neuromuscular junction formation

26
Neuromuscular Junction Formation-A great game for Agrin/LrP4/Musk signalling Abstract One of the greatest virtues of high vertebrates and mammals lies in their capability of performing complex and sophisticated tasks rendered by their behavioural changes according to environmental alterations. A well-orchestrated nervous system and locomotion system are two keys to this feature. The synapses, which are the fundamental function units of nervous system, have empowered the nervous system a speed of communication by electrical signals that is incomparable to anything on the planet. Neuromuscular junctions (NMJs) are special synapses formed between motoneurons and muscle fibres, making them the bridging structures between central nervous system (CNS) and muscular system. For this reason, the formation of NMJs has been an important research interest. Furthermore, abnormal NMJ formation encompasses a broad spectrum of diseases affecting the neuromotor system, making it an attractive therapeutic target. This review will focus on the underlying molecular mechanism and associated diseases of NMJ formation.

Upload: qilong-wu

Post on 14-Apr-2017

34 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Neuromuscular Junction Formation

Neuromuscular Junction Formation-A great game for Agrin/LrP4/Musk signalling

AbstractOne of the greatest virtues of high vertebrates and mammals lies in their capability of performing complex and sophisticated tasks rendered by their behavioural changes according to environmental alterations. A well-orchestrated nervous system and locomotion system are two keys to this feature. The synapses, which are the fundamental function units of nervous system, have empowered the nervous system a speed of communication by electrical signals that is incomparable to anything on the planet. Neuromuscular junctions (NMJs) are special synapses formed between motoneurons and muscle fibres, making them the bridging structures between central nervous system (CNS) and muscular system. For this reason, the formation of NMJs has been an important research interest. Furthermore, abnormal NMJ formation encompasses a broad spectrum of diseases affecting the neuromotor system, making it an attractive therapeutic target. This review will focus on the underlying molecular mechanism and associated diseases of NMJ formation.

Page 2: Neuromuscular Junction Formation

IntroductionSynapses are fundamental units of neuronal circuitry, which is the building block of nerve network of nerve network. A typical synapse comprises three elements. Namely, the pre-synaptic apparatus, post-synaptic apparatus and the intermediate synaptic cleft (Figure 1). In a neuromuscular junction (NMJ), which is a special type of synapse formed between motoneuron (the pre-synaptic apparatus) and muscle fibre (the post-synaptic apparatus), neurotransmitters, which are hormones of the nervous system, are synthesised and released from the pre-synaptic nerve terminals into the synaptic cleft where neurotransmitters diffuse and ultimately bind to specific receptors on post-synaptic fibre, causing cellular changes (i.e. transcriptional changes, opening of ion channels etc.) that subsequently lead to contraction (excitatory synapses) or relaxation (inhibitory synapses) of target muscle. The appearance of NMJs is believed to begins between embryonic development stage of day 12 to day 13.5 (1,2). At this stage, motoneurons that aggregate in the spinal cord stretch out and form growing axons in an effort to guide themselves to target muscle fibres. While nerve terminals play an active role in the path-exploring and guidance process, it is now being increasingly appreciated that post-synaptic muscle fibres and extra-synaptic glial cells (e.g. Schwann cells in peripheral nervous system, PNS) play indispensable roles in synapse formation and the subsequent events, including synapse maturation and synaptic plasticity is post-neonatal life. In addition to the interplay between those three elements (i.e. nerve terminal, muscle fibre and glial cell), the molecular interactions are regarded causative and deserve serious investigations. Owing to their large size and abundance, NMJs have fulfilled neurobiologist’s desire for a research model that renders them the possibility to study NMJ formation and the associated diseases (i.e. congenital myasthenia syndrome and myasthenia gravis) extensively. Indeed, research in the past decades has significantly extended our understandings. In particular, the discovery of Agrin/Lrp4/Musk signalling in NMJ formation has been exciting and

Page 3: Neuromuscular Junction Formation

inspiring novel research that tries to identify therapeutic targets for neuromotor system diseases.

This review will begin with an assertion of NMJ formation at cellular and molecular level followed by an explanation of how are Wnt and other molecules involved. The associated diseases (i.e. congenital myasthenia syndrome and myasthenia gravis) and the potential therapeutic strategies will be discussed.

NMJ formation at Cellular levelThe formation of NMJs at cellular level is largely attributable to interactions between the growing nerve terminal, muscle fibre and glial cell. At the very stage, motoneurones are arranged into different groups according to their specific locations in the spinal cord (3,4). Those neurones will innervate different target muscle. When innervation process is triggered, motoneurones start growing out trajectories from their growth zones, which are guided to take up the path of travelling to specific muscle fibres. The guidance process is believed to engage both diffusible and in-diffusible molecules, including extracellular matrix (ECM) molecules (e.g. netrins) and adhesion molecules (e.g. cadheirns and immunoglobulins) (5,6), both of which can either attract or repel the growth zone, resulting in the formation of a characteristic physical path by which an axon migrates to its target muscle/cell. There are four physical forces generated in such a process. Namely, chemo-attraction/repulsion, and contact attraction/repulsion (7). It is worth noting that majority of those molecules are bifunctional with few exceptions, meaning many of them can be either attractants or repellents, depending on the specific receptors expressed on growth zone (7,8).

The destination of every individual motoneuron is a muscle fibre, which, according to in vivo and in vitro experiments, shows acetylcholine

Page 4: Neuromuscular Junction Formation

receptor (AchR) clusters prior to the arrival of nerve terminal (9,10). This peculiar phenomenon is referred to as pre-patterning, describing the presence of aneural AchRs and challenging the conventional notion of nerve terminal’s leading role in post-synaptic AchR clustering. For example, previous research on embryonic diaphragm innervation has shown that centralisation of post-synaptic AchRs is a biological event that takes place posterior to innervation (11), indicating the importance of nerve terminal in pre-patterning. However, an in vivo study shows that AchR clusters are present before nerve terminal arrives (9,10) . This notion is further backed-up by the same observations made in mutant mice lacking phrenic or motor nerves (12), suggesting autonomy of post-synaptic muscle fibre in pre-patterning. Further research show that both motoneuron and muscle fibre play important roles in pre-patterning and that whichever predominates depends on specific species (11,13–15) . Nevertheless, there are some issues I would like to address. First, those experiments were done using different muscle types and in different system (i.e. in vivo and in vitro), putting doubts on the credential. Second, in the mutant mice lacking phrenic and motor nerve experiments, a absence of innervation does not necessitate a mutual absence of synaptogenic factors, which could have been produced from the neurones or glial cells and have spatially-restricted impacts on the aggregation of AchRs even before the arrival of nerve terminals (see more in next section).

The formation of NMJs is subsequently followed by synapses maturation, a process that involves changes in both pre-synaptic and post-synaptic structures. Specifically, the maturation of synaptic apparatus is characterised by changes in the quantity, stability and distribution of post-synaptic AchRs (16). The exact underlying molecular mechanisms are well elucidated and will be discussed in the next section. The maturation of pre-synaptic apparatus is largely attributable to increases in nerve ramification, active zone and the distribution and type of calcium channels expressed (to facilitate neurotransmitter release) (16). Moreover, the

Page 5: Neuromuscular Junction Formation

number of nerve terminal that forms NMJs with muscle fibre decreases significantly. This process is necessary and important for increasing the specificity and strengthening the power of inter-neuronal communication. At early stage of innervation, a given nerve terminal contacts multiple muscle fibres (17–19). This kind of NMJ is unspecific and is unable to induce muscle contraction. By the end of synapse maturation, a given motoneuron terminal establishes only one NMJ with a given muscle fibre (note that a muscle fibre can receive innervation from several motoneurones) (20). Unlike post-synaptic maturation, the molecular mechanisms that underline pre-synaptic maturation remains largely elusive. Nevertheless, there has been research showing that laminins, which are muscle-derived heterotrimeric glycoproteins comprising three different subunits, including alpha, beta and gamma subunit (21,22). Laminins are believed to be able to interact with different types of calcium ion channels at different development stages to modulate the release of neurotransmitters (23–25). This notion is further backed-up by research carried out on laminin mutant mice which show reduction in synaptic activity (26–28).

Mature synapses (NMJs) do not stay in a standstill state. Rather, they are subjected to a dynamic environment which requires constant modulation at both transcriptional and molecular level. This scenario is called synaptic plasticity. Although it is true that nerve terminal and muscle fibre are the two responsible elements of synaptic plasticity, extrasynaptic glial cells also play an active role. First of all, there are solid evidence demonstrating the bilateral interactions between Schwann cell (glial cell in PNS) and motoneuron, which produces Nrg4, the motoneuron-derived synaptogenic factor. Nrg4 binds to Erbb4, the receptor expressed on Schwann cell, favouring its proliferation and survival. Secondly, Schwann cells produce diffusible factors, including beta-transforming growth factor (beta-TGF), axotactin and Wnt (Wingless in Drosophila), all of them act in a retrograde manner to facilitate the assembly of NMJs (11,29–32). An interesting novel observation has been made on glial cell in NMJ

Page 6: Neuromuscular Junction Formation

formation. Acknowledging its phagocytic nature, glial cell has been shown to be able to facilitate NMJ formation by engulfing fragmented synapses generated during synapse maturation (33–35).

NMJ formation at molecular level---Agrin/Lrp4/Musk signalling The identification of Agrin/Lrp4/Musk signalling pathway is considered as a prominent breakthrough in NMJ research because it enables neurobiologists, for the first time, to understand and elucidate NMJ assembly at molecular level. Agrins are motoneuron-derived glycoproteins that were originally identified for its ability to induce AchR clustering (36). Agrin’s role in NMJ assembly is further confirmed by the observation of mice carrying mutations in gene-coding agrin have defects in NMJ complicated by a lack of post-synaptic AchRs (37). Furthermore, ectopic expression of agrin is able to induce AchR cluster formation in murine muscle tissues (38). An intriguing observation has also been made in agrin mutant mice which surprisingly is able to assemble aneural AchR prior to nerve terminal arrival, indicating that agrin is neither involved in nor is important for pre-patterning (9,10). The exact mechanism by which agrin promotes AchR clustering has been shown to via counteracting the inhibitory effects of electrical activity and released acetylcholine (Ach). It has been shown that increased electrical activities in muscle fibres accompanied by Ach release causes reduction in Ach subunits expression and increase in AchR degradation, indicating a negative role of Ach in AchR clustering and therefore in NMJ assembly (11,39).

The introduction of Musk (muscle-specific kinase), a muscle-derived receptor tyrosine kinase, was originally made due to its profuseness in synapse-rich organ (40). Agrin is not able to induce post-synaptic AchR clustering in Musk-/- mice (41), indicating a connection between those two molecules that probably suggest an “agonist-receptor relationship”. Further research confirmed this plausible relationship by showing that

Page 7: Neuromuscular Junction Formation

muscle fibre was able to restore agrin responsiveness when exogenous wild-type Musk was expressed (42,43). In addition, ectopically expressed Musk is capable of inducing synapse formation in the absence of agrins (44), suggesting the presence of other agonists or rather, a more sophisticated mechanism of activation. Crystal structure studies revealed agrin does not directly interact with Musk and that there is a mediating co-receptor (45,46), low-density lipoprotein receptor 4 (Lrp4) that serves as a connector. Lrp4 belongs to Lrp receptor family (47) and is required for agrin-induced Musk activation.

The protein structure of Lrp4 characterised by a large extracellular domain, a single transmembrane domain and a small intracellular domain that has no known kinase-associated activity (48–50). Lrp4-/- mice die at birth because of respiratory retardation that resemble those of Musk-/- mice, indicating an important role for Lrp4 in embryonic NMJ assembly (51). Interestingly, it has been shown that the extracellular domain of Lrp4 is able to self-associate and then interact with the extracellular domain of Musk (44,47), causing basal Musk activation that is relatively less potent than what is induced by agrin. This may explain why Musk induces synaptogenesis in the absence of agrin. Given the intrinsic biochemical property of Musk as a receptor tyrosine kinase, it is not surprising to suggest that Musk mediated downstream signalling cascade involves many kinase-induced protein phosphorylation. Indeed, the downstream effector molecules include downstream-of-tyrosine-kinase-7 (Dok7), which is essential for Musk’s interaction with downstream molecules (52), and rapsyn, which is a scaffold molecule that interacts with AchR and is important for its trafficking (53–56). Others are summarised in somewhere else (11).

How does exactly Musk activation leads to AchR clustering? Research has suggested that this process involves rearrangement of cytoskeleton. Specifically, activated Musk interacts with tyrosine kinase Abl (57) and metalloenzyme geranylgeranyl transferase I (GGT I) (58), both of which

Page 8: Neuromuscular Junction Formation

ultimately contribute to Rho GTPase activation (59,60). Rho GTPase has several downstream target molecules, including the serine/threonine kinase Pak1, which stabilises actin by phosphorylating cortactin (61), a substrate of Src kinase enriched at sites where heavy actin assembly occurs (62,63). Moreover, Pak1 counteracts the effect of actin depolymerising factors (ADFs) (64).

Wnt-signalling as a novel player

Recent proceedings in NMJ assembly research has revealed a novel player, the Wnt signalling pathway.

Wnt signalling pathway is probably one of the most investigated signalling pathway in biology owing to its pivotal role in a broad range of developmental biology events, including body axis patterning, limb development and somitogenesis (65–71). Its role in NMJ assembly was originally discovered for its participation in regulation of synaptogenesis of developing rodent cerebellar granular neuron that uses Wnt7a as a retrograde signal to influence the growth of axon (72). Subsequent research using invertebrates (i.e. Drosophila) as animal models has shown that Wingless (the Drosophila orthologue of Wnt) mutation causes a reduction in synapse button number (73). Furthermore, other Wnt isoforms shown positive effects in promoting pre-and post-synaptic apparatus differentiation (74,75). The role that Wnt plays in vertebrate NMJ formation is largely attributable to two aspects. First, a solid body of evidence has been laid down showing that the cysteine-rich domain (CRD) in Musk, the receptor for agrin, shows homology with the Wnt-binding domain of Frizzled’s (76,77). Second, previous research has also shown that beta-catenin, which is the key signal molecule of Wnt signalling cascade, participates in NMJ assembly by regulating actin polymerisation

Page 9: Neuromuscular Junction Formation

(78). This is indicative of the possibility of Wnt as a ligand for Musk. Indeed, the CRD of zebrafish Mush interacts with Wnt11r, contributing to formation of aneural AchR clusters. Wnt presumably binds to Musk expressed in muscle fibres prior to the arrival of nerve terminal, thereby pre-patterning the post-synaptic apparatus. Notably, it would be worth and interesting to investigate whether ligand Wnt family interacts with the low-density lipoprotein receptor family. And, if so, how? Potential approaches include protein co-immunoprecipitation.

Neuromuscular DiseasesMyasthenia Gravis (MG) is a collection of neuromuscular disease characterised by a variety of pathophysiological background, genetic pre-deposition, and responses to treatment. Generally speaking, the clinical manifestation of MG begins with muscle weakness in the face, neck and jaw which then becomes generalised involving skeletal muscles of the whole body, especially that of upper and lower limbs. MG is classified into three groups depending on their respective pathophysiological causes. Namely, the AchR MG, Musk MG and congenital myasthenia syndrome (CMS). The former two groups are autoimmune diseases caused by auto-destruction of neuromuscular junctions (NMJs) mediated by autoantibodies. CMS is a rare inherited condition and is largely attributable to genetic pre-depositions (although postnatal environmental exposure can’t be excluded). Next, I will compare AchR and Musk MG as an example so as to demonstrate how and why should them be evaluated and treated differently. First of all, AchR and Musk MGs have very similar symptoms except that Musk MG rarely affects the ocular muscles (muscles in the eye) (79–81). Secondly, different types of autoantibodies are present. In AchR MG, the antibodies are mostly seen as IgG1 and IgG3 subclasses, whereas in Musk MG, it is IgG4 that is most commonly detected (82,83). Furthermore, it is believed that AchR type of MG has deeper involvement with CD4+ T cells as supported by the observation that thymectomy (removal of the entire thymus) actually is beneficial

Page 10: Neuromuscular Junction Formation

(84). In contrast, Musk type of MG shows more of B cell. An increased frequency of B10 cells (85), which are major cells responsible for immune-tolerance (86–88) has been observed in Musk MG patients. In addition to B-cell and T-cell, those two types of MG differ also in genetic background (84,85).

Owing to their different pathophysiological causes, Musk and AchR MG respond differently to treatments. For example, Musk MG patients have moderate responses (89) to inhibitors of acetylcholinesterase, which is an enzyme responsible for acetylcholine degradation (90). However, Musk MG patients respond well to immunosuppressant corticosteroid, plasma exchange (filtering the autoantibodies out) and pharmaceutical drug rituximab (monoclonal antibody originally used for Non-Hodgkin Lymphoma treatment) (84,91,92). It works by binding to Fc-receptor on immune cell, thereby activating complement system, leading to the death of targeted immune cells. Musk MG patient show less sign of thymic hyperplasia, thymectomy is therefore less effective than it is in AchR type of MG (81).

Page 11: Neuromuscular Junction Formation

ConclusionNeuromuscular junctions (NMJs) are specialised synapses formed between motor neuronal terminals and muscle fibres. They are essential coordinators of the nervous system and skeletal system, conferring high vertebrates the ability to perform complex tasks and great adaptability. Because of their abundancy and large size, NMJs have served as a fabulous model for neurobiologists to explore the molecular mechanisms underlying NMJ assembly. Research in the past decades have been proved fruitful, especially with the identification of agrin/Lrp4/Musk signalling cascade. New excitement has also been brought into this field with the appreciation of Wnt signalling pathway as a novel player in NMJ assembly. However, there are many more issues to be addressed. First of all, research investigating pre-patterning has shown contradictory results (9–12). This is probably because different experimental systems (in vivo and in vitro) and different animal models were used (vertebrates and invertebrates). Secondly, how is Wnt signalling pathway cooperated into Lrp4/Musk signalling? Does it interact with Lrp4 or other members of low-density lipoprotein receptor family? If it does, does it activate the canonical or non-canonical signalling pathway? Owing to its indispensable role in the motility of physical body, NMJ assembly retardation could lead to serious diseases known as neuromuscular diseases (represented by myasthenia gravis). Fortunately, research results have yield a great deal of clinical implications. Targeted molecular therapeutic strategies are now available for neuromuscular treatment with minimised side effects.

Page 12: Neuromuscular Junction Formation
Page 13: Neuromuscular Junction Formation

Reference:

1. Bevan S, Steinbach JH. The distribution of alpha-bungarotoxin binding sites of mammalian skeletal muscle developing in vivo. J Physiol [Internet]. 1977;267(1):195–213. Available from: http://www.ncbi.nlm.nih.gov/pubmed/874836

2. Creazzo TL, Sohal GS. Neural control of embryonic acetylcholine receptor and skeletal muscle. Cell Tissue Res. 1983;228(1):1–12.

3. Landmesser LT. The acquisition of motoneuron subtype identity and motor circuit formation. International Journal of Developmental Neuroscience. 2001. p. 175–82.

4. Jessell TM. Neuronal specification in the spinal cord: inductive signals and transcriptional codes. Nat Rev Genet [Internet]. 2000;1(1):20–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11262869

5. Mitchell KJ, Doyle L. JL, Serafini T, Kennedy TE, Tessier-Lavigne M, Goodman CS, et al. Genetic analysis of Netrin genes in Drosophila: Netrins guide CNS commissural axons and peripheral motor axons. Neuron. 1996;17(2):203–15.

6. Rutishauser U. Adhesion molecules of the nervous system. Curr Opin Neurobiol. 1993;3(5):709–15.

7. Tessier-Lavigne M, Goodman CS. The molecular biology of axon guidance. Science [Internet]. 1996;274(5290):1123–33. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8895455

8. Tessier-Lavigne M. Axon guidance by diffusible repellants and attractants. Current Opinion in Genetics and Development. 1994. p. 596–601.

9. Lin W, Burgess RW, Dominguez B, Pfaff SL, Sanes JR, Lee KF. Distinct roles of nerve and muscle in postsynaptic differentiation of the neuromuscular synapse. Nature. 2001;410(6832):1057–64.

10. Yang X, Arber S, William C, Li L, Tanabe Y, Jessell TM, et al. Patterning of muscle acetylcholine receptor gene expression in the absence of motor innervation. Neuron. 2001;30(2):399–410.

11. Wu H, Xiong WC, Mei L. To build a synapse: signaling pathways in neuromuscular junction assembly. Development [Internet]. 2010;137(7):1017–33. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2835321&tool=pmcentrez&rendertype=abstract

12. Yang X, Li W, Prescott ED, Burden SJ, Wang JC. DNA topoisomerase IIbeta and neural development. Science. 2000;287(5450):131–4.

13. Panzer JA, Gibbs SM, Dosch R, Wagner D, Mullins MC, Granato M, et al. Neuromuscular synaptogenesis in wild-type and mutant zebrafish. Dev Biol. 2005;285(2):340–57.

14. Keshishian H, Broadie K, Chiba A, Bate M. The drosophila neuromuscular junction: a model system for studying synaptic development and function. Annu Rev Neurosci [Internet]. 1996;19:545–75. Available from: http://eutils.ncbi.nlm.nih.gov/entrez/eutils/elink.fcgi?dbfrom=pubmed&id=8833454&retmode=ref&cmd=prlinks\npapers3://publication/doi/10.1146/annurev.ne.19.030196.002553

15. Gautam M, Noakes PG, Mudd J, Nichol M, Chu GC, Sanes JR, et al. Failure of postsynaptic specialization to develop at neuromuscular junctions of rapsyn-deficient mice [Internet]. Nature. 1995. p. 232–6. Available from:

Page 14: Neuromuscular Junction Formation

http://www.ncbi.nlm.nih.gov/pubmed/7675108\nhttp://www.nature.com/nature/journal/v377/n6546/pdf/377232a0.pdf

16. Darabid H, Perez-Gonzalez AP, Robitaille R. Neuromuscular synaptogenesis: coordinating partners with multiple functions. Nat Rev Neurosci [Internet]. 2014 Nov [cited 2016 Jul 29];15(October):1–16. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25493308

17. Tapia JC, Wylie JD, Kasthuri N, Hayworth KJ, Schalek R, Berger DR, et al. Pervasive Synaptic Branch Removal in the Mammalian Neuromuscular System at Birth. Neuron. 2012;74(5):816–29.

18. Wyatt RM, Balice-Gordon RJ. Activity-dependent elimination of neuromuscular synapses. Journal of Neurocytology. 2003. p. 777–94.

19. Chung WS, Barres BA. Selective remodeling: Refining neural connectivity at the neuromuscular junction. PLoS Biology. 2009;7(8).

20. Heckman CJ, Enoka RM. Motor unit. Compr Physiol. 2012;2(4):2629–82. 21. Durbeej M. Laminins. Cell Tissue Res [Internet]. 2010;339(1):259–68. Available from:

http://www.ncbi.nlm.nih.gov/pubmed/19693542\nhttp://link.springer.com/article/10.1007/s00441-009-0838-2

22. Durbeej M. Laminins. Cell and Tissue Research. 2010. p. 259–68. 23. Singhal N, Martin PT. Role of extracellular matrix proteins and their receptors in the

development of the vertebrate neuromuscular junction. Dev Neurobiol. 2011;71(11):982–1005.

24. Urbano FJ, Rosato-Siri MD, Uchitel OD. Calcium channels involved in neurotransmitter release at adult, neonatal and P/Q-type deficient neuromuscular junctions (Review). Mol Membr Biol [Internet]. 2002;19(4):293–300. Available from: http://www.ncbi.nlm.nih.gov/pubmed/12512776

25. Nishimune H, Sanes JR, Carlson SS. A synaptic laminin-calcium channel interaction organizes active zones in motor nerve terminals. Nature [Internet]. 2004;432(7017):580–7. Available from: http://eutils.ncbi.nlm.nih.gov/entrez/eutils/elink.fcgi?dbfrom=pubmed&id=15577901&retmode=ref&cmd=prlinks\npapers3://publication/doi/10.1038/nature03112

26. Patton BL, Cunningham JM, Thyboll J, Kortesmaa J, Westerblad H, Edström L, et al. Properly formed but improperly localized synaptic specializations in the absence of laminin alpha4. Nat Neurosci [Internet]. 2001;4(6):597–604. Available from: http://www.nature.com/doifinder/10.1038/88414\npapers3://publication/doi/10.1038/88414

27. Noakes PG, Gautam M, Mudd J, Sanes JR, Merlie JP. Aberrant differentiation of neuromuscular junctions in mice lacking s-laminin/laminin beta 2. Nature. 1995;374(6519):258–62.

28. Knight D, Tolley LK, Kim DK, Lavidis NA, Noakes PG. Functional analysis of neurotransmission at beta 2-laminin deficient terminals. J Physiol [Internet]. 2003;546(3):789–800. Available from: <Go to ISI>://WOS:000183570000014

29. Gyorkos AM, McCullough MJ, Spitsbergen JM. Glial cell line-derived neurotrophic factor (GDNF) expression and NMJ plasticity in skeletal muscle following endurance exercise. Neuroscience. 2014;257:111–8.

30. Arbour D, Tremblay E, Martineau É, Julien J-P, Robitaille R. Early and Persistent Abnormal Decoding by Glial Cells at the Neuromuscular Junction in an ALS Model. J

Page 15: Neuromuscular Junction Formation

Neurosci [Internet]. 2015;35(2):688–706. Available from: http://www.jneurosci.org/content/35/2/688\nhttp://www.jneurosci.org/content/35/2/688.short\nhttp://www.ncbi.nlm.nih.gov/pubmed/25589763

31. Brink DL, Gilbert M, Xie X, Petley-Ragan L, Auld VJ. Glial processes at the Drosophila larval neuromuscular junction match synaptic growth. PLoS One. 2012;7(5).

32. Kerr KS, Fuentes-Medel Y, Brewer C, Barria R, Ashley J, Abruzzi KC, et al. Glial wingless/Wnt regulates glutamate receptor clustering and synaptic physiology at the Drosophila neuromuscular junction. J Neurosci [Internet]. 2014;34(8):2910–20. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24553932

33. Sugiura Y, Lin W. Neuron-glia interactions: the roles of Schwann cells in neuromuscular synapse formation and function. Biosci Rep [Internet]. 2011;31(5):295–302. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=4573580&tool=pmcentrez&rendertype=abstract

34. Shklyar B, Sellman Y, Shklover J, Mishnaevski K, Levy-Adam F, Kurant E. Developmental regulation of glial cell phagocytic function during Drosophila embryogenesis. Dev Biol. 2014;393(2):255–69.

35. Ko CP, Robitaille R. Perisynaptic schwann cells at the neuromuscular synapse: Adaptable, multitasking glial cells. Cold Spring Harb Perspect Biol. 2015;7(10).

36. McMahan UJ. The agrin hypothesis. In: Cold Spring Harbor Symposia on Quantitative Biology. 1990. p. 407–18.

37. Gautam M, Noakes PG, Moscoso L, Rupp F, Scheller RH, Merlie JP, et al. Defective neuromuscular synaptogenesis in agrin-deficient mutant mice. Cell. 1996;85(4):525–35.

38. Jones G, Meier T, Lichtsteiner M, Witzemann V, Sakmann B, Brenner HR. Induction by agrin of ectopic and functional postsynaptic-like membrane in innervated muscle. Proc Natl Acad Sci U S A [Internet]. 1997;94(6):2654–9. Available from: http://eutils.ncbi.nlm.nih.gov/entrez/eutils/elink.fcgi?dbfrom=pubmed&id=9122251&retmode=ref&cmd=prlinks\npapers2://publication/uuid/B8FD64F7-132C-41C0-87A0-7B63539955E9

39. Salpeter MM, Cooper DL, Levitt-Gilmour T. Degradation rates of acetylcholine receptors can be modified in the postjunctional plasma membrane of the vertebrate neuromuscular junction. J Cell Biol. 1986;103(4):1399–403.

40. Jennings CG, Dyer SM, Burden SJ. Muscle-specific trk-related receptor with a kringle domain defines a distinct class of receptor tyrosine kinases. Proc Natl Acad Sci U S A. 1993;90(7):2895–9.

41. Glass DJ, Bowen DC, Stitt TN, Radziejewski C, Bruno J, Ryan TE, et al. Agrin acts via a MuSK receptor complex. Cell. 1996;85(4):513–23.

42. Herbst R, Burden SJ. The juxtamembrane region of MuSK has a critical role in agrin-mediated signaling. EMBO J [Internet]. 2000;19(1):67–77. Available from: http://emboj.embopress.org/content/19/1/67.abstract

43. Zhou H, Glass DJ, Yancopoulos GD, Sanes JR. Distinct domains of MuSK mediate its abilities to induce and to associate with postsynaptic specializations. J Cell Biol. 1999;146(5):1133–46.

44. Kim N, Burden SJ. MuSK controls where motor axons grow and form synapses. Nat Neurosci [Internet]. 2008;11(1):19–27. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?

Page 16: Neuromuscular Junction Formation

artid=2923649&tool=pmcentrez&rendertype=abstract45. Zong Y, Zhang B, Gu S, Lee K, Zhou J, Yao G, et al. Structural basis of agrin-LRP4-MuSK

signaling. Genes Dev. 2012;26(3):247–58. 46. Zhang W, Coldefy AS, Hubbard SR, Burden SJ. Agrin binds to the N-terminal region of

Lrp4 protein and stimulates association between Lrp4 and the first immunoglobulin-like domain in muscle-specific kinase (MuSK). J Biol Chem. 2011;286(47):40624–30.

47. Zhang B, Luo S, Wang Q, Suzuki T, Xiong WC, Mei L. LRP4 Serves as a Coreceptor of Agrin. Neuron. 2008;60(2):285–97.

48. Johnson EB, Hammer RE, Herz J. Abnormal development of the apical ectodermal ridge and polysyndactyly in Megf7-deficient mice. Hum Mol Genet. 2005;14(22):3523–38.

49. Tian QB, Suzuki T, Yamauchi T, Sakagami H, Yoshimura Y, Miyazawa S, et al. Interaction of LDL receptor-related protein 4 (LRP4) with postsynaptic scaffold proteins via its C-terminal PDZ domain-binding motif, and its regulation by Ca2+/calmodulin-dependent protein kinase II. Eur J Neurosci. 2006;23(11):2864–76.

50. Lu Y, Tian QB, Endo S, Suzuki T. A role for LRP4 in neuronal cell viability is related to apoE-binding. Brain Res. 2007;1177:19–28.

51. Weatherbee SD, Anderson K V, Niswander LA. LDL-receptor-related protein 4 is crucial for formation of the neuromuscular junction. Development [Internet]. 2006;133(24):4993–5000. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17119023

52. Okada K, Inoue A, Okada M, Murata Y, Kakuta S, Jigami T, et al. The muscle protein Dok-7 is essential for neuromuscular synaptogenesis. Science. 2006;312(5781):1802–5.

53. Chen F, Qian L, Yang ZH, Huang Y, Ngo ST, Ruan NJ, et al. Rapsyn Interaction with Calpain Stabilizes AChR Clusters at the Neuromuscular Junction. Neuron. 2007;55(2):247–60.

54. Luo S, Zhang B, Dong X ping, Tao Y, Ting A, Zhou Z, et al. HSP90?? Regulates Rapsyn Turnover and Subsequent AChR Cluster Formation and Maintenance. Neuron. 2008;60(1):97–110.

55. Dobbins GC, Luo S, Yang Z, Xiong WC, Mei L. alpha-Actinin interacts with rapsyn in agrin-stimulated AChR clustering. Mol Brain [Internet]. 2008;1:18. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2621155&tool=pmcentrez&rendertype=abstract

56. Cossins J, Burke G, Maxwell S, Spearman H, Man S, Kuks J, et al. Diverse molecular mechanisms involved in AChR deficiency due to rapsyn mutations. Brain. 2006;129(10):2773–83.

57. Finn AJ, Feng G, Pendergast AM. Postsynaptic requirement for Abl kinases in assembly of the neuromuscular junction. Nat Neurosci. 2003;6(7):717–23.

58. Luo ZG, Je HS, Wang Q, Yang F, Dobbins GC, Yang ZH, et al. Implication of geranylgeranyltransferase I in synapse formation. Neuron. 2003;40(4):703–17.

59. Weston C, Yee B, Hod E, Prives J. Agrin-induced acetylcholine receptor clustering is mediated by the small guanosine triphosphatases Rac and Cdc42. J Cell Biol. 2000;150(1):205–12.

60. Weston C, Gordon C, Teressa G, Hod E, Ren XD, Prives J. Cooperative regulation by Rac and Rho of agrin-induced acetylcholine receptor clustering in muscle cells. J Biol Chem. 2003;278(8):6450–5.

Page 17: Neuromuscular Junction Formation

61. Webb BA, Zhou S, Eves R, Shen L, Jia L, Mak AS. Phosphorylation of cortactin by p21-activated kinase. Arch Biochem Biophys. 2006;456(2):183–93.

62. Weaver AM, Karginov A V., Kinley AW, Weed SA, Li Y, Parsons JT, et al. Cortactin promotes and stabilizes Arp2/3-induced actin filament network formation. Curr Biol. 2001;11(5):370–4.

63. Weaver AM, Heuser JE, Karginov A V., Lee W lih, Parsons JT, Cooper JA. Interaction of cortactin and N-WASp with Arp2/3 complex. Curr Biol. 2002;12(15):1270–8.

64. Delorme-Walker VD, Peterson JR, Chernoff J, Waterman CM, Danuser G, DerMardirossian C, et al. Pak1 regulates focal adhesion strength, myosin IIA distribution, and actin dynamics to optimize cell migration. J Cell Biol. 2011;193(7):1289–303.

65. Engert S, Burtscher I, Liao WP, Dulev S, Schotta G, Lickert H. Wnt/β-catenin signalling regulates Sox17 expression and is essential for organizer and endoderm formation in the mouse. Development [Internet]. 2013;140(15):3128–38. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23824574

66. Hobmayer B, Rentzsch F, Kuhn K, Happel CM, von Laue CC, Snyder P, et al. WNT signalling molecules act in axis formation in the diploblastic metazoan Hydra. Nature. 2000;407(6801):186–9.

67. Jager M, Dayraud C, Mialot A, Qu??innec E, Le Guyader H, Manuel M. Evidence for involvement of Wnt signalling in body polarities, cell proliferation, and the neuro-sensory system in an adult ctenophore. PLoS One. 2013;8(12).

68. Parr BA, Shea MJ, Vassileva G, McMahon AP. Mouse Wnt genes exhibit discrete domains of expression in the early embryonic CNS and limb buds. Development. 1993;119(1):247–61.

69. Church VL, Francis-West P. Wnt signalling during limb development. Int J Dev Biol. 2002;46(7):927–36.

70. Bajard L, Morelli LG, Ares S, Pécréaux J, Jülicher F, Oates AC. Wnt-regulated dynamics of positional information in zebrafish somitogenesis. Development [Internet]. 2014;141(6):1381–91. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3943186&tool=pmcentrez&rendertype=abstract

71. Jensen PB, Pedersen L, Krishna S, Jensen MH. A wnt oscillator model for somitogenesis. Biophys J. 2010;98(6):943–50.

72. Hall AC, Lucas FR, Salinas PC. Axonal remodeling and synaptic differentiation in the cerebellum is regulated by WNT-7a signaling. Cell [Internet]. 2000;100(5):525–35. Available from: http://www.cell.com/article/S0092867400806893/fulltext

73. Packard M, Koo ES, Gorczyca M, Sharpe J, Cumberledge S, Budnik V. The Drosophila Wnt, wingless, provides an essential signal for pre- and postsynaptic differentiation. Cell. 2002;111(3):319–30.

74. Ataman B, Ashley J, Gorczyca M, Ramachandran P, Fouquet W, Sigrist SJ, et al. Rapid Activity-Dependent Modifications in Synaptic Structure and Function Require Bidirectional Wnt Signaling. Neuron. 2008;57(5):705–18.

75. Stamatakou E, Salinas PC. Postsynaptic assembly: A role for Wnt signaling. Dev Neurobiol. 2014;74(8):818–27.

76. Messeant J, Dobbertin a., Girard E, Delers P, Manuel M, Mangione F, et al. MuSK Frizzled-Like Domain Is Critical for Mammalian Neuromuscular Junction Formation and Maintenance. J Neurosci [Internet]. 2015;35(12):4926–41. Available from:

Page 18: Neuromuscular Junction Formation

http://www.jneurosci.org/cgi/doi/10.1523/JNEUROSCI.3381-14.201577. Stiegler AL, Burden SJ, Hubbard SR. Crystal Structure of the Frizzled-Like Cysteine-Rich

Domain of the Receptor Tyrosine Kinase MuSK. J Mol Biol. 2009;393(1):1–9. 78. Li X-M, Dong X-P, Luo S-W, Zhang B, Lee D-H, Ting AKL, et al. Retrograde regulation of

motoneuron differentiation by muscle beta-catenin. Nat Neurosci. 2008;11(3):262–8. 79. Sanders DB, El-Salem K, Massey JM, McConville J, Vincent a. Clinical aspects of MuSK

antibody positive seronegative MG. Neurology. 2003;60(12):1978–80. 80. Evoli A, Tonali PA, Padua L, Lo Monaco M, Scuderi F, Batocchi AP, et al. Clinical

correlates with anti-MuSK antibodies in generalized seronegative myasthenia gravis. Brain. 2003;126(10):2304–11.

81. Deymeer F, Gungor-Tuncer O, Ylmaz V, Parman Y, Serdaroglu P, Ozdemir C, et al. Clinical comparison of anti-MuSK- vs anti-AChR-positive and seronegative myasthenia gravis. Neurology. 2007;68(8):609–11.

82. Koneczny I, Cossins J, Vincent A. The role of muscle-specific tyrosine kinase (MuSK) and mystery of MuSK myasthenia gravis. Journal of Anatomy. 2014. p. 29–35.

83. Tsonis AI, Zisimopoulou P, Lazaridis K, Tzartos J, Matsigkou E, Zouvelou V, et al. MuSK autoantibodies in myasthenia gravis detected by cell based assay - A multinational study. J Neuroimmunol. 2015;284:10–7.

84. Evoli A, Padua L. Diagnosis and therapy of myasthenia gravis with antibodies to muscle-specific kinase. Autoimmunity Reviews. 2013. p. 931–5.

85. Guptill JT, Yi JS, Sanders DB, Guidon AC, Juel VC, Massey JM, et al. Characterization of B cells in muscle-specific kinase antibody myasthenia gravis. Neurol Neuroimmunol neuroinflammation [Internet]. 2015 Apr [cited 2016 Aug 1];2(2):e77. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25745635

86. Candando KM, Lykken JM, Tedder TF. B10 cell regulation of health and disease. Immunol Rev. 2014;259(1):259–72.

87. Yoshizaki A, Miyagaki T, DiLillo DJ, Matsushita T, Horikawa M, Kountikov EI, et al. Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions. Nature [Internet]. 2012;491(7423):264–8. Available from: http://dx.doi.org/10.1038/nature11501

88. Daien CI, Gailhac S, Mura T, Audo R, Combe B, Hahne M, et al. Regulatory B10 cells are decreased in patients with rheumatoid arthritis and are inversely correlated with disease activity. Arthritis Rheumatol. 2014;66(8):2037–46.

89. Hatanaka Y, Hemmi S, Morgan MB, Scheufele ML, Claussen GC, Wolfe GI, et al. Nonresponsiveness to anticholinesterase agents in patients with MuSK-antibody-positive MG. Neurology [Internet]. 2005 Nov 8 [cited 2016 Aug 2];65(9):1508–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16275854

90. Colovic MB, Krstic DZ, Lazarevic-Pasti TD, Bondzic AM, Vasic VM. Acetylcholinesterase Inhibitors: Pharmacology and Toxicology. Curr Neuropharmacol [Internet]. 2013;11(3):315–35. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3648782&tool=pmcentrez&rendertype=abstract\nhttp://www.eurekaselect.com/openurl/content.php?genre=article&issn=1570-159X&volume=11&issue=3&spage=315

91. Hain B, Jordan K, Deschauer M, Zierz S. Successful treatment of MuSK antibody-positive myasthenia gravis with rituximab. Muscle and Nerve. 2006;33(4):575–80.

92. MAYO CLINIC [Internet]. Available from: http://www.mayoclinic.org/drugs-

Page 19: Neuromuscular Junction Formation

supplements/rituximab-intravenous-route/description/drg-20068057