neuromuscular disease modeling on a chip · neuromuscular disease modeling on a chip jeffrey w....

14
REVIEW Neuromuscular disease modeling on a chip Jeffrey W. Santoso 1 and Megan L. McCain 1,2, * ABSTRACT Organs-on-chips are broadly defined as microfabricated surfaces or devices designed to engineer cells into microscale tissues with native- like features and then extract physiologically relevant readouts at scale. Because they are generally compatible with patient-derived cells, these technologies can address many of the human relevance limitations of animal models. As a result, organs-on-chips have emerged as a promising new paradigm for patient-specific disease modeling and drug development. Because neuromuscular diseases span a broad range of rare conditions with diverse etiology and complex pathophysiology, they have been especially challenging to model in animals and thus are well suited for organ-on-chip approaches. In this Review, we first briefly summarize the challenges in neuromuscular disease modeling with animal models. Next, we describe a variety of existing organ-on-chip approaches for neuromuscular tissues, including a survey of cell sources for both muscle and nerve, and two- and three-dimensional neuromuscular tissue-engineering techniques. Although researchers have made tremendous advances in modeling neuromuscular diseases on a chip, the remaining challenges in cell sourcing, cell maturity, tissue assembly and readout capabilities limit their integration into the drug development pipeline today. However, as the field advances, models of healthy and diseased neuromuscular tissues on a chip, coupled with animal models, have vast potential as complementary tools for modeling multiple aspects of neuromuscular diseases and identifying new therapeutic strategies. KEY WORDS: Skeletal muscle, Motor neurons, Amyotrophic lateral sclerosis, Induced pluripotent stem cells, Tissue engineering, Microfluidic devices Introduction Neuromuscular diseases collectively affect 160 per 100,000 people worldwide and are generally characterized by progressive motor impairment and muscular atrophy (Deenen et al., 2015). Although these conditions have diverse etiologies, they each affect one or more components of the motor unit (see Box 1, Fig. 1). For decades, animal models, especially humanized mice (De Giorgio et al., 2019; Nair et al., 2019; Aartsma-Rus and van Putten, 2020), have been the gold standard for neuromuscular disease modeling. More recently, non-mammalian models, such as fruit flies (Lloyd and Taylor, 2010), Caenorhabditis elegans (Sleigh and Sattelle, 2010) and zebrafish (Babin et al., 2014), have also been used for neuromuscular disease modeling. Although these simpler models are limited by their lower conservation with human genetics, anatomy and physiology compared to mice, they are beneficial because of their lower cost, rapid growth rate, tractable anatomy and ease of genetic manipulation. In general, animal models capture important hallmarks of their human disease counterparts and thus are invaluable for understanding disease progression on an organ- and organism-level scale. However, disease phenotypes in animals can vary widely from humans in terms of progression, severity and other characteristics (De Giorgio et al., 2019; Aartsma-Rus and van Putten, 2020; Babin et al., 2014). Another limitation of animal models is that it is difficult, if not impossible, to recapitulate the genotypic heterogeneity and allelic variation observed in individuals with neuromuscular diseases without generating an unreasonable number of animal strains (Juneja et al., 2019; Morrice et al., 2018). Even monogenic neuromuscular diseases, such as spinal muscular atrophy (SMA), are difficult to model in animals due to patient-specific genotypic features. SMA is an autosomal recessive disease caused by inactivating mutations in the SMN1 gene, which encodes the survival of motor neuron (SMN) protein (Li, 2017). SMN plays a role in protein homeostasis, cytoskeletal assembly, endocytosis, metabolism and many other processes in motor neurons (Chaytow et al., 2018). SMN shortage or dysfunction causes deficits in axonogenesis, migration, electrophysiology and many other features, leading to neuromuscular junction (NMJ) degeneration and motor neuron death (Laird et al., 2016; McGovern et al., 2015). A second gene, SMN2, also produces SMN, but at 20% of the levels transcribed from fully functional SMN1 (Bowerman et al., 2017; Jedrzejowska et al., 2009). SMA has been modeled in mice (Hsieh-Li et al., 2000), Drosophila (Spring et al., 2019), zebrafish (McWhorter et al., 2003) and C. elegans (Briese et al., 2009) by deleting the endogenous Smn gene and overexpressing the human SMN2 gene. However, the severity and progression of SMA largely depends on the number of SMN2 copies in a patient (Bowerman et al., 2017; Jedrzejowska et al., 2009), a patient-specific feature of the disease that is nearly impossible to faithfully recapitulate in animals. The only treatment options for SMA are the gene therapy drugs Spinraza (Dangouloff and Servais, 2019) and Zolgensma (Zuroske, 2019), both of which are extremely expensive and thus impractical for many individuals. Compared to SMA, several neuromuscular diseases have a more heterogeneous genetic etiology, which is even more challenging to model in animals. For example, Charcot-Marie-Tooth (CMT) diseases have been linked to 870 mutations in over 80 genes (McCorquodale et al., 2016), such as PMP22, MPZ, GJB1 or MFN2 (Morena et al., 2019; Saporta et al., 2011). This genetic heterogeneity partially explains the wide range of age of onset and disease symptoms, which usually involve involuntary contraction of limbs and loss of sensation due to axon demyelination. CMT has been modeled in zebrafish and other animal models by introducing a mutation in a single gene known to cause a specific subtype of CMT disease, such as mfn2 (Chapman et al., 2013) or prps1 (Pei et al., 2016). However, owing to the vast genetic heterogeneity of CMT 1 Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA. 2 Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA. *Author for correspondence ([email protected]) M.L.M., 0000-0003-1908-6783 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed. 1 © 2020. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867 Disease Models & Mechanisms

Upload: others

Post on 19-Oct-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

REVIEW

Neuromuscular disease modeling on a chipJeffrey W. Santoso1 and Megan L. McCain1,2,*

ABSTRACTOrgans-on-chips are broadly defined as microfabricated surfaces ordevices designed to engineer cells into microscale tissues with native-like features and then extract physiologically relevant readouts at scale.Because they are generally compatible with patient-derived cells, thesetechnologies can address many of the human relevance limitations ofanimal models. As a result, organs-on-chips have emerged as apromising new paradigm for patient-specific diseasemodeling and drugdevelopment. Because neuromuscular diseases span a broad range ofrare conditions with diverse etiology and complex pathophysiology, theyhave been especially challenging to model in animals and thus are wellsuited for organ-on-chip approaches. In this Review, we first brieflysummarize the challenges in neuromuscular disease modeling withanimal models. Next, we describe a variety of existing organ-on-chipapproaches for neuromuscular tissues, including a survey of cellsources for both muscle and nerve, and two- and three-dimensionalneuromuscular tissue-engineering techniques. Although researchershavemade tremendous advances inmodeling neuromuscular diseaseson a chip, the remaining challenges in cell sourcing, cell maturity, tissueassembly and readout capabilities limit their integration into the drugdevelopment pipeline today. However, as the field advances, models ofhealthy and diseased neuromuscular tissues on a chip, coupled withanimal models, have vast potential as complementary tools formodeling multiple aspects of neuromuscular diseases and identifyingnew therapeutic strategies.

KEY WORDS: Skeletal muscle, Motor neurons, Amyotrophic lateralsclerosis, Induced pluripotent stem cells, Tissue engineering,Microfluidic devices

IntroductionNeuromuscular diseases collectively affect 160 per 100,000 peopleworldwide and are generally characterized by progressive motorimpairment and muscular atrophy (Deenen et al., 2015). Althoughthese conditions have diverse etiologies, they each affect one ormore components of the motor unit (see Box 1, Fig. 1). For decades,animal models, especially humanized mice (De Giorgio et al., 2019;Nair et al., 2019; Aartsma-Rus and van Putten, 2020), have been thegold standard for neuromuscular disease modeling. More recently,non-mammalian models, such as fruit flies (Lloyd and Taylor,2010), Caenorhabditis elegans (Sleigh and Sattelle, 2010) andzebrafish (Babin et al., 2014), have also been used for

neuromuscular disease modeling. Although these simpler modelsare limited by their lower conservation with human genetics,anatomy and physiology compared to mice, they are beneficialbecause of their lower cost, rapid growth rate, tractable anatomy andease of genetic manipulation. In general, animal models captureimportant hallmarks of their human disease counterparts and thusare invaluable for understanding disease progression on an organ-and organism-level scale. However, disease phenotypes in animalscan vary widely from humans in terms of progression, severity andother characteristics (De Giorgio et al., 2019; Aartsma-Rus and vanPutten, 2020; Babin et al., 2014).

Another limitation of animal models is that it is difficult, if notimpossible, to recapitulate the genotypic heterogeneity and allelicvariation observed in individuals with neuromuscular diseases withoutgenerating an unreasonable number of animal strains (Juneja et al.,2019; Morrice et al., 2018). Even monogenic neuromuscular diseases,such as spinal muscular atrophy (SMA), are difficult to model inanimals due to patient-specific genotypic features. SMA is anautosomal recessive disease caused by inactivating mutations in theSMN1 gene, which encodes the survival of motor neuron (SMN)protein (Li, 2017). SMN plays a role in protein homeostasis,cytoskeletal assembly, endocytosis, metabolism and many otherprocesses in motor neurons (Chaytow et al., 2018). SMN shortageor dysfunction causes deficits in axonogenesis, migration,electrophysiology and many other features, leading to neuromuscularjunction (NMJ) degeneration and motor neuron death (Laird et al.,2016; McGovern et al., 2015). A second gene, SMN2, also producesSMN, but at ∼20% of the levels transcribed from fully functionalSMN1 (Bowerman et al., 2017; Jedrzejowska et al., 2009). SMA hasbeenmodeled inmice (Hsieh-Li et al., 2000),Drosophila (Spring et al.,2019), zebrafish (McWhorter et al., 2003) andC. elegans (Briese et al.,2009) by deleting the endogenous Smn gene and overexpressing thehuman SMN2 gene. However, the severity and progression of SMAlargely depends on the number of SMN2 copies in a patient(Bowerman et al., 2017; Jedrzejowska et al., 2009), a patient-specificfeature of the disease that is nearly impossible to faithfully recapitulatein animals. The only treatment options for SMA are the gene therapydrugs Spinraza (Dangouloff and Servais, 2019) and Zolgensma(Zuroske, 2019), both of which are extremely expensive and thusimpractical for many individuals.

Compared to SMA, several neuromuscular diseases have a moreheterogeneous genetic etiology, which is even more challenging tomodel in animals. For example, Charcot-Marie-Tooth (CMT)diseases have been linked to 870 mutations in over 80 genes(McCorquodale et al., 2016), such as PMP22,MPZ, GJB1 orMFN2(Morena et al., 2019; Saporta et al., 2011). This genetic heterogeneitypartially explains the wide range of age of onset and diseasesymptoms, which usually involve involuntary contraction of limbsand loss of sensation due to axon demyelination. CMT has beenmodeled in zebrafish and other animal models by introducing amutation in a single gene known to cause a specific subtype of CMTdisease, such as mfn2 (Chapman et al., 2013) or prps1 (Pei et al.,2016). However, owing to the vast genetic heterogeneity of CMT

1Laboratory for Living Systems Engineering, Department of BiomedicalEngineering, USC Viterbi School of Engineering, University of Southern California,Los Angeles, CA 90089, USA. 2Department of Stem Cell Biology and RegenerativeMedicine, Keck School of Medicine of USC, University of Southern California, LosAngeles, CA 90033, USA.

*Author for correspondence ([email protected])

M.L.M., 0000-0003-1908-6783

This is an Open Access article distributed under the terms of the Creative Commons AttributionLicense (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use,distribution and reproduction in any medium provided that the original work is properly attributed.

1

© 2020. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 2: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

diseases, it is infeasible to generate animal models that represent allmutations (Juneja et al., 2019). Largely due to a lack of modularmodel systems, CMT diseases still lack clinical data supporting anyeffective treatment beyond physical therapy and pain management(McCorquodale et al., 2016). Mouse models of CMT have alsodemonstrated that impaired development of the NMJ precedessynaptic deficits (Sleigh et al., 2013; Spaulding et al., 2016),suggesting that microscale models of the motor unit might be usefulfor elucidating the pathophysiology of this broad group of diseases.Amyotrophic lateral sclerosis (ALS) is another neuromuscular

disease that introduces unique challenges for modeling in animals

because it can be either inherited (10%) or sporadic (90%) (Boylan,2015). In ALS, over 50 genes either directly cause motor neurondeath or alter key functions, such as vesicle trafficking, axonalstructure and cytoskeletal stability (Boylan, 2015; Seminary et al.,2018; Shi et al., 2018). The most commonly affected genes areC9ORF72, SOD1, TARDBP and FUS, usually occurring in somekind of combination (Lattante et al., 2015; Nguyen et al., 2018).Animal models of ALS have been generated by expressing amutated version of one of these human genes in mice (Ripps et al.,1995; Zhang et al., 1997; Devoy et al., 2017), Drosophila (Sahinet al., 2017; Watson et al., 2008; Perry et al., 2017; Xu et al., 2013),zebrafish (Shaw et al., 2018; Lissouba et al., 2018) and C. elegans(Oeda et al., 2001; Wang et al., 2009). Additionally, environmentalfactors, such as pesticides, flame retardants and military-relatedtrauma, have been correlated to ALS (Su et al., 2016). However, thesmall number of clinical cases and limited model systems makeassigning causality from environmental factors very difficult. Thenatural process of aging has also been tied to ALS, probably becauseof the aggregation of misfolded proteins and oxidative stress (Jangand Van Remmen, 2011; Turner et al., 2012). Owing, in large part,to the many causes and complex pathophysiologies of ALS, theonly therapies are the anti-glutamatergic compound riluzole and theantioxidant edaravone, both of which only assuage symptoms andextend survival for a few months (Nguyen et al., 2018).

Neuromuscular diseases can also be caused by factors external tothe motor unit. For example, myasthenia gravis (MG) is a sporadicautoimmune disease in which auto-antibodies selectively destroyacetylcholine receptors, causing a reduction in NMJ signaltransmission (Phillips and Vincent, 2016). A mouse model of MGhas been developed by injecting rat acetylcholine receptors intomice, which then triggered the development of auto-antibodies totheir own acetylcholine receptors (Granato et al., 1976). However,MG cannot be modeled in simple organisms such asDrosophila andC. elegans because they lack an adaptive immune system. Thecauses of MG are still mostly unknown and treatment is limited toacetylcholinesterase inhibitors, immunosuppressants or athymectomy (Gilhus, 2016), highlighting the need for additionalpredictive model systems to develop more targeted therapies.

Collectively, these examples highlight that neuromuscular diseasesare very diverse and are characterized by many complex genetic andnon-genetic etiologies and pathophysiologies. These complexitiesintroduce many challenges for developing comprehensive animalmodels. Thus, new disease models that are more efficient and predictiveare essential for accelerating our mechanistic knowledge of thesediseases, as well as the discovery of effective therapies. Integratingpatient-derived cells with microfabricated in vitro platforms, known asorgans-on-chips, is an emerging solution to fill the gaps of animalmodels and holds promise for patient-specific neuromuscular diseasemodeling and drug development. As discussed below, these platformsare often developed using animal cells or cell lines that are easy to scale,and that can provide important proof-of-concept and basic physiologicalinformation. To address issues of human relevance, animal cells or celllines can then be replaced with patient-derived cells, which can beacquired from a variety of sources. In the next section, we describe thecell sources for neuromuscular disease models, which can ultimately beintegrated into the two- (2D) and three-dimensional (3D) engineeredtissue platforms described in the following sections.

Cell sources for in vitro models of neuromuscular tissuesIn vitro models can mitigate many of the limitations of animalmodels described above, such as human relevance and scalability.However, the usefulness of any in vitro model is highly dependent

Box 1. Structure and physiology of the motor unitAll voluntary movements are controlled by a collection of motor units,each of which comprises a single motor neuron and all the muscle fibersthat it innervates (Fig. 1). Motor neurons have a soma that resides in themotor cortex, brain stem or spinal cord, and a single myelinated axon thatforms specialized synapses, known as neuromuscular junctions (NMJs),on muscle fibers. Muscle fibers are elongated multi-nucleated cells thatare packed with myofibrils, each of which is an interconnected chain ofcontractile sarcomere units. Multiple muscle fibers are bundled togetherand wrapped in connective tissue to form a muscle.Contraction of a motor unit begins when signals from the central nervoussystem trigger an action potential in the motor neuron, which induces theaxon to release the neurotransmitter acetylcholine into the synaptic cleftof the NMJ. Acetylcholine binds to acetylcholine receptors on themembrane of the muscle fiber, which depolarizes the membrane andinitiates an action potential. The muscle fiber then propagates this actionpotential along its length, triggering the entry of extracellular calciumthrough voltage-sensitive ion channels in the membrane andsubsequently a large release of calcium from the sarcoplasmicreticulum. This increase in cytosolic calcium enables the heads ofmyosin filaments to pull on actin filaments, shortening the sarcomere andultimately contracting the muscle fiber in an ATP-demanding process.Depending on the frequency of the action potential transmitted by themotor neuron, the muscle fiber undergoes either a singular or sustainedcontraction, referred to as twitch or tetanus, respectively. Lastly, the freeacetylcholine in the NMJ is broken down by acetylcholinesterase,cytosolic calcium is transported back into the sarcoplasmic reticulum,and the membrane potential of the muscle fiber returns to resting levels,thus causing muscle relaxation (reviewed by Hall and Hall, 2015).

Collagen

Laminin

Acetylcholine vesicle

Acetylcholine receptor

Muscle fiber

Key

Motor neuron

Neuromuscular junction

Fig. 1. Schematic of the neuromuscular junction. Multi-nucleated musclefibers are innervated by myelinated motor neurons at neuromuscular junctions(NMJs). At the NMJ, motor neurons release acetylcholine vesicles. Theneurotransmitter acetylcholine binds to acetylcholine receptors on the membraneof the muscle fiber, causing membrane depolarization and muscle contraction.

2

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 3: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

on the source and structural and functional maturity of its cells. Thisis especially complex when modeling neuromuscular tissues, whichconsist of both muscle cells and motor neurons. In this section, wewill describe the types of muscle and motor neuron cell typesavailable today for in vitro models, and weigh up their advantagesand disadvantages.

Skeletal muscle cellsGenerating skeletal muscle tissue in vitro is amulti-step process. First,mononuclear skeletal myoblasts are seeded on a standard culturesurface that is often coated with extracellular matrix (ECM) proteins,such as collagen or laminin. The myoblasts are then expanded in ahigh-serum growthmedium until they reach confluence. Themediumis then substituted with a low-serum differentiation medium thattriggers the fusion of myoblasts into multi-nucleated myotubes, the invitro surrogate to muscle fibers (Neville et al., 1997). As illustrated inFig. 2, several different sources of myoblasts are currently available,each with distinct advantages and disadvantages that are important toconsider when engineering neuromuscular disease models.Primary myoblasts are harvested from embryonic or adult animals,

such as chicks (Urja et al., 2018; Vallette et al., 1986) or mice (Hindiet al., 2017), by excising muscle tissue and either enzymaticallydigesting it to a cell suspension or collecting the cells that migrate fromcultured tissue explants (Vaughan and Lamia, 2019). Myoblasts are

then purified using simple pre-plating steps or more sophisticatedtechniques, such as magnetic cell sorting (Sincennes et al., 2017;Spinazzola and Gussoni, 2017). Human primary myoblasts can beisolated from muscle tissue collected during a surgical procedure or byneedle biopsy (Joyce et al., 2012), and similarly processed and purified.

The structural and functional properties of myotubes differentiatedfrom primary myoblasts closely recapitulate those of native muscle,such as a high density of myofibrils and spontaneous contractilebehavior (Pimentel et al., 2017). However, primary myoblasts canonly be passaged a few times before their growth rate and myogeniccapacity decline, leading to limited supply and passage-dependentvariability. These supply and variability issues are especiallyproblematic for human primary myoblasts, which are generallyisolated from relatively small muscle biopsies. Moreover, primarymyoblasts can vary widely in purity and functional maturitydepending on the isolation and purification methods, and thecharacteristics of the subject (Cheng et al., 2014; Soriano-Arroquiaet al., 2017). This issue is further exacerbated by data indicating abeneficial role for fibroblasts in myotube function (Rao et al., 2013),raising questions about the ideal purity of primary myoblasts for invitro studies and adding further variability to the performance ofmyotubes differentiated from primary myoblasts.

Compared to primary myoblasts, immortalized myoblast cell linesare a more convenient source of cells that are relatively pure and easy

Immortalized cell lines

Pluripotent stem cell derivatives

Immortalized myoblasts

Skeletal muscle Motor neurons

Mousemotor neuron

Mouseneuroblastoma

NSC-34

Reprogrammingfactors

hiPSCs

hESCs

Spontaneousmutation or

viral transduction

Hybridization

Primary cells

Human(muscle only)

Mouse Chick

Fig. 2. Muscle and motor neuron cell sources for in vitro models. Myoblasts and motor neurons available for in vitro models fall into three categories:immortalized cell lines, primary cells and pluripotent stem cell derivatives. Immortalized cell lines recapitulate the basic properties of the original cell type and areinexpensive and easy to expand in culture. However, the immortalization process causes de-differentiation and a loss of important structural and functionalfeatures. Primary cells are usually the most mature and physiologically relevant cell source but are relatively costly and difficult to obtain, and have a limited abilityto expand in culture. Pluripotent stem cells can be widely expanded in culture and then differentiated into myoblasts or motor neurons. Induced pluripotent stemcells have the additional advantage of patient specificity. However, pluripotent stem cell derivatives are generally heterogeneous and immature. hESC, humanembryonic stem cell; hiPSC, human induced pluripotent stem cell.

3

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 4: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

to expand. Themost commonmyoblast cell line is C2C12, which wasisolated from 2-month-old mouse muscle in 1977 (Yaffe and Saxel,1977). Another common cell line is L6, which was isolated fromnewborn rat muscle in 1968 (Yaffe, 1968). Both C2C12 and L6 cellsproliferate rapidly with a generation time of ∼24 h and fuse intomulti-nucleated contractile myotubes (McMahon et al., 1994; Öberget al., 2011). Thus, they are a convenient model system forinvestigating processes such as myotube fusion (Zhao et al., 2015)or degeneration (Menconi et al., 2008). These cells are alsocompatible with gene transfection (Balcı and Dinçer, 2009), whichis useful for establishing the functions of genetic variants in myoblastgrowth or fusion (Prinsen and Veerkamp, 1998), or introducingdisease-relevant mutations (Liang et al., 2016).Myotubes differentiated from cell lines have lower levels of

structural and functional maturity, and distinct metabolic propertiescompared to myotubes differentiated from primary myoblasts(Abdelmoez et al., 2019; Robinson et al., 2019), probablybecause the immortalization process causes some amount of de-differentiation and loss of myogenic properties that continue todecline with increasing passage number. Thus, although myoblastcell lines are a reproducible and cost-effective cell source comparedto primary myoblasts, myotubes differentiated from cell lines havelimited relevance to native muscle tissue. Furthermore, acommercialized human myoblast cell line does not currently exist,raising further concerns about the translatability of data collectedusing common myoblast cell lines, which are derived from rodents.However, primary human myoblasts have been immortalized by theforced expression of a telomerase subunit and cyclin dependentkinase 4, which blocks a stress pathway (Mamchaoui et al., 2011).Myotubes generated from these cell lines produce relatively maturesarcomeres (Morris et al., 2020) and form NMJs when co-culturedwith motor neurons (Saini et al., 2019), suggesting that they couldbecome a promising cell source.To overcome the limitations of primary myoblasts and myoblast

cell lines, several protocols for deriving myogenic cells from humanembryonic stem cells (hESCs) or human induced pluripotent stemcells (hiPSCs) have recently emerged (Salani et al., 2012). Thesecells have the advantages of human origin and essentially limitlesssupply, as hESCs and hiPSCs can be expanded in culture for manypassages without loss of functionality. hESCs and hiPSCs are alsocompatible with gene editing techniques, such as CRISPR/Cas9,which can be used to introduce or correct select disease-relevantmutations (Shi et al., 2018; Young et al., 2016). Because hiPSCs arereprogrammed from somatic cells, such as skin fibroblasts, hiPSC-derived myogenic cells can be used to generate patient-specificmyotubes, which makes them especially desirable for modelinginherited neuromuscular diseases.One approach for generating myogenic progenitors from hiPSCs is

to overexpress master regulators of myogenic differentiation, such asMYOD1 (Abujarour et al., 2014; Rao et al., 2018) or PAX7 (Darabiet al., 2012). A similar reprogramming process has also been used todirectly transdifferentiate other cell types, such as fibroblasts, intomyogenic progenitors (Boularaoui et al., 2018; Ito et al., 2017; Lattanziet al., 1998). A second approach, known as directed differentiation,guides hiPSCs through native-like myogenic developmental pathwaysby sequentially adding small molecules that activate or suppressspecific signaling pathways (Chal et al., 2016, 2015; Maffioletti et al.,2015; Shelton et al., 2016; van der Wal et al., 2018; Xi et al., 2017).Directed differentiation is generally slower than transdifferentiation,but the resultingmyogenic progenitors are thought to be a closer matchto native myoblasts because they follow a more natural differentiationprocess (Jiwlawat et al., 2018).

Although impressive progress has been made in derivingmyogenicprogenitors from hiPSCs, most current protocols generally suffer fromwide variability and low efficiency (Jiwlawat et al., 2018). Theseissues limit cell yield and purity. However, protocols forcryopreserving and expanding hiPSC-derived myogenic progenitorsare being developed (van der Wal et al., 2018), which helps mitigateissues with differentiation variability and throughput. Despite thesepractical limitations related to cell differentiation, hESC- and hiPSC-derived myogenic progenitors successfully fuse into myotubes thatcontain myofibrils and exhibit key functional behaviors, such ascalcium cycling and contractility (Rao et al., 2018; Skoglund et al.,2014). However, myofibrils in hESC- and hiPSC-derived myotubesstill have immature features compared to native muscle fibers ormyotubes derived from primary myoblasts (Lainé et al., 2018).

The structural and functional immaturity of myotubes probablycontributes to the stunted maturation of NMJs that form betweenhiPSC-derived myotubes and motor neurons. However, severalapproaches for maturing hiPSC-derived myotubes are underdevelopment, such as identifying small molecules that boostmaturation (Selvaraj et al., 2019) or applying other strategiesdiscussed below. Additionally, the maturation of C2C12 myotubeshas been improved by applying biophysical cues, such as mechanicalstretch (Chang et al., 2016; Heher et al., 2015) or electricalstimulation (Ito et al., 2014; Nedachi et al., 2008), which mighthave similar benefits for hiPSC-derived myotubes. Thus, althoughhiPSC-derived myotubes have significant potential as an essentiallylimitless source of patient-specific myotubes, researchers need toenhance the differentiation efficiency and maturity of these cells toimprove their throughput and relevance for modeling neuromusculardiseases in vitro.

Motor neuronsCompared to myoblasts, fewer sources of motor neurons exist forin vitro models. Because most motor neurons stem from the spinalcord and project onto muscle fibers, it is not possible to isolate intactprimary motor neurons from humans. However, primary motorneurons can be isolated from embryonic or adult mice by extractingand digesting spinal cord tissue and using density gradientseparation to isolate motor neurons from supporting cell types,such as astrocytes and other glial cells (Beaudet et al., 2015; Gingraset al., 2007). Although rodents are a viable source of primary motorneurons, these cells are not human, which limits their relevance forneuromuscular disease modeling. Furthermore, the cell yield isrelatively low and cannot be increased with passaging becausemotor neurons are terminally differentiated and non-proliferative.

Because motor neurons do not proliferate, true motor neuron celllines do not exist. However, a hybrid mouse cell line (NSC-34) hasbeen generated by fusing neuroblastoma cells with embryonic motorneurons. NSC-34 cells retain the proliferative properties of the tumorcells while also exhibiting select neuronal properties, such asacetylcholine synthesis, neurotransmitter release and neurofilamentproteins (Cashman et al., 1992). This cell line has been used tomeasure the neurotoxicity of drugs (Maier et al., 2013) and receptortrafficking (Matusica et al., 2008), and has also been transfected tointroduce mutations relevant to ALS (Gomes et al., 2010; Pinto et al.,2017). Similar tomyoblast cell lines, the disadvantage of these cells istheir non-human origin and limited relevance to native motorneurons. For example, these cells do not replicate glutamate-mediatedexcitotoxicity (Madji Hounoum et al., 2016), questioning their abilityto replicate key features of neuromuscular diseases.

Human motor neurons can also be derived from hESCs and hiPSCsvia reprogramming or directed differentiation. hESCs and hiPSCs have

4

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 5: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

been reprogrammed intomotor neurons byoverexpressingNGN2 (alsoknown asNEUROG2), ISL1 and LHX3 (Goto et al., 2017; Hester et al.,2011; Lee et al., 2012). Human fibroblasts have also beentransdifferentiated into motor neurons by overexpressing eight genes(Son et al., 2011). Several directed differentiation methods have alsobeen established, which entail dosing hESCs or hiPSCs with acombination of neurotrophic factors, retinoic acid, sonic hedgehog andNotch inhibitors (Du et al., 2015; Hu and Zhang, 2009; Li et al.,2008b; Qu et al., 2014; Shimojo et al., 2015). Motor neurons have alsobeen differentiated from the human fetal spinal cord stem cell line NSI-566RSC (Guo et al., 2010), which serves as another relativelyaccessible source of human motor neurons.Similar to other stem cell derivatives, stem cell-derived motor

neurons can be limited by cell heterogeneity, varying differentiationefficiency and stunted maturation (Ichida et al., 2018). Althoughmouse motor neurons derived from transdifferentiated fibroblasts ordirectly differentiated iPSCs have a transcriptome that is similar toprimary motor neurons (Ichida et al., 2018), how the structural andfunctional properties of these cells compare to their primarycounterpart is mostly unknown. Despite the limited functionalcharacterization of these cells, hiPSC-derived motor neurons havealready been shown to be a promising cell source for patient-specificmodeling of neuromuscular diseases such as ALS (Dimos et al., 2008;Sances et al., 2016; Sareen et al., 2013; Shi et al., 2018) and SMA(Fuller et al., 2016;Murdocca et al., 2016). Thus, hiPSC-derivedmotorneurons are likely to contribute to the development of new therapies forthese diseases that account for the genotype of the patient.

Engineered in vitro models of neuromuscular tissuesIn addition to cell source, another important consideration for in vitromodel development is the configuration of the cells, such that thecultured tissue is anatomically relevant and integrated with assays tomeasure functional phenotypes. Initial approaches for engineeringneuromuscular tissues in vitro entailed simply seeding dissociatedmotor neurons (Daniels et al., 2000; Das et al., 2010; Guo et al., 2011;Kengaku et al., 1991; Son et al., 2011; Umbach et al., 2012) or spinalcord explants (Askanas et al., 1987; Braun et al., 1997) on top of a 2Dlayer of myotubes attached to a conventional culture surface. Over thecourse of several days, the neurons extend axons and formNMJswiththe myotubes that successfully exhibit functional post-synapticpotentials. However, image analysis has revealed blotchycolocalization of pre- and post-synaptic markers and pooracetylcholine receptor clustering in these simple co-culturescompared to native NMJs (Das et al., 2010; Umbach et al., 2012).This limited synaptic maturity brings into question the ability of theseculture systems to accurately model disease-relevant phenotypes.The relatively stunted NMJ development in conventional co-

cultures could be attributed to many factors. First, without spatialorganization cues, myoblasts fuse into branched myotubes withrandom orientations that poorly recapitulate the architecture ofnative muscle fibers (Bettadapur et al., 2016; Denes et al., 2019),which can limit the formation of elongated myofibrils and maturesarcomeres. Second, myotubes often delaminate from conventionalculture surfaces within ∼2 weeks as they generate increasingamounts of mechanical stress (Wang et al., 2012; Sun et al., 2013).This can probably be attributed to both the high stiffness ofconventional culture substrates and the limited number of cell-adhesive molecules presented on their surfaces (Bettadapur et al.,2016). Limited culture lifetime is especially problematic forengineering neuromuscular tissues because NMJ maturationprobably requires longer than 2 weeks. Third, in situ, motorneuron soma are located in the spinal cord and only the axons of

motor neurons physically interact with muscle fibers. Thus, seedingmotor neurons on top of myotubes is not anatomically relevant andmight alter the physiology of one or both cell types.

Simple mixed co-cultures also suffer from technical problemsthat limit data collection. For example, measuring forces generatedby cells is not possible on most, if not all, conventional culturesubstrates, precluding quantitative assessment of muscle forceproduction due to motor neuron stimulation. This is a key functionalreadout in animal models, with high relevance to the severity ofneuromuscular disease (Bonetto et al., 2015). A second limitation isthat mixed co-cultures afford minimal independent control oranalysis of each cell type, as the cells are cultured in the samemedium and any electrical stimulation or drug treatment reachesboth cell types simultaneously. Similarly, isolating material fromeach cell type independently to measure changes in gene or proteinexpression, which is often important for establishing diseasemechanisms as well as drug effects, is challenging.

To overcome these diverse biological and technical challenges,researchers have developed several types of tunable culture surfacesand microfabricated devices to engineer more sophisticatedneuromuscular tissues in vitro. These surfaces and devices areusually also integrated with assays for quantifying structural andfunctional tissue phenotypes. In particular, when coupled with thehiPSC-derived cell sources described above, these platforms, knownas organs-on-chips or microphysiological systems, have immensepotential for advancing neuromuscular disease modeling and drugdevelopment. In this section, we describe both 2D and 3D engineeredtissue models.

Engineered 2D models of neuromuscular tissuesEngineering a neuromuscular tissue in vitro depends on first culturingmature and stable myotubes. In native skeletal muscle fibers, theECM plays a key role in tissue development and physiology bybinding to integrin receptors and providing biomechanical support asthe muscle fibers contract (Gillies and Lieber, 2011). The ECM isalso a rich source of biochemical cues that regulate behaviors such asadhesion, proliferation and differentiation. Furthermore, the ECMplays an active role in skeletal muscle disease, injury and aging, withfibrosis and subsequent tissue stiffening contributing to diminishedmuscle function (Mann et al., 2011). Owing to the documentedimportance of the ECM in native muscle fibers, several types oftunable culture substrates that mimic aspects of native muscle ECMhave been developed, as described below. However, anotherimportant feature of in vitro models of neuromuscular tissues is theability to measure muscle contractility in response to motor neuronstimulation. Thus, we also describe later in this section howengineered substrates have been integrated with contractility assays.

Natural biomaterial substratesHydrogels synthesized from natural polymers are popular culturesubstrates due to their biocompatibility, although they can suffer frombatch-to-batch variability. Collagen hydrogels are routinely used asculture substrates for myoblasts and myotubes due to their intrinsicbioactivity (Palade et al., 2019), as we also discuss above. Alignedmyotubes have been fabricated on collagen hydrogels by embeddingtopographical features into the hydrogel (Kim et al., 2017) or usingultrasound to pattern the cells acoustically (Armstrong et al., 2018).Gelatin, a partially hydrolyzed form of collagen, is also crosslinkedinto thermostable hydrogels by either mixing gelatin polymers withenzymatic crosslinking agents (Bettadapur et al., 2016; Denes et al.,2019; Suh et al., 2017) or methacrylating gelatin polymers such thatthey are compatible with photopolymerization techniques (Hosseini

5

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 6: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

et al., 2012; Sun et al., 2018). Crosslinking increases the stiffness ofthe hydrogel and reduces its degradability (Sun et al., 2018), whichcan be advantageous for in vitro neuromuscular models that need tobe stable for several weeks.To promote myotube alignment on gelatin hydrogels, the surface

can be micromolded with polydimethylsiloxane (PDMS) (a siliconeelastomer) stamps with ridges several micrometers in size(Bettadapur et al., 2016; Chal et al., 2016; Denes et al., 2019;Hosseini et al., 2012). PDMS stamps are fabricated by casting PDMSon silicon wafer templates made using photolithography, which cangenerate feature sizes of ∼1 µm (Suh et al., 2017). Probably due totheir enhanced bioactivity, micromolded gelatin hydrogels can extendthe culture lifetime and maturation of C2C12 myotubes compared tosynthetic culture surfaces (Bettadapur et al., 2016; Denes et al., 2019).Carbon nanotubes have also been embedded into methacrylatedgelatin hydrogels to enhance myotube maturation by increasingelectrical conductivity (Ahadian et al., 2015; Ramón-Azcón et al.,2013). To better mimic the basement membrane of muscle,micromolded gelatin hydrogels have also been crosslinked with alayer of laminin, which improves the adherence, morphology andelectrophysiology of myotubes and neural cells (Besser et al., 2020).

Synthetic biomaterial substratesSynthetic biomaterials are advantageous culture substrates becausetheir mechanical and biochemical properties are highly controllableand reproducible. For example, polyethylene glycol andpolyacrylamide (PA) are both biologically inert hydrophilicpolymers that can be crosslinked into hydrogels with elastic modulituned to match the developing, healthy or fibrotic muscle tissuematrices (Engler et al., 2004). Another synthetic biomaterial that isimplemented as a culture surface is the aforementioned PDMS. Theelasticity of PDMS can be easily tuned to physiological or pathologicalvalues by altering the ratio of base to crosslinker (Wang et al., 2014) orblending different formulations of PDMS (Palchesko et al., 2012).To achieve consistent cell adhesion, researchers must functionalize

the synthetic substratewith ECMproteins. This is generally consideredan advantage because ECM ligand type and concentration can bespecified. Because collagen accounts for up to 10% of the dry weightof muscle (Gillies and Lieber, 2011), several studies have fabricatedsubstrates for C2C12 cultures by transferring collagen onto PDMS(Duffy et al., 2016) or PA hydrogels (Engler et al., 2004; Li et al.,2008a). Because the basement membrane of muscle fibers is enrichedin laminin and fibronectin, synthetic substrates functionalized witheither of these glycoproteins also promote myoblast adhesion andfusion into myotubes (Duffy et al., 2016; Gilbert et al., 2010;Palchesko et al., 2012; Ziemkiewicz et al., 2018).Synthetic biomaterials are also compatible with many

micropatterning techniques that can be used to introduce microscalefeatures on the surface to spatially control cell adhesion and alignment(Falconnet et al., 2006), as shown in Fig. 3. For example, PDMSstamps generated using the same photolithography techniquesdescribed above can be used to transfer ECM proteins onto a surfacein a process known as microcontact printing (Qin et al., 2010). Thisprocess has been used to prescribe myotube alignment on Petri dishes(Bajaj et al., 2011), PDMS-coated surfaces (Bettadapur et al., 2016;Jiwlawat et al., 2019; Nesmith et al., 2016; Palchesko et al., 2012; Sunet al., 2013) and PA hydrogels (Li et al., 2008a). Photolithography hasalso been used to selectively expose strips of a PA hydrogel to UVlight, which activates only the exposed regions for collagen bindingand thus myoblast adhesion (Engler et al., 2004). Myotubes have alsobeen aligned on substrates with nanoscale ridges fabricated usingelectron beam lithography, in which electrons are scanned in a defined

pattern on awafer coatedwith a light-sensitive photoresist (Wang et al.,2012). Another alternative is solvent-assisted capillary forcelithography, in which a polymer solution is molded on a siliconwafer with features at the hundreds of nanometers scale (Yang et al.,2014).

Integrated contractility assaysMicropatterned synthetic culture substrates are especially compatiblewith assays that quantify myotube contractility because themechanical properties of the substrate are well defined, andmyotube architecture can be controlled to increase the magnitudeand reproducibility of contractile force production. MicropatternedPA gels are widely used as a substrate for traction force microscopy, atechnique that quantifies forces generated by cells by tracking thedisplacement of fluorescent beads embedded in the hydrogel.Although traction force microscopy is more commonly used forcardiac myocytes (Ariyasinghe et al., 2017; McCain et al., 2012;Pasqualini et al., 2018; Ribeiro et al., 2015), it has also been used toquantify forces generated by micropatterned C2C12 myotubes (Liet al., 2008a).

Contractility can also be quantified by culturing myotubes onflexible cantilevers, as in the muscular thin film (MTF) assay. TheMTF assay entails first spin coating a glass coverslip with a layer ofpoly(N-isopropylacrylamide) (PNIPAAm), a temperature-sensitivepolymer, followed by a layer of PDMS (Feinberg et al., 2007). ThePDMS is then laser-cut into arrays of cantilevers with dimensionsranging from 1 mm to 5 mm (Agarwal et al., 2013), microcontactprinted with lines of fibronectin, and used to culture myotubes.After the desired culture period, the muscle-PDMS cantilevers,referred to as MTFs, are released by reducing the temperature from37°C to 25°C to solubilize the PNIPAAm. Electrodes are then usedto stimulate myotube contraction, which causes cantilever bending.Contractile stress is calculated based on the radius of curvature ofeach MTF (Grosberg et al., 2011). The MTF assay has beensuccessfully used to measure twitch and tetanus forces generated byC2C12 myotubes (Sun et al., 2013) and primary human myotubes(Nesmith et al., 2016). Microfabricated silicon cantilevers withdimensions of <1 mm have also been used as a culture substrate forprimary rat myotubes, and the contractile stresses in this system aremeasured based on the myotube-induced deflection of laser light(Smith et al., 2014a; Wilson et al., 2010). These compact lasersystems are advantageous for multiplexing, which increases testingthroughput and scalability for drug screening applications (Smithet al., 2014b).

Engineered co-cultures of skeletal muscle and motor neuronsMicrofabricated surfaces have also been developed to improvemixed co-cultures of myotubes and motor neurons. For example,photopolymerization techniques have been used to fabricate PAhydrogels with alternating soft and stiff stripes that mimic therigidity of nervous tissue and muscle tissue, respectively (Happeet al., 2017). On these surfaces, myoblasts preferentially migrateonto the stiffer stripes and fuse into aligned myotubes. When co-cultured with motor neurons, myotubes on mechanically patternedhydrogels exhibited increased acetylcholine receptor clusteringcompared to myotubes co-cultured on uniform hydrogels (Happeet al., 2017).

NMJ maturation has also been achieved in mixed co-cultures byapplying electrical stimulation using a bioreactor (Charoensooket al., 2017). This approach could be further refined by transfectingone or both cell types with channelrhodopsin, a membrane channelthat is activated by blue light (Fig. 3). Because chronic optogenetic

6

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 7: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

Microfabricated surfaces for controllingECM rigidity and myotube alignment

Optogenetic stimulationof myotubes and/or

motor neurons

Compartmentalized culture devicesto chemically and electrically isolate

myotubes and motor neurons

ChR2

Mixed co-culture

470 nm

Neuromuscularjunction

Skeletal muscle

Glass coverslipNeuronal bodies

Microchannels(for axon migration)

PDMS frame

Microcontact printing of fibronectin onto PDMS

10x10PDMS stamp

Fibronectin

PDMS-coated coverslip

Aligned myotubes

Micromolding of gelatin hydrogels

10x10 PDMSstamp

Activated coverslip

Tape

Aligned myotubesGelatin

Mechanical patterning of photosensitive polyacrylamide hydrogels

Aligned myotubes

Methacrylatedcoverslip

2nd PA gel

Photomask

UVUV

1st PA gel Cured PA gel

Mechanically patterned hydrogel

Fig. 3. Engineered 2D neuromuscular tissues. Conventional approaches for engineering neuromuscular tissues in vitro entailed mixed co-cultures (center).New advances to improve the architecture and assaying capabilities of 2D neuromuscular tissues include microfabricated surfaces (top), compartmentalizedculture devices (right), and integration of optogenetics (left). ChR2, channelrhodopsin-2; ECM, extracellular matrix; PA, polyacrylamide; PDMS,polydimethylsiloxane; UV, ultraviolet.

7

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 8: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

stimulation of myotubes can improve maturity (Rangarajan et al.,2014), a similar strategy applied to co-cultures could be a relativelynon-invasive approach for maturation. Transfecting motor neuronswith channelrhodopsin is also a powerful experimental tool formixed co-cultures because it enables users to stimulate only motorneurons and therefore more clearly identify responses in the musclethat are driven specifically by motor neurons (Lin et al., 2019;Steinbeck et al., 2016).Compartmentalized culture devices have also beenmicrofabricated

to physically isolate motor neurons and myotubes into separatechambers (Fig. 3). These chambers are connected by microchannelsthat are permissive to axons, but not cell bodies, to allow thecontrolled formation of NMJs in the myotube chamber (Santhanamet al., 2018; Taylor et al., 2003). Because the chambers are chemicallyisolated, these devices allow each cell type to be cultured in its ownmedium, which may boost viability. Furthermore, drugs or othersmall molecules can be selectively added to one or both chambers

(Santhanam et al., 2018), which can be useful for establishing drugmechanisms. Structural and functional analyses are also easier inthese devices compared to mixed co-cultures because NMJs form inrelatively prescribed locations and cells in each chamber can beelectrically stimulated independently.

Engineered 3D models of neuromuscular tissuesAlthough engineered 2D neuromuscular tissues have manyadvantages from an assay perspective, they fundamentally lackthe bundle-like architecture and cell-ECM interactions of nativemuscle fibers. To address this, researchers have developed severalapproaches to engineer miniature 3Dmuscle bundles (Fig. 4). Thesetypes of approaches were first reported in the late 1990s and entailedinjecting primary myoblasts mixed in an ECM pre-polymer solutioninto a rectangular chamber with patches of stainless-steel screening(Shansky et al., 1997) or Velcro (Powell et al., 1999) at itslongitudinal ends. As the myoblasts fused into myotubes, they

Matrigel

Thrombin

Fibrinogen

Myoblasts

A

B

Velcro/nylon frame

Gel filling port

Reservoir

Mediumchannel

Gel region

Vacuuminlet

Vacuumchannel

Musclestrip

Hydrogel

Neurosphere

D7 D14

Tuj1

/F-a

ctin

/DAP

I

Tuj1

/F-a

ctin

/DAP

I

Fig. 4. Engineered 3D neuromuscular tissues. (A) Aligned 3Dmuscle bundles are engineered bymixing myoblasts in an ECM pre-polymer solution of Matrigel,thrombin and fibrinogen, and casting it into a microfabricated support structure, such as a Velcro frame. Scale bars: 50 mm (left image); 50 µm (right image).Adapted from Madden et al. (2015). (B) Compartmentalized fluidic devices have been microfabricated to controllably co-culture 3D muscle bundles and motorneuron spheroids, and generated NMJs after 14 days in culture. D7, day 7; D14, day 14; DAPI, 4′,6-diamidino-2-phenylindole; nAChR, nicotinic acetylcholinereceptor; SAA, sarcomeric alpha-actinin; Tuj1, neuron-specific class III β-tubulin. Scale bars: 2 mm (left image); 10 µm (right image). Adapted with permissionfrom Osaki et al. (2018) and Uzel et al. (2016). The images in this figure are not published under the terms of the CC-BY license of this article. For permission toreuse, please see Madden et al. (2015), Osaki et al. (2018) and Uzel et al. (2016).

8

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 9: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

detached from the bottom surface but remained embedded in theECM and anchored by the screening or Velcro, forming anelongated 3D muscle bundle with aligned myotubes.Over the past two decades, approaches for engineering 3Dmuscle

bundles have been advanced and refined. Several different types ofculture chambers with anchor points have been fabricated(Costantini et al., 2017a; Smith et al., 2016), including Velcro andnylon frames (Davis et al., 2017, 2019; Madden et al., 2015; Raoet al., 2018; Smith et al., 2016; Zhang et al., 2018) andmicrofabricated chambers with pillars (Osaki et al., 2018, 2020;Uzel et al., 2016). Testing of multiple ECM solutions has alsorevealed that fibrin hydrogels are optimal for encapsulatingmyotubes due to their strength (Hinds et al., 2011; Pollot et al.,2018), although these hydrogel compositions are not necessarilyphysiological. To improve assay capabilities, contractile forces havebeen measured in 3D muscle bundles with custom force transducers(Davis et al., 2019; Madden et al., 2015; Rao et al., 2018) or bytracking the displacement of pillars (Osaki et al., 2018; Uzel et al.,2016). Similar to 2D tissues, biophysical cues, such as mechanicalstretch (Powell et al., 2002) and optogenetic stimulation (Mills et al.,2019), or addition of fibroblasts (Dennis et al., 2001) have also beenshown to mature 3D muscle bundles.Microfluidic devices have also been fabricated to engineer and

maintain 3D muscle bundles (Agrawal et al., 2017; Shimizu et al.,2015). These systems are advantageous because they continuouslyperfuse freshmedia to the engineered tissues, which probably improvesviability compared to static culture. Furthermore, microfluidic devicescan be used to screen drugs at a higher throughput and can be linked toother microfluidic organ-on-chip systems to capture organ-organinteractions, and mimic organism-level responses (Novak et al., 2020).One common approach to innervate 3D muscle bundles is to

directly seed them with spheroids of motor neurons (Afshar Bakooshliet al., 2019; Morimoto et al., 2013; Smith et al., 2016). These systemshave demonstrated that the resulting NMJs are functional but still haverelatively diffuse acetylcholine receptor clustering (Morimoto et al.,2013). Acetylcholine clustering in 3D muscle bundles has beenadvanced by adding the basement membrane components agrin andlaminin (Wang et al., 2013), which could help improveNMJ formationin these co-cultures. Despite their limited maturity, these 3Dneuromuscular tissues show reduced contractility in response to serafrom MG patients (Morimoto et al., 2013), recapitulating thepathological response in MG and demonstrating their promise formodeling complex neuromuscular diseases.Similar to 2D models, compartmentalized microdevices have

been developed to culture motor neuron spheroids and engineeredmuscle bundles in separate compartments connected by axon-permissive channels (Fig. 4) (Osaki et al., 2018, 2020; Uzel et al.,2016). In these studies, the muscle bundles were attached to flexiblepillars and the motor neurons were optogenetically modified. Withthis combination of technologies, the users could quantify musclecontractility as a function of motor neuron stimulation, a key readoutof NMJ function. This type of device was also used to capture NMJdegeneration in tissues generated using hiPSC-derived motorneurons from an ALS patient. Importantly, the application of twoALS drug candidates, bosutinib and rapamycin, to this modelreduced muscle atrophy and dysfunction (Osaki et al., 2018),demonstrating how this type of approach has the potential forpatient-specific disease modeling and drug screening.

ConclusionsRecently developed approaches to model healthy and diseasedneuromuscular tissues on a chip have the potential to capture the

vastly heterogeneous genotypes and phenotypes of individuals witha variety of neuromuscular disorders. Newer technologies, such as3D bioprinting, which is a form of additive manufacturing that usescells and other biomaterials as ‘inks’ to print living structures (Choiet al., 2016; Costantini et al., 2017b; Kang et al., 2016; Kim et al.,2020), will probably further advance these models. However,in vitro models of neuromuscular tissues are far from achievingadult-like maturity, especially when based on hiPSC-derivedmuscle cells and motor neurons. Furthermore, in vitro models ofneuromuscular tissues lack the supporting cells known to beimportant regulators of NMJs in health and disease, such asSchwann cells (Santosa et al., 2018). Most in vitro models alsocurrently lack immune cells, despite the established role ofneuroinflammation in many neuromuscular diseases (Mäureret al., 2002; Thonhoff et al., 2018). However, researchers havebegun developing models that integrate immune cells, such asmacrophages (Juhas et al., 2018), to probe the role of the immunesystem in muscle injury and repair. Given these limitations, in vitromodels are most powerful when implemented hand-in-hand withanimal models, which have less human relevance but moreadvanced motor unit structure and physiology. Together, thesecomplementary model systems are likely to pave the way for moreeffective and personalized therapies for these debilitating diseases.

Competing interestsThe authors declare no competing or financial interests.

FundingThis project was supported by the University of Southern California Viterbi School ofEngineering, Women in Science and Engineering, University of Southern California,an Amyotrophic Lateral Sclerosis Association Starter Grant (18-IIA-401 toM.L.M.), aRose Hills Foundation Innovator Grant to M.L.M., and a National ScienceFoundation Graduate Research Fellowship Grant (DGE 1418060 to J.W.S.).

ReferencesAartsma-Rus, A. and van Putten, M. (2020). The use of genetically humanized

animal models for personalized medicine approaches. Dis. Model. Mech. 13,dmm041673. doi:10.1242/dmm.041673

Abdelmoez, A. M., Sardon Puig, L., Smith, J. A. B., Gabriel, B. M., Savikj, M.,Dollet, L., Chibalin, A. V., Krook, A., Zierath, J. R. and Pillon, N. J. (2019).Comparative profiling of skeletal muscle models reveals heterogeneity oftranscriptome and metabolism. Am. J. Physiol. Cell Physiol. 318, C615-C626.doi:10.1152/ajpcell.00540.2019

Abujarour, R., Bennett, M., Valamehr, B., Lee, T. T., Robinson, M., Robbins, D.,Le, T., Lai, K. and Flynn, P. (2014). Myogenic differentiation of musculardystrophy-specific induced pluripotent stem cells for use in drug discovery. StemCells Transl. Med. 3, 149-160. doi:10.5966/sctm.2013-0095

Afshar Bakooshli, M., Lippmann, E. S., Mulcahy, B., Iyer, N., Nguyen, C. T.,Tung, K., Stewart, B. A., van denDorpel, H., Fuehrmann, T., Shoichet, M. et al.(2019). A 3D culture model of innervated human skeletal muscle enables studiesof the adult neuromuscular junction. eLife 8, e44530. doi:10.7554/eLife.44530

Agarwal, A., Goss, J. A., Cho, A., McCain, M. L. and Parker, K. K. (2013).Microfluidic heart on a chip for higher throughput pharmacological studies. Lab.Chip 13, 3599-3608. doi:10.1039/c3lc50350j

Agrawal, G., Aung, A. and Varghese, S. (2017). Skeletal muscle-on-a-chip: an invitro model to evaluate tissue formation and injury. Lab. Chip 17, 3447-3461.doi:10.1039/C7LC00512A

Ahadian, S., Ramon-Azcon, J., Estili, M., Liang, X., Ostrovidov, S., Shiku, H.,Ramalingam, M., Nakajima, K., Sakka, Y., Bae, H. et al. (2015). Hybridhydrogels containing vertically aligned carbon nanotubes with anisotropicelectrical conductivity for muscle myofiber fabrication. Sci. Rep. 4, 4271. doi:10.1038/srep04271

Ariyasinghe, N. R., Reck, C. H., Viscio, A. A., Petersen, A. P., Lyra-Leite, D. M.,Cho, N. and McCain, M. L. (2017). Engineering micromyocardium to delineatecellular and extracellular regulation of myocardial tissue contractility. Integr. Biol.9, 730-741. doi:10.1039/C7IB00081B

Armstrong, J. P. K., Puetzer, J. L., Serio, A., Guex, A. G., Kapnisi, M., Breant, A.,Zong, Y., Assal, V., Skaalure, S. C., King, O. et al. (2018). Engineeringanisotropic muscle tissue using acoustic cell patterning. Adv. Mater. 30, 1802649.doi:10.1002/adma.201802649

Askanas, V., Kwan, H., Alvarez, R. B., Engel, W. K., Kobayashi, T., Martinuzzi, A.and Hawkins, E. F. (1987). De novo neuromuscular junction formation on human

9

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 10: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

muscle fibres cultured in monolayer and innervated by foetal rat spinal cord:ultrastructural and ultrastructural-cytochemical studies. J. Neurocytol. 16,523-537. doi:10.1007/BF01668506

Babin, P. J., Goizet, C. and Raldua, D. (2014). Zebrafish models of human motorneuron diseases: advantages and limitations.Prog. Neurobiol. 118, 36-58. doi:10.1016/j.pneurobio.2014.03.001

Bajaj, P., Reddy, B., Jr, Millet, L., Wei, C., Zorlutuna, P., Bao, G. and Bashir, R.(2011). Patterning the differentiation of C2C12 skeletal myoblasts. Integr. Biol. 3,897-909. doi:10.1039/c1ib00058f

Balci, B. and Dinçer, P. (2009). Efficient transfection of mouse-derived C2C12myoblasts using a matrigel basement membrane matrix. Biotechnol. J. 4,1042-1045. doi:10.1002/biot.200800269

Beaudet, M.-J., Yang, Q., Cadau, S., Blais, M., Bellenfant, S., Gros-Louis, F. andBerthod, F. (2015). High yield extraction of pure spinal motor neurons, astrocytesandmicroglia from single embryo and adult mouse spinal cord.Sci. Rep. 5, 16763.doi:10.1038/srep16763

Besser, R. R., Bowles, A. C., Alassaf, A., Carbonero, D., Claure, I., Jones, E.,Reda, J., Wubker, L., Batchelor, W., Ziebarth, N. et al. (2020). Enzymaticallycrosslinked gelatin–laminin hydrogels for applications in neuromuscular tissueengineering. Biomaterials Sci. 8, 591-606. doi:10.1039/C9BM01430F

Bettadapur, A., Suh, G. C., Geisse, N. A., Wang, E. R., Hua, C., Huber, H. A.,Viscio, A. A., Kim, J. Y., Strickland, J. B. and McCain, M. L. (2016). Prolongedculture of aligned skeletal myotubes on micromolded gelatin hydrogels. Sci. Rep.6, 28855. doi:10.1038/srep28855

Bonetto, A., Andersson, D. C. and Waning, D. L. (2015). Assessment of musclemass and strength in mice.BoneKEy Rep. 4, 732. doi:10.1038/bonekey.2015.101

Boularaoui, S. M., Abdel-Raouf, K. M. A., Alwahab, N. S. A., Kondash, M. E.,Truskey, G. A., Teo, J. C. M. and Christoforou, N. (2018). Efficienttransdifferentiation of human dermal fibroblasts into skeletal muscle. J. TissueEng. Regen. Med. 12, e918-e936. doi:10.1002/term.2415

Bowerman, M., Becker, C. G., Yanez-Mun oz, R. J., Ning, K., Wood, M. J. A.,Gillingwater, T. H., Talbot, K. and Consortium, U. S. R. (2017). Therapeuticstrategies for spinal muscular atrophy: SMN and beyond. Dis. Model. Mech. 10,943-954. doi:10.1242/dmm.030148

Boylan, K. (2015). Familial amyotrophic lateral sclerosis. Neurol. Clin. 33, 807-830.doi:10.1016/j.ncl.2015.07.001

Braun, S., Croizat, B., Lagrange, M.-C., Poindron, P. and Warter, J.-M. (1997).Degeneration of cocultures of spinal muscular atrophy muscle cells and rat spinalcord explants is not due to secreted factors and cannot be prevented byneurotrophins. Muscle Nerve 20, 953-960. doi:10.1002/(SICI)1097-4598(199708)20:8<953::AID-MUS4>3.0.CO;2-5

Briese, M., Esmaeili, B., Fraboulet, S., Burt, E. C., Christodoulou, S., Towers,P. R., Davies, K. E. and Sattelle, D. B. (2009). Deletion of smn-1, theCaenorhabditis elegans ortholog of the spinal muscular atrophy gene, results inlocomotor dysfunction and reduced lifespan. Hum. Mol. Genet. 18, 97-104.doi:10.1093/hmg/ddn320

Cashman, N. R., Durham, H. D., Blusztajn, J. K., Oda, K., Tabira, T., Shaw, I. T.,Dahrouge, S. and Antel, J. P. (1992). Neuroblastoma×spinal cord (NSC) hybridcell lines resemble developing motor neurons. Dev. Dyn. 194, 209-221. doi:10.1002/aja.1001940306

Chal, J., Oginuma, M., Al Tanoury, Z., Gobert, B., Sumara, O., Hick, A.,Bousson, F., Zidouni, Y., Mursch, C., Moncuquet, P. et al. (2015).Differentiation of pluripotent stem cells to muscle fiber to model Duchennemuscular dystrophy. Nat. Biotechnol. 33, 962-969. doi:10.1038/nbt.3297

Chal, J., Al Tanoury, Z., Hestin, M., Gobert, B., Aivio, S., Hick, A., Cherrier, T.,Nesmith, A. P., Parker, K. K. and Pourquie, O. (2016). Generation of humanmuscle fibers and satellite-like cells from human pluripotent stem cells in vitro.Nat.Protoc. 11, 1833-1850. doi:10.1038/nprot.2016.110

Chang, Y.-J., Chen, Y.-J., Huang, C.-W., Fan, S.-C., Huang, B.-M., Chang, W.-T.,Tsai, Y.-S., Su, F.-C. and Wu, C.-C. (2016). Cyclic stretch facilitates myogenesisin C2C12 myoblasts and rescues thiazolidinedione-inhibited myotube formation.Front. Bioeng. Biotechnol. 4, 27. doi:10.3389/fbioe.2016.00027

Chapman, A. L., Bennett, E. J., Ramesh, T. M., De Vos, K. J. and Grierson, A. J.(2013). Axonal transport defects in a mitofusin 2 loss of function model of charcot-marie-tooth disease in zebrafish. PLoS ONE 8, e67276. doi:10.1371/journal.pone.0067276

Charoensook, S. N., Williams, D. J., Chakraborty, S., Leong, K. W. and Vunjak-Novakovic, G. (2017). Bioreactor model of neuromuscular junction with electricalstimulation for pharmacological potency testing. Integr. Biol. 9, 956-967. doi:10.1039/C7IB00144D

Chaytow, H., Huang, Y.-T., Gillingwater, T. H. and Faller, K. M. E. (2018). The roleof survival motor neuron protein (SMN) in protein homeostasis. Cell. Mol. Life Sci.75, 3877-3894. doi:10.1007/s00018-018-2849-1

Cheng, C. S., El-Abd, Y., Bui, K., Hyun, Y.-E., Hughes, R. H., Kraus, W. E. andTruskey, G. A. (2014). Conditions that promote primary human skeletal myoblastculture and muscle differentiation in vitro. Am. J. Physiol. Cell Physiol. 306,C385-C395. doi:10.1152/ajpcell.00179.2013

Choi, Y.-J., Kim, T. G., Jeong, J., Yi, H.-G., Park, J.W., Hwang,W. andCho, D.-W.(2016). 3D cell printing of functional skeletal muscle constructs using skeletal

muscle-derived bioink. Adv. Healthc. Mater. 5, 2636-2645. doi:10.1002/adhm.201600483

Costantini, M., Testa, S., Fornetti, E., Barbetta, A., Trombetta, M., Cannata,S. M., Gargioli, C. and Rainer, A. (2017a). Engineering muscle networks in 3Dgelatin methacryloyl hydrogels: influence of mechanical stiffness and geometricalconfinement. Front. Bioeng. Biotechnol. 5, 22. doi:10.3389/fbioe.2017.00022

Costantini, M., Testa, S., Mozetic, P., Barbetta, A., Fuoco, C., Fornetti, E.,Tamiro, F., Bernardini, S., Jaroszewicz, J., Swieszkowski, W. et al. (2017b).Microfluidic-enhanced 3D bioprinting of aligned myoblast-laden hydrogels leadsto functionally organized myofibers in vitro and in vivo. Biomaterials 131, 98-110.doi:10.1016/j.biomaterials.2017.03.026

Dangouloff, T. and Servais, L. (2019). Clinical evidence supporting early treatmentof patients with spinal muscular atrophy: current perspectives. Ther. Clin. RiskManagement 15, 1153-1161. doi:10.2147/TCRM.S172291

Daniels, M. P., Lowe, B. T., Shah, S., Ma, J., Samuelsson, S. J., Lugo, B., Parakh,T. and Uhm, C.-S. (2000). Rodent nerve-muscle cell culture system for studies ofneuromuscular junction development: refinements and applications. Microsc.Res. Tech. 49, 26-37. doi:10.1002/(SICI)1097-0029(20000401)49:1<26::AID-JEMT4>3.0.CO;2-8

Darabi, R., Arpke, R. W., Irion, S., Dimos, J. T., Grskovic, M., Kyba, M. andPerlingeiro, R. C. R. (2012). Human ES- and iPS-derived myogenic progenitorsrestore DYSTROPHIN and improve contractility upon transplantation in dystrophicmice. Cell Stem Cell 10, 610-619. doi:10.1016/j.stem.2012.02.015

Das, M., Rumsey, J. W., Bhargava, N., Stancescu, M. and Hickman, J. J. (2010).A defined long-term in vitro tissue engineered model of neuromuscular junctions.Biomaterials 31, 4880-4888. doi:10.1016/j.biomaterials.2010.02.055

Davis, B. N. J., Santoso, J. W., Walker, M. J., Cheng, C. S., Koves, T. R., Kraus,W. E. and Truskey, G. A. (2017). Human, tissue-engineered, skeletal musclemyobundles to measure oxygen uptake and assess mitochondrial toxicity. TissueEng. C Methods 23, 189-199. doi:10.1089/ten.tec.2016.0264

Davis, B. N. J., Santoso, J. W., Walker, M. J., Oliver, C. E., Cunningham, M. M.,Boehm, C. A., Dawes, D., Lasater, S. L., Huffman, K., Kraus,W. E. et al. (2019).Modeling the effect of TNF-α upon drug-induced toxicity in human, tissue-engineeredmyobundles.Ann. Biomed. Eng. 47, 1596-1610. doi:10.1007/s10439-019-02263-8

De Giorgio, F., Maduro, C., Fisher, E. M. C. and Acevedo-Arozena, A. (2019).Transgenic and physiological mousemodels give insights into different aspects ofamyotrophic lateral sclerosis. Dis. Model. Mech. 12, dmm037424. doi:10.1242/dmm.037424

Deenen, J. C. W., Horlings, C. G. C., Verschuuren, J. J. G. M., Verbeek, A. L. M.and van Engelen, B. G. M. (2015). The epidemiology of neuromusculardisorders: a comprehensive overview of the literature. J. Neuromuscul. Dis. 2,73-85. doi:10.3233/JND-140045

Denes, L. T., Riley, L. A., Mijares, J. R., Arboleda, J. D., McKee, K., Esser, K. A.and Wang, E. T. (2019). Culturing C2C12 myotubes on micromolded gelatinhydrogels accelerates myotube maturation. Skelet. Muscle 9, 17. doi:10.1186/s13395-019-0203-4

Dennis, R. G., Kosnik, P. E., Gilbert, M. E. and Faulkner, J. A. (2001). Excitabilityand contractility of skeletal muscle engineered from primary cultures and cell lines.Am. J. Physiol. Cell Physiol. 280, C288-C295. doi:10.1152/ajpcell.2001.280.2.C288

Devoy, A., Kalmar, B., Stewart, M., Park, H., Burke, B., Noy, S. J., Redhead, Y.,Humphrey, J., Lo, K., Jaeger, J. et al. (2017). Humanized mutant FUS drivesprogressive motor neuron degeneration without aggregation in ‘FUSDelta14’knockin mice. Brain 140, 2797-2805. doi:10.1093/brain/awx248

Dimos, J. T., Rodolfa, K. T., Niakan, K. K., Weisenthal, L. M., Mitsumoto, H.,Chung, W., Croft, G. F., Saphier, G., Leibel, R., Goland, R. et al. (2008).Induced pluripotent stem cells generated from patients with ALS can bedifferentiated into motor neurons. Science 321, 1218-1221. doi:10.1126/science.1158799

Du, Z.-W., Chen, H., Liu, H., Lu, J., Qian, K., Huang, C. T.-L., Zhong, X., Fan, F.and Zhang, S.-C. (2015). Generation and expansion of highly pure motor neuronprogenitors from human pluripotent stem cells. Nat. Commun. 6, 6626. doi:10.1038/ncomms7626

Duffy, R. M., Sun, Y. and Feinberg, A. W. (2016). Understanding the role of ECMprotein composition and geometric micropatterning for engineering humanskeletal muscle. Ann. Biomed. Eng. 44, 2076-2089. doi:10.1007/s10439-016-1592-8

Engler, A. J., Griffin, M. A., Sen, S., Bonnemann, C. G., Sweeney, H. L. andDischer, D. E. (2004). Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological implications for soft or stiff microenvironments. J. CellBiol. 166, 877-887. doi:10.1083/jcb.200405004

Falconnet, D., Csucs, G., Grandin, H. M. and Textor, M. (2006). Surfaceengineering approaches to micropattern surfaces for cell-based assays.Biomaterials 27, 3044-3063. doi:10.1016/j.biomaterials.2005.12.024

Feinberg, A. W., Feigel, A., Shevkoplyas, S. S., Sheehy, S., Whitesides, G. M.and Parker, K. K. (2007). Muscular thin films for building actuators and poweringdevices. Science 317, 1366-1370. doi:10.1126/science.1146885

Fuller, H. R., Mandefro, B., Shirran, S. L., Gross, A. R., Kaus, A. S., Botting,C. H., Morris, G. E. and Sareen, D. (2016). Spinal muscular atrophy patient iPSC-

10

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 11: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

derived motor neurons have reduced expression of proteins important in neuronaldevelopment. Front. Cell. Neurosci. 9, 506. doi:10.3389/fncel.2015.00506

Gilbert, P. M., Havenstrite, K. L., Magnusson, K. E. G., Sacco, A., Leonardi,N. A., Kraft, P., Nguyen, N. K., Thrun, S., Lutolf, M. P. and Blau, H. M. (2010).Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture.Science 329, 1078-1081. doi:10.1126/science.1191035

Gilhus, N. E. (2016). Myasthenia gravis. N. Engl. J. Med. 375, 2570-2581. doi:10.1056/NEJMra1602678

Gillies, A. R. and Lieber, R. L. (2011). Structure and function of the skeletal muscleextracellular matrix. Muscle Nerve 44, 318-331. doi:10.1002/mus.22094

Gingras, M., Gagnon, V., Minotti, S., Durham, H. D. and Berthod, F. (2007).Optimized protocols for isolation of primary motor neurons, astrocytes andmicroglia from embryonic mouse spinal cord. J. Neurosci. Methods 163, 111-118.doi:10.1016/j.jneumeth.2007.02.024

Gomes, C., Escrevente, C. and Costa, J. (2010). Mutant superoxide dismutase 1overexpression in NSC-34 cells: effect of trehalose on aggregation, TDP-43localization and levels of co-expressed glycoproteins. Neurosci. Lett. 475,145-149. doi:10.1016/j.neulet.2010.03.065

Goto, K., Imamura, K., Komatsu, K., Mitani, K., Aiba, K., Nakatsuji, N., Inoue, M.,Kawata, A., Yamashita, H., Takahashi, R. et al. (2017). Simple derivation ofspinal motor neurons from ESCs/iPSCs using Sendai virus vectors. Mol. Ther.Methods Clin. Dev. 4, 115-125. doi:10.1016/j.omtm.2016.12.007

Granato, D. A., Fulpius, B. W. and Moody, J. F. (1976). Experimental myastheniain Balb/c mice immunized with rat acetylcholine receptor from rat denervatedmuscle. Proc. Natl. Acad. Sci. USA 73, 2872-2876. doi:10.1073/pnas.73.8.2872

Grosberg, A., Alford, P.W., McCain, M. L. and Parker, K. K. (2011). Ensembles ofengineered cardiac tissues for physiological and pharmacological study: heart ona chip. Lab. Chip 11, 4165-4173. doi:10.1039/c1lc20557a

Guo, X., Johe, K., Molnar, P., Davis, H. and Hickman, J. (2010). Characterizationof a human fetal spinal cord stem cell line, NSI-566RSC, and its induction tofunctional motoneurons. J. Tissue Eng. Regen. Med. 4, 181-193. doi:10.1002/term.223

Guo, X., Gonzalez, M., Stancescu, M., Vandenburgh, H. H. and Hickman, J. J.(2011). Neuromuscular junction formation between human stem cell-derivedmotoneurons and human skeletal muscle in a defined system. Biomaterials 32,9602-9611. doi:10.1016/j.biomaterials.2011.09.014

Hall, J. E. and Hall, M. E. (2015). Guyton and Hall Textbook of Medical Physiology,13th edn. Saunders.

Happe, C. L., Tenerelli, K. P., Gromova, A. K., Kolb, F. and Engler, A. J. (2017).Mechanically patterned neuromuscular junctions-in-a-dish have improvedfunctional maturation. Mol. Biol. Cell 28, 1950-1958. doi:10.1091/mbc.e17-01-0046

Heher, P., Maleiner, B., Pruller, J., Teuschl, A. H., Kollmitzer, J., Monforte, X.,Wolbank, S., Redl, H., Runzler, D. and Fuchs, C. (2015). A novel bioreactor forthe generation of highly aligned 3D skeletal muscle-like constructs throughorientation of fibrin via application of static strain. Acta Biomater. 24, 251-265.doi:10.1016/j.actbio.2015.06.033

Hester, M. E., Murtha, M. J., Song, S. W., Rao, M., Miranda, C. J., Meyer, K., Tian,J., Boulting, G., Schaffer, D. V., Zhu, M. X. et al. (2011). Rapid and efficientgeneration of functional motor neurons from human pluripotent stem cells usinggene delivered transcription factor codes.Mol. Ther. 19, 1905-1912. doi:10.1038/mt.2011.135

Hindi, L., McMillan, J. D., Afroze, D., Hindi, S. M. and Kumar, A. (2017). Isolation,culturing, and differentiation of primary myoblasts from skeletal muscle of adultmice. Bio Protoc. 7, e2248. doi:10.21769/BioProtoc.2248

Hinds, S., Bian, W., Dennis, R. G. and Bursac, N. (2011). The role of extracellularmatrix composition in structure and function of bioengineered skeletal muscle.Biomaterials 32, 3575-3583. doi:10.1016/j.biomaterials.2011.01.062

Hosseini, V., Ahadian, S., Ostrovidov, S., Camci-Unal, G., Chen, S., Kaji, H.,Ramalingam, M. and Khademhosseini, A. (2012). Engineered contractileskeletal muscle tissue on a microgrooved methacrylated gelatin substrate. TissueEng. Part A 18, 2453-2465. doi:10.1089/ten.tea.2012.0181

Hsieh-Li, H. M., Chang, J.-G., Jong, Y.-J., Wu, M.-H., Wang, N. M., Tsai, C. H. andLi, H. (2000). A mouse model for spinal muscular atrophy. Nat. Genet. 24, 66-70.doi:10.1038/71709

Hu, B.-Y. and Zhang, S.-C. (2009). Differentiation of spinal motor neurons frompluripotent human stem cells. Nat. Protoc. 4, 1295-1304. doi:10.1038/nprot.2009.127

Ichida, J. K., Staats, K. A., Davis-Dusenbery, B. N., Clement, K., Galloway, K. E.,Babos, K. N., Shi, Y., Son, E. Y., Kiskinis, E., Atwater, N. et al. (2018).Comparative genomic analysis of embryonic, lineage-converted and stem cell-derived motor neurons. Development 145, dev168617. doi:10.1242/dev.168617

Ito, A., Yamamoto, Y., Sato, M., Ikeda, K., Yamamoto, M., Fujita, H., Nagamori,E., Kawabe, Y. and Kamihira, M. (2014). Induction of functional tissue-engineered skeletal muscle constructs by defined electrical stimulation. Sci.Rep. 4, 4781. doi:10.1038/srep04781

Ito, N., Kii, I., Shimizu, N., Tanaka, H. and Takeda, S. (2017). Directreprogramming of fibroblasts into skeletal muscle progenitor cells bytranscription factors enriched in undifferentiated subpopulation of satellite cells.Sci. Rep. 7, 8097. doi:10.1038/s41598-017-08232-2

Jang, Y. C. and Van Remmen, H. (2011). Age-associated alterations of theneuromuscular junction. Exp. Gerontol. 46, 193-198. doi:10.1016/j.exger.2010.08.029

Jedrzejowska, M., Milewski, M., Zimowski, J., Borkowska, J., Kostera-Pruszczyk, A., Sielska, D., Jurek, M. and Hausmanowa-Petrusewicz, I.(2009). Phenotype modifiers of spinal muscular atrophy: the number of SMN2gene copies, deletion in the NAIP gene and probably gender influence the courseof the disease. Acta Biochim. Pol. 56, 103-108. doi:10.18388/abp.2009_2521

Jiwlawat, N., Lynch, E., Jeffrey, J., Van Dyke, J. M. and Suzuki, M. (2018).Current progress and challenges for skeletal muscle differentiation from humanpluripotent stem cells using transgene-free approaches. Stem Cells Int. 2018,6241681. doi:10.1155/2018/6241681

Jiwlawat, N., Lynch, E. M., Napiwocki, B. N., Stempien, A., Ashton, R. S., Kamp,T. J., Crone, W. C. and Suzuki, M. (2019). Micropatterned substrates withphysiological stiffness promote cell maturation and Pompe disease phenotype inhuman induced pluripotent stem cell-derived skeletal myocytes. Biotechnol.Bioeng. 116, 2377-2392. doi:10.1002/bit.27075

Joyce, N. C., Oskarsson, B. and Jin, L.-W. (2012). Muscle biopsy evaluation inneuromuscular disorders. Phys. Med. Rehabil. Clin. N Am. 23, 609-631. doi:10.1016/j.pmr.2012.06.006

Juhas, M., Abutaleb, N., Wang, J. T., Ye, J., Shaikh, Z., Sriworarat, C., Qian, Y.and Bursac, N. (2018). Incorporation of macrophages into engineered skeletalmuscle enables enhanced muscle regeneration. Nat. Biomed. Eng. 2, 942-954.doi:10.1038/s41551-018-0290-2

Juneja, M., Burns, J., Saporta, M. A. and Timmerman, V. (2019). Challenges inmodelling the Charcot-Marie-Tooth neuropathies for therapy development.J. Neurol. Neurosurg. Psychiatr. 90, 58-67. doi:10.1136/jnnp-2018-318834

Kang, H.-W., Lee, S. J., Ko, I. K., Kengla, C., Yoo, J. J. and Atala, A. (2016). A 3Dbioprinting system to produce human-scale tissue constructs with structuralintegrity. Nat. Biotechnol. 34, 312-319. doi:10.1038/nbt.3413

Kengaku, M., Kawata, A., Kawashima, S. and Nakane, M. (1991). Role offibronectin in the inhibitory effect of TGF-beta on choline acetyltransferase activityin co-cultures of spinal cord neurons and myotubes. Brain Res. Dev. Brain Res.61, 281-284. doi:10.1016/0165-3806(91)90144-8

Kim, M., Kim, W. J. and Kim, G. H. (2017). Topologically micropatterned collagenand poly(ε-caprolactone) struts fabricated using the poly(vinyl alcohol) fibrillation/leaching process to develop efficiently engineered skeletal muscle tissue. ACSAppl. Material. Interfaces 9, 43459-43469. doi:10.1021/acsami.7b14192

Kim, W. J., Lee, H., Lee, J. U., Atala, A., Yoo, J. J., Lee, S. J. and Kim, G. H.(2020). Efficient myotube formation in 3D bioprinted tissue construct bybiochemical and topographical cues. Biomaterials 230, 119632. doi:10.1016/j.biomaterials.2019.119632

Laine, J., Skoglund, G., Fournier, E. and Tabti, N. (2018). Development of theexcitation-contraction coupling machinery and its relation to myofibrillogenesis inhuman iPSC-derived skeletal myocytes.Skelet. Muscle 8, 1. doi:10.1186/s13395-017-0147-5

Laird, A. S., Mackovski, N., Rinkwitz, S., Becker, T. S. and Giacomotto, J.(2016). Tissue-specific models of spinal muscular atrophy confirm a critical role ofSMN in motor neurons from embryonic to adult stages. Hum. Mol. Genet. 25,1728-1738. doi:10.1093/hmg/ddw044

Lattante, S., Ciura, S., Rouleau, G. A. and Kabashi, E. (2015). Defining thegenetic connection linking amyotrophic lateral sclerosis (ALS) with frontotemporaldementia (FTD). Trends Genet. 31, 263-273. doi:10.1016/j.tig.2015.03.005

Lattanzi, L., Salvatori, G., Coletta, M., Sonnino, C., Cusella De Angelis, M. G.,Gioglio, L., Murry, C. E., Kelly, R., Ferrari, G., Molinaro, M. et al. (1998). Highefficiency myogenic conversion of human fibroblasts by adenoviral vector-mediated MyoD gene transfer. An alternative strategy for ex vivo gene therapyof primary myopathies. J. Clin. Invest. 101, 2119-2128. doi:10.1172/JCI1505

Lee, S., Cuvillier, J. M., Lee, B., Shen, R., Lee, J. W. and Lee, S.-K. (2012). Fusionprotein Isl1-Lhx3 specifies motor neuron fate by inducing motor neuron genes andconcomitantly suppressing the interneuron programs. Proc. Natl. Acad. Sci. USA109, 3383-3388. doi:10.1073/pnas.1114515109

Li, W. (2017). How do SMA-linked mutations of SMN1 lead to structural/functionaldeficiency of the SMA protein?. PLoS ONE 12, e0178519. doi:10.1371/journal.pone.0178519

Li, B., Lin, M., Tang, Y., Wang, B. and Wang, J. H.-C. (2008a). A novel functionalassessment of the differentiation of micropatterned muscle cells. J. Biomech. 41,3349-3353. doi:10.1016/j.jbiomech.2008.09.025

Li, X.-J., Hu, B.-Y., Jones, S. A., Zhang, Y.-S., LaVaute, T., Du, Z.-W. and Zhang,S.-C. (2008b). Directed differentiation of ventral spinal progenitors and motorneurons from human embryonic stem cells by small molecules. Stem Cells 26,886-893. doi:10.1634/stemcells.2007-0620

Liang, R., Dong, W., Shen, X., Peng, X., Aceves, A. G. and Liu, Y. (2016).Modeling myotonic dystrophy 1 in C2C12 myoblast cells. J. Vis. Exp., e54078.doi:10.3791/54078

Lin, C.-Y., Yoshida, M., Li, L.-T., Ikenaka, A., Oshima, S., Nakagawa, K., Sakurai,H., Matsui, E., Nakahata, T. and Saito, M. K. (2019). iPSC-derived functionalhuman neuromuscular junctions model the pathophysiology of neuromusculardiseases. JCI Insight 4, e124299. doi:10.1172/jci.insight.124299

11

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 12: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

Lissouba, A., Liao, M., Kabashi, E. and Drapeau, P. (2018). Transcriptomicanalysis of zebrafish TDP-43 transgenic lines. Front. Mol. Neurosci. 11, 463.doi:10.3389/fnmol.2018.00463

Lloyd, T. E. and Taylor, J. P. (2010). Flightless flies: Drosophila models ofneuromuscular disease. Ann. N. Y. Acad. Sci. 1184, e1-e20. doi:10.1111/j.1749-6632.2010.05432.x

Madden, L., Juhas, M., Kraus, W. E., Truskey, G. A. and Bursac, N. (2015).Bioengineered human myobundles mimic clinical responses of skeletal muscle todrugs. eLife 4, e04885. doi:10.7554/eLife.04885

Madji Hounoum, B., Vourc’h, P., Felix, R., Corcia, P., Patin, F., Gueguinou, M.,Potier-Cartereau, M., Vandier, C., Raoul, C., Andres, C. R. et al. (2016). NSC-34 motor neuron-like cells are unsuitable as experimental model for glutamate-mediated excitotoxicity. Front. Cell. Neurosci. 10, 118. doi:10.3389/fncel.2016.00118

Maffioletti, S. M., Gerli, M. F. M., Ragazzi, M., Dastidar, S., Benedetti, S.,Loperfido, M., VandenDriessche, T., Chuah, M. K. and Tedesco, F. S. (2015).Efficient derivation and inducible differentiation of expandable skeletal myogeniccells from human ES and patient-specific iPS cells. Nat. Protoc. 10, 941-958.doi:10.1038/nprot.2015.057

Maier, O., Bohm, J., Dahm, M., Bruck, S., Beyer, C. and Johann, S. (2013).Differentiated NSC-34 motoneuron-like cells as experimental model forcholinergic neurodegeneration. Neurochem. Int. 62, 1029-1038. doi:10.1016/j.neuint.2013.03.008

Mamchaoui, K., Trollet, C., Bigot, A., Negroni, E., Chaouch, S., Wolff, A.,Kandalla, P. K., Marie, S., Di Santo, J., St Guily, J. L. et al. (2011). Immortalizedpathological human myoblasts: towards a universal tool for the study ofneuromuscular disorders. Skelet. Muscle 1, 34. doi:10.1186/2044-5040-1-34

Mann, C. J., Perdiguero, E., Kharraz, Y., Aguilar, S., Pessina, P., Serrano, A. L.and Mun oz-Canoves, P. (2011). Aberrant repair and fibrosis development inskeletal muscle. Skelet. Muscle 1, 21. doi:10.1186/2044-5040-1-21

Matusica, D., Fenech, M. P., Rogers, M.-L. and Rush, R. A. (2008).Characterization and use of the NSC-34 cell line for study of neurotrophinreceptor trafficking. J. Neurosci. Res. 86, 553-565. doi:10.1002/jnr.21507

Maurer, M., Toyka, K. V. and Gold, R. (2002). Immune mechanisms in acquireddemyelinating neuropathies: lessons from animal models. Neuromuscul. Disord.12, 405-414. doi:10.1016/S0960-8966(01)00302-9

McCain, M. L., Desplantez, T., Geisse, N. A., Rothen-Rutishauser, B., Oberer,H., Parker, K. K. and Kleber, A. G. (2012). Cell-to-cell coupling in engineeredpairs of rat ventricular cardiomyocytes: relation between Cx43immunofluorescence and intercellular electrical conductance. Am. J. Physiol.Heart Circ. Physiol. 302, H443-H450. doi:10.1152/ajpheart.01218.2010

McCorquodale, D., Pucillo, E. M. and Johnson, N. E. (2016). Management ofCharcot-Marie-Tooth disease: improving long-term care with a multidisciplinaryapproach. J. Multidiscip. Healthc. 9, 7-19. doi:10.2147/JMDH.S69979

McGovern, V. L., Iyer, C. C., Arnold, W. D., Gombash, S. E., Zaworski, P. G.,Blatnik, A. J., III, Foust, K. D. and Burghes, A. H. M. (2015). SMN expression isrequired in motor neurons to rescue electrophysiological deficits in the SMNΔ7mouse model of SMA. Hum. Mol. Genet. 24, 5524-5541. doi:10.1093/hmg/ddv283

McMahon, D. K., Anderson, P. A., Nassar, R., Bunting, J. B., Saba, Z., Oakeley,A. E. and Malouf, N. N. (1994). C2C12 cells: biophysical, biochemical, andimmunocytochemical properties. Am. J. Physiol. 266, C1795-C1802. doi:10.1152/ajpcell.1994.266.6.C1795

McWhorter, M. L., Monani, U. R., Burghes, A. H. M. and Beattie, C. E. (2003).Knockdown of the survival motor neuron (Smn) protein in zebrafish causesdefects in motor axon outgrowth and pathfinding. J. Cell Biol. 162, 919-931.doi:10.1083/jcb.200303168

Menconi, M., Gonnella, P., Petkova, V., Lecker, S. and Hasselgren, P.-O. (2008).Dexamethasone and corticosterone induce similar, but not identical, musclewasting responses in cultured L6 and C2C12 myotubes. J. Cell. Biochem. 105,353-364. doi:10.1002/jcb.21833

Mills, R. J., Parker, B. L., Monnot, P., Needham, E. J., Vivien, C. J., Ferguson, C.,Parton, R. G., James, D. E., Porrello, E. R. and Hudson, J. E. (2019).Development of a human skeletal micro muscle platform with pacing capabilities.Biomaterials 198, 217-227. doi:10.1016/j.biomaterials.2018.11.030

Morena, J., Gupta, A. and Hoyle, J. C. (2019). Charcot-marie-tooth: frommolecules to therapy. Int. J. Mol. Sci. 20, 3419. doi:10.3390/ijms20143419

Morimoto, Y., Kato-Negishi, M., Onoe, H. and Takeuchi, S. (2013). Three-dimensional neuron–muscle constructs with neuromuscular junctions.Biomaterials 34, 9413-9419. doi:10.1016/j.biomaterials.2013.08.062

Morrice, J. R., Gregory-Evans, C. Y. and Shaw, C. A. (2018). Animal models ofamyotrophic lateral sclerosis: A comparison of model validity. Neural Regener.Res. 13, 2050-2054. doi:10.4103/1673-5374.241445

Morris, T. A., Naik, J., Fibben, K. S., Kong, X., Kiyono, T., Yokomori, K. andGrosberg, A. (2020). Striated myocyte structural integrity: Automated analysis ofsarcomeric z-discs. PLoS Comput. Biol. 16, e1007676. doi:10.1371/journal.pcbi.1007676

Murdocca, M., Ciafre, S. A., Spitalieri, P., Talarico, R. V., Sanchez, M., Novelli, G.and Sangiuolo, F. (2016). SMA human iPSC-derived motor neurons show

perturbed differentiation and reduced miR-335-5p expression. Int. J. Mol. Sci. 17,1231. doi:10.3390/ijms17081231

Nair, R. R., Corrochano, S., Gasco, S., Tibbit, C., Thompson, D., Maduro, C., Ali,Z., Fratta, P., Arozena, A. A., Cunningham, T. J. et al. (2019). Uses forhumanised mouse models in precision medicine for neurodegenerative disease.Mamm. Genome 30, 173-191. doi:10.1007/s00335-019-09807-2

Nedachi, T., Fujita, H. and Kanzaki, M. (2008). Contractile C2C12 myotube modelfor studying exercise-inducible responses in skeletal muscle. Am. J. Physiol.Endocrinol. Metab. 295, E1191-E1204. doi:10.1152/ajpendo.90280.2008

Nesmith, A. P., Wagner, M. A., Pasqualini, F. S., O’Connor, B. B., Pincus, M. J.,August, P. R. and Parker, K. K. (2016). A human in vitro model of Duchennemuscular dystrophy muscle formation and contractility. J. Cell Biol. 215, 47-56.doi:10.1083/jcb.201603111

Neville, C., Rosenthal, N., McGrew,M., Bogdanova, N. andHauschka, S. (1997).Chapter 5 Skeletal muscle cultures. Methods Cell Biol. 52, 85-116. doi:10.1016/S0091-679X(08)60375-1

Nguyen, H. P., Van Broeckhoven, C. and van der Zee, J. (2018). ALS genes in thegenomic Era and their implications for FTD. Trends Genet. 34, 404-423. doi:10.1016/j.tig.2018.03.001

Novak, R., Ingram, M., Marquez, S., Das, D., Delahanty, A., Herland, A., Maoz,B. M., Jeanty, S. S. F., Somayaji, M. R., Burt, M. et al. (2020). Robotic fluidiccoupling and interrogation of multiple vascularized organ chips. Nat. Biomed.Eng. 4, 407-420. doi:10.1038/s41551-019-0497-x

Oberg, A. I., Dehvari, N. and Bengtsson, T. (2011). β-Adrenergic inhibition ofcontractility in L6 skeletal muscle cells. PLoS ONE 6, e22304. doi:10.1371/journal.pone.0022304

Oeda, T., Shimohama, S., Kitagawa, N., Kohno, R., Imura, T., Shibasaki, H. andIshii, N. (2001). Oxidative stress causes abnormal accumulation of familialamyotrophic lateral sclerosis-related mutant SOD1 in transgenic Caenorhabditiselegans. Hum. Mol. Genet. 10, 2013-2023. doi:10.1093/hmg/10.19.2013

Osaki, T., Uzel, S. G. M. and Kamm, R. D. (2018). Microphysiological 3D model ofamyotrophic lateral sclerosis (ALS) from human iPS-derived muscle cells andoptogenetic motor neurons. Sci. Adv. 4, eaat5847. doi:10.1126/sciadv.aat5847

Osaki, T., Uzel, S. G. M. and Kamm, R. D. (2020). On-chip 3D neuromuscularmodel for drug screening and precision medicine in neuromuscular disease. Nat.Protoc. 15, 421-449. doi:10.1038/s41596-019-0248-1

Palade, J., Pal, A., Rawls, A., Stabenfeldt, S. and Wilson-Rawls, J. (2019).Molecular analysis of muscle progenitor cells on extracellular matrix coatings andhydrogels. Acta Biomater. 97, 296-309. doi:10.1016/j.actbio.2019.08.019

Palchesko, R. N., Zhang, L., Sun, Y. and Feinberg, A. W. (2012). Development ofpolydimethylsiloxane substrates with tunable elastic modulus to study cellmechanobiology in muscle and nerve. PLoS ONE 7, e51499. doi:10.1371/journal.pone.0051499

Pasqualini, F. S., Agarwal, A., O’Connor, B. B., Liu, Q., Sheehy, S. P. andParker,K. K. (2018). Traction force microscopy of engineered cardiac tissues. PLoS ONE13, e0194706. doi:10.1371/journal.pone.0194706

Pei, W., Xu, L., Varshney, G. K., Carrington, B., Bishop, K., Jones, M. P., Huang,S. C., Idol, J., Pretorius, P. R., Beirl, A. et al. (2016). Additive reductions inzebrafish PRPS1 activity result in a spectrum of deficiencies modeling severalhuman PRPS1-associated diseases. Sci. Rep. 6, 29946. doi:10.1038/srep29946

Perry, S., Han, Y., Das, A. and Dickman, D. (2017). Homeostatic plasticity can beinduced and expressed to restore synaptic strength at neuromuscular junctionsundergoing ALS-related degeneration. Hum. Mol. Genet. 26, 4153-4167. doi:10.1093/hmg/ddx304

Phillips, W. D. and Vincent, A. (2016). Pathogenesis of myasthenia gravis: updateon disease types, models, and mechanisms. F1000Research 5, 1513. doi:10.12688/f1000research.8206.1

Pimentel, M. R., Falcone, S., Cadot, B. and Gomes, E. R. (2017). In vitrodifferentiation of mature myofibers for live imaging. J. Vis. Exp. e55141. doi:10.3791/55141

Pinto, S., Cunha, C., Barbosa, M., Vaz, A. R. and Brites, D. (2017). Exosomesfrom NSC-34 cells transfected with hSOD1-G93A are enriched in miR-124 anddrive alterations in microglia phenotype. Front. Neurosci. 11, 213. doi:10.3389/fnins.2017.00273

Pollot, B. E., Rathbone, C. R.,Wenke, J. C. andGuda, T. (2018). Natural polymerichydrogel evaluation for skeletal muscle tissue engineering. J. Biomed. Mater.Res. B Appl. Biomater. 106, 672-679. doi:10.1002/jbm.b.33859

Powell, C., Shansky, J., Del Tatto, M., Forman, D. E., Hennessey, J., Sullivan, K.,Zielinski, B. A. and Vandenburgh, H. H. (1999). Tissue-engineered humanbioartificial muscles expressing a foreign recombinant protein for gene therapy.Hum. Gene. Ther. 10, 565-577. doi:10.1089/10430349950018643

Powell, C. A., Smiley, B. L., Mills, J. and Vandenburgh, H. H. (2002). Mechanicalstimulation improves tissue-engineered human skeletal muscle. Am. J. Physiol.Cell Physiol. 283, C1557-C1565. doi:10.1152/ajpcell.00595.2001

Prinsen, F. M. C. and Veerkamp, H. J. (1998). Transfection of L6 myoblasts withadipocyte fatty acid-binding protein cDNA does not affect fatty acid uptake butdisturbs lipid metabolism and fusion. Biochem. J. 329, 265-273. doi:10.1042/bj3290265

Qin, D., Xia, Y. and Whitesides, G. M. (2010). Soft lithography for micro- andnanoscale patterning. Nat. Protoc. 5, 491-502. doi:10.1038/nprot.2009.234

12

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 13: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

Qu, Q., Li, D., Louis, K. R., Li, X., Yang, H., Sun, Q., Crandall, S. R., Tsang, S.,Zhou, J., Cox, C. L. et al. (2014). High-efficiencymotor neuron differentiation fromhuman pluripotent stem cells and the function of Islet-1. Nat. Commun. 5, 3449.doi:10.1038/ncomms4449

Ramon-Azcon, J., Ahadian, S., Estili, M., Liang, X., Ostrovidov, S., Kaji, H.,Shiku, H., Ramalingam, M., Nakajima, K., Sakka, Y. et al. (2013).Dielectrophoretically aligned carbon nanotubes to control electrical andmechanical properties of hydrogels to fabricate contractile muscle myofibers.Adv. Mater. 25, 4028-4034. doi:10.1002/adma.201301300

Rangarajan, S., Madden, L. and Bursac, N. (2014). Use of flow, electrical, andmechanical stimulation to promote engineering of striated muscles. Ann. Biomed.Eng. 42, 1391-1405. doi:10.1007/s10439-013-0966-4

Rao, N., Evans, S., Stewart, D., Spencer, K. H., Sheikh, F., Hui, E. E. andChristman, K. L. (2013). Fibroblasts influence muscle progenitor differentiationand alignment in contact independent and dependent manners in organized co-culture devices. Biomed. Microdevices 15, 161-169. doi:10.1007/s10544-012-9709-9

Rao, L., Qian, Y., Khodabukus, A., Ribar, T. and Bursac, N. (2018). Engineeringhuman pluripotent stem cells into a functional skeletal muscle tissue. Nat.Commun. 9, 126. doi:10.1038/s41467-017-02636-4

Ribeiro, A. J. S., Ang, Y.-S., Fu, J.-D., Rivas, R. N., Mohamed, T. M. A., Higgs,G. C., Srivastava, D. and Pruitt, B. L. (2015). Contractility of singlecardiomyocytes differentiated from pluripotent stem cells depends onphysiological shape and substrate stiffness. Proc. Natl. Acad. Sci. USA 112,12705-12710. doi:10.1073/pnas.1508073112

Ripps, M. E., Huntley, G. W., Hof, P. R., Morrison, J. H. and Gordon, J. W. (1995).Transgenic mice expressing an altered murine superoxide dismutase geneprovide an animal model of amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci.USA 92, 689-693. doi:10.1073/pnas.92.3.689

Robinson, M. M., Sather, B. K., Burney, E. R., Ehrlicher, S. E., Stierwalt, H. D.,Franco, M. C. and Newsom, S. A. (2019). Robust intrinsic differences inmitochondrial respiration and H2O2 emission between L6 and C2C12 cells.Am. J. Physiol. Cell Physiol. 317, C339-C347. doi:10.1152/ajpcell.00343.2018

⍰ahin, A., Held, A., Bredvik, K., Major, P., Achilli, T.-M., Kerson, A. G., Wharton,K., Stilwell, G. and Reenan, R. (2017). Human SOD1 ALS mutations in aDrosophila knock-in model cause severe phenotypes and reveal dosage-sensitive gain- and loss-of-function components. Genetics 205, 707-723.doi:10.1534/genetics.116.190850

Saini, J., Faroni, A., Abd Al Samid, M., Reid, A. J., Lightfoot, A. P., Mamchaoui,K., Mouly, V., Butler-Browne, G., McPhee, J. S., Degens, H. et al. (2019).Simplified in vitro engineering of neuromuscular junctions between rat embryonicmotoneurons and immortalized human skeletal muscle cells. Stem Cells CloningAdv. Appl. 12, 1-9. doi:10.2147/SCCAA.S187655

Salani, S., Donadoni, C., Rizzo, F., Bresolin, N., Comi, G. P. and Corti, S. (2012).Generation of skeletal muscle cells from embryonic and induced pluripotent stemcells as an in vitro model and for therapy of muscular dystrophies. J. Cell. Mol.Med. 16, 1353-1364. doi:10.1111/j.1582-4934.2011.01498.x

Sances, S., Bruijn, L. I., Chandran, S., Eggan, K., Ho, R., Klim, J. R., Livesey,M. R., Lowry, E., Macklis, J. D., Rushton, D. et al. (2016). Modeling ALS withmotor neurons derived from human induced pluripotent stem cells. Nat. Neurosci.19, 542-553. doi:10.1038/nn.4273

Santhanam, N., Kumanchik, L., Guo, X., Sommerhage, F., Cai, Y., Jackson, M.,Martin, C., Saad, G., McAleer, C. W., Wang, Y. et al. (2018). Stem cell derivedphenotypic human neuromuscular junction model for dose response evaluation oftherapeutics. Biomaterials 166, 64-78. doi:10.1016/j.biomaterials.2018.02.047

Santosa, K. B., Keane, A. M., Jablonka-Shariff, A., Vannucci, B. and Snyder-Warwick, A. K. (2018). Clinical relevance of terminal Schwann cells: anoverlooked component of the neuromuscular junction. J. Neurosci. Res. 96,1125-1135. doi:10.1002/jnr.24231

Saporta, A. S. D., Sottile, S. L., Miller, L. J., Feely, S. M. E., Siskind, C. E. andShy, M. E. (2011). Charcot-Marie-Tooth disease subtypes and genetic testingstrategies. Ann. Neurol. 69, 22-33. doi:10.1002/ana.22166

Sareen, D., O’Rourke, J. G., Meera, P., Muhammad, A. K. M. G., Grant, S.,Simpkinson, M., Bell, S., Carmona, S., Ornelas, L., Sahabian, A. et al. (2013).Targeting RNA foci in iPSC-derived motor neurons from ALS patients with aC9ORF72 repeat expansion. Sci. Transl. Med. 5, 208ra149. doi:10.1126/scitranslmed.3007529

Selvaraj, S., Mondragon-Gonzalez, R., Xu, B., Magli, A., Kim, H., Laine, J., Kiley,J., McKee, H., Rinaldi, F., Aho, J. et al. (2019). Screening identifies smallmolecules that enhance the maturation of human pluripotent stem cell-derivedmyotubes. eLife 8, e47970, doi:10.7554/eLife.47970

Seminary, E. R., Sison, S. L. andEbert, A. D. (2018). Modeling protein aggregationand the heat shock response in ALS iPSC-derived motor neurons. Front.Neurosci. 12, 86. doi:10.3389/fnins.2018.00086

Shansky, J., Del Tatto, M., Chromiak, J. and Vandenburgh, H. (1997). Asimplifiedmethod for tissue engineering skeletal muscle organoids in vitro. In VitroCell. Dev. Biol. Anim. 33, 659-661. doi:10.1007/s11626-997-0118-y

Shaw, M. P., Higginbottom, A., McGown, A., Castelli, L. M., James, E.,Hautbergue, G. M., Shaw, P. J. and Ramesh, T. M. (2018). Stable transgenicC9orf72 zebrafish model key aspects of the ALS/FTD phenotype and reveal novel

pathological features. Acta Neuropathol. Commun. 6, 125. doi:10.1186/s40478-018-0629-7

Shelton, M., Kocharyan, A., Liu, J., Skerjanc, I. S. and Stanford, W. L. (2016).Robust generation and expansion of skeletal muscle progenitors and myocytesfrom human pluripotent stem cells. Methods 101, 73-84. doi:10.1016/j.ymeth.2015.09.019

Shi, Y., Lin, S., Staats, K. A., Li, Y., Chang, W.-H., Hung, S.-T., Hendricks, E.,Linares, G. R., Wang, Y., Son, E. Y. et al. (2018). Haploinsufficiency leads toneurodegeneration in C9ORF72 ALS/FTD human induced motor neurons. Nat.Med. 24, 313. doi:10.1038/nm.4490

Shimizu, K., Araki, H., Sakata, K., Tonomura, W., Hashida, M. and Konishi, S.(2015). Microfluidic devices for construction of contractile skeletal musclemicrotissues. J. Biosci. Bioeng. 119, 212-216. doi:10.1016/j.jbiosc.2014.07.003

Shimojo, D., Onodera, K., Doi-Torii, Y., Ishihara, Y., Hattori, C., Miwa, Y.,Tanaka, S., Okada, R., Ohyama, M., Shoji, M. et al. (2015). Rapid, efficient, andsimple motor neuron differentiation from human pluripotent stem cells. Mol. Brain8, 79. doi:10.1186/s13041-015-0172-4

Sincennes, M. C., Wang, Y. X. and Rudnicki, M. A. (2017). Primary mousemyoblast purification using magnetic cell separation. Methods Mol. Biol. 1556,41-50. doi:10.1007/978-1-4939-6771-1_3

Skoglund, G., Laine, J., Darabi, R., Fournier, E., Perlingeiro, R. and Tabti, N.(2014). Physiological and ultrastructural features of human induced pluripotentand embryonic stem cell-derived skeletal myocytes in vitro. Proc. Natl. Acad. Sci.USA 111, 8275-8280. doi:10.1073/pnas.1322258111

Sleigh, J. N. and Sattelle, D. B. (2010). C. elegans models of neuromusculardiseases expedite translational research. Transl. Neurosci. 1, 214-227. doi:10.2478/v10134-010-0032-9

Sleigh, J. N., Grice, S. J., Burgess, R. W., Talbot, K. and Cader, M. Z. (2013).Neuromuscular junction maturation defects precede impaired lower motor neuronconnectivity in Charcot-Marie-Tooth type 2D mice. Hum. Mol. Genet. 23,2639-2650. doi:10.1093/hmg/ddt659

Smith, A. S., Long, C. J., McAleer, C., Bobbitt, N., Srinivasan, B. and Hickman,J. J. (2014a). Utilization of microscale silicon cantilevers to assess cellularcontractile function in vitro. J. Vis. Exp. e51866.

Smith, A. S. T., Long, C. J., Pirozzi, K., Najjar, S., McAleer, C., Vandenburgh,H. H. and Hickman, J. J. (2014b). A multiplexed chip-based assay system forinvestigating the functional development of human skeletal myotubes in vitro.J. Biotechnol. 185, 15-18. doi:10.1016/j.jbiotec.2014.05.029

Smith, A. S. T., Passey, S. L., Martin, N. R. W., Player, D. J., Mudera, V.,Greensmith, L. and Lewis, M. P. (2016). Creating interactions between tissue-engineered skeletal muscle and the peripheral nervous system. Cells TissuesOrgans 202, 143-158. doi:10.1159/000443634

Son, E. Y., Ichida, J. K., Wainger, B. J., Toma, J. S., Rafuse, V. F.,Woolf, C. J. andEggan, K. (2011). Conversion of mouse and human fibroblasts into functionalspinal motor neurons. Cell Stem Cell 9, 205-218. doi:10.1016/j.stem.2011.07.014

Soriano-Arroquia, A., Clegg, P. D., Molloy, A. P. and Goljanek-Whysall, K.(2017). Preparation and culture of myogenic precursor cells/primary myoblastsfrom skeletal muscle of adult and aged humans. J. Vis. Exp. 55047. doi:10.3791/51866

Spaulding, E. L., Sleigh, J. N., Morelli, K. H., Pinter, M. J., Burgess, R. W. andSeburn, K. L. (2016). Synaptic deficits at neuromuscular junctions in two mousemodels of charcot–marie–tooth type 2d. J. Neurosci. 36, 3254-3267. doi:10.1523/JNEUROSCI.1762-15.2016

Spinazzola, J. M. and Gussoni, E. (2017). Isolation of primary human skeletalmuscle cells. Bio. Protoc. 7. doi:10.21769/BioProtoc.2591

Spring, A. M., Raimer, A. C., Hamilton, C. D., Schillinger, M. J. and Matera, A. G.(2019). Comprehensive modeling of spinal muscular atrophy in Drosophilamelanogaster. Front. Mol. Neurosci. 12, 113. doi:10.3389/fnmol.2019.00113

Steinbeck, J. A., Jaiswal, M. K., Calder, E. L., Kishinevsky, S., Weishaupt, A.,Toyka, K. V., Goldstein, P. A. and Studer, L. (2016). Functional connectivityunder optogenetic control allows modeling of human neuromuscular disease.CellStem Cell 18, 134-143. doi:10.1016/j.stem.2015.10.002

Su, F.-C., Goutman, S. A., Chernyak, S., Mukherjee, B., Callaghan, B. C.,Batterman, S. and Feldman, E. L. (2016). Association of environmental toxinswith amyotrophic lateral sclerosis. JAMA Neurol. 73, 803-811. doi:10.1001/jamaneurol.2016.0594

Suh, G. C., Bettadapur, A., Santoso, J.W. andMcCain, M. L. (2017). Fabrication ofmicromolded gelatin hydrogels for long-term culture of aligned skeletal myotubes.Methods Mol. Biol. 1668, 147-163. doi:10.1007/978-1-4939-7283-8_11

Sun, Y., Duffy, R., Lee, A. and Feinberg, A.W. (2013). Optimizing the structure andcontractility of engineered skeletal muscle thin films.Acta Biomater. 9, 7885-7894.doi:10.1016/j.actbio.2013.04.036

Sun, M., Sun, X., Wang, Z., Guo, S., Yu, G. and Yang, H. (2018). Synthesis andproperties of Gelatin Methacryloyl (GelMA) hydrogels and their recentapplications in load-bearing tissue. Polymers 10, 1290. doi:10.3390/polym10111290

Taylor, A. M., Rhee, S. W., Tu, C. H., Cribbs, D. H., Cotman, C. W. and Jeon, N. L.(2003). Microfluidic multicompartment device for neuroscience research.Langmuir 19, 1551-1556. doi:10.1021/la026417v

13

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms

Page 14: Neuromuscular disease modeling on a chip · Neuromuscular disease modeling on a chip Jeffrey W. Santoso1 and Megan L. McCain1,2,* ABSTRACT Organs-on-chips are broadly defined as microfabricated

Thonhoff, J. R., Simpson, E. P. and Appel, S. H. (2018). Neuroinflammatorymechanisms in amyotrophic lateral sclerosis pathogenesis. Curr. Opin. Neurol.31, 635-639. doi:10.1097/WCO.0000000000000599

Turner, M. R., Barnwell, J., Al-Chalabi, A. and Eisen, A. (2012). Young-onsetamyotrophic lateral sclerosis: historical and other observations. Brain 135,2883-2891. doi:10.1093/brain/aws144

Umbach, J. A., Adams, K. L., Gundersen, C. B. and Novitch, B. G. (2012).Functional neuromuscular junctions formed by embryonic stem cell-derivedmotorneurons. PLoS ONE 7, e36049. doi:10.1371/journal.pone.0036049

Urja, V., Khaire, K., Balakrishnan, S. and Uggini, G. K. (2018). Chick embryoniccells as a source for generating in vitro model of muscle cell dystrophy. In VitroCell. Dev. Biol. Anim. 54, 756-769. doi:10.1007/s11626-018-0297-8

Uzel, S. G. M., Platt, R. J., Subramanian, V., Pearl, T. M., Rowlands, C. J., Chan,V., Boyer, L. A., So, P. T. C. and Kamm, R. D. (2016). Microfluidic device for theformation of optically excitable, three-dimensional, compartmentalized motorunits. Sci. Adv. 2, e1501429. doi:10.1126/sciadv.1501429

Vallette, F. M., Vigny, M. and Massoulie, J. (1986). Muscular differentiation ofchicken myotubes in a simple defined synthetic culture medium and in serumsupplemented media: expression of the molecular forms of acetylcholinesterase.Neurochem. Int. 8, 121-133. doi:10.1016/0197-0186(86)90109-9

van der Wal, E., Herrero-Hernandez, P., Wan, R., Broeders, M., In ‘t Groen,S. L. M., van Gestel, T. J. M., van Ijcken, W. F. J., Cheung, T. H., van der Ploeg,A. T., Schaaf, G. J. et al. (2018). Large-scale expansion of human iPSC-derivedskeletal muscle cells for disease modeling and cell-based therapeutic strategies.Stem Cell Rep. 10, 1975-1990. doi:10.1016/j.stemcr.2018.04.002

Vaughan, M. and Lamia, K. A. (2019). Isolation and differentiation of primarymyoblasts from mouse skeletal muscle explants. J. Vis. Exp. e60310. doi:10.3791/60310

Wang, J., Farr, G. W., Hall, D. H., Li, F., Furtak, K., Dreier, L. and Horwich, A. L.(2009). An ALS-linkedmutant SOD1 produces a locomotor defect associated withaggregation and synaptic dysfunction when expressed in neurons ofCaenorhabditis elegans. PLoS Genet. 5, e1000350. doi:10.1371/journal.pgen.1000350

Wang, P.-Y., Thissen, H. and Tsai, W.-B. (2012). The roles of RGD and groovedtopography in the adhesion, morphology, and differentiation of C2C12 skeletalmyoblasts. Biotechnol. Bioeng. 109, 2104-2115. doi:10.1002/bit.24452

Wang, L., Shansky, J. and Vandenburgh, H. (2013). Induced formation andmaturation of acetylcholine receptor clusters in a defined 3D bio-artificial muscle.Mol. Neurobiol. 48, 397-403. doi:10.1007/s12035-013-8412-z

Wang, Z., Volinsky, A. A. and Gallant, N. D. (2014). Crosslinking effect onpolydimethylsiloxane elastic modulus measured by custom-built compressioninstrument. J. Appl. Polym. Sci. 131. doi:10.1002/app.41050

Watson, M. R., Lagow, R. D., Xu, K., Zhang, B. and Bonini, N. M. (2008). Adrosophila model for amyotrophic lateral sclerosis reveals motor neuron damageby humanSOD1. J. Biol. Chem. 283, 24972-24981. doi:10.1074/jbc.M804817200

Wilson, K., Das, M., Wahl, K. J., Colton, R. J. and Hickman, J. (2010).Measurement of contractile stress generated by cultured rat muscle on silicon

cantilevers for toxin detection andmuscle performance enhancement.PLoSONE5, e11042. doi:10.1371/journal.pone.0011042

Xi, H., Fujiwara, W., Gonzalez, K., Jan, M., Liebscher, S., Van Handel, B.,Schenke-Layland, K. and Pyle, A. D. (2017). In vivo human somitogenesisguides somite development from hPSCs. Cell Rep. 18, 1573-1585. doi:10.1016/j.celrep.2017.01.040

Xu, Z., Poidevin, M., Li, X., Li, Y., Shu, L., Nelson, D. L., Li, H., Hales, C. M.,Gearing, M., Wingo, T. S. et al. (2013). Expanded GGGGCC repeat RNAassociated with amyotrophic lateral sclerosis and frontotemporal dementiacauses neurodegeneration. Proc. Natl. Acad. Sci. USA 110, 7778-7783. doi:10.1073/pnas.1219643110

Yaffe, D. (1968). Retention of differentiation potentialities during prolongedcultivation of myogenic cells. Proc. Natl. Acad. Sci. USA 61, 477-483. doi:10.1073/pnas.61.2.477

Yaffe, D. and Saxel, O. (1977). Serial passaging and differentiation of myogeniccells isolated from dystrophic mouse muscle. Nature 270, 725-727. doi:10.1038/270725a0

Yang, H. S., Ieronimakis, N., Tsui, J. H., Kim, H. N., Suh, K.-Y., Reyes, M. andKim, D.-H. (2014). Nanopatterned muscle cell patches for enhanced myogenesisand dystrophin expression in a mouse model of muscular dystrophy. Biomaterials35, 1478-1486. doi:10.1016/j.biomaterials.2013.10.067

Young, C. S., Hicks, M. R., Ermolova, N. V., Nakano, H., Jan, M., Younesi, S.,Karumbayaram, S., Kumagai-Cresse, C., Wang, D., Zack, J. A. et al. (2016). Asingle CRISPR-Cas9 deletion strategy that targets the majority of DMD patientsrestores dystrophin function in hiPSC-derived muscle cells. Cell Stem Cell 18,533-540. doi:10.1016/j.stem.2016.01.021

Zhang, B., Tu, P., Abtahian, F., Trojanowski, J. Q. and Lee, V. M.-Y. (1997).Neurofilaments and orthograde transport are reduced in ventral root axons oftransgenic mice that express human SOD1 with a G93A mutation. J. Cell Biol.139, 1307-1315. doi:10.1083/jcb.139.5.1307

Zhang, X., Hong, S., Yen, R., Kondash, M., Fernandez, C. E. and Truskey, G. A.(2018). A system to monitor statin-induced myopathy in individual engineeredskeletal muscle myobundles. Lab. Chip 18, 2787-2796. doi:10.1039/C8LC00654G

Zhao, Q., Yang, S. T., Wang, J. J., Zhou, J., Xing, S. S., Shen, C. C., Wang, X. X.,Yue, Y. X., Song, J., Chen, M. et al. (2015). TNF alpha inhibits myogenicdifferentiation of C2C12 cells through NF-κB activation and impairment of IGF-1signaling pathway.Biochem. Biophys. Res. Commun. 458, 790-795. doi:10.1016/j.bbrc.2015.02.026

Ziemkiewicz, N., Talovic, M., Madsen, J., Hill, L., Scheidt, R., Patel, A., Haas, G.,Marcinczyk, M., Zustiak, S. P. and Garg, K. (2018). Laminin-111 functionalizedpolyethylene glycol hydrogels support myogenic activity in vitro. Biomed. Mater.13, 065007. doi:10.1088/1748-605X/aad915

Zuroske, T. (2019). Upcoming market catalysts in Q2 2019. Nat. Rev. Drug Discov.18, 244. doi:10.1038/d41573-019-00048-1

14

REVIEW Disease Models & Mechanisms (2020) 13, dmm044867. doi:10.1242/dmm.044867

Disea

seModels&Mechan

isms