F-fluorodopamine PET scanning. stress does not increase with age in healthy humans. Am. J. Physiol. Heart Circ.
J. Nucl. Med. 44, 1599–1603. Physiol. 267, H344–H353.
Li, W., Lam, M.T., Notani, D., 2014. Enhancer RNAs. Cell Cycle 13, 3151–3152. Nishiguchi, S., Yamada, M., Shirooka, H., Nozaki, Y., Fukutani, N., Tashiro, Y.,
Li, W., Notani, D., Rosenfeld, M.G., 2016. Enhancers as non-coding RNA transcription Hirata, H., Yamaguchi, M., Tasaka, S., Matsushita, T., Matsubara, K., Tsuboyama, T.,
units: recent insights and future perspectives. Nat. Rev. Genet. 17, 207–223. Aoyama, T., 2016. Sarcopenia as a risk factor for cognitive deterioration in
Lindquist, S., Stangel, M., 2011. Update on treatment options for Lambert-Eaton community-dwelling older adults: a 1-Year prospective study. J. Am. Med. Dir.
myasthenic syndrome: focus on use of amifampridine. Neuropsychiatr. Dis. Treat. 7, Assoc. 17, 372.e375-372.e378.
341–349. O’Suilleabhain, P., Low, P.A., Lennon, V.A., 1998. Autonomic dysfunction in the
Lipsitz, L.A., Novak, V., 2008. Aging and Autonomic Function, third ed. Mayo Lambert-Eaton myasthenic syndrome: serologic and clinical correlates. Neurology
Foundation, Rochester. 50, 88–93.
Low, P.A., Bennarroch, E.E., 2008. Clinical Autonomic Disorders, 3rd ed. Lippincott Ohara, O., Gahara, Y., Miyake, T., Teraoka, H., Kitamura, T., 1993. Neurofilament
Williams & Wilkins. deficiency in quail caused by nonsense mutation in neurofilament-L gene. J. Cell
Lynch, G.S., Ryall, J.G., 2008. Role of beta-adrenoceptor signaling in skeletal muscle: Biol. 121, 387–395.
implications for muscle wasting and disease. Physiol. Rev. 88, 729–767. Okada, K., Inoue, A., Okada, M., Murata, Y., Kakuta, S., Jigami, T., Kubo, S., Shiraishi, H.,
Lynch, N.A., Metter, E.J., Lindle, R.S., Fozard, J.L., Tobin, J.D., Roy, T.A., Fleg, J.L., Eguchi, K., Motomura, M., Akiyama, T., Iwakura, Y., Higuchi, O., Yamanashi, Y.,
Hurley, B.F., 1999. Muscle quality. I. Age-associated differences between arm and 2006. The muscle protein Dok-7 is essential for neuromuscular synaptogenesis.
leg muscle groups. J. Appl. Physiol. 86, 188–194. Science 312, 1802–1805.
Machado, J., Silveira, W.A., Gonçalves, D.A., Schavinski, A.Z., Khan, M.M., Zanon, N.M., Okamoto, L.E., Biaggioni, I., 2012. Chronic fatigue syndrome and the autonomic nervous
Diaz, M.B., Rudolf, R., Kettelhut, I.C., Navegantes, L.C., 2019. А− Calcitonin gene- system. In: Robertson, D., Biaggioni, I., Burnstock, G., Low, P.A., Paton, J.F.R. (Eds.),
related peptide inhibits autophagy and calpain systems and maintains the stability of Primer on the Autonomic Nervous System, 3rd ed. Academic, Amsterdam,
neuromuscular junction in denervated muscles. Mol. Metab. 28, 91–106. pp. 531–534.
Madisen, L., Mao, T., Koch, H., Zhuo, J.-m., Berenyi, A., Fujisawa, S., Hsu, Y.-W.A., Ooi, W., Barrett, S., Hossain, M., Kelley-Gagnon, M., Lipsitz, L.A., 1997. PAtterns of
Garcia, Iii, A.J., Gu, X., Zanella, S., Kidney, J., Gu, H., Mao, Y., Hooks, B.M., orthostatic blood pressure change and their clinical correlates in a frail, elderly
Boyden, E.S., Buzsáki, G., Ramirez, J.M., Jones, A.R., Svoboda, K., Han, X., population. JAMA 277, 1299–1304.
Turner, E.E., Zeng, H., 2012. A toolbox of Cre-dependent optogenetic transgenic Orimo, S., Kanazawa, T., Nakamura, A., Uchihara, T., Mori, F., Kakita, A.,
mice for light-induced activation and silencing. Nat. Neurosci. 15 (5), 793–802. Wakabayashi, K., Takahashi, H., 2007. Degeneration of cardiac sympathetic nerve
Maeda, K., Akagi, J., 2016. Sarcopenia is an independent risk factor of dysphagia in can occur in multiple system atrophy. Acta Neuropathol. 113, 81–86.
hospitalized older people. Geriatr. Gerontol. Int. 16, 515–521. Owino, W., Yang, S.Y., Goldspink, G., 2001. Age-related loss of skeletal muscle function
Maegawa, S., Hinkal, G., Kim, H.S., Shen, L., Zhang, L., Zhang, J., Zhang, N., Liang, S., and the inability to express the autocrine form of insulin-like growth factor-1 (MGF)
Donehower, L.A., Issa, J.P., 2010. Widespread and tissue specific age-related DNA in response to mechanical overload. FEBS Lett. 505, 259–263.
methylation changes in mice. Genome Res. 20, 332–340. Palmer, G.J., Ziegler, M.G., Lake, C.R., 1978. Response of norepinephrine and blood
Masiero, E., Agatea, L., Mammucari, C., Blaauw, B., Loro, E., Komatsu, M., Metzger, D., pressure to stress increases with age. J. Gerontol. 33, 482–487.
Reggiani, C., Schiaffino, S., Sandri, M., 2009. Autophagy is required to maintain Pascualya, M., Petrie, E.C., Brodkin, K., Peskind, E.R., Veith, R.C., Raskind, M.A., 1999.
muscle mass. Cell Metab. 10, 507–515. Effects of advanced aging on plasma catecholamine responses to the cold pressor
Mayeuf-Louchart, A., Hardy, D., Thorel, Q., Roux, P., Gueniot, L., Briand, D., test. Neurobiol. Aging 20, 637–642.
Mazeraud, A., Bougle, A., Shorte, S.L., Staels, B., Chretien, F., Duez, H., Payne, A.M., Delbono, O., 2004. Neurogenesis of excitation-contraction uncoupling in
Danckaert, A., 2018. MuscleJ: a high-content analysis method to study skeletal aging skeletal muscle. Exerc. Sport Sci. Rev. 32, 36–40.
muscle with a new Fiji tool. Skelet. Muscle 8 (1), 25. Payne, A.M., Messi, M.L., Zheng, Z., Delbono, O., 2006a. Motor neuron targeting of IGF-1
Mazzeo, R.S., Rajkumar, C., Jennings, G., Esler, M., 1997. Norepinephrine spillover at attenuates age-related external Ca2+-dependent skeletal muscle contraction in
rest and during submaximal exercise in young and old subjects. J. Appl. Physiol. 82, senescent mice. J. Physiol. 570, 283–294.
1869–1874. Payne, A.M., Zheng, Z., Messi, M.L., Milligan, C.E., Gonzalez, E., Delbono, O., 2006b.
Mei, H., Ho, M.K.C., Yung, L.Y., Wu, Z., Ip, N.Y., Wong, Y.H., 2011. Expression of Gαz in Motor neurone targeting of IGF-1 prevents specific force decline in ageing mouse
C2C12 cells restrains myogenic differentiation. Cell. Signal. 23, 389–397. muscle. J. Physiol. 570, 283–294.
Meitzen, J., Perry, A.N., Westenbroek, C., Hedges, V.L., Becker, J.B., Mermelstein, P.G., Pellegrino, M.J., Parrish, D.C., Zigmond, R.E., Habecker, B.A., 2011. Cytokines inhibit
2013. Enhanced striatal β1-Adrenergic receptor expression following hormone loss norepinephrine transporter expression by decreasing Hand2. Mol. Cell. Neurosci. 46,
in adulthood is programmed by both early sexual differentiation and puberty: a 671–680.
study of humans and rats. Endocrinology 154, 1820–1831. Personius, K.E., Balice-Gordon, R.J., 2000. Activity-dependent editing of neuromuscular
Messéant, J., Ezan, J., Delers, P., Glebov, K., Marchiol, C., Lager, F., Renault, G., synaptic connections. Brain Res. Bull. 53, 513–522.
Tissir, F., Montcouquiol, M., Sans, N., Legay, C., Strochlic, L., 2017. Wnt proteins Pertl, C., Eblenkamp, M., Pertl, A., Pfeifer, S., Wintermantel, E., Lochmuller, H.,
contribute to neuromuscular junction formation through distinct signaling Walter, M.C., Krause, S., Thirion, C., 2013. A new web-based method for automated
pathways. Development 144, 1712–1724. analysis of muscle histology. BMC Musculoskelet. Disord. 14, 26.
Messi, M.L., Li, T., Wang, Z.M., Marsh, A.P., Nicklas, B., Delbono, O., 2016. Resistance Pette, D., Staron, R.S., 2001. Transitions of muscle fiber phenotypic profiles. Histochem.
training enhances skeletal muscle innervation without modifying the number of Cell Biol. 115, 359–372.
satellite cells or their myofiber association in obese older adults. J. Gerontol. A Biol. Pfleger, J., Gresham, K., Koch, W.J., 2019. G protein-coupled receptor kinases as
Sci. Med. Sci. 71, 1273–1280. therapeutic targets in the heart. Nat. Rev. Cardiol. 16, 612–622.
Miazaki, M., Viana, M.P., Yang, Z., Comin, C.H., Wang, Y., da, F.C.L., Xu, X., 2015. Phillips, R.J., Powley, T.L., 2001. As the gut ages: timetables for aging of innervation
Automated high-content morphological analysis of muscle fiber histology. Comput. vary by organ in the Fischer 344 rat. J. Comp. Neurol. 434, 358–377.
Biol. Med. 63, 28–35.

14
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

Phillips, R.J., Powley, T.L., 2007. Innervation of the gastrointestinal tract: patterns of Sakaguchi, T., Okada, M., Kitamura, T., Kawasaki, K., 1993. Reduced diameter and
aging. Auton. Neurosci. 136, 1–19. conduction velocity of myelinated fibers in the sciatic nerve of a neurofilament-
Picca, A., Calvani, R., Leeuwenburgh, C., Coelho-Junior, H.J., Bernabei, R., Landi, F., deficient mutant quail. Neurosci. Lett. 153, 65–68.
Marzetti, E., 2019. Targeting mitochondrial quality control for treating sarcopenia: Sakellariou, G.K., Lightfoot, A.P., Earl, K.E., Stofanko, M., McDonagh, B., 2017. Redox
lessons from physical exercise. Expert Opin. Ther. Targets 23, 153–160. homeostasis and age-related deficits in neuromuscular integrity and function.
Pigna, E., Renzini, A., Greco, E., Simonazzi, E., Fulle, S., Mancinelli, R., Moresi, V., J. Cachexia Sarcopenia Muscle 8, 881–906.
Adamo, S., 2018. HDAC4 preserves skeletal muscle structure following long-term Sakuma, K., Kinoshita, M., Ito, Y., Aizawa, M., Aoi, W., Yamaguchi, A., 2016. p62/
denervation by mediating distinct cellular responses. Skelet. Muscle 8, 6. SQSTM1 but not LC3 is accumulated in sarcopenic muscle of mice. J. Cachexia
Powley, T.L., 2008. Central control of autonomic functions: organization of the Sarcopenia Muscle 7, 204–212.
autonomic nervous system. In: Squire, L., Berg, D., Bloom, F., du Lac, S., Gosh, A., Salminen, A.L., Brandt, A., Samuelsson, K., Toytari, O., Malmivaara, A., 2009. Mobility
Spitzer, N. (Eds.), Fundamental Neuroscience, third ed. Elsevier Inc., Burlington, devices to promote activity and participation: a systematic review. J. Rehabil. Med.
MA. USA, pp. 807–928. 41, 697–706.
Purves, D., Augustine, G.J., Fitzpatrick, D., Hall, W.C., LaMantia, A.S., White, L.E., 2011. Salvatori, S., Damiani, E., Zorzato, F., Volpe, P., Pierobon, S., Quaglino, D., Salviati, G.,
Neuroscience, 5th ed. Sinauer Associates, Inc., Massachusetts, USA. Margreth, A., 1988. Denervation-induced proliferative changes of triads in rabbit
Purves-Smith, F.M., Sgarioto, N., Hepple, R.T., 2014. Fiber typing in aging muscle. Exerc. skeletal muscle. Muscle Nerve 11, 1246–1259.
Sport Sci. Rev. 42, 45–52. Santulli, G., Iaccarino, G., 2013. Pinpointing beta adrenergic receptor in ageing
Radovanovic, D., Peikert, K., Lindström, M., Domellöf, F.P., 2015. Sympathetic pathophysiology: victim or executioner? Evidence from crime scenes. Immun.
innervation of human muscle spindles. J. Anat. 226, 542–548. Ageing 10 (1), 10.
Rauen, T., Hedrich, C.M., Tenbrock, K., Tsokos, G.C., 2013. cAMP responsive element Savitt, J.M., Jang, S.S., Mu, W., Dawson, V.L., Dawson, T.M., 2005. Bcl-x is required for
modulator: a critical regulator of cytokine production. Trends Mol. Med. 19, proper development of the mouse substantia nigra. J. Neurosci. 25, 6721–6728.
262–269. Schiaffino, S., 2018. Muscle fiber type diversity revealed by anti-myosin heavy chain
Renganathan, M., Sonntag, W.E., Delbono, O., 1997. L-type Ca2+ channel-insulin-like antibodies. FEBS J. 285, 3688–3694.
growth factor-1 receptor signaling impairment in aging rat skeletal muscle. Biochem. Schiaffino, S., Reggiani, C., 1994. Myosin isoforms in mammalian skeletal muscle.
Biophys. Res. Commun. 235, 784–789. J. Appl. Physiol. 77, 493–501.
Rengo, G., Pagano, G., Vitale, D.F., Formisano, R., Komici, K., Petraglia, L., Parisi, V., Schiaffino, S., Reggiani, C., 1996. Molecular diversity of myofibrillar proteins: gene
Femminella, G.D., de Lucia, C., Paolillo, S., Cannavo, A., Attena, E., Pellegrino, T., regulation and functional significance. Physiol. Rev. 76, 371–423.
Dellegrottaglie, S., Memmi, A., Trimarco, B., Cuocolo, A., Filardi, P.P., Leosco, D., Schiaffino, S., Dyar, K.A., Ciciliot, S., Blaauw, B., Sandri, M., 2013. Mechanisms
Ferrara, N., 2016. Impact of aging on cardiac sympathetic innervation measured by regulating skeletal muscle growth and atrophy. FEBS J. 280, 4294–4314.
123I-mIBG imaging in patients with systolic heart failure. Eur. J. Nucl. Med. Mol. Schmidt, M., Lin, S., Pape, M., Ernsberger, U., Stanke, M., Kobayashi, K., Howard, M.J.,
Imaging 43, 2392–2400. Rohrer, H., 2009. The bHLH transcription factor Hand2 is essential for the
Révész, D., Verhoeven, J.E., Milaneschi, Y., de Geus, E.J.C.N., Wolkowitz, O.M., maintenance of noradrenergic properties in differentiated sympathetic neurons. Dev.
Penninx, B.W.J.H., 2014. Dysregulated physiological stress systems and accelerated Biol. 329, 191–200.
cellular aging. Neurobiol. Aging 35, 1422–1430. Seibenhener, M.L., Babu, J.R., Geetha, T., Wong, H.C., Krishna, N.R., Wooten, M.W.,
Ricci, A., Mariotta, S., Greco, S., Pallone, G., Papale, M., Bisetti, A., 1997. Age-related 2004. Sequestosome 1/p62 is a polyubiquitin chain binding protein involved in
changes of the noradrenergic innervation of rat tracheo-bronchial tree and ubiquitin proteasome degradation. Mol. Cell. Biol. 24, 8055–8068.
pulmonary vasculature. Mech. Ageing Dev. 99, 245–255. Sekulic, A., Hudson, C.C., Homme, J.L., Yin, P., Otterness, D.M., Karnitz, L.M.,
Ristow, M., 2014. Unraveling the truth about antioxidants: mitohormesis explains ROS- Abraham, R.T., 2000. A direct linkage between the phosphoinositide 3-kinase-AKT
induced health benefits. Nat. Med. 20, 709–711. signaling pathway and the mammalian target of rapamycin in mitogen-stimulated
Robertson, D., Biaggioni, I., Burnstock, G., Low, P.A., Paton, J.F.R., 2012. Primer on the and transformed cells. Cancer Res. 60, 3504–3513.
Autonomic Nervous System, third ed. Elsevier Inc., London, UK. Silveira, W.A., Goncalves, D.A., Graca, F.A., Andrade-Lopes, A.L., Bergantin, L.B.,
Robinson, T.E., 1978. Electrical stimulation of the brain stem in freely moving rats: I. Zanon, N.M., Godinho, R.O., Kettelhut, I.C., Navegantes, L.C., 2014. Activating
Effects on behavior. Physiol. Behav. 21, 223–231. cAMP/PKA signaling in skeletal muscle suppresses the ubiquitin-proteasome-
Rodovalho, G.V., Drummond, L.R., Coimbra, C.C., 2020. Involvement of brainstem dependent proteolysis: implications for sympathetic regulation. J. Appl. Physiol. 117
noradrenergic system in cutaneous heat loss during exercise. Brain Res. Bull. 164, (1985), 11–19.
372–379. Smith, L.R., Barton, E.R., 2014. SMASH - semi-automatic muscle analysis using
Rodrigues, A.C.Z., Messi, M.L., Wang, Z.-M., Abba, M.C., Pereyra, A., Birbrair, A., segmentation of histology: a MATLAB application. Skelet. Muscle 4, 21.
Zhang, T., O’Meara, M., Kwan, P., Lopez, E.I.S., Willis, M.S., Mintz, A., Files, D.C., Sonjak, V., Jacob, K., Morais, J.A., Rivera-Zengotita, M., Spendiff, S., Spake, C.,
Furdui, C., Oppenheim, R.W., Delbono, O., 2018. The sympathetic nervous system Taivassalo, T., Chevalier, S., Hepple, R.T., 2019. Fidelity of muscle fibre
regulates skeletal muscle motor innervation and acetylcholine receptor stability. reinnervation modulates ageing muscle impact in elderly women. J. Physiol. 597
Acta Physiol. 225, e13195. (19), 5009–5023.
Rodrigues, A.Z.C., Wang, Z.-M., Messi, M.L., Delbono, O., 2019. Sympathomimetics Srikanthan, P., Hevener, A.L., Karlamangla, A.S., 2010. Sarcopenia exacerbates obesity-
regulate neuromuscular junction transmission through TRPV1, P/Q- and N-type Ca2 associated insulin resistance and dysglycemia: findings from the national health and
+ channels. Mol. Cell. Neurosci. 95, 59–70. nutrition examination survey III. PLoS One 5, e10805.
Rodrigues, A.C.Z., Wang, Z.-M., Messi, M.L., Bonilla, H.J., Liu, L., Freeman, W.M., Stanzel, S., Stubbusch, J., Pataskar, A., Howard, M.J., Deller, T., Ernsberger, U.,
Delbono, O., 2020. Heart and neural crest derivative 2-induced preservation of Tiwari, V.K., Rohrer, H., Tsarovina, K., 2016. Distinct roles of Hand2 in developing
sympathetic neurons attenuates sarcopenia with aging. J. Cachexia Sarcopenia and adult autonomic neurons. Dev. Neurobiol. 76 (10), 1111–1124.
Muscle 11. Straka, T., Vita, V., Prokshi, K., Horner, S.J., Khan, M.M., Pirazzini, M., Williams, M.P.I.,
Rohrer, H., 2011. Transcriptional control of differentiation and neurogenesis in Hafner, M., Zaglia, T., Rudolf, R., 2018. Postnatal development and distribution of
autonomic ganglia. Eur. J. Neurosci. 34, 1563–1573. sympathetic innervation in mouse skeletal muscle. Int. J. Mol. Sci. 19.
Rommel, C., Bodine, S.C., Clarke, B.A., Rossman, R., Nunez, L., Stitt, T.N., Straub, V., Campbell, K.P., 1997. Muscular dystrophies and the dystrophin-glycoprotein
Yancopoulos, G.D., Glass, D.J., 2001. Mediation of IGF-1-induced skeletal myotube complex. Curr. Opin. Neurol. 10, 168–175.
hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat. Cell Biol. Studenski, S., Perera, S., Patel, K., Rosano, C., Faulkner, K., Inzitari, M., Brach, J.,
3, 1009–1013. Chandler, J., Cawthon, P., Connor, E.B., Nevitt, M., Visser, M., Kritchevsky, S.,
Rosenbaum, D.M., Rasmussen, S.G., Kobilka, B.K., 2009. The structure and function of G- Badinelli, S., Harris, T., Newman, A.B., Cauley, J., Ferrucci, L., Guralnik, J., 2011.
protein-coupled receptors. Nature 459, 356–363. Gait speed and survival in older adults. JAMA 305, 50–58.
Rosenberg, 1989. Summary comments. Am. J. Clin. Nutr. 50, 1231–1233. Tachi, N., Ohya, K., Chiba, S., Nihira, H., Minagawa, K., 1995. Muscle involvement in
Rubinsztein, D.C., Marino, G., Kroemer, G., 2011. Autophagy and aging. Cell 146, congenital insensitivity to pain with anhidrosis. Pediatr. Neurol. 12, 264–266.
682–695. Taetzsch, T., Valdez, G., 2018. NMJ maintenance and repair in aging. Curr. Opin.
Rudolf, R., Bogomolovas, J., Strack, S., Choi, K.R., Khan, M.M., Wagner, A., Brohm, K., Physiol. 4, 57–64.
Hanashima, A., Gasch, A., Labeit, D., Labeit, S., 2013. Regulation of nicotinic Tang, H., Goldman, D., 2006. Activity-dependent gene regulation in skeletal muscle is
acetylcholine receptor turnover by MuRF1 connects muscle activity to endo/ mediated by a histone deacetylase (HDAC)-Dach2-myogenin signal transduction
lysosomal and atrophy pathways. Age (Dordr) 35, 1663–1674. cascade. Proc. Natl. Acad. Sci. U. S. A. 103, 16977–16982.
Ruegg, M.A., Bixby, J.L., 1998. Agrin orchestrates synaptic differentiation at the Tang, S., Wong, H.-C., Wang, Z.M., Huang, Y., Zou, J., Zhuo, Y., Pennati, A., Gadda, G.,
vertebrate neuromuscular junction. Trends Neurosci. 21, 22–27. Delbono, O., Yang, J.J., 2011. Design and application of a class of sensors to monitor
Ryall, J.G., Lynch, G.S., 2008. The potential and the pitfalls of beta-adrenoceptor Ca2+ dynamics in high Ca2+ concentration cellular compartments. Proc. Natl.
agonists for the management of skeletal muscle wasting. Pharmacol. Ther. 120, Acad. Sci. 16265–16270, 2011 Sep 27. Epub 2011 Sep 13.
219–232. Tang, H., Shrager, J.B., Goldman, D., 2019. Rapamycin protects aging muscle. Aging
Ryall, J.G., Schertzer, J.D., Alabakis, T.M., Gehrig, S.M., Plant, D.R., Lynch, G.S., 2008. (Albany NY) 11, 5868–5870.
Intramuscular {beta}2-agonist administration enhances early regeneration and Tchkonia, T., Zhu, Y., van Deursen, J., Campisi, J., Kirkland, J.L., 2013. Cellular
functional repair in rat skeletal muscle after myotoxic injury. J. Appl. Physiol. 105 senescence and the senescent secretory phenotype: therapeutic opportunities.
(1), 165–172. J. Clin. Invest. 123, 966–972.
Rychlik, J.L., Hsieh, M., Eiden, L.E., Lewis, E.J., 2005. Phox2 and dHAND transcription Thoma, A., Lightfoot, A.P., 2018. NF-kB and inflammatory cytokine signalling: role in
factors select shared and unique target genes in the noradrenergic cell type. J. Mol. skeletal muscle atrophy. Adv. Exp. Med. Biol. 1088, 267–279.
Neurosci. 27, 281–292. Tieland, M., Trouwborst, I., Clark, B.C., 2018. Skeletal muscle performance and ageing.
Ryu, Y.K., Collins, S.E., Ho, H.-Y.H., Zhao, H., Kuruvilla, R., 2013. An autocrine Wnt5a- J. Cachexia Sarcopenia Muscle 9, 3–19.
Ror signaling loop mediates sympathetic target innervation. Dev. Biol. 377, 79–89.

15
O. Delbono et al. Ageing Research Reviews 67 (2021) 101305

Tournadre, A., Vial, G., Capel, F., Soubrier, M., Boirie, Y., 2019. Sarcopenia. Joint Bone Wang, Z.-M., Messi, M.L., Delbono, O., 2002. Sustained overexpression of IGF-1 prevents
Spine 86, 309–314. age-dependent decrease in charge movement and intracellular calcium in mouse
Trimmer, J.S., Cooperman, S.S., Tomiko, S.A., Zhou, J.Y., Crean, S.M., Boyle, M.B., skeletal muscle. Biophys. J. 82, 1338–1344.
Kallen, R.G., Sheng, Z.H., Barchi, R.L., Sigworth, F.J., et al., 1989. Primary structure Wang, Z.M., Zheng, Z., Messi, M.L., Delbono, O., 2005. Extension and magnitude of
and functional expression of a mammalian skeletal muscle sodium channel. Neuron denervation in skeletal muscle from ageing mice. J. Physiol. 565, 757–764.
3, 33–49. Wang, Z.-M., Rodrigues, A.C.Z., Messi, M.L., Delbono, O., 2020a. Aging blunts
Tsarovina, K., Reiff, T., Stubbusch, J., Kurek, D., Grosveld, F.G., Parlato, R., Schütz, G., sympathetic neuron regulation of motoneurons synaptic vesicle release mediated by
Rohrer, H., 2010. The Gata3 transcription factor is required for the survival of β1- and α2B-Adrenergic receptors in geriatric mice. J. Gerontol. Ser. A 75,
embryonic and adult sympathetic neurons. J. Neurosci. 30, 10833–10843. 1473–1480.
Tseng, B.S., Marsh, D.R., Hamilton, M.T., Booth, F.W., 1995. Strength and aerobic Wang, Z.M., Messi, M.L., Grinevich, V., Budygin, E., Delbono, O., 2020b. Postganglionic
training attenuate muscle wasting and improve resistance to the development of sympathetic neurons, but not locus coeruleus optostimulation, activates
disability with aging. J. Gerontol. A Biol. Sci. Med. Sci. 50, 113–119. Spec No. neuromuscular transmission in the adult mouse in vivo. Mol. Cell. Neurosci. 109,
Tsentsevitsky, A.N., Kovyazina, I.V., Bukharaeva, E.A., 2019. Diverse effects of 103563.
noradrenaline and adrenaline on the quantal secretion of acetylcholine at the mouse Weatherbee, S.D., Anderson, K.V., Niswander, L.A., 2006. LDL-receptor-related protein 4
neuromuscular junction. Neuroscience 423, 162–171. is crucial for formation of the neuromuscular junction. Development 133,
Upadhya, B., Haykowsky, M.J., Eggebeen, J., Kitzman, D.W., 2015. Exercise intolerance 4993–5000.
in heart failure with preserved ejection fraction: more than a heart problem. Weindruch, R., 1995. Interventions based on the possibility that oxidative stress
J. Geriatr. Cardiol. 12, 294–304. contributes to sarcopenia. J. Gerontol. A Biol. Sci. Med. Sci. 50, 157–161.
Urasawa, K., Murakami, T., Yasuda, H., 1991. Age-related alterations in adenylyl cyclase Wen, Y., Murach, K.A., Vechetti Jr., I.J., Fry, C.S., Vickery, C., Peterson, C.A.,
system of rat hearts. Jpn. Circ. J. 55, 676–684. McCarthy, J.J., Campbell, K.S., 2018. MyoVision: software for automated high-
Urbancheck, M.G., Picken, E.B., Kalliainen, L.K., Kuzon, W.M., 2001. Specific force content analysis of skeletal muscle immunohistochemistry. J. Appl. Physiol. 124
deficit in skeletal muscles of old rats is partially explained by the existence of (1985), 40–51.
denervated muscle fibers. J. Gerontol. Biol. Sci. 56A, B191–B198. White, Z., White, R.B., McMahon, C., Grounds, M.D., Shavlakadze, T., 2016. High
Vainshtein, A., Tryon, L.D., Pauly, M., Hood, D.A., 2015. Role of PGC-1alpha during mTORC1 signaling is maintained, while protein degradation pathways are perturbed
acute exercise-induced autophagy and mitophagy in skeletal muscle. Am. J. Physiol. in old murine skeletal muscles in the fasted state. Int. J. Biochem. Cell Biol. 78,
Cell Physiol. 308, C710–719. 10–21.
Valdez, G., Tapia, J.C., Kang, H., Clemenson Jr., G.D., Gage, F.H., Lichtman, J.W., Wokke, J.H., Jennekens, F.G., van den Oord, C.J., Veldman, H., Smit, L.M., Leppink, G.J.,
Sanes, J.R., 2010. Attenuation of age-related changes in mouse neuromuscular 1990. Morphological changes in the human end plate with age. J. Neurol. Sci. 95,
synapses by caloric restriction and exercise. Proc. Natl. Acad. Sci. U. S. A. 107, 291–310.
14863–14868. Xiang, H.-B., Liu, C., Liu, T.-T., Xiong, J., 2014. Central circuits regulating the
Vasilaki, A., Richardson, A., Van Remmen, H., Brooks, S.V., Larkin, L., McArdle, A., sympathetic outflow to lumbar muscles in spinally transected mice by retrograde
Jackson, M.J., 2017. Role of nerve-muscle interactions and ROS in regulation of transsynaptic transport. Int. J. Clin. Exp. Pathol. 7, 2987–2997.
muscle proteostasis with aging. J. Physiol. 595, 6409–6415. Xu, Z., Feng, X., Dong, J., Wang, Z.-M., Lee, J., Furdui, C., Clark Files, D., Beavers, K.M.,
Veith, R.C., Featherstone, J.A., Linares, O.A., Halter, J.B., 1986. Age differences in Kritchevsky, S., Milligan, C., Jin, J.-P., Delbono, O., Zhang, T., 2017. Cardiac
plasma norepinephrine kinetics in humans. J. Gerontol. 41, 319–324. troponin T and fast skeletal muscle denervation in ageing. J. Cachexia Sarcopenia
Vitale, G., Cesari, M., Mari, D., 2016. Aging of the endocrine system and its potential Muscle 8, 808–823.
impact on sarcopenia. Eur. J. Intern. Med. 35, 10–15. Zhang, C., Goto, N., Suzuki, M., Ke, M., 1996. Age-related reductions in number and size
von Haehling, S., Morley, J.E., Anker, S.D., 2010. An overview of sarcopenia: facts and of anterior horn cells at C6 level of the human spinal cord. Okajimas Folia Anatomica
numbers on prevalence and clinical impact. J. Cachexia Sarcopenia Muscle 1, Japonica 73, 171–177.
129–133. Zheng, Z., Wang, Z.M., Delbono, O., 2002. Insulin-like growth factor-1 increases skeletal
Wang, Y., Hekimi, S., 2015. Mitochondrial dysfunction and longevity in animals: muscle DHPR alpha 1S transcriptional activity by acting on the cAMP-response
untangling the knot. Science 350, 1204–1207. element-binding protein element of the promoter region. J. Biol. Chem. 277,
Wang, Z.-M., Messi, M.L., Delbono, O., 2000. L-type Ca2+ channel charge movement and 50535–50542.
intracellular Ca2+ in skeletal muscle fibers from aging mice. Biophys. J. 78, Zhu, Y., Armstrong, J.L., Tchkonia, T., Kirkland, J.L., 2014. Cellular senescence and the
1947–1954. senescent secretory phenotype in age-related chronic diseases. Curr. Opin. Clin.
Nutr. Metab. Care 17, 324–328.

16

You might also like