Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Dec 10.
Published in final edited form as: Respir Physiol Neurobiol. 2008 Dec 10;164(1-2):252–262. doi: 10.1016/j.resp.2008.07.018

Trophic Factor Expression in Phrenic Motor Neurons

Carlos B Mantilla 1, Gary C Sieck 1
PMCID: PMC2642900  NIHMSID: NIHMS78659  PMID: 18708170

Abstract

The function of a motor neuron and the muscle fibers it innervates (i.e., a motor unit) determines neuromotor output. Unlike other skeletal muscles, respiratory muscles (e.g., the diaphragm, DIAm) must function from birth onwards in sustaining ventilation. DIAm motor units are capable of both ventilatory and non-ventilatory behaviors, including expulsive behaviors important for airway clearance. There is significant diversity in motor unit properties across different types of motor units in the DIAm. The mechanisms underlying the development and maintenance of motor unit diversity in respiratory muscles (including the DIAm) are not well understood. Recent studies suggest that trophic factor influences contribute to this diversity. Remarkably little is known about the expression of trophic factors and their receptors in phrenic motor neurons. This review will focus on the contribution of trophic factors to the establishment and maintenance of motor unit diversity in the DIAm, during development and in response to injury or disease.

Keywords: Respiratory muscles, diaphragm, skeletal muscle, development, neurotrophin, neuregulin, glial cell line-derived neurotrophic factor

1. Introduction

The function of a motor neuron and the muscle fibers it innervates (i.e., a motor unit) are important determinants of respiratory neuromotor output. Respiratory muscles, unlike other skeletal muscles, must function from birth onwards and commonly display high levels of activity. Indeed, as the major inspiratory muscle in mammals, the diaphragm muscle (DIAm) is uniquely active and rat DIAm motor units display a daily duty cycle (ratio of active to inactive time) ∼ 35% (Kong et al., 1986). In contrast, rat hind limb muscles display duty cycles ranging from ∼2% for the extensor digitorum longus (EDL) muscle to ∼14% for the soleus muscle (Hensbergen et al., 1997). DIAm motor units are involved in a number of ventilatory and non-ventilatory behaviors, including vocalization, swallowing, and expulsive behaviors involved in airway clearance (Sieck et al., 1989a). At present, the mechanisms underlying the development and regulation of motor unit diversity (necessary to provide for this broad range of motor behaviors) are not well understood. Trophic factor influences may contribute to this diversity and this review will focus on the expression of trophic factors and their effects on phrenic motor neurons during development and in response to injury or disease.

Phrenic motor neurons innervating the DIAm are located within lamina IX of cervical spinal cord segments C3-C5 in rats (Prakash et al., 2000; Song et al., 2000), C4-C6 in cats (Webber et al., 1979), C5-C7 in ferrets (Yates et al., 1999), and C3-C5 in humans (Keswani et al., 1955). Phrenic motor neurons receive rhythmic excitatory drive from premotor neurons located in the medulla (Feldman et al., 1985; Ellenberger et al., 1988). Thus, trophic factors influencing phrenic motor neurons might derive from peripheral, segmental or central sources (Fig. 1). Peripheral sources include DIAm fibers, Schwann cells in the phrenic nerve and circulating (systemic) factors. Segmental sources include surrounding glia, interneurons and afferent neurons with their cell bodies in neighboring segments of the spinal cord and in dorsal root ganglia. Central sources derive predominantly from supraspinal sources such as the rostral ventral respiratory group in rats (Ellenberger et al., 1990; Dobbins et al., 1994) or the ventral and dorsal respiratory groups and medial reticular formation in cats and ferrets (Feldman et al., 1985; Yates et al., 1999). Any and all of these sources may provide trophic support to or derive trophic support from phrenic motor neurons.

Figure 1.

Figure 1

Schematic representation of a motor unit (i.e., a motoneuron and the muscle fibers it innervates) and the potential sources of trophic factor influence on motor units. Trophic factors and their receptors (in italics) are noted for each source. A ? sign is used if expression at that site is not reported consistently. *, denotes expression mainly following injury or disease. Parentheses indicate expression by only a subset of motor neurons. See text for details. BDNF, brain-derived neurotrophic factor; GDNF, glial cell line-derived neurotrophic factor; GFR, GDNF family receptor; NRG, neuregulin; NT, neurotrophin; p75NTR, p75 neurotrophin receptor; RET, rearranged in transfection receptor; Trk, tropomyosin-related kinase receptor.

Members of a number of trophic factor families are now known to exert trophic influences on motor neurons (Oppenheim, 1996). What is less clear is the cellular substrate for trophic effects of these factors, either acting directly on motor neurons or indirectly via effects on neighboring cells in the environment surrounding motor neurons. In this review, we will explore the contribution of trophic factors to the establishment and maintenance of motor unit diversity.

2. Classification of DIAm motor units

Neural control of the DIAm is based on recruitment and frequency coding of motor units, which in the adult DIAm display considerable diversity in their mechanical, histochemical and biochemical properties (Fournier et al., 1988; Sieck, 1991; Sieck, 1994; Su et al., 1997; Butler et al., 1999). This heterogeneity controls the range of muscle force generation required during different motor behaviors. Indeed, the combined contractile and fatigue properties of a motor unit pool determine the limits under which a muscle can respond to the varying mechanical demands placed upon it. Clearly these motor demands change during development and DIAm motor units must adapt or remodel to accommodate these changing demands.

Motor units are commonly classified into different types according to the mechanical and fatigue properties of their muscle fibers (Burke, 1981; Fournier et al., 1988; Sieck et al., 1989b): 1) slow-twitch, fatigue resistant (type S), 2) fast-twitch, fatigue resistant (type FR), 3) fast-twitch, fatigue-intermediate (type FInt), and 4) fast-twitch, fatigable (type FF). This diversity is manifest in structural and functional differences across muscle fibers, classified histochemically or according to myosin heavy chain (MHC) isoform expression (Johnson et al., 1994; Sieck et al., 1996; Geiger et al., 2000). The innervation ratio (i.e., the number of muscle fibers innervated by a motor neuron) varies across muscles, ranging from 10 or less in hand and eye muscles to hundreds in limb and postural muscles (Sieck, 1988). Within a muscle, innervation ratio is greater at type FInt and FF motor units compared to type S and FR units. In muscles of mixed composition, differences in fiber size also exist across motor units with fibers of type FInt and FF units (expressing MHC2X and MHC2B isoforms) displaying greater cross-sectional area than fibers of type S and FR units (expressing MHCSlow and MHC2A isoforms, respectively). Specific force (i.e., force per unit cross-sectional area) varies across fibers, with that of fibers expressing MHC2X and/or MHC2B being greater than that of fibers expressing MHCSlow or MHC2A (Geiger et al., 2000). Together, the greater innervation ratio, larger fiber size and greater specific force contribute to greater forces being generated by type FInt and FF motor units compared to type S and FR units.

Motor unit composition is fundamental in determining the functional capacity of the DIAm in accomplishing different motor behaviors. The overall force generated by a muscle results from the recruitment of motor units in an orderly fashion (Sieck et al., 1989a; Butler et al., 1999). Consistent with the “size principle” (Henneman et al., 1965; Gordon et al., 2004), recruitment of motor units generally matches their mechanical and fatigue properties: type S and FR motor units are recruited first, followed by type FInt and FF units. Forces generated during most sustained motor behaviors in the DIAm only require recruitment of type S and FR motor units. Only during high force, short duration motor behaviors does recruitment of more fatigable type FInt and FF units become necessary (Sieck et al., 1989a). The mechanisms underlying the development and maintenance of motor unit diversity are not well understood. Trophic influences may play an important role in defining motor unit diversity.

3. Trophic factors and their receptors

In very general terms, trophic factors stimulate proliferation, promote cell survival and differentiation, and regulate cell function. Many trophic factors were first identified by their roles in sustaining survival of specific neuronal populations during development (Hamburger, 1992). However, trophic factors are now known to exert additional effects including regulation of synaptic efficacy in the adult. The abundance of trophic factors in the nervous system suggests that they exhibit both unique roles as well as overlapping effects on their target cells, for instance motor neurons (Oppenheim, 1996; Baloh et al., 2000; Sendtner et al., 2000; Buonanno et al., 2001).

The sites of action of trophic factors have not been unambiguously ascertained. For instance, trophic factors could be released by motor neuron dendrites or cell bodies and act retrogradely on interneurons, descending afferent axons or surrounding glia (Yan et al., 1993; Rind et al., 2005). Alternatively, trophic factors could be transported anterogradely in motor axons and affect distal targets, including skeletal muscle and perisynaptic Schwann cells (Russell et al., 2000; Mantilla et al., 2004b; Hellyer et al., 2006). Several trophic factors and/or their receptors display widespread expression across motor units and will be discussed briefly (Fig. 1).

3.1. Neurotrophin family

The family of neurotrophins includes nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3) and NT-4 (Reichardt, 2006). Neurotrophins exert their effects through a low affinity receptor (p75) or a family of high affinity tropomyosin-related kinase (Trk) receptors. Trk receptors exhibit the following selectivity: TrkA preferentially binds NGF, TrkB binds BDNF and NT-4, and TrkC binds NT-3. Neurotrophins are synthesized as immature pro-neurotrophins, which are cleaved by furin and other proteases to their mature form (Lee et al., 2001b). Pro-neurotrophins such as pro-NGF and pro-BDNF induce apoptosis by activating a p75 receptor-sortilin complex (Lee et al., 2001b; Teng et al., 2005), but their physiological roles are poorly understood (Jansen et al., 2007).

The expression of neurotrophins and Trk receptors has been extensively studied, particularly during development (Funakoshi et al., 1993; Timmusk et al., 1993; Griesbeck et al., 1995; Erickson et al., 1996; Ip et al., 2001). Neurotrophins are produced by motor neurons (Henderson et al., 1993; Koliatsos et al., 1993). In fact, BDNF and NT-3 immunoreactivity is present in large neurons (possibly phrenic motor neurons) in the cervical ventral spinal cord (Johnson et al., 2000). The neurotrophins BDNF, NT-3 and NT-4 are also expressed in skeletal muscle (Henderson et al., 1993; Griesbeck et al., 1995), but it is unclear whether there are differences in neurotrophin expression across motor unit types (Funakoshi et al., 1995; Sakuma et al., 2001; Vernon et al., 2004).

Alternative splicing of Trk receptor mRNA yields both full-length (catalytically active) and truncated forms lacking the intracellular tyrosine kinase domain (Barbacid, 1994). Neurotrophin binding to Trk receptors is thought to result in receptor dimerization and transactivation at the cell surface, with subsequent internalization (Reichardt, 2006). Recently, TrkB receptors have also been implicated in neurotrophin-independent signaling via interactions with Gs-coupled receptors and transactivation of intracellular TrkB (Lee et al., 2001a; Rajagopal et al., 2004; Golder et al., 2008).

The receptor TrkB is present at both pre- and postsynaptic sites of neuromuscular junctions at both slow and fast muscle fibers, as well as in perisynaptic Schwann cells (Frisen et al., 1993; Funakoshi et al., 1993; Escandon et al., 1994; Gonzalez et al., 1999). It is unclear whether there are differences in expression of the full-length vs. truncated forms of neurotrophin receptors in these different components of the neuromuscular junction (Funakoshi et al., 1995; Gonzalez et al., 1999; Sakuma et al., 2001).

3.2. Transforming growth factor family (TGFβ)

The TGFβ family of trophic factors includes glial cell line-derived neurotrophic factor (GDNF), neurturin, artemin and persephin (Baloh et al., 2000; Takahashi, 2001; Sariola et al., 2003). Members of the TGFβ family bind GDNF family receptors (GFR), which signal by dimerizing with the rearranged in transfection (RET) receptor tyrosine kinase. There are four glycosyl-phosphatidylinositol-linked GFR receptors (GFRα1-4), which are membrane-anchored and bind their ligands with different affinities: GDNF preferentially binds to GFRα1, neurturin to GFRα2, artemin to GFRα3 and persephin to GFRα4 (Baloh et al., 2000; Takahashi, 2001). However, GDNF can also bind GFRα2, and neurturin can bind GFRα1, albeit with lower affinities (Sariola et al., 2003). The receptors GFRα1 and RET show widespread expression in cranial and spinal motor neurons, but only a small subset express GFRα2 (Golden et al., 1999; Oppenheim et al., 2000). GDNF and neurturin are expressed in skeletal muscle and Schwann cells both during development and in the adult (Henderson et al., 1994; Nguyen et al., 1998; Suzuki et al., 1998). GDNF produced by Schwann cells can be transported retrogradely and anterogradely within motor neurons (Russell et al., 2000). Given that the RET receptor is not expressed in peripheral targets such as Schwann cells or muscle fibers in the tongue or DIAm (Russell et al., 2000), it is not clear whether peripherally delivered GDNF acts only axon terminals themselves (Nguyen et al., 1998; Yang et al., 2004). Recent studies indicate that GFRα1 might act independent of RET by complexing with neural cell-adhesion molecule NCAM (Paratcha et al., 2003; Sariola et al., 2003), but whether this effect occurs postsynaptically in muscle fibers has not been explored.

3.3. Epidermal growth factor (EGF) family

Neuregulins (NRG) belong to the larger EGF family of trophic factors and activate receptor tyrosine kinases of the ErbB family (Zhu et al., 1995; Trinidad et al., 2000). Four NRG genes exist (NRG1-4) with alternative splicing resulting in multiple NRG isoforms, all of which share EGF-like binding domains (Falls et al., 1993; Buonanno et al., 2001). There are four members in the ErbB receptor family: epidermal growth factor (EGF) receptor (EGFR, also referred to as ErbB1), ErbB2/Neu, ErbB3 and ErbB4 (Yarden et al., 2001). A number of ligands bind to ErbB receptors including all NRG isoforms but with varying affinities and differential signaling effects (Jones et al., 1999). The ErbB1 receptor does not appreciably bind either NRG-1 or NRG-2, consistent with the very limited effects of its prototypical ligand (EGF) in the central nervous system (Falls, 2003). Both NRG-1 and NRG-2 bind to the extracellular domain of ErbB3 or ErbB4 receptors leading to dimerization preferentially with ErbB2 (Graus-Porta et al., 1997; Olayioye et al., 1998). Although the ErbB2 receptor shows no ligand binding affinity itself, upon heterodimerization it greatly increases NRG binding affinity of the ErbB receptor complex (Jones et al., 1999). For example, ErbB2/ErbB3 or ErbB2/ErbB4 heterodimers display ∼100-fold higher affinity for NRG-1β than ErbB3 or ErbB4 homodimers (Sliwkowski et al., 1994; Jones et al., 1999). Similarly, NRG-2 binds ErbB2/ErbB4 heterodimers with greatest affinity (Tzahar et al., 1996; Jones et al., 1999). Consequently, cells co-expressing ErbB2 with ErbB3 or ErbB4 would preferentially form ErbB2/ErbB3 or ErbB2/ErbB4 heterodimers after being exposed to NRG-1, and ErbB2/ErbB4 heterodimers following NRG-2. Importantly, distinct signaling pathways are activated depending on the ErbB receptor complex formed by NRG binding (Li et al., 2007).

Previous studies have shown that NRG-1 (both α and β isoforms) and NRG-2β are expressed by spinal motor neurons (Buonanno et al., 2001; Falls, 2003; Rimer et al., 2004). Schwann cells also express both NRG-1 and NRG-2, whereas muscle fibers reportedly express NRG-1 (Falls et al., 1993). ErbB2, ErbB3 and ErbB4 are expressed in spinal motor neurons (Ricart et al., 2006) and mature skeletal muscle (Moscoso et al., 1995; Zhu et al., 1995; Lebrasseur et al., 2003), including rat DIAm (Hellyer et al., 2006). The functional implications of the different NRG isoforms are incompletely understood, at least in part because of the widespread and seemingly redundant expression of NRG isoforms and ErbB receptors.

3.4. Other trophic factors

Members of several different families of trophic factors and cytokines have received considerable attention primarily as survival factors for motor neurons (Oppenheim, 1996; Sendtner et al., 2000; Brunet et al., 2007). In this regard, individual trophic factors likely exhibit both unique and overlapping effects on survival that are difficult to ascertain unequivocally. Indeed, a number of receptor complexes have been shown to interact with one or more trophic factors to elicit beneficial survival effects. Trophic factors such as vascular endothelial growth factor (Storkebaum et al., 2005), basic fibroblast growth factor (Teng et al., 1999), insulin-like growth factors (Neff et al., 1993), cardiotrophin-1 (Oppenheim et al., 2001), ciliary-neurotrophic factor - CNTF (Oppenheim et al., 1991) and leukemia inhibitory factor (Li et al., 1995) are known to exert significant survival effects both in culture and in vivo. In fact, trophic factors have been used in the treatment of human motor neuron diseases such as amyotrophic lateral sclerosis (Miller et al., 1996; Borasio et al., 1998; Ochs et al., 2000). Unfortunately, these studies had limited success. Although of interest, discussion of trophic factors in the context of motor neuron degeneration is outside the scope of this review. At present, there is insufficient information to determine whether any of these trophic factors may play a role in defining motor unit properties. Thus, these other trophic factors will not be discussed further.

4. Role of trophic factors during development

Although a number of growth factors are known to be involved in motor neuron survival and formation of the neuromuscular junction (Oppenheim, 1996; Sariola et al., 2003), far less is known about the role of trophic factors in the growth of motor neurons or muscle fibers, or in their specialization into motor unit types. For instance, trophic factors may contribute to motor unit type differences via selective expression of specific factors, their receptors or individual signaling pathway components. These are important aspects that are poorly understood.

Phrenic motor neurons exhibit significant growth during postnatal development (Cameron et al., 1990; Cameron et al., 1991; Prakash et al., 2000). For example, rat phrenic motor neurons increase in both soma and dendrite dimensions between postnatal day 21 and adulthood, with significant and proportionate growth of muscle fibers (Prakash et al., 2000). This proportionate growth indicates that even during postnatal development, it is important to match motor neuron and muscle fiber size. In addition, there is greater heterogeneity in phrenic motor neuron size in the adult, with the variance in the distribution of total surface area increasing ∼4-fold (Prakash et al., 2000). The characteristic heterogeneity of motor unit types develops following weaning in the rat DIAm when motor unit innervation ratio is fixed (Sieck et al., 1991; Prakash et al., 1993; Mantilla et al., 2008). Before weaning, it is difficult to determine whether synapse elimination and/or secondary myogenesis contribute to changes in motor unit size.

Synapse elimination may affect motor neuron size via an effect on innervation ratio, assuming that trophic support depends on the number of muscle fibers innervated by a motor neuron. For instance, increased innervation ratio resulting from partial denervation of hindlimb muscles and concomitant sprouting of spared axons is associated with increases in motor neuron size (Tissenbaum et al., 1991). In rat DIAm, synapse elimination continues through postnatal day 14; thus, if anything, innervation ratio is reduced during the postnatal period, and this would tend to decrease phrenic motor neuron size - just opposite to what is observed (Cameron et al., 1990; Cameron et al., 1991; Prakash et al., 2000). Postnatal phrenic motor neuron growth may thus depend on specific trophic influences not solely related to innervation ratio. Similarly, it seems reasonable to assume that most trophic effects derive from sources providing the greatest number of synaptic inputs to motor neurons, but it is possible that trophic influence does not correspond with the sheer number of synaptic connections onto motor neurons. This issue deserves further study.

Whether differences in descending excitatory input, activation of specific muscle groups or other activity-related effects play a role in the specialization of motor unit types is not presently known. Early in development, phrenic motor neurons are activated by rhythmic motor patterns generated in the spinal cord (Greer et al., 1992). Subsequently, fetal respiratory movements appear and they increase in frequency and regularity during the prenatal period. It is possible that the growth and maturation of motor neurons depends on these rhythmic motor patterns, either directly via activity-dependent effects or indirectly via neurotransmitter- or trophic factor-mediated effects. During postnatal development, DIAm motor unit activity changes as functional demands shift to include a wider range of ventilatory and non-ventilatory behaviors (Greer et al., 1992; Greer et al., 2006; Mantilla et al., 2008). Accordingly, activation of nearly all DIAm motor units is necessary in neonates during resting ventilation in order to overcome their reduced lung compliance and chest wall stiffness. The more homogeneous and smaller size of phrenic motor neurons in the early postnatal period is consistent with increased excitability (Henneman et al., 1965; Su et al., 1997), and can facilitate synchronous recruitment of DIAm motor units. As neonates mature, their lung compliance and chest wall stiffness increase, and more selective activation of DIAm motor units likely occurs during resting ventilation. Indeed, in the adult rat DIAm only 25-30% of motor units are activated during eupneic breathing (Sieck et al., 1989a; Sieck et al., 1996). Recruitment of additional motor units is necessary only when engaging in more forceful motor behaviors (e.g., coughing, sneezing). Clearly, trophic factor influences may act in an activity-dependent fashion, further complicating unambiguous interpretation of any developmental effects on motor units. Studies using targeted deletions of specific trophic factors or their receptors in the absence of changes in activity levels might help elucidate these issues. Several important questions remain to be answered, including: What are critical trophic influences on motor neurons? Are there developmental windows for a specific form of trophic support? Do trophic factors present in motor neurons exert an effect directly on motor neurons or only indirectly via an effect on the environment surrounding motor neurons?

4.1. Trophic factor effects on motor neuron elimination

The importance of target-derived factors for motor neuron survival was initially revealed by axotomy and limb bud ablation experiments demonstrating increased motor neuron loss during the so-called postnatal period of programmed motor neuron cell death (Hamburger, 1992; Oppenheim, 1996; Sendtner et al., 2000). However, it is not clear whether all motor neuron pools, specifically phrenic motor neurons, are similarly affected by deprivation of target-derived trophic support (Oppenheim, 1986). Regardless, a number of trophic factor families have demonstrated survival-promoting effects both in avian and mammalian model systems (Oppenheim, 1996; Brunet et al., 2007). The survival effects of individual trophic factors on motor neurons during this period of naturally occurring cell death are difficult to study explicitly given the abundance of trophic factors and their likely overlapping effects (Oppenheim, 1996; Sendtner et al., 2000; Brunet et al., 2007).

Several lines of evidence suggest that neurotrophins are dispensable for maintenance of motor neuron numbers during the postnatal period of motor neuron elimination, despite demonstrated survival-promoting effects of neurotrophins like BDNF (Oppenheim et al., 1992; Brunet et al., 2007) or the widespread distribution of neurotrophins and Trk receptors (Funakoshi et al., 1993; Timmusk et al., 1993; Griesbeck et al., 1995; Erickson et al., 1996; Ip et al., 2001). During postnatal development, mRNA expression for BDNF and NT-3 gradually decreases in both the rat soleus and gastrocnemius muscles, although NT-4 expression increases (Funakoshi et al., 1995). Importantly, mice genetically deficient in NT4 have normal numbers of spinal motor neurons (Conover et al., 1995). Although there is motor neuron loss in knockout mice lacking BDNF, it occurs only during periods of naturally occurring cell death, and likely results from reduced trophic support from peripheral tissues rather than a direct effect on motor neuron survival (Ernfors et al., 1994; Erickson et al., 1996).

In contrast, GDNF seems necessary for survival of certain populations of spinal motor neurons (Garces et al., 2000; Oppenheim et al., 2000; Takahashi, 2001). Indeed, GDNF is one of the most potent survival factors for cultured embryonic spinal motor neurons (Henderson et al., 1994; Brunet et al., 2007). For example, whereas RET is expressed by nearly all motor neurons early in embryological development, GFRα1 and GFRα2 display distinct expression patterns with motor neurons expressing one or both receptors (Oppenheim et al., 2000; Homma et al., 2003). Most GDNF-dependent motor neurons require GFRα1 for their survival response to GDNF or neurturin (Garces et al., 2000). Loss of neurturin or GFRα2 does not seemingly result in altered trophic response or motor neuron loss (Heuckeroth et al., 1999; Rossi et al., 1999). In a recent study, motor neuron loss in GDNF-, RET- and GFRα1-deficient mice was restricted to muscle spindle-innervating motor neurons (Gould et al., 2008). Whether a subset of phrenic motor neurons is specifically dependent on GDNF is presently unclear.

NRG-1 effects on acetylcholine receptor (AChR) clustering in muscle fibers have been extensively studied (Sandrock et al., 1997; Sanes et al., 2001; Falls, 2003), however much less is known about other possible effects of NRG-1 on motor neurons. Recently, NRG-1β was shown to inhibit apoptosis of cultured rat motor neurons in a phosphatidylinositol-3-kinase (PI3K)-dependent manner (Ricart et al., 2006). Importantly, NRG-1β acted synergistically with GDNF to promote motor neuron survival. In contrast, the neurotrophins BDNF and NGF, acting via the p75 receptor, were pro-apoptotic and completely abolished NRG-1 pro-survival effects by activating the c- Jun N-terminal kinase mitogen-activated protein kinase (MAPK) pathway with induction of neuronal nitric oxide synthase and peroxynitrite formation. Although not specifically examined in phrenic motor neurons, spinal motor neurons in rats and mice express both neurotrophin and ErbB receptors (Rimer et al., 1998; Copray et al., 2000; Pearson et al., 2004), thus providing the substrate for such an effect in vivo. Whether a similar interaction between different trophic factor pathways exists during periods of motor neuron loss (e.g., during postnatal development) is yet to be determined. The interaction between these and other trophic factor families on motor neuron survival is still poorly understood. Trophic factor effects may depend on the specific model used in the assessment of motor neuron survival (Brunet et al., 2007).

4.2. Trophic factors and motor unit specialization

Beyond their possible effects in regulating motor neuron survival during postnatal development, trophic factors may also contribute to the coordinated postnatal growth of motor neurons and muscle fibers (Mantilla et al., 2008). Several studies have examined differences in expression of trophic factors and their receptors in the neonate and in the adult (Golden et al., 1999; Buck et al., 2000; Ip et al., 2001; Hess et al., 2007). Whether differences exist in neurotrophin expression across motor neurons innervating different motor unit types is controversial. Postnatal expression of NT-4 mRNA increased in hindlimb muscles (Funakoshi et al., 1995), with higher expression in the soleus (predominantly expressing MHCSlow) than in the gastrocnemius (predominantly expressing MHC2X and/or MHC2B). In agreement, NT-4 immunoreactivity was limited to muscle fibers histochemically classified as belonging to type S motor units (Funakoshi et al., 1995). However, when motor neuron size was used to differentiate mRNA expression of neurotrophin and Trk receptors in adult hindlimb motor neurons, large variations in BDNF, NT-3, NT-4, TrkB and TrkC expression were observed, with no specific correlation with cell size or fast vs. slow motor unit composition (Copray et al., 2000). The heterogeneity in neurotrophin mRNA expression could be related, at least in part, to differences in muscle activation history between hindlimb muscles and unrelated to motor unit type or cell size. It is presently unknown if fibers in mixed muscles such as the DIAm display differential expression of neurotrophins or their receptors.

The coordinated postnatal development of all components of DIAm motor units (Mantilla et al., 2008) suggests that studying the mechanisms underlying muscle fiber growth and/or fiber type specialization may yield important clues to the origin and maintenance of motor unit diversity. For example, NRG-1 may constitute a nerve-derived trophic influence for muscle fiber growth during perinatal development. NRG-1 is expressed by motor neurons during embryonic and early postnatal development (Sandrock et al., 1997), and induces acetylcholine receptor (AChR) expression at embryonic and perhaps adult neuromuscular junctions likely via its effect on Schwann cells (Jo et al., 1995). In addition, NRG-1 induced phosphorylation of ErbB receptors results in downstream activation of the PI3K/Akt intracellular pathway, which is a critical regulator of muscle protein synthesis and hypertrophy (Bodine et al., 2001). All members of the ErbB receptor family can signal through PI3K, and the ErbB2/ErbB3 heterodimer is a very strong activator of PI3K given the multiple consensus sites on ErbB3 (Hellyer et al., 1998; Citri et al., 2003). Indeed, NRG-1 induces protein synthesis in the postnatal DIAm (Hellyer et al., 2006). It is currently unknown whether there are differences in NRG subtypes or ErbB receptor expression during development or across motor unit types, and whether NRG signaling contributes to the differential postnatal growth of DIAm fibers.

Although most studies have examined NRG-1 effects on neuromuscular junction formation (Sandrock et al., 1997; Buonanno et al., 2001; Sanes et al., 2001), other members of the NRG family are likely involved. In mice with tissue-specific deletions of NRG-1 expression (obtained using the CRE recombinase system), inactivating NRG-1 in motor neurons, muscle fibers or both did not prevent AChR clustering (Jaworski et al., 2006). In agreement, mice lacking skeletal muscle ErbB2 and ErbB4 receptors displayed normal neuromuscular junction development and maintenance (Escher et al., 2005). Both motor neurons and terminal Schwann cells express NRG-2, which is localized around perisynaptic Schwann cells at neuromuscular junctions (Rimer et al., 2004). In addition, NRG-2 induced AChR transcription in cultured myotubes expressing ErbB4 either singly or in combination with ErbB2 or ErbB3, but not in myotubes expressing only ErbB2 and ErbB3. Taken together, these results highlight the complexity of determining unique effects of a specific trophic factor family in vivo. The seemingly redundant expression of trophic factors (e.g., NRG-1 and NRG-2) and receptor isoforms (e.g., ErbB2, ErbB3 and ErbB4) makes unequivocal interpretation of trophic effects difficult when exogenous factors are administered or when expression of a single trophic factor or receptor is mutated or down-regulated.

5. Role of trophic factors in motor unit plasticity

In a series of studies, Mitchell and colleagues examined the role of neurotrophins and their receptors in the plasticity of phrenic motor output (Kinkead et al., 1998; Johnson et al., 2000; Baker-Herman et al., 2004; Golder et al., 2008). Cervical dorsal rhizotomy augments the serotonin-dependent long-term facilitation induced by intermittent hypoxia (Kinkead et al., 1998), and increases BDNF and NT-3 expression in the cervical spinal cord with no change in GDNF expression (Johnson et al., 2000). Intermittent hypoxia-induced long-term facilitation of phrenic motor output requires new BDNF synthesis in the spinal cord, and application of BDNF to the cervical spinal cord is sufficient to elicit a similar change in phrenic motor output (Baker-Herman et al., 2004). In addition, transactivation of TrkB receptors induced by adenosine 2a receptor ligands is sufficient to elicit facilitation of phrenic motor output (Golder et al., 2008). Importantly, the exact cellular substrate(s) for these forms of phrenic motor neuron plasticity are still unclear. Whether increased neurotrophins derive from motoneurons exerting an autocrine effect on TrkB receptors present on the motor neurons themselves, or act indirectly on neighboring cells to augment neurotrophin release (Canossa et al., 1997) or induce production of other factors (Reichardt, 2006) deserve further investigation.

Neurotrophins have important synaptic effects, even in the adult. For instance, neurotrophins can directly modulate synaptic efficacy (Kang et al., 1995; Kafitz et al., 1999), and neurotrophin synthesis and release are regulated by neuronal activity in the hippocampus (Schinder et al., 2000; Poo, 2001). BDNF may stimulate synapsin I phosphorylation via activation of a MAPK pathway (Jovanovic et al., 2000), and therefore, regulate neurotransmitter release (Hilfiker et al., 1999). In adult rat DIAm-phrenic nerve preparations, the neurotrophins BDNF and NT-4 improved neuromuscular transmission, likely via TrkB activation (Mantilla et al., 2004b). Isometric contractile properties of the DIAm were unaffected by either neurotrophin treatment or TrkB inhibition, suggesting a presynaptic effect of BDNF and NT-4 on neuromuscular transmission. In agreement, BDNF potentiates both spontaneous and evoked synaptic activity in cultured amphibian neuromuscular junctions (Lohof et al., 1993), an effect that is greatly facilitated by presynaptic depolarization (Boulanger et al., 1999). Neuromuscular junctions formed onto amphibian myocytes overexpresing NT-4 showed both higher levels of spontaneous synaptic activity and enhanced evoked synaptic transmission when compared to synapses formed onto neighboring myocytes not overexpressing NT-4 (Wang et al., 1997). Taken together, these results provide converging evidence for an important presynaptic effect of BDNF and NT-4 acting via TrkB receptors on regulation of synaptic vesicle release and neuromuscular transmission. In cultured amphibian neuromuscular junctions, NT-3 also potentiates postsynaptic currents, acting via PI3K, phospholipase C-γ and increased inositol 1,4,5-triphosphate (IP3)-induced Ca2+ release (Lohof et al., 1993; Yang et al., 2001). Whether neurotrophins have different effects depending on motor unit type has not been directly explored, but it is tempting to speculate that neurotrophins might underlie fiber type differences in synaptic efficacy at the neuromuscular junction (Mantilla et al., 2004a; Ermilov et al., 2007; Rowley et al., 2007).

GDNF may play an important role in neuromuscular junction plasticity both during development and in the adult. Anterogradely transported GDNF modulates presynaptic vesicle release, increasing spontaneous neurotransmitter release in neonatal mouse neuromuscular junctions (Ribchester et al., 1998). In transgenic mice overexpressing GDNF, axonal branching of motor neurons is increased (Nguyen et al., 1998). Furthermore, exogenous administration of GDNF during the first postnatal week increased neuromuscular junction remodeling, reducing synapse elimination, and thus increasing innervation ratio and motor unit size (Keller-Peck et al., 2001). In adult rodents, muscle activity may influence GDNF expression. For example, treadmill training increased GDNF content whereas hindlimb unloading decreased GDNF in both rat soleus and gastrocnemius muscles (Wehrwein et al., 2002). The magnitude of changes in GDNF expression was similar across these muscles, suggesting that motor unit type might not be as important in determining GDNF expression. However, the level of activity of these hindlimb muscles is low when compared to respiratory muscles such as the DIAm (Kong et al., 1986; Hensbergen et al., 1997). It is currently unknown if motor unit type differences in GDNF expression are present in the DIAm.

Whether multiple trophic factors (e.g., BDNF and GDNF) modulate neurotransmitter release at individual synapses is presently unknown and clearly deserves to be explored. It is tempting to speculate that these trophic factors exert non-overlapping functions related to motor unit type rather than truly redundant effects, but this hypothesis has not been examined directly.

6. Role of trophic factors in neural regeneration

6.1. Nerve injury

Neurotrophin effects on motor neuron survival and axon regeneration following peripheral nerve injury have been studied extensively (Sakuma et al., 2001; Mousavi et al., 2002; Boyd et al., 2003). In neonatal rats, sciatic nerve axotomy leads to atrophy of denervated muscles including the soleus, tibialis anterior and EDL muscles with associated motor neuron loss and reduced muscle fiber number and cross-sectional area. Fibers expressing MHC2B seem particularly susceptible to axotomy (Mousavi et al., 2002). Administering BDNF, NT-3 or NT-4 in combination with CNTF at the site of nerve transection reduces motor neuron loss and prevents axotomy-induced muscle atrophy (Mousavi et al., 2002; Mousavi et al., 2004). In postnatal rats, loss of MHC2B-expressing muscle fibers in the EDL and tibialis muscles is prevented by BDNF (Mousavi et al., 2004), but not by NT-3 or NT-4 (Mousavi et al., 2002). In adults, NT-4 supports type S motor units in the soleus muscle, preventing muscle atrophy and muscle fiber loss (Simon et al., 2003). Interestingly, it seems that neurotrophins both promote and inhibit axon sprouting depending on the receptor they activate. In particular, neurotrophin-induced p75-activation inhibits axon sprouting of axotomized adult motor neurons, whereas TrkB activation promotes neurite outgrowth (Boyd et al., 2003). Indeed, BDNF or NT-4 may facilitate and inhibit axon outgrowth when administered at low and high doses, respectively, given the different neurotrophin affinity of these receptors (Reichardt, 2006). Neural regeneration post-nerve injury may thus depend not only on the availability of specific neurotrophins (e.g., NT-4 for type S or BDNF for type FF motor units), but also on the balance of receptors expressed across motor units types. Further studies are needed to clarify the possible roles of neurotrophins in the survival of motor neurons and muscle fibers both during development and in the adult, and specifically in models using muscles of mixed motor unit composition such as the DIAm.

Members of the GDNF family of trophic factors do not seem to play a role in presynaptic sprouting post-nerve transection. Interestingly, skeletal muscles express both GDNF and neurturin (Henderson et al., 1994; Nguyen et al., 1998; Suzuki et al., 1998). GDNF mRNA expression is increased in partially denervated human skeletal muscle obtained from patients with peripheral neuropathy or amyotrophic lateral sclerosis (Lie et al., 1998). However, in a recent study, mice with a targeted deletion of RET kinase activity in cranial motor neurons displayed normal hypoglossal motor neuron sprouting and neuromuscular junction re-innervation following axotomy (Baudet et al., 2008). Exogenous treatment with GDNF increased the number of motor axons at neuromuscular junctions only when administered during an early developmental time window (first postnatal week), not when treatment was begun afterwards (Keller-Peck et al., 2001). These results highlight a possible developmental role of GDNF on axon sprouting, in contrast to a more limited role in the adult. However, further studies are needed, specifically with quantification of GDNF and neurturin levels in Schwann cells and muscle fibers, as well determination of GFRα1-4 receptor expression in axon terminals.

NRGs may have a complex role in supporting motor units following nerve or muscle injury. NRG-1 isoform expression is reduced in facial motor neurons post-axotomy, gradually returning to control levels as re-innervation occurs (Kerber et al., 2003). However, mice with a specific ErbB2 deletion in adult Schwann cells (obtained using an inducible CRE recombinase system) do not display deficits in Schwann cell proliferation following sciatic nerve transection (Atanasoski et al., 2006), suggesting that NRGs may not directly regulate nerve repair in the adult. It is not clear if a minimum level of NRG/ErbB signaling in Schwann cells is necessary for axon sprouting and myelination following nerve injury. With bupivacaine injection into the tibialis anterior muscle in adult rats, muscle NRG-1 expression decreased initially (Hirata et al., 2007). After 4-6 days, NRG-1 mRNA and protein levels gradually increased, being immunohistochemically localized to differentiating satellite cells. Importantly, spinal cord NRG-1 protein (not mRNA) levels showed a similar temporal expression profile post-injury. Taken together, these results suggest that following muscle injury, regenerating satellite cells increase their expression of NRG-1 which can then act both on neighboring muscle cells to promote muscle fiber growth and on motor neurons (likely via retrograde transport) to promote motor neuron survival and outgrowth (sprouting) of spared axons. These possibilities remain to be explored explicitly. Additional studies are also needed to determine the signaling pathways induced by NRG-1 to promote muscle and neural regeneration post-injury.

6.2. Spinal cord injury

A number of studies have reported changes in the expression of trophic factors such as NGF, BDNF, NT-3, NT-4 and GDNF following spinal cord injury (Bennett et al., 1999; Dougherty et al., 2000; Satake et al., 2000; Widenfalk et al., 2001). Most of these studies examined mRNA expression rather than protein expression, and limited temporal expression profiles. For instance, after mechanical injuries of the midthoracic spinal cord in adult rats (either via compression or complete transection), GDNF mRNA expression increased in meningeal cells and glia at the injury site, whereas p75 mRNA increased in neurons and surrounding microglia (Widenfalk et al., 2001). More prominently, GFRα1 and truncated TrkB receptor mRNA expression increased throughout the spinal cord white matter by 6 weeks post-injury, being localized primarily to astrocytes (Widenfalk et al., 2001). Following a midthoracic compression injury, the number of astrocytes and microglia displaying BDNF immunoreactivity increased over time at the injury site but not at more distant sites (Dougherty et al., 2000; Ikeda et al., 2001). Detailed, systematic examinations of motor neuron expression of trophic factors or their receptors are not available, especially for motor neurons in pools below the level of injury. Such studies would be particularly meaningful in the case of phrenic motor neurons given that these motor neurons receive excitatory respiratory drive primarily from medullary premotor neurons (Feldman et al., 1985; Ellenberger et al., 1990; Dobbins et al., 1994; Yates et al., 1999). Importantly, comparisons across experimental models (e.g., cervical vs. thoracic levels, mechanical vs. traumatic injury), animal species, and timing of the measurements is complicated by differences in the cellular substrates and even the direction of the change in trophic factor expression post-injury.

Understanding the cellular substrates involved in post-spinal injury changes in expression of trophic factors or their receptors seems critical to the development of rational therapeutic interventions that would enhance functional recovery. Several studies showed that delivery of combinations of trophic factors (either via chronic infusion, adenoviral transfection or stem cell grafts) increases motor neuron survival and axon sprouting within the injured cord (Bregman et al., 1997; Novikova et al., 2002; Lu et al., 2003; Ruitenberg et al., 2004; Iarikov et al., 2007). Unfortunately, increased neuronal survival or axonal sprouting does not readily translate into functional recovery. In recent studies, significant reorganization of spinal cord circuits involved in locomotor behaviors was observed following spinal cord injury (Cai et al., 2006; Courtine et al., 2008) and was enhanced by a number of interventions including BDNF, NT-3 or GDNF treatment (Dolbeare et al., 2003; Iarikov et al., 2007). Whether respiratory circuits involved in the control of DIAm activity undergo similar reorganization post-injury and whether trophic factors promote recovery of rhythmic respiratory activity have not been studied to date.

Another aspect that deserves further attention is that of trophic support of cells in the vicinity of motor neurons, which may thus indirectly affect motor unit properties. For example, motor neuron-derived NT-3 serves as a survival factor for spinal interneurons (Bechade et al., 2002). In this study, a p75-conjugated immunotoxin was used to induce motor neuron loss, which then increased apoptosis of interneurons in organotypic cultures of embryonic rat spinal cord. Exogenous application of NT-3 rescued interneurons from apoptosis, whereas quenching NT-3 with a soluble TrkC receptor induced even greater interneuron loss (Bechade et al., 2002). Conversely, pro-NGF secreted by astrocytes promotes motor neuron apoptosis via activation of the p75 receptor (Domeniconi et al., 2007). The p75 receptor is expressed in spinal motor neurons primarily up to the second post-natal week, following which its expression is normally downregulated (Yan et al., 1988). However, motor neuron expression of p75 increases following both spinal cord injury (Widenfalk et al., 2001) or peripheral nerve injury (Koliatsos et al., 1991; Xie et al., 2003), possibly rendering adult motor neurons susceptible to p75-induced apoptosis again. Complex trophic interactions likely occur within the spinal cord in the environment surrounding phrenic motor neurons, where motor neuron-derived trophic factors can affect presynaptic terminals, interneurons and surrounding glial cells. Trophic factors from any of these sources can also directly exert both positive and negative effects on motor neurons depending on the specific context of trophic factor receptor expression post-injury.

7. Summary/Conclusions

The DIAm is a highly active muscle, with a duty cycle in most species ∼35% (Kong et al., 1986; Sieck, 1988; Butler et al., 1999), and must be active from birth onwards. Phrenic motor neurons provide innervation to the DIAm and show remarkable diversity in functional properties. Given this remarkable and unique activation history, DIAm motor units may be particularly responsive to trophic influences and/or inactivity. Most studies have used immunohistochemical methods and in situ hybridization to localize trophic factor expression to large neurons in the ventral spinal cord (including cervical segments). Other studies have determined protein and mRNA expression in ventral horn of the cervical spinal cord. Clearly, these results need to be interpreted with caution when extending the findings to specific cellular substrates within the phrenic motor nucleus. Future studies using novel techniques, e.g., laser capture microdissection where retrogradely-labeled phrenic motor neurons can be uniquely assessed for mRNA measurements, hold great promise in providing cell-based measurements of trophic factor or receptor expression. Detailed and accurate knowledge of the network of trophic interactions at all levels of the motor unit might provide new directions in the study of respiratory motor control.

Acknowledgments

Supported by NIH grants HL 37680 and AR 51173 and the Mayo Foundation.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errorsmaybe discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  1. Atanasoski S, Scherer SS, Sirkowski E, Leone D, Garratt AN, Birchmeier C, Suter U. ErbB2 signaling in Schwann cells is mostly dispensable for maintenance of myelinated peripheral nerves and proliferation of adult Schwann cells after injury. J. Neurosci. 2006;26:2124–2131. doi: 10.1523/JNEUROSCI.4594-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Baker-Herman TL, Fuller DD, Bavis RW, Zabka AG, Golder FJ, Doperalski NJ, Johnson RA, Watters JJ, Mitchell GS. BDNF is necessary and sufficient for spinal respiratory plasticity following intermittent hypoxia. Nat. Neurosci. 2004;7:48–55. doi: 10.1038/nn1166. [DOI] [PubMed] [Google Scholar]
  3. Baloh RH, Enomoto H, Johnson EM, Jr., Milbrandt J. The GDNF family ligands and receptors - implications for neural development. Curr. Opin. Neurobiol. 2000;10:103–110. doi: 10.1016/s0959-4388(99)00048-3. [DOI] [PubMed] [Google Scholar]
  4. Barbacid M. The Trk family of neurotrophin receptors. J. Neurobiol. 1994;25:1386–1403. doi: 10.1002/neu.480251107. [DOI] [PubMed] [Google Scholar]
  5. Baudet C, Pozas E, Adameyko I, Andersson E, Ericson J, Ernfors P. Retrograde signaling onto Ret during motor nerve terminal maturation. J. Neurosci. 2008;28:963–975. doi: 10.1523/JNEUROSCI.4489-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bechade C, Mallecourt C, Sedel F, Vyas S, Triller A. Motoneuron-derived neurotrophin-3 is a survival factor for PAX2-expressing spinal interneurons. J. Neurosci. 2002;22:8779–8784. doi: 10.1523/JNEUROSCI.22-20-08779.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bennett AD, Taglialatela G, Perez-Polo R, Hulsebosch CE. NGF levels decrease in the spinal cord and dorsal root ganglion after spinal hemisection. Neuroreport. 1999;10:889–893. doi: 10.1097/00001756-199903170-00040. [DOI] [PubMed] [Google Scholar]
  8. Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, Zlotchenko E, Scrimgeour A, Lawrence JC, Glass DJ, Yancopoulos GD. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat. Cell Biol. 2001;3:1014–1019. doi: 10.1038/ncb1101-1014. [DOI] [PubMed] [Google Scholar]
  9. Borasio GD, Robberecht W, Leigh PN, Emile J, Guiloff RJ, Jerusalem F, Silani V, Vos PE, Wokke JH, Dobbins T, European ALS/IGF-I Study Group A placebo-controlled trial of insulin-like growth factor-I in amyotrophic lateral sclerosis. Neurology. 1998;51:583–586. doi: 10.1212/wnl.51.2.583. [DOI] [PubMed] [Google Scholar]
  10. Boulanger L, Poo MM. Presynaptic depolarization facilitates neurotrophin-induced synaptic potentiation. Nat. Neurosci. 1999;2:346–351. doi: 10.1038/7258. [DOI] [PubMed] [Google Scholar]
  11. Boyd JG, Gordon T. Glial cell line-derived neurotrophic factor and brain-derived neurotrophic factor sustain the axonal regeneration of chronically axotomized motoneurons in vivo. Exp. Neurol. 2003;183:610–619. doi: 10.1016/s0014-4886(03)00183-3. [DOI] [PubMed] [Google Scholar]
  12. Bregman BS, McAtee M, Dai HN, Kuhn PL. Neurotrophic factors increase axonal growth after spinal cord injury and transplantation in the adult rat. Exp. Neurol. 1997;148:475–494. doi: 10.1006/exnr.1997.6705. [DOI] [PubMed] [Google Scholar]
  13. Brunet N, Tarabal O, Portero-Otin M, Oppenheim RW, Esquerda JE, Caldero J. Survival and death of mature avian motoneurons in organotypic slice culture: trophic requirements for survival and different types of degeneration. J. Comp. Neurol. 2007;501:669–690. doi: 10.1002/cne.21157. [DOI] [PubMed] [Google Scholar]
  14. Buck CR, Seburn KL, Cope TC. Neurotrophin expression by spinal motoneurons in adult and developing rats. J. Comp. Neurol. 2000;416:309–318. doi: 10.1002/(sici)1096-9861(20000117)416:3<309::aid-cne3>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
  15. Buonanno A, Fischbach GD. Neuregulin and ErbB receptor signaling pathways in the nervous system. Curr. Opin. Neurobiol. 2001;11:287–296. doi: 10.1016/s0959-4388(00)00210-5. [DOI] [PubMed] [Google Scholar]
  16. Burke RE. Motor units: anatomy, physiology and functional organization. In: Peachey LD, editor. Handbook of Physiology. The Nervous System. Motor Control. Vol. 3. Am Physiol Soc; Bethesda, MD: 1981. pp. 345–422. [Google Scholar]
  17. Butler JE, McKenzie DK, Gandevia SC. Discharge properties and recruitment of human diaphragmatic motor units during voluntary inspiratory tasks. J. Physiol. 1999;518:907–920. doi: 10.1111/j.1469-7793.1999.0907p.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cai LL, Courtine G, Fong AJ, Burdick JW, Roy RR, Edgerton VR. Plasticity of functional connectivity in the adult spinal cord. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 2006;361:1635–1646. doi: 10.1098/rstb.2006.1884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Cameron WE, Brozanski BS, Guthrie RD. Postnatal development of phrenic motoneurons in the cat. Dev. Brain Res. 1990;51:142–145. doi: 10.1016/0165-3806(90)90269-5. [DOI] [PubMed] [Google Scholar]
  20. Cameron WE, He F, Kalipatnapu P, Jodkowski JS, Guthrie RD. Morphometric analysis of phrenic motoneurons in the cat during postnatal development. J. Comp. Neurol. 1991;314:763–776. doi: 10.1002/cne.903140409. [DOI] [PubMed] [Google Scholar]
  21. Canossa M, Griesbeck O, Berninger B, Campana G, Kolbeck R, Thoenen H. Neurotrophin release by neurotrophins: implications for activity-dependent neuronal plasticity. Proc. Natl. Acad. Sci. U S A. 1997;94:13279–13286. doi: 10.1073/pnas.94.24.13279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Citri A, Skaria KB, Yarden Y. The deaf and the dumb: the biology of ErbB-2 and ErbB-3. Exp. Cell Res. 2003;284:54–65. doi: 10.1016/s0014-4827(02)00101-5. [DOI] [PubMed] [Google Scholar]
  23. Conover JC, Erickson JT, Katz DM, Bianchi LM, Poueymirou WT, McClain J, Pan L, Helgren M, Ip NY, Boland P, Friedman B, Wiegand S, Vejsada R, Kato AC, Dechiara TM, Yancopoulos GD. Neuronal deficits, not involving motor neurons, in mice lacking BDNF and/or NT4. Nature. 1995;375:235–238. doi: 10.1038/375235a0. [DOI] [PubMed] [Google Scholar]
  24. Copray S, Kernell D. Neurotrophins and trk-receptors in adult rat spinal motoneurons: differences related to cell size but not to ‘slow/fast’ specialization. Neurosci. Lett. 2000;289:217–220. doi: 10.1016/s0304-3940(00)01305-7. [DOI] [PubMed] [Google Scholar]
  25. Courtine G, Song B, Roy RR, Zhong H, Herrmann JE, Ao Y, Qi J, Edgerton VR, Sofroniew MV. Recovery of supraspinal control of stepping via indirect propriospinal relay connections after spinal cord injury. Nat. Med. 2008;14:69–74. doi: 10.1038/nm1682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Dobbins EG, Feldman JL. Brainstem network controlling descending drive to phrenic motoneurons in rat. J. Comp. Neurol. 1994;347:64–86. doi: 10.1002/cne.903470106. [DOI] [PubMed] [Google Scholar]
  27. Dolbeare D, Houle JD. Restriction of axonal retraction and promotion of axonal regeneration by chronically injured neurons after intraspinal treatment with glial cell line-derived neurotrophic factor (GDNF) J. Neurotrauma. 2003;20:1251–1261. doi: 10.1089/089771503770802916. [DOI] [PubMed] [Google Scholar]
  28. Domeniconi M, Hempstead BL, Chao MV. Pro-NGF secreted by astrocytes promotes motor neuron cell death. Mol. Cell Neurosci. 2007;34:271–279. doi: 10.1016/j.mcn.2006.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Dougherty KD, Dreyfus CF, Black IB. Brain-derived neurotrophic factor in astrocytes, oligodendrocytes, and microglia/macrophages after spinal cord injury. Neurobiol. Dis. 2000;7:574–585. doi: 10.1006/nbdi.2000.0318. [DOI] [PubMed] [Google Scholar]
  30. Ellenberger HH, Feldman JL. Monosynaptic transmission of respiratory drive to phrenic motoneurons from brainstem bulbospinal neurons in rats. J. Comp. Neurol. 1988;269:47–57. doi: 10.1002/cne.902690104. [DOI] [PubMed] [Google Scholar]
  31. Ellenberger HH, Feldman JL, Goshgarian HG. Ventral respiratory group projections to phrenic motoneurons: electron microscopic evidence for monosynaptic connections. J. Comp. Neurol. 1990;302:707–714. doi: 10.1002/cne.903020403. [DOI] [PubMed] [Google Scholar]
  32. Erickson JT, Conover JC, Borday V, Champagnat J, Barbacid M, Yancopoulos G, Katz DM. Mice lacking brain-derived neurotrophic factor exhibit visceral sensory neuron losses distinct from mice lacking NT4 and display a severe developmental deficit in control of breathing. J. Neurosci. 1996;16:5361–5371. doi: 10.1523/JNEUROSCI.16-17-05361.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Ermilov LG, Mantilla CB, Rowley KL, Sieck GC. Safety factor for neuromuscular transmission at type-identified diaphragm fibers. Muscle Nerve. 2007;35:800–803. doi: 10.1002/mus.20751. [DOI] [PubMed] [Google Scholar]
  34. Ernfors P, Lee KF, Jaenisch R. Mice lacking brain-derived neurotrophic factor develop with sensory deficits. Nature. 1994;368:147–150. doi: 10.1038/368147a0. [DOI] [PubMed] [Google Scholar]
  35. Escandon E, Soppet D, Rosenthal A, Mendoza-Ramirez J-L, Szonyi E, Burton LE, Henderson CE, Parada LF, Nikolics K. Regulation of neurotrophin receptor expression during embryonic and postnatal development. J. Neurosci. 1994;14:2054–2068. doi: 10.1523/JNEUROSCI.14-04-02054.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Escher P, Lacazette E, Courtet M, Blindenbacher A, Landmann L, Bezakova G, Lloyd KC, Mueller U, Brenner HR. Synapses form in skeletal muscles lacking neuregulin receptors. Science. 2005;308:1920–1923. doi: 10.1126/science.1108258. [DOI] [PubMed] [Google Scholar]
  37. Falls DL. Neuregulins: functions, forms, and signaling strategies. Exp. Cell Res. 2003;284:14–30. doi: 10.1016/s0014-4827(02)00102-7. [DOI] [PubMed] [Google Scholar]
  38. Falls DL, Rosen KM, Corfas G, Lane WS, Fischbach GD. ARIA, a protein that stimulates acetylcholine receptor synthesis, is a member of the neu ligand family. Cell. 1993;72:801–815. doi: 10.1016/0092-8674(93)90407-h. [DOI] [PubMed] [Google Scholar]
  39. Feldman JL, Loewy AD, Speck DF. Projections from the ventral respiratory group to phrenic and intercostal motoneurons in cat: an autoradiographic study. J. Neurosci. 1985;5:1993–2000. doi: 10.1523/JNEUROSCI.05-08-01993.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Fournier M, Sieck GC. Mechanical properties of muscle units in the cat diaphragm. J. Neurophysiol. 1988;59:1055–1066. doi: 10.1152/jn.1988.59.3.1055. [DOI] [PubMed] [Google Scholar]
  41. Frisen J, Verge VM, Fried K, Risling M, Persson H, Trotter J, Hokfelt T, Lindholm D. Characterization of glial trkB receptors: differential response to injury in the central and peripheral nervous systems. Proc. Natl. Acad. Sci. U S A. 1993;90:4971–4975. doi: 10.1073/pnas.90.11.4971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Funakoshi H, Belluardo N, Arenasa E, Yamamoto Y, Casabona A, Persson H, Ibanez CF. Muscle-derived neurotrophin-4 as an activity-dependent trophic signal for adult motor neurons. Science. 1995;268:1495–1499. doi: 10.1126/science.7770776. [DOI] [PubMed] [Google Scholar]
  43. Funakoshi H, Frisen J, Barbany G, Zachrisson O, Verge VM, Persson H. Differential expression of mRNAs for neurotrophins and their receptors after axotomy. J. Cell. Biol. 1993;123:455–465. doi: 10.1083/jcb.123.2.455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Garces A, Haase G, Airaksinen MS, Livet J, Filippi P, deLapeyriere O. GFRalpha 1 is required for development of distinct subpopulations of motoneuron. J. Neurosci. 2000;20:4992–5000. doi: 10.1523/JNEUROSCI.20-13-04992.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Geiger PC, Cody MJ, Macken RL, Sieck GC. Maximum specific force depends on myosin heavy chain content in rat diaphragm muscle fibers. J. Appl. Physiol. 2000;89:695–703. doi: 10.1152/jappl.2000.89.2.695. [DOI] [PubMed] [Google Scholar]
  46. Golden JP, DeMaro JA, Osborne PA, Milbrandt J, Johnson EM., Jr. Expression of neurturin, GDNF, and GDNF family-receptor mRNA in the developing and mature mouse. Exp. Neurol. 1999;158:504–528. doi: 10.1006/exnr.1999.7127. [DOI] [PubMed] [Google Scholar]
  47. Golder FJ, Ranganathan L, Satriotomo I, Hoffman M, Lovett-Barr MR, Watters JJ, Baker-Herman TL, Mitchell GS. Spinal adenosine A2a receptor activation elicits long-lasting phrenic motor facilitation. J. Neurosci. 2008;28:2033–2042. doi: 10.1523/JNEUROSCI.3570-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Gonzalez M, Ruggiero FP, Chang Q, Shi YJ, Rich MM, Kraner S, Balice-Gordon RJ. Disruption of Trkb-mediated signaling induces disassembly of postsynaptic receptor clusters at neuromuscular junctions. Neuron. 1999;24:567–583. doi: 10.1016/s0896-6273(00)81113-7. [DOI] [PubMed] [Google Scholar]
  49. Gordon T, Thomas CK, Munson JB, Stein RB. The resilience of the size principle in the organization of motor unit properties in normal and reinnervated adult skeletal muscles. Can. J. Physiol. Pharmacol. 2004;82:645–661. doi: 10.1139/y04-081. [DOI] [PubMed] [Google Scholar]
  50. Gould TW, Yonemura S, Oppenheim RW, Ohmori S, Enomoto H. The neurotrophic effects of glial cell line-derived neurotrophic factor on spinal motoneurons are restricted to fusimotor subtypes. J. Neurosci. 2008;28:2131–2146. doi: 10.1523/JNEUROSCI.5185-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Graus-Porta D, Beerli RR, Daly JM, Hynes NE. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J. 1997;16:1647–1655. doi: 10.1093/emboj/16.7.1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Greer JJ, Funk GD, Ballanyi K. Preparing for the first breath: prenatal maturation of respiratory neural control. J. Physiol. 2006;570:437–444. doi: 10.1113/jphysiol.2005.097238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Greer JJ, Smith JC, Feldman JL. Respiratory and locomotor patterns generated in the fetal rat brain stem-spinal cord in vitro. J. Neurophysiol. 1992;67:996–999. doi: 10.1152/jn.1992.67.4.996. [DOI] [PubMed] [Google Scholar]
  54. Griesbeck O, Parsadanian AS, Sendtner M, Thoenen H. Expression of neurotrophins in skeletal muscle: quantitative comparison and significance for motoneuron survival and maintenance of function. J. Neurosci. Res. 1995;42:21–33. doi: 10.1002/jnr.490420104. [DOI] [PubMed] [Google Scholar]
  55. Hamburger V. History of the discovery of neuronal death in embryos. J. Neurobiol. 1992;23:1116–1123. doi: 10.1002/neu.480230904. [DOI] [PubMed] [Google Scholar]
  56. Hellyer NJ, Cheng K, Koland JG. ErbB3 (HER3) interaction with the p85 regulatory subunit of phosphoinositide 3-kinase. Biochem. J. 1998;333(Pt 3):757–763. doi: 10.1042/bj3330757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hellyer NJ, Mantilla CB, Park EW, Zhan WZ, Sieck GC. Neuregulin-dependent protein synthesis in C2C12 myotubes and rat diaphragm muscle. Am. J. Physiol. Cell. Physiol. 2006;291:C1056–1061. doi: 10.1152/ajpcell.00625.2005. [DOI] [PubMed] [Google Scholar]
  58. Henderson CE, Camu W, Mettling C, Gouin A, Poulsen K, Karihaloo M, Rullamas J, Evans T, McMahon SB, Armanini MP, Berkemeier L, Phillips HS, Rosenthal A. Neurotrophins promote motor neuron survival and are present in embryonic limb bud. Nature. 1993;363:266–270. doi: 10.1038/363266a0. [DOI] [PubMed] [Google Scholar]
  59. Henderson CE, Phillips HS, Pollack RA, Davies AM, Lemeulle C, Armanini M, Simpson LC, Moffet B, Vandlen RA, Koliatsos VE, Rosenthal A. GDNF: A potent survival factor for motoneurons present in peripheral nerve and muscle. Science. 1994;266:1062–1064. doi: 10.1126/science.7973664. [DOI] [PubMed] [Google Scholar]
  60. Henneman E, Somjen G, Carpenter DO. Functional significance of cell size in spinal motoneurons. J. Neurophysiol. 1965;28:560–580. doi: 10.1152/jn.1965.28.3.560. [DOI] [PubMed] [Google Scholar]
  61. Hensbergen E, Kernell D. Daily durations of spontaneous activity in cat’s ankle muscles. Exp. Brain Res. 1997;115:325–332. doi: 10.1007/pl00005701. [DOI] [PubMed] [Google Scholar]
  62. Hess DM, Scott MO, Potluri S, Pitts EV, Cisterni C, Balice-Gordon RJ. Localization of TrkC to Schwann cells and effects of neurotrophin-3 signaling at neuromuscular synapses. J. Comp. Neurol. 2007;501:465–482. doi: 10.1002/cne.21163. [DOI] [PubMed] [Google Scholar]
  63. Heuckeroth RO, Enomoto H, Grider JR, Golden JP, Hanke JA, Jackman A, Molliver DC, Bardgett ME, Snider WD, Johnson EM, Jr., Milbrandt J. Gene targeting reveals a critical role for neurturin in the development and maintenance of enteric, sensory, and parasympathetic neurons. Neuron. 1999;22:253–263. doi: 10.1016/s0896-6273(00)81087-9. [DOI] [PubMed] [Google Scholar]
  64. Hilfiker S, Pieribone VA, Czernik AJ, Kao HT, Augustine GJ, Greengard P. Synapsins as regulators of neurotransmitter release. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 1999;354:269–279. doi: 10.1098/rstb.1999.0378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hirata M, Sakuma K, Okajima S, Fujiwara H, Inashima S, Yasuhara M, Kubo T. Increased expression of neuregulin-1 in differentiating muscle satellite cells and in motoneurons during muscle regeneration. Acta Neuropathol. 2007;113:451–459. doi: 10.1007/s00401-007-0198-5. [DOI] [PubMed] [Google Scholar]
  66. Homma S, Yaginuma H, Vinsant S, Seino M, Kawata M, Gould T, Shimada T, Kobayashi N, Oppenheim RW. Differential expression of the GDNF family receptors RET and GFRalpha1, 2, and 4 in subsets of motoneurons: a relationship between motoneuron birthdate and receptor expression. J. Comp. Neurol. 2003;456:245–259. doi: 10.1002/cne.10529. [DOI] [PubMed] [Google Scholar]
  67. Iarikov DE, Kim BG, Dai HN, McAtee M, Kuhn PL, Bregman BS. Delayed transplantation with exogenous neurotrophin administration enhances plasticity of corticofugal projections after spinal cord injury. J. Neurotrauma. 2007;24:690–702. doi: 10.1089/neu.2006.0172. [DOI] [PubMed] [Google Scholar]
  68. Ikeda O, Murakami M, Ino H, Yamazaki M, Nemoto T, Koda M, Nakayama C, Moriya H. Acute up-regulation of brain-derived neurotrophic factor expression resulting from experimentally induced injury in the rat spinal cord. Acta Neuropathol. (Berl) 2001;102:239–245. doi: 10.1007/s004010000357. [DOI] [PubMed] [Google Scholar]
  69. Ip FC, Cheung J, Ip NY. The expression profiles of neurotrophins and their receptors in rat and chicken tissues during development. Neurosci. Lett. 2001;301:107–110. doi: 10.1016/s0304-3940(01)01603-2. [DOI] [PubMed] [Google Scholar]
  70. Jansen P, Giehl K, Nyengaard JR, Teng K, Lioubinski O, Sjoegaard SS, Breiderhoff T, Gotthardt M, Lin F, Eilers A, Petersen CM, Lewin GR, Hempstead BL, Willnow TE, Nykjaer A. Roles for the pro-neurotrophin receptor sortilin in neuronal development, aging and brain injury. Nat. Neurosci. 2007;10:1449–1457. doi: 10.1038/nn2000. [DOI] [PubMed] [Google Scholar]
  71. Jaworski A, Burden SJ. Neuromuscular synapse formation in mice lacking motor neuron- and skeletal muscle-derived Neuregulin-1. J. Neurosci. 2006;26:655–661. doi: 10.1523/JNEUROSCI.4506-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Jo SA, Zhu X, Marchionni MA, Burden SJ. Neuregulins are concentrated at nerve-muscle synapses and activate ACh-receptor gene expression. Nature. 1995;373:158–161. doi: 10.1038/373158a0. [DOI] [PubMed] [Google Scholar]
  73. Johnson BD, Wilson LE, Zhan WZ, Watchko JF, Daood MJ, Sieck GC. Contractile properties of the developing diaphragm correlate with myosin heavy chain phenotype. J. Appl. Physiol. 1994;77:481–487. doi: 10.1152/jappl.1994.77.1.481. [DOI] [PubMed] [Google Scholar]
  74. Johnson RA, Okragly AJ, Haak-Frendscho M, Mitchell GS. Cervical dorsal rhizotomy increases brain-derived neurotrophic factor and neurotrophin-3 expression in the ventral spinal cord. J. Neurosci. 2000;20:77. doi: 10.1523/JNEUROSCI.20-10-j0005.2000. RC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Jones JT, Akita RW, Sliwkowski MX. Binding specificities and affinities of egf domains for ErbB receptors. FEBS Lett. 1999;447:227–231. doi: 10.1016/s0014-5793(99)00283-5. [DOI] [PubMed] [Google Scholar]
  76. Jovanovic JN, Czernik AJ, Fienberg AA, Greengard P, Sihra TS. Synapsins as mediators of BDNF-enhanced neurotransmitter release. Nat. Neurosci. 2000;3:323–329. doi: 10.1038/73888. [DOI] [PubMed] [Google Scholar]
  77. Kafitz KW, Rose CR, Thoenen H, Konnerth A. Neurotrophin-evoked rapid excitation through TrkB receptors. Nature. 1999;401:918–921. doi: 10.1038/44847. [DOI] [PubMed] [Google Scholar]
  78. Kang H, Schuman EM. Long-lasting neurotrophin-induced enhancement of synaptic transmission in the adult hippocampus. Science. 1995;267:1658–1662. doi: 10.1126/science.7886457. [DOI] [PubMed] [Google Scholar]
  79. Keller-Peck CR, Feng G, Sanes JR, Yan Q, Lichtman JW, Snider WD. Glial cell line-derived neurotrophic factor administration in postnatal life results in motor unit enlargement and continuous synaptic remodeling at the neuromuscular junction. J. Neurosci. 2001;21:6136–6146. doi: 10.1523/JNEUROSCI.21-16-06136.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Kerber G, Streif R, Schwaiger FW, Kreutzberg GW, Hager G. Neuregulin-1 isoforms are differentially expressed in the intact and regenerating adult rat nervous system. J. Mol. Neurosci. 2003;21:149–165. doi: 10.1385/JMN:21:2:149. [DOI] [PubMed] [Google Scholar]
  81. Keswani NH, Hollinshead WH. The phrenic nucleus. III. Organization of the phrenic nucleus in the spinal cord of the cat and man. Proc. Staff Meet. Mayo Clin. 1955;30:566–577. [PubMed] [Google Scholar]
  82. Kinkead R, Zhan WZ, Prakash YS, Bach KB, Sieck GC, Mitchell GS. Cervical dorsal rhizotomy enhances serotonergic innervation of phrenic motoneurons and serotonin-dependent long-term facilitation of respiratory motor output in rats. J. Neurosci. 1998;18:8436–8443. doi: 10.1523/JNEUROSCI.18-20-08436.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Koliatsos VE, Clatterbuck RE, Winslow JW, Cayouette MH, Price DL. Evidence that brain-derived neurotrophic factor is a trophic factor for motor neurons in vivo. Neuron. 1993;10:359–367. doi: 10.1016/0896-6273(93)90326-m. [DOI] [PubMed] [Google Scholar]
  84. Koliatsos VE, Crawford TO, Price DL. Axotomy induces nerve growth factor receptor immunoreactivity in spinal motor neurons. Brain Res. 1991;549:297–304. doi: 10.1016/0006-8993(91)90471-7. [DOI] [PubMed] [Google Scholar]
  85. Kong FJ, Berger AJ. Firing properties and hypercapnic responses of single phrenic motor axons in the rat. J. Appl. Physiol. 1986;61:1999–2004. doi: 10.1152/jappl.1986.61.6.1999. [DOI] [PubMed] [Google Scholar]
  86. Lebrasseur NK, Cote GM, Miller TA, Fielding RA, Sawyer DB. Regulation of neuregulin/ErbB signaling by contractile activity in skeletal muscle. Am. J. Physiol. Cell Physiol. 2003;284:C1149–1155. doi: 10.1152/ajpcell.00487.2002. [DOI] [PubMed] [Google Scholar]
  87. Lee FS, Chao MV. Activation of Trk neurotrophin receptors in the absence of neurotrophins. Proc. Natl. Acad. Sci. U S A. 2001a;98:3555–3560. doi: 10.1073/pnas.061020198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Lee R, Kermani P, Teng KK, Hempstead BL. Regulation of cell survival by secreted proneurotrophins. Science. 2001b;294:1945–1948. doi: 10.1126/science.1065057. [DOI] [PubMed] [Google Scholar]
  89. Li M, Sendtner M, Smith A. Essential function of LIF receptor in motor neurons. Nature. 1995;378:724–727. doi: 10.1038/378724a0. [DOI] [PubMed] [Google Scholar]
  90. Li Z, Mei Y, Liu X, Zhou M. Neuregulin-1 only induces transphosphorylation between ErbB receptor heterodimer partners. Cell Signal. 2007;19:466–471. doi: 10.1016/j.cellsig.2006.07.020. [DOI] [PubMed] [Google Scholar]
  91. Lie DC, Weis J. GDNF expression is increased in denervated human skeletal muscle. Neurosci. Lett. 1998;250:87–90. doi: 10.1016/s0304-3940(98)00434-0. [DOI] [PubMed] [Google Scholar]
  92. Lohof AM, Ip NY, Poo MM. Potentiation of developing neuromuscular synapses by the neurotrophins NT-3 and BDNF. Nature. 1993;363:350–353. doi: 10.1038/363350a0. [DOI] [PubMed] [Google Scholar]
  93. Lu P, Jones LL, Snyder EY, Tuszynski MH. Neural stem cells constitutively secrete neurotrophic factors and promote extensive host axonal growth after spinal cord injury. Exp. Neurol. 2003;181:115–129. doi: 10.1016/s0014-4886(03)00037-2. [DOI] [PubMed] [Google Scholar]
  94. Mantilla CB, Rowley KL, Fahim MA, Zhan WZ, Sieck GC. Synaptic vesicle cycling at type-identified diaphragm neuromuscular junctions. Muscle Nerve. 2004a;30:774–783. doi: 10.1002/mus.20173. [DOI] [PubMed] [Google Scholar]
  95. Mantilla CB, Sieck GC. Key aspects of phrenic motoneuron and diaphragm muscle development during the perinatal period. J. Appl. Physiol. 2008;104:1818–1827. doi: 10.1152/japplphysiol.01192.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Mantilla CB, Zhan WZ, Sieck GC. Neurotrophins improve neuromuscular transmission in the adult rat diaphragm. Muscle Nerve. 2004b;29:381–386. doi: 10.1002/mus.10558. [DOI] [PubMed] [Google Scholar]
  97. Miller RG, Petajan JH, Bryan WW, Armon C, Barohn RJ, Goodpasture JC, Hoagland RJ, Parry GJ, Ross MA, Stromatt SC, rhCNTF ALS Study Group A placebo-controlled trial of recombinant human ciliary neurotrophic (rhCNTF) factor in amyotrophic lateral sclerosis. Ann. Neurol. 1996;39:256–260. doi: 10.1002/ana.410390215. [DOI] [PubMed] [Google Scholar]
  98. Moscoso LM, Chu GC, Gautam M, Noakes PG, Merlie JP, Sanes JR. Synapse-associated expression of an acetylcholine receptor-inducing protein, ARIA/heregulin, and its putative receptors, ErbB2 and ErbB3, in developing mammalian muscle. Dev. Biol. 1995;172:158–169. doi: 10.1006/dbio.1995.0012. [DOI] [PubMed] [Google Scholar]
  99. Mousavi K, Miranda W, Parry DJ. Neurotrophic factors enhance the survival of muscle fibers in EDL, but not SOL, after neonatal nerve injury. Am. J. Physiol. Cell Physiol. 2002;283:C950–959. doi: 10.1152/ajpcell.00081.2002. [DOI] [PubMed] [Google Scholar]
  100. Mousavi K, Parry DJ, Jasmin BJ. BDNF rescues myosin heavy chain IIB muscle fibers after neonatal nerve injury. Am. J. Physiol. Cell Physiol. 2004;287:C22–29. doi: 10.1152/ajpcell.00583.2003. [DOI] [PubMed] [Google Scholar]
  101. Neff NT, Prevette D, Houenou LJ, Lewis ME, Glicksman MA, Yin QW, Oppenheim RW. Insulin-like growth factors: putative muscle-derived trophic agents that promote motoneuron survival. J. Neurobiol. 1993;24:1578–1588. doi: 10.1002/neu.480241203. [DOI] [PubMed] [Google Scholar]
  102. Nguyen QT, Prasadanian AS, Snider WD, Lichtman JW. Hyperinnervation of neuromuscular junctions caused by GDNF overexpression in muscle. Science. 1998;279:1725–1729. doi: 10.1126/science.279.5357.1725. [DOI] [PubMed] [Google Scholar]
  103. Novikova LN, Novikov LN, Kellerth JO. Differential effects of neurotrophins on neuronal survival and axonal regeneration after spinal cord injury in adult rats. J. Comp. Neurol. 2002;452:255–263. doi: 10.1002/cne.10381. [DOI] [PubMed] [Google Scholar]
  104. Ochs G, Penn RD, York M, Giess R, Beck M, Tonn J, Haigh J, Malta E, Traub M, Sendtner M, Toyka KV. A phase I/II trial of recombinant methionyl human brain derived neurotrophic factor administered by intrathecal infusion to patients with amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Other Motor Neuron Disord. 2000;1:201–206. doi: 10.1080/14660820050515197. [DOI] [PubMed] [Google Scholar]
  105. Olayioye MA, Graus-Porta D, Beerli RR, Rohrer J, Gay B, Hynes NE. ErbB-1 and ErbB-2 acquire distinct signaling properties dependent upon their dimerization partner. Mol. Cell Biol. 1998;18:5042–5051. doi: 10.1128/mcb.18.9.5042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Oppenheim RW. The absence of significant postnatal motoneuron death in the brachial and lumbar spinal cord of the rat. J. Comp. Neurol. 1986;246:281–286. doi: 10.1002/cne.902460211. [DOI] [PubMed] [Google Scholar]
  107. Oppenheim RW. Neurotrophic survival molecules for motoneurons: an embarrassment of riches. Neuron. 1996;17:195–197. doi: 10.1016/s0896-6273(00)80151-8. [DOI] [PubMed] [Google Scholar]
  108. Oppenheim RW, Houenou LJ, Parsadanian AS, Prevette D, Snider WD, Shen L. Glial cell line-derived neurotrophic factor and developing mammalian motoneurons: regulation of programmed cell death among motoneuron subtypes. J. Neurosci. 2000;20:5001–5011. doi: 10.1523/JNEUROSCI.20-13-05001.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Oppenheim RW, Prevette D, Qin-Wei Y, Collins F, MacDonald J. Control of embryonic motoneuron survival in vivo by ciliary neurotrophic factor. Science. 1991;251:1616–1618. doi: 10.1126/science.2011743. [DOI] [PubMed] [Google Scholar]
  110. Oppenheim RW, Wiese S, Prevette D, Armanini M, Wang S, Houenou LJ, Holtmann B, Gotz R, Pennica D, Sendtner M. Cardiotrophin-1, a muscle-derived cytokine, is required for the survival of subpopulations of developing motoneurons. J. Neurosci. 2001;21:1283–1291. doi: 10.1523/JNEUROSCI.21-04-01283.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Oppenheim RW, Yin QW, Prevette D, Yan Q. Brain-derived neurotrophic factor rescues developing avian motoneurons from cell death. Nature. 1992;360:755–757. doi: 10.1038/360755a0. [DOI] [PubMed] [Google Scholar]
  112. Paratcha G, Ledda F, Ibanez CF. The neural cell adhesion molecule NCAM is an alternative signaling receptor for GDNF family ligands. Cell. 2003;113:867–879. doi: 10.1016/s0092-8674(03)00435-5. [DOI] [PubMed] [Google Scholar]
  113. Pearson RJ, Jr., Carroll SL. ErbB transmembrane tyrosine kinase receptors are expressed by sensory and motor neurons projecting into sciatic nerve. J. Histochem. Cytochem. 2004;52:1299–1311. doi: 10.1177/002215540405201006. [DOI] [PubMed] [Google Scholar]
  114. Poo MM. Neurotrophins as synaptic modulators. Nat. Rev. Neurosci. 2001;2:24–32. doi: 10.1038/35049004. [DOI] [PubMed] [Google Scholar]
  115. Prakash YS, Fournier M, Sieck GC. Effects of prenatal undernutrition on developing rat diaphragm. J. Appl. Physiol. 1993;75:1044–1052. doi: 10.1152/jappl.1993.75.3.1044. [DOI] [PubMed] [Google Scholar]
  116. Prakash YS, Mantilla CB, Zhan WZ, Smithson KG, Sieck GC. Phrenic motoneuron morphology during rapid diaphragm muscle growth. J. Appl. Physiol. 2000;89:563–572. doi: 10.1152/jappl.2000.89.2.563. [DOI] [PubMed] [Google Scholar]
  117. Rajagopal R, Chen ZY, Lee FS, Chao MV. Transactivation of Trk neurotrophin receptors by G-protein-coupled receptor ligands occurs on intracellular membranes. J. Neurosci. 2004;24:6650–6658. doi: 10.1523/JNEUROSCI.0010-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Reichardt LF. Neurotrophin-regulated signalling pathways. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 2006;361:1545–1564. doi: 10.1098/rstb.2006.1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Ribchester RR, Thomson D, Haddow LJ, Ushkaryov YA. Enhancement of spontaneous transmitter release at neonatal mouse neuromuscular junctions by the glial cell line-derived neurotrophic factor (GDNF) J. Physiol. 1998;512:635–641. doi: 10.1111/j.1469-7793.1998.635bd.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Ricart K,J, Pearson R,J, Viera L, Cassina P, Kamaid A, Carroll SL, Estevez AG. Interactions between beta-neuregulin and neurotrophins in motor neuron apoptosis. J. Neurochem. 2006;97:222–233. doi: 10.1111/j.1471-4159.2006.03739.x. [DOI] [PubMed] [Google Scholar]
  121. Rimer M, Cohen I, Lomo T, Burden SJ, McMahan UJ. Neuregulins and erbB receptors at neuromuscular junctions and at agrin-induced postsynaptic-like apparatus in skeletal muscle. Mol. Cell Neurosci. 1998;12:1–15. doi: 10.1006/mcne.1998.0695. [DOI] [PubMed] [Google Scholar]
  122. Rimer M, Prieto AL, Weber JL, Colasante C, Ponomareva O, Fromm L, Schwab MH, Lai C, Burden SJ. Neuregulin-2 is synthesized by motor neurons and terminal Schwann cells and activates acetylcholine receptor transcription in muscle cells expressing ErbB4. Mol. Cell Neurosci. 2004;26:271–281. doi: 10.1016/j.mcn.2004.02.002. [DOI] [PubMed] [Google Scholar]
  123. Rind HB, Butowt R, von Bartheld CS. Synaptic targeting of retrogradely transported trophic factors in motoneurons: comparison of glial cell line-derived neurotrophic factor, brain-derived neurotrophic factor, and cardiotrophin-1 with tetanus toxin. J. Neurosci. 2005;25:539–549. doi: 10.1523/JNEUROSCI.4322-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Rossi J, Luukko K, Poteryaev D, Laurikainen A, Sun YF, Laakso T, Eerikainen S, Tuominen R, Lakso M, Rauvala H, Arumae U, Pasternack M, Saarma M, Airaksinen MS. Retarded growth and deficits in the enteric and parasympathetic nervous system in mice lacking GFR alpha2, a functional neurturin receptor. Neuron. 1999;22:243–252. doi: 10.1016/s0896-6273(00)81086-7. [DOI] [PubMed] [Google Scholar]
  125. Rowley KL, Mantilla CB, Ermilov LG, Sieck GC. Synaptic vesicle distribution and release at rat diaphragm neuromuscular junctions. J. Neurophysiol. 2007;98:478–487. doi: 10.1152/jn.00251.2006. [DOI] [PubMed] [Google Scholar]
  126. Ruitenberg MJ, Blits B, Dijkhuizen PA, te Beek ET, Bakker A, van Heerikhuize JJ, Pool CW, Hermens WT, Boer GJ, Verhaagen J. Adeno-associated viral vector-mediated gene transfer of brain-derived neurotrophic factor reverses atrophy of rubrospinal neurons following both acute and chronic spinal cord injury. Neurobiol. Dis. 2004;15:394–406. doi: 10.1016/j.nbd.2003.11.018. [DOI] [PubMed] [Google Scholar]
  127. Russell FD, Koishi K, Jiang Y, McLennan IS. Anterograde axonal transport of glial cell line-derived neurotrophic factor and its receptors in rat hypoglossal nerve. Neuroscience. 2000;97:575–580. doi: 10.1016/s0306-4522(00)00079-8. [DOI] [PubMed] [Google Scholar]
  128. Sakuma K, Watanabe K, Sano M, Uramoto I, Nakano H, Li YJ, Kaneda S, Sorimachi Y, Yoshimoto K, Yasuhara M, Totsuka T. A possible role for BDNF, NT-4 and TrkB in the spinal cord and muscle of rat subjected to mechanical overload, bupivacaine injection and axotomy. Brain Res. 2001;907:1–19. doi: 10.1016/s0006-8993(01)02288-0. [DOI] [PubMed] [Google Scholar]
  129. Sandrock AW, Jr., Dryer SE, Rosen KM, Gozani SN, Kramer R, Theill LE, Fischbach GD. Maintenance of acetylcholine receptor number by neuregulins at the neuromuscular junction in vivo. Science. 1997;276:599–603. doi: 10.1126/science.276.5312.599. [DOI] [PubMed] [Google Scholar]
  130. Sanes JR, Lichtman JW. Induction, assembly, maturation and maintenance of a postsynaptic apparatus. Nat. Rev. Neurosci. 2001;2:791–805. doi: 10.1038/35097557. [DOI] [PubMed] [Google Scholar]
  131. Sariola H, Saarma M. Novel functions and signalling pathways for GDNF. J Cell Sci. 2003;116:3855–3862. doi: 10.1242/jcs.00786. [DOI] [PubMed] [Google Scholar]
  132. Satake K, Matsuyama Y, Kamiya M, Kawakami H, Iwata H, Adachi K, Kiuchi K. Up-regulation of glial cell line-derived neurotrophic factor (GDNF) following traumatic spinal cord injury. Neuroreport. 2000;11:3877–3881. doi: 10.1097/00001756-200011270-00054. [DOI] [PubMed] [Google Scholar]
  133. Schinder AF, Poo M. The neurotrophin hypothesis for synaptic plasticity. Trends Neurosci. 2000;23:639–645. doi: 10.1016/s0166-2236(00)01672-6. [DOI] [PubMed] [Google Scholar]
  134. Sendtner M, Pei G, Beck M, Schweizer U, Wiese S. Developmental motoneuron cell death and neurotrophic factors. Cell Tissue Res. 2000;301:71–84. doi: 10.1007/s004410000217. [DOI] [PubMed] [Google Scholar]
  135. Sieck GC. Diaphragm muscle: structural and functional organization. Clin. Chest Med. 1988;9:195–210. [PubMed] [Google Scholar]
  136. Sieck GC. Neural control of the inspiratory pump. NIPS. 1991;6:260–264. [Google Scholar]
  137. Sieck GC. Physiological effects of diaphragm muscle denervation and disuse. Clin. Chest Med. 1994;15:641–659. [PubMed] [Google Scholar]
  138. Sieck GC, Fournier M. Diaphragm motor unit recruitment during ventilatory and nonventilatory behaviors. J. Appl. Physiol. 1989a;66:2539–2545. doi: 10.1152/jappl.1989.66.6.2539. [DOI] [PubMed] [Google Scholar]
  139. Sieck GC, Fournier M, Blanco CE. Diaphragm muscle fatigue resistance during postnatal development. J. Appl. Physiol. 1991;71:458–464. doi: 10.1152/jappl.1991.71.2.458. [DOI] [PubMed] [Google Scholar]
  140. Sieck GC, Fournier M, Enad JG. Fiber type composition of muscle units in the cat diaphragm. Neurosci. Lett. 1989b;97:29–34. doi: 10.1016/0304-3940(89)90134-1. [DOI] [PubMed] [Google Scholar]
  141. Sieck GC, Fournier M, Prakash YS, Blanco CE. Myosin phenotype and SDH enzyme variability among motor unit fibers. J. Appl. Physiol. 1996;80:2179–2189. doi: 10.1152/jappl.1996.80.6.2179. [DOI] [PubMed] [Google Scholar]
  142. Simon M, Porter R, Brown R, Coulton GR, Terenghi G. Effect of NT-4 and BDNF delivery to damaged sciatic nerves on phenotypic recovery of fast and slow muscles fibres. Eur. J. Neurosci. 2003;18:2460–2466. doi: 10.1046/j.1460-9568.2003.02978.x. [DOI] [PubMed] [Google Scholar]
  143. Sliwkowski MX, Schaefer G, Akita RW, Lofgren JA, Fitzpatrick VD, Nuijens A, Fendly BM, Cerione RA, Vandlen RL, Carraway KL., 3rd Coexpression of erbB2 and erbB3 proteins reconstitutes a high affinity receptor for heregulin. J. Biol. Chem. 1994;269:14661–14665. [PubMed] [Google Scholar]
  144. Song A, Ashwell KW, Tracey DJ. Development of the rat phrenic nucleus and its connections with brainstem respiratory nuclei. Anat. Embryol. (Berl) 2000;202:159–177. doi: 10.1007/s004290000096. [DOI] [PubMed] [Google Scholar]
  145. Storkebaum E, Lambrechts D, Dewerchin M, Moreno-Murciano MP, Appelmans S, Oh H, Van Damme P, Rutten B, Man WY, De Mol M, Wyns S, Manka D, Vermeulen K, Van Den Bosch L, Mertens N, Schmitz C, Robberecht W, Conway EM, Collen D, Moons L, Carmeliet P. Treatment of motoneuron degeneration by intracerebroventricular delivery of VEGF in a rat model of ALS. Nat. Neurosci. 2005;8:85–92. doi: 10.1038/nn1360. [DOI] [PubMed] [Google Scholar]
  146. Su CK, Mellen NM, Feldman JL. Intrinsic and extrinsic factors affecting phrenic motoneuronal excitability in neonatal rats. Brain Res. 1997;774:62–68. doi: 10.1016/s0006-8993(97)81688-5. [DOI] [PubMed] [Google Scholar]
  147. Suzuki H, Hase A, Miyata Y, Arahata K, Akazawa C. Prominent expression of glial cell line-derived neurotrophic factor in human skeletal muscle. J. Comp. Neurol. 1998;402:303–312. [PubMed] [Google Scholar]
  148. Takahashi M. The GDNF/RET signaling pathway and human diseases. Cytokine Growth Factor Rev. 2001;12:361–373. doi: 10.1016/s1359-6101(01)00012-0. [DOI] [PubMed] [Google Scholar]
  149. Teng HK, Teng KK, Lee R, Wright S, Tevar S, Almeida RD, Kermani P, Torkin R, Chen ZY, Lee FS, Kraemer RT, Nykjaer A, Hempstead BL. ProBDNF induces neuronal apoptosis via activation of a receptor complex of p75NTR and sortilin. J. Neurosci. 2005;25:5455–5463. doi: 10.1523/JNEUROSCI.5123-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Teng YD, Mocchetti I, Taveira-DaSilva AM, Gillis RA, Wrathall JR. Basic fibroblast growth factor increases long-term survival of spinal motor neurons and improves respiratory function after experimental spinal cord injury. J. Neurosci. 1999;19:7037–7047. doi: 10.1523/JNEUROSCI.19-16-07037.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Timmusk T, Belluardo N, Metsis M, Persson H. Widespread and developmentally regulated expression of neurotrophin-4 messenger RNA in rat brain and peripheral tissues. Eur. J. Neurosci. 1993;5:605–613. doi: 10.1111/j.1460-9568.1993.tb00526.x. [DOI] [PubMed] [Google Scholar]
  152. Tissenbaum HA, Parry DJ. The effect of partial denervation of tibialis anterior muscle on the number and sizes of motoneurons in the motor nucleus of normal and dystrophic mice. Can. J. Physiol. Pharmacol. 1991;69:1769–1773. doi: 10.1139/y91-261. [DOI] [PubMed] [Google Scholar]
  153. Trinidad JC, Fischbach GD, Cohen JB. The Agrin/MuSK signaling pathway is spatially segregated from the neuregulin/ErbB receptor signaling pathway at the neuromuscular junction. J. Neurosci. 2000;20:8762–8770. doi: 10.1523/JNEUROSCI.20-23-08762.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Tzahar E, Waterman H, Chen X, Levkowitz G, Karunagaran D, Lavi S, Ratzkin BJ, Yarden Y. A hierarchical network of interreceptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor. Mol. Cell Biol. 1996;16:5276–5287. doi: 10.1128/mcb.16.10.5276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Vernon EM, Oppenheim RW, Johnson JE. Distinct muscle targets do not vary in the developmental regulation of brain-derived neurotrophic factor. J. Comp. Neurol. 2004;470:317–329. doi: 10.1002/cne.20018. [DOI] [PubMed] [Google Scholar]; J. Comp. Neurol. 2004;470:330–337. doi: 10.1002/cne.20018. [DOI] [PubMed] [Google Scholar]
  156. Wang XH, Poo MM. Potentiation of developing synapses by postsynaptic release of neurotrophin-4. Neuron. 1997;19:825–835. doi: 10.1016/s0896-6273(00)80964-2. [DOI] [PubMed] [Google Scholar]
  157. Webber CL, Wurster RD, Chung JM. Cat phrenic nucleus architecture as revealed by horseradish peroxidase mapping. Exp. Brain Res. 1979;35:395–406. doi: 10.1007/BF00236759. [DOI] [PubMed] [Google Scholar]
  158. Wehrwein EA, Roskelley EM, Spitsbergen JM. GDNF is regulated in an activity-dependent manner in rat skeletal muscle. Muscle Nerve. 2002;26:206–211. doi: 10.1002/mus.10179. [DOI] [PubMed] [Google Scholar]
  159. Widenfalk J, Lundstromer K, Jubran M, Brene S, Olson L. Neurotrophic factors and receptors in the immature and adult spinal cord after mechanical injury or kainic acid. J. Neurosci. 2001;21:3457–3475. doi: 10.1523/JNEUROSCI.21-10-03457.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Xie Y, Yao Z, Chai H, Wong WM, Wu W. Expression and role of low-affinity nerve growth factor receptor (p75) in spinal motor neurons of aged rats following axonal injury. Dev. Neurosci. 2003;25:65–71. doi: 10.1159/000071469. [DOI] [PubMed] [Google Scholar]
  161. Yan Q, Elliott JL, Matheson C, Sun J, Zhang L, Mu X, Rex KL, Snider WD. Influences of neurotrophins on mammalian motoneurons in vivo. J. Neurobiol. 1993;24:1555–1577. doi: 10.1002/neu.480241202. [DOI] [PubMed] [Google Scholar]
  162. Yan Q, Johnson EM. An immunohistochemical study of nerve growth factor receptor in developing rats. J. Neurosci. 1988;8:3841–3498. doi: 10.1523/JNEUROSCI.08-09-03481.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Yang F, He X, Feng L, Mizuno K, Liu XW, Russell J, Xiong WC, Lu B. PI-3 kinase and IP3 are both necessary and sufficient to mediate NT3-induced synaptic potentiation. Nat. Neurosci. 2001;4:19–28. doi: 10.1038/82858. [DOI] [PubMed] [Google Scholar]
  164. Yang LX, Nelson PG. Glia cell line-derived neurotrophic factor regulates the distribution of acetylcholine receptors in mouse primary skeletal muscle cells. Neuroscience. 2004;128:497–509. doi: 10.1016/j.neuroscience.2004.06.067. [DOI] [PubMed] [Google Scholar]
  165. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol. Cell Biol. 2001;2:127–137. doi: 10.1038/35052073. [DOI] [PubMed] [Google Scholar]
  166. Yates BJ, Smail JA, Stocker SD, Card JP. Transneuronal tracing of neural pathways controlling activity of diaphragm motoneurons in the ferret. Neuroscience. 1999;90:1501–1513. doi: 10.1016/s0306-4522(98)00554-5. [DOI] [PubMed] [Google Scholar]
  167. Zhu X, Lai C, Thomas S, Burden SJ. Neuregulin receptors, erbB3 and erbB4, are localized at neuromuscular synapses. EMBO J. 1995;14:5842–5848. doi: 10.1002/j.1460-2075.1995.tb00272.x. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES