Transcript
Page 1: Recent advances using zebrafish animal models for muscle disease drug discovery

1. Introduction

2. Zebrafish as a model for muscle

development and disease

3. Pharmacological therapies for

DMD studied in zebrafish

4. Additional zebrafish muscle

disease models and

pharmacological studies

5. Analyses of zebrafish muscle

disease and assessment of

pharmacological rescue

6. Using zebrafish to study

pharmacological approaches

to muscle disease gene

modification

7. Conclusion

8. Expert opinion

Review

Recent advances using zebrafishanimal models for muscle diseasedrug discoveryLisa MavesCenter for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute,

and Department of Pediatrics, University of Washington, Seattle, WA, USA

Introduction: Animal models have enabled great progress in the discovery

and understanding of pharmacological approaches for treating muscle

diseases like Duchenne muscular dystrophy.

Areas covered: With this article, the author provides the reader with a

description of the zebrafish animal model, which has been employed to

identify and study pharmacological approaches to muscle disease. In particu-

lar, the author focuses on how both large-scale chemical screens and targeted

drug treatment studies have established zebrafish as an important model for

muscle disease drug discovery.

Expert opinion: There are a number of opportunities arising for the use of

zebrafish models for further developing pharmacological approaches to

muscle diseases, including studying drug combination therapies and utilizing

genome editing to engineer zebrafish muscle disease models. It is the author’s

particular belief that the availability of a wide range of zebrafish transgenic

strains for labeling immune cell types, combined with live imaging and

drug treatment of muscle disease models, should allow for new elegant

studies demonstrating how pharmacological approaches might influence

inflammation and the immune response in muscle disease.

Keywords: birefringence, drug screen, Duchenne muscular dystrophy, muscle function,

muscle structure, myopathy, zebrafish

Expert Opin. Drug Discov. (2014) 9(9):1033-1045

1. Introduction

Myopathies are muscle diseases in which dysfunction of muscle fibers results inmuscle weakness. The diversity of inherited myopathies, which include the muscu-lar dystrophies, is reflected in the diversity of genetic mutations that are implicatedin these diseases [1]. No cures are currently available for inherited myopathies.Gaining increased understanding of the cellular and molecular mechanisms under-lying the muscle defects seen in inherited myopathies will help provide insight intopotential therapies [2,3]. Here, I review how the zebrafish animal model hasbeen employed to both identify, and study the mechanisms of, pharmacologicalapproaches to muscle disease therapies.

Many animal models, including mice, dogs, pigs, fruit flies, worms and zebra-fish, have contributed to our understanding of the genetic basis of, and molecularand cellular mechanisms behind, inherited muscle diseases [4-9]. In particular, onemuscle disease that is well characterized in animal models is Duchenne musculardystrophy (DMD). DMD is the most common and severe form of musculardystrophy, affecting about 1 in 3500 males, and is caused by mutations in theX-linked DMD gene, which encodes dystrophin [10]. Dystrophin contributes to

10.1517/17460441.2014.927435 © 2014 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X 1033All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 2: Recent advances using zebrafish animal models for muscle disease drug discovery

the multi-protein dystrophin--glycoprotein complex (DGC),also known as the dystrophin-associated protein complex,which links the extracellular matrix with the cytoskeletonand is critical for muscle cell membrane stability and cellsignaling [2,11]. Many types of muscular dystrophy are causedby mutations in genes that encode components of theDGC [2,11]. Muscular dystrophies are characterized by pro-gressive muscle degeneration and progressive loss of musclefunction. Mechanisms that contribute to muscle degenera-tion in the muscular dystrophies include muscle membraneinstability, disrupted calcium homeostasis and oxidativestress [2,11]. The congenital myopathies (sometimes alsoreferred to as ‘inherited myopathies’), such as nemalinemyopathy, are characterized by muscle weakness but do notusually show progressive muscle degeneration. Congenitalmyopathies typically show characteristic structural defects inthe contractile apparatus that lead to reduced myofiber con-tractile function [5]. Disruption of excitation--contractioncoupling appears to be a common pathological mechanismin the congenital myopathies [3,5].Animal models have also contributed to the development of

therapeutic approaches for muscle diseases. For DMD inparticular, gene therapy and stem cell-mediated therapeuticstrategies hold tremendous promise, but these approaches stillface many obstacles [12,13]. Therefore, many different pharma-cological therapies are currently being pursued [14-17]. Oneadvantage of pharmacological therapy is that systemicallydelivered drugs could reach all muscle groups, including theheart, which undergoes cardiomyopathy in DMD patients[13-15]. Animal models from fruit flies [18] to dogs [19] havehelped advance our knowledge of potential pharmacologicaltherapies for DMD. The zebrafish animal model, in particular,offers many advantages for drug discovery and for understand-ing drug mechanisms of action not only for DMD therapiesbut for therapies for other muscle diseases as well.

2. Zebrafish as a model for muscledevelopment and disease

2.1 Zebrafish model organism advantagesThe zebrafish, Danio rerio, offers several advantages as amodel organism for human disease and drug discovery [20].Zebrafish can be produced readily in large numbers suchthat hundreds of embryos can be obtained in a single day.The embryos are transparent and develop rapidly outside ofthe mother, allowing the earliest stages of development to beexamined. Also, zebrafish are readily manipulated by bothgenetic and chemical approaches (Figure 1). I will discussgenetic manipulations below (Sections 2.3 and 2.4). Forchemical approaches, zebrafish embryos raised in a petri dishcan readily absorb drugs that are simply added to the embryobath (Figure 2) [21]. Embryos can be raised in 96- or 384-wellplates, allowing for high-throughput chemical screening (Fig-ure 1) [21]. Significantly, drugs identified through screening inthe zebrafish have led to clinical trials [22,23], underscoring therelevance of pharmacological studies in zebrafish to humandisease. In addition to these general advantages, there are sev-eral additional characteristics that have made zebrafish an out-standing model for human muscle disease, as I describe below.

2.2 Zebrafish skeletal muscle features and motor

behaviorsOne advantage of studying zebrafish muscle is that develop-ment of the main body musculature is rapid, such that by24 h post-fertilization, all 30 segmental blocks of muscle, ormyotomes, are present and easily visible along the trunk andtail of the embryo. The transparency of the zebrafish embryohas facilitated cellular observations of early muscle cell line-ages, cellular migrations and muscle fiber morphogenesis[24-26]. Many of the molecular events of skeletal muscle devel-opment and differentiation are conserved between zebrafishand mammals [25,26]. Structurally, zebrafish skeletal muscle isvery similar to human muscle, and zebrafish share the samemuscle disease genes with humans, including genes encodingcomponents of the sarcomere, the DGC and excitation--contraction coupling [6,27-32]; (see Section 2.4 Zebrafish muscledisease models).

The mechanisms of skeletal muscle repair and regenerationare similar in zebrafish and mammals. Defective muscle mem-brane repair can lead to myopathy, and both zebrafish andmammals share requirements for dysferlin and associatedproteins such as annexins for sarcolemmal repair [33,34].Pax7-expressing skeletal muscle stem cells, or satellite cells,are required for mammalian skeletal muscle regeneration [35].Zebrafish have Pax7-expressing cells that migrate into areas ofmuscle damage [36]. Whether these Pax7-expressing cells func-tion as true ‘satellite cells’ and contribute to new muscle fibersduring zebrafish muscle regeneration has not yet been defini-tively shown [37]. However, zebrafish muscle does appear toregenerate from proliferative stem cell-like cells and not

Article highlights.

. The zebrafish animal model offers many advantages fordrug discovery and for understanding drug mechanismsof action for muscle disease therapies.

. Zebrafish has served as a vertebrate high-throughputscreening platform for new drug therapies for Duchennemuscular dystrophy.

. Pharmacological studies in zebrafish muscle diseasemodels have helped provide new insights intomechanisms of drug action, therapeutic targets anddisease pathogenesis.

. Zebrafish is an exceptional animal model for the rangeof extremely simple to sophisticated approaches forassessing muscle structure and function.

. The use of new approaches, such as genome editingtechnologies, will further enhance the use of zebrafishfor muscle disease drug discovery.

This box summarizes key points contained in the article.

L. Maves

1034 Expert Opin. Drug Discov. (2014) 9(9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 3: Recent advances using zebrafish animal models for muscle disease drug discovery

from de-differentiated fibers [37]. These studies help supportthe idea that investigating the mechanisms of zebrafish muscleresponse to injury and disease will inform our understandingof human muscle.

Early motor behaviors during zebrafish embryonic andlarval stages have been well characterized and can be easilyobserved and measured [38-41]. The first movements begin atabout 17 h post-fertilization as spontaneous contractions.

Forward geneticscreening

Injectmorpholinos

InjectCRISPR/Cas9

mRNAs

Gene knock-down

dmd+/+

dmd-/-

Genome editing

Zebrafishmuscle disease

models

In vivo imaging

High-throughputdrug screening

Time-lapse larval movement/muscle functions studies

Figure 1. Zebrafish offer many advantages as an animal model for muscle diseases. There are several ways to generate

zebrafish muscle disease models, including using forward genetic screens and birefringence, injecting antisense morpholinos

into zebrafish one-cell embryos to cause targeted gene knock-down, and injecting CRISPR/Cas9 mRNAs to induce targeted

gene editing. Zebrafish muscle disease models, such as dmd mutants, can show disruptions in the muscle birefringence

pattern (center figure). Zebrafish muscle disease models can be used in high-throughput drug screens to identify compounds

that ameliorate the muscle disease phenotype, for in vivo muscle fiber imaging for muscle structure analyses, and for muscle

function analyses, such as time-lapse tracking of larval movements in petri dishes.DMD: Duchenne muscular dystrophy.

4 dayspost fertilization

1 day post fertilization

+DMSO

Birefringence

Phalloidin

+drug

Zebrafish embryosfrom dmd+/-X dmd+/-cross

Zebrafish embryosfrom dmd+/-X dmd+/-cross

3 days incubation

dmd+/+ or dmd+/- dmd+/+ or dmd+/-dmd-/- dmd-/-

Figure 2. Example of a strategy for testingpharmacological rescue of a zebrafishmuscle diseasemodel. In this example, embryos

are collected from dmd+/- parents and raised in awater bath in petri dishes. Drugs, and vehicle control such as dimethyl sulfoxide

(DMSO) can be added to the water bath at any time, typically at 1 day post-fertilization [54,55,59,62]. Following incubation, larvae

can be scored formuscle lesions using birefringence (shown in lowmagnification views; [39,54]) ormuscle stains such as phalloidin

(larval trunkmyotomes shown inhighermagnificationviews; [62,73]).At4dayspost-fertilization, about25%of larvae fromDMSO-

control treated dmd+/-crosses show disruptions in the muscle fiber pattern, whereas lower frequencies of larvae with muscle

defects appear upon treatment with drugs that improve the dmd phenotype (Table 1). Arrows point to larvae with disrupted

birefringence pattern. Phalloidin images were previously published in [62] and appear with permission of PLOS Currents.DMD: Duchenne muscular dystrophy.

Recent advances using zebrafish animal models for muscle disease drug discovery

Expert Opin. Drug Discov. (2014) 9(9) 1035

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 4: Recent advances using zebrafish animal models for muscle disease drug discovery

At about 24 h, embryos will coil in response to touch. Atabout 26 h, embryos exhibit swimming movements inresponse to touch, and by 96 h the larva is freely swimming.These stereotyped movements have formed the basis forgenetic screens for zebrafish mutant strains with muscledefects, as I describe next, and for characterization of zebrafishmuscle disease models (see Section 5.2 Assessing musclefunction).

2.3 Zebrafish genetic screens for muscle defectsThe rapid development of zebrafish and the ability to readilyobtain large numbers of animals have facilitated forwardgenetic screens that have significantly contributed to the useof zebrafish as an animal model for human muscle disease.Some of the earliest genetic screens in zebrafish usedembryonic motility to identify loci required for muscle func-tion [39,42]. The stereotypic touch-evoked escape response inzebrafish larvae has formed the basis of several genetic screensin an effort to identify genes involved in motor control ofbehavior [39,40,43,44]. Additional zebrafish forward geneticscreens have utilized muscle birefringence, a measure of skele-tal muscle structural integrity (see Section 5.1 Assessing mus-cle structure; Figure 1), to identify genes relevant to skeletalmuscle disease [40,45]. Many of the genes responsible for thephenotypes in the mutant strains recovered from these screenshave been identified, and most are orthologs of humanmuscular dystrophy and myopathy genes [28,30,45-48].

2.4 Zebrafish muscle disease modelsThese genetic screens and the resulting mutant zebrafish strainshave thus provided a wealth of zebrafish models of humanmuscle diseases. Additional approaches to generating zebrafishmodels of humanmuscle diseases have been the use of antisensemorpholinos (Figure 1) or, in cases where the human diseaseresults from a dominant mutation or misexpression, transgeniczebrafish or transient overexpression in zebrafish embryos.Recent reviews provide tables with comprehensive lists ofknown zebrafish models of human muscle diseases [6,32,49].Genome editing technology now opens up new approaches togenerating zebrafish muscle disease models, as I will discussfurther below (Section 8 Expert opinion; Figure 1). The identi-fication of zebrafish mutant strains for human muscle diseasegenes has revealed that zebrafish represent excellent models ofhuman myopathies [32,50]. For example, zebrafish dmd mutantsshow severe muscle pathology and motor defects by 4 days anddie by about 30 days post-fertilization, while mouse DMD(mdx) mutants are viable and have only mild muscle defects[28,50,51].The identification of zebrafish models of human muscle

diseases has provided opportunities to use these models fordirectly testing genetic and pharmacological approaches forameliorating the skeletal muscle defects associated with eachdisease. For example, studies of zebrafish acetylcholinesterase(ache) mutants and acetylcholine (ACh) receptor (nic1 andsop) mutants were among the earliest to show that zebrafish

models can be employed for investigating genetic and phar-macological suppression of myopathies [43,52,53]. ACh is themajor neurotransmitter in the nervous system. ACh bindspost-synaptic ACh receptors (AChR) on muscle cells and isdegraded by acetylcholinesterase (AChE). Zebrafish achemutant embryos show progressive motor and muscle struc-tural defects [43]. The ache mutant muscle phenotype can besuppressed by loss of the nic1 AChR or by treating acheembryos with the drug eserine, suggesting that eserine actsnot only as a known AChE inhibitor but also an antagonistfor the AChR [43,52]. Additional studies showed that the sopAChR mutation could suppress the myopathy observed inzebrafish dmd mutants [53]. Taken together, these studiesshowed that zebrafish can serve as a screening system for drugsthat can suppress myopathies and also showed that modifiersof the zebrafish dmd mutant phenotype could be discoveredand readily examined in easily accessible young animals.

3. Pharmacological therapies for DMDstudied in zebrafish

Recent pharmacological approaches, including large-scaledrug screens as well as targeted compound tests, that havedemonstrated the rescue of the zebrafish dmd mutant arereviewed below. These examples illustrate how zebrafishembryos can serve as a vertebrate high-throughput screeningplatform for new drug therapies for DMD. These examplesalso illustrate how zebrafish can be used to examine the mech-anism of action of specific drugs and bridge the gap betweencell culture and mammalian models. Table 1 provides a listof these drug studies in zebrafish dmd models, and Figure 2

illustrates a drug treatment strategy.

3.1 Large-scale drug screensTo establish zebrafish as a significant vertebrate model foridentifying pharmacological approaches for DMD, the Kun-kel laboratory has performed large-scale drug screens for smallmolecules capable of suppressing the zebrafish dystrophin-null(dmd, also referred to as sapje) mutant phenotype [54,55]. Inzebrafish, the dmd gene is not sex chromosome linked, as inhumans and mice, and shows autosomal recessive inheri-tance [31]. With the zebrafish dmd mutant strain, about 25%of the larvae from a cross of heterozygote carriers show thedmd muscle lesion phenotype [28,29,54]. These muscle lesionscan be observed through a simple, high-throughput birefrin-gence assay in 4-day-old larvae (Figure 2; [54]; see also Section5.1 Assessing muscle structure). In their initial large-scalescreen, Kawahara et al. tested 1120 small molecules fromthe Prestwick library of bioreactive compounds approved forhuman use [54]. First, 140 chemical pools were tested andselected if < 7.5% of the progeny exhibited muscle lesions,compared with 25% in the untreated control embryos. Inthe second step, the selected pools were separated and48 chemicals were tested individually. Seven chemicals fromthis two-step screen rescue the dmd mutant muscle lesion

L. Maves

1036 Expert Opin. Drug Discov. (2014) 9(9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 5: Recent advances using zebrafish animal models for muscle disease drug discovery

phenotype [54]. The chemical that promoted the highest long-term survival in dmd fish was aminophylline, a non-selectivephosphodiesterase (PDE) inhibitor [54]. They further tested aseries of other PDE inhibitors and found that sildenafil, aPDE5 inhibitor, improved muscle lesions in zebrafish dmdlarvae [54]. Because PDE5 inhibitors, including sildenafil,have been shown to ameliorate the mouse mdx model [56-58],this study showed that zebrafish dmd drug screening is highlyrelevant to identifying potential mammalian DMD therapies.

More recently, Kawahara et al. have expanded their drugscreening on zebrafish dmd models with an additional 1520chemicals, identifying 8 more compounds, including sildena-fil [55]. Out of the 15 total drugs identified from both screens,6 compounds target heme oxygenase signaling [55]. Thesecompounds improve the zebrafish dmd muscle phenotypethrough upregulation of Heme oxygenase 1 (Hmox1; [55]).The authors further show that sildenafil treatments in mdxmice, as in the dmd fish, increase Hmox1 expression [55].These studies thus reveal Hmox1 as a novel potential targetfor DMD therapy. These large-scale screens from the Kunkellaboratory highlight the potential of zebrafish models foridentifying new therapeutic compounds and targets as wellas for understanding the molecular mechanisms behindmuscle disease.

3.2 Targeted drug studiesTwo recent studies have directly tested whether compoundsthat ameliorate the mouse mdx model can also ameliorate

the zebrafish dmd model. In one study, Winder et al. testedthe proteosomal inhibitor MG132, because it had been previ-ously shown that inhibition of the proteasome by MG132 wasable to restore components of the DGC in both mdxmice andDMD patient extracts and also showed signs of amelioratingdisease symptoms in these models [59-61]. When zebrafishdmd mutant larvae are exposed to MG132, reduced muscledamage, as assessed by birefringence (see below Section5.1 Assessing muscle structure), is observed [59]. In a secondstudy, my laboratory tested the histone deacetylase (HDAC)inhibitor Trichostatin A (TSA), because it and other HDACinhibitors can ameliorate the mdx model [17,62,63]. We showedthat TSA robustly rescues muscle fiber damage in the zebra-fish dmd mutant [62]. These studies further underscore thatzebrafish is an appropriate model for testing pharmacologicaltherapies for DMD.

The ability of TSA to ameliorate muscular dystrophy in themouse mdx model may work through more than one mecha-nism. Initial studies of TSA-treated mdx mice suggested thatTSA acted through promoting upregulation of follistatinexpression in satellite cells [64]. A recent study, however, showedthat fibro-adipogenic progenitor cells mediate the ability ofTSA to ameliorate muscular dystrophy in young mdx mice[63]. The recent zebrafish TSA studies now provide an additionalmodel system for further mechanistic analysis of how HDACinhibitors function to ameliorate dystrophic muscle.

One limitation of the zebrafish dmd mutant model forchemical screening is that phenotypic rescue can be assessed

Table 1. Zebrafish muscle disease models with drug therapies studied in zebrafish.

Zebrafish

gene

Protein product Human disease model Type of zebrafish model

treated

Drug therapy Ref.

ache Acetylcholinesterase Unknown Loss of function mutant Eserine [52]

col6a1 Collagen VI Ullrich congenital musculardystrophy, Bethlemmyopathy

Morpholino Cyclosporine A [68]

dag1 Dystroglycan 1 Limb-girdle musculardystrophy

Morpholino Nicotinamide AdenineDinucleotide (NAD+),Emergen-C

[73]

dmd Dystrophin Duchenne musculardystrophy, Becker musculardystrophy

Two loss of functionmutants, Morpholino

15 chemicals from screens [54,54]

Loss of function mutant MG132 [59]

Loss of function mutant,Morpholino

Trichostatin A [62]

Loss of function mutant Splice-altering morpholinos [88]

Loss of function mutant Ataluren [94]

dnm2 Dynamin 2 Centronuclear myopathy,dominant

Overexpression of humanDNM2 mutant RNA

Edrophonium [76]

itga7 Integrin, a 7 Congenital musculardystrophy with integrindefect

Morpholino NAD+, Emergen-C [73]

lmna Lamin A Multiple types of musculardystrophy

Overexpression of mutantlmna

Farnesyltransferase inhibitorL-744832

[101]

ryr1b Ryanodinereceptor 1(skeletal)

RYR1-related congenitalmyopathies

Loss of function mutant N-acetylcysteine [71]

Recent advances using zebrafish animal models for muscle disease drug discovery

Expert Opin. Drug Discov. (2014) 9(9) 1037

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 6: Recent advances using zebrafish animal models for muscle disease drug discovery

on only the 25% of larvae in the clutch that are mutant, while75% of the larvae are phenotypically normal [54]. Further-more, to accurately assess rescue, the mutant individualsmust be genotyped. Therefore, my laboratory developed arobust zebrafish dmd antisense morpholino (dmd-MO)knock-down model that closely resembles the zebrafish dmdmutant phenotype and achieves almost 100% penetrance [62].We showed that this dmd-MO model is useful for identifyingsmall molecules that rescue the dmd phenotype by showingthat TSA can rescue muscle fiber damage similarly in bothdmd-MO and dmd mutant larvae (Figure 2; [62]). Thus,dmd-MO larvae could be employed as a rapid and robustmodel for initial chemical screening. Candidate drugs thatrescue dmd-MO larvae can subsequently be retested on dmdmutants. I discuss further advantages and disadvantages ofusing morpholinos below (Section 8 Expert opinion).

4. Additional zebrafish muscle diseasemodels and pharmacological studies

Here, I would like to highlight examples where pharmacolog-ical studies in other zebrafish muscle disease models havehelped provide new insights into mechanisms of drug action,therapeutic targets and disease pathogenesis. Table 1 providesa list of these and other examples.

4.1 Collagen VI myopathies and cyclosporine A

treatmentDeficiencies in collagen VI, an essential component of theextracellular matrix in skeletal muscle, lead to Ullrich congen-ital muscular dystrophy (UCMD) and Bethlem myopathy(BM) [65]. Collagen VI myopathies share a common patho-genesis of mitochondrial dysfunction. Mitochondrial defectsand associated apoptosis seen in the mouse Col6a1-/- mousemodel and in muscle cell cultures from UCMD patients canbe rescued by treatment with the proton pump modifiercyclosporine A (CsA; [66,67]). In order to better understandthese myopathies and the mechanism of CsA action, Telferet al. generated zebrafish models of the collagen VI myopa-thies [68]. Antisense morpholinos designed to target eitherexon 9 or exon 13 of the zebrafish col6a1 gene caused motordefects and skeletal muscle structural defects, includingabnormal mitochondria, consistent with either a mild BM-like (exon 13) or severe UCMD-like (exon 9) phenotype [68].Treatment with CsA improves the mitochondrial and apopto-sis defects and improves the motor deficits in the UCMD-likezebrafish [68]. However, CsA does not improve muscle fiberintegrity observed using birefringence, suggesting that CsAacts ‘downstream’ of muscle membrane integrity. Reducingapoptosis through p53 suppression does not improve themotor phenotype of UCDM fish, suggesting that CsA is notworking solely through reducing mitochondrial-mediatedapoptosis [68]. These studies raise the question of whetherthe ability of CsA to improve motor function in UCMD

fish is related to other effects of improving mitochondrialfunction or is due to additional roles of CsA. Because themouse Col6a1-/- model shows no clear functional defects [69],the zebrafish UCMD-like model can serve for further studiesof muscle structural and functional pharmacological rescue.

4.2 RYR-1-related myopathy and oxidative stressMutations in the skeletal muscle ryanodine receptor (RYR1), acalcium release channel needed for excitation--contractioncoupling, are associated with several congenital myopathies(RYR1-related myopathies; [70]). To identify novel diseasemechanisms in RYR1-related myopathies, Dowling et al.used the zebrafish relatively relaxed (ryr) mutant, a zebrafishmodel of RYR1-related myopathies [30,71]. ryr zebrafish carrya mutation in ryr1b and show decreased RyR1 levels, impairedexcitation--contraction coupling in fast twitch muscles, and aslow-swimming phenotype. RNA expression analysis of zebra-fish ryr mutant larvae reveals significant changes in severalpathways associated with cellular stress, in particular increasedexpression of markers of oxidative stress [71]. Treatment of ryrzebrafish with the antioxidant N-acetylcysteine rescues theswimming defects and improves the muscle histologicalappearance [71]. This study further showed that myotubesderived from patients with RYR1-related myopathies also dis-play increased oxidative stress and that N-acetylcysteine couldreduce oxidative stress and improve survival of cultured patientmyotubes [71]. Thus, the zebrafish RYR1-related myopathymodel identified oxidative stress as an important pathophysio-logical mechanism in RYR1-related myopathies. Treatmentwith N-acetylcysteine now represents a potential therapy forRYR1-related myopathies.

4.3 Vitamin rescue of dystroglycanopathy and

muscle fiber adhesionGenetic mutations that disrupt adhesion of muscle fibers totheir extracellular environment can lead to several myopa-thies [72,73]. The receptors used by muscle fibers to adhere tothe basement membrane in the extracellular matrix are theDGC (see Section 1 Introduction) and integrin a7b1 hetero-dimers. Weakened links between these receptors and the base-ment membrane can make muscle fibers more susceptible todamage and degeneration. Because their previous studiesshow that the ubiquitous coenzyme nicotinamide adeninedinucleotide (NAD+) helps promote basement membraneorganization in zebrafish muscle, Goody et al. tested whetherNAD+ treatments could improve muscle fiber attachment inzebrafish muscular dystrophy models [73,74]. Indeed, they findthat in zebrafish lacking either dystroglycan or integrin a 7,basement membrane organization, muscle fiber attachmentand motility are all significantly improved by treating embryoswith NAD+ [73]. An NAD+ precursor, niacin (vitamin B3), issimilarly able to rescue zebrafish embryos lacking dystroglycanor integrin a 7 [73]. The niacin source chosen was Emergen-C,an over-the-counter nutritional supplement powder that easily

L. Maves

1038 Expert Opin. Drug Discov. (2014) 9(9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 7: Recent advances using zebrafish animal models for muscle disease drug discovery

dissolves in the zebrafish embryo medium. Interestingly, theEmergen-C concentration that rescues the dystroglycan-nullzebrafish is calculated to be roughly equivalent to what theconcentration of niacin would be in an adult human’sbloodstream after consuming an Emergen-C packet [73]. Fur-thermore, Goody et al. used the NAD+ treatments to helpdemonstrate the requirement for a novel skeletal muscle adhe-sion receptor, integrin a 6 [73]. Altogether, this study combinesmany advantages of zebrafish (cell biology, imaging, geneticsand pharmacological treatments), to support the ideas thatmodulation of muscle fiber--extracellular matrix adhesionmay be a therapeutic avenue for many myopathies and thatvitamin supplementation warrants further investigation.

4.4 Dynamin-2-related centronuclear myopathy and

neuromuscular transmissionCentronuclear myopathies (CNM) are characterized by mus-cle weakness and abnormal centralized nuclei and are mostcommonly caused by dominant mutations in dynamin-2(DNM2; [75]). The mechanisms by which DNM2 mutationslead to myopathy are not well understood. Gibbs et al. gener-ated a zebrafish model for DNM2--CNM by overexpressingone of the most common and severe DNM2 mutations(DNM2-S619L; [76,77];). They find that overexpression ofDNM2-S619L in zebrafish embryos impairs motor behavior,causes perinuclear muscle structural abnormalities anddecreases AChR clustering in muscle fibers [76], and alsocauses defects in T-tubule and sarcoplasmic reticulum forma-tion and in excitation--contraction coupling [78]. Clinical stud-ies of patients with DNM2--CNM mutations also suggestdefects in neuromuscular transmission [76]. Treating theDNM2--CNM zebrafish larvae with an AChE inhibitorgreatly improved their motor behavior [76]. These resultssuggest a new mechanism of neuromuscular junction defectsin DNM2--CNM pathology.

5. Analyses of zebrafish muscle disease andassessment of pharmacological rescue

The examples presented above of both large-scale drug screen-ing and targeted drug studies have not only identified classes ofdrugs that ameliorate zebrafish dmd and other myopathies, butalso identified optimal approaches for analyzing and quantify-ing zebrafish muscle disease and its rescue. The zebrafishsystem is an exceptional animal model for the range ofextremely simple to sophisticated approaches for assessingmuscle structure and function. Here, I discuss studies ofzebrafish muscle structure and function that have enhancedzebrafish as a model for pharmacological approaches tomuscle disease.

5.1 Assessing muscle structureA critical aspect of using zebrafish as a model for pharmaco-logical approaches to muscle disease is the ability to

quantitatively assess muscle structural damage and rescue bydrug treatment. The Kunkel group has successfully used asimple birefringence assay through a stereomicroscope forlarge-scale drug screening with the zebrafish dmdmodel [54,55].The highly organized sarcomere pattern of zebrafish skeletalmuscle appears bright under polarized light, but zebrafishwith muscle disease show darkened disruptions in this bire-fringence due to muscle fiber disorganization, detachment ordegeneration (Figures 1 and 2; [6,32,39,40]). Imaging birefrin-gence in zebrafish larvae can be rapid, is noninvasive, andcan be performed on live or on fixed specimens [62,79]. How-ever, birefringence is very dependent on orientation ofthe larva between two polarizing filters, and improper align-ment of larvae may prevent correct scoring of the musclephenotype. Also, overall birefringence may not allow for dis-tinguishing subtle effects of a drug treatment on muscle struc-ture. Three studies have addressed these issues by describinghighly quantitative approaches to birefringence. Selected areasof muscle birefringence images taken from a stereomicroscopecan be analyzed for mean pixel intensity using freely availablesoftware [79]. The Currie group described a polarizing lightmicroscope system that rapidly generates an unbiased birefrin-gence map of a fish larva that is independent of the specimenorientation [80]. Subsequent measuring of the pixel brightnessallows quantification of the birefringence. Also, zebrafish dmdmuscle damage can be quantified by subjecting gray-scalebirefringence intensities over the length of the larva to Fourieranalysis [59].

In addition to birefringence, skeletal muscle can be readilyvisualized in whole zebrafish larvae using dyes, stains or trans-genic skeletal muscle fluorescent reporters. Evans Blue Dye, avital stain that only enters cells with damaged membranes,can reveal sarcolemmal damage in dystrophic zebrafish [28,48].Immunocytochemistry, histology and electron microscopycan provide insight into muscle fiber and sarcomere structuredefects in zebrafish myopathy models [27,48,53,68,73]. Skeletalmuscle actin labeling with the transgene acta1a:gfp or withphalloidin, or muscle myosin labeling with immunocytochem-istry, can reveal muscle lesions in dystrophic zebrafish(e.g., Figure 2; [28,48,54,62,73]). The Henry group used phalloidinstaining to quantitate dystrophic muscle lesions by scoring thepercent of myotomes (muscle segments) affected per larva [73].The Henry group has also used two-dimensional wavelettransform modulus maxima, a measure of muscle fiber organi-zation, in combination with phalloidin staining and immuno-cytochemistry, to quantify changes in muscle structure inzebrafish muscular dystrophy models [73,81]. Elegant high-resolution imaging of the damage response in myofibers inlive zebrafish larvae has been used to identify a novel sequentialsarcolemmal repair process [34]. Thus, the zebrafish system hasa range of simple to sophisticated approaches for assessingmuscle structure.

For large-scale chemical screening, it is ideal to optimizethe cost and speed of the screening approach while still allow-ing for reliable phenotypic scoring. Assessment of the muscle

Recent advances using zebrafish animal models for muscle disease drug discovery

Expert Opin. Drug Discov. (2014) 9(9) 1039

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 8: Recent advances using zebrafish animal models for muscle disease drug discovery

birefringence pattern, in which larvae are simply scored asaffected versus unaffected using a stereomicroscope, providesa measurement of dmd phenotypic rescue, following treat-ment with the drug TSA, that is comparable to the morequantitative, but time-consuming, approach of countingmyotome lesions per larva, visualized with muscle actin label-ing using the confocal microscope [62]. This study supportsthe large-scale birefringence screening approach used by theKunkel group [54,55] as an easy and reliable assessment ofdrug-treatment rescue of dystrophic zebrafish larvae.

5.2 Assessing muscle functionAnother critical aspect of using zebrafish as a model forpharmacological approaches to muscle disease is the abilityto quantitatively assess rescue of muscle function followingdrug treatment. Zebrafish are quite amenable to assessingmuscle function, and, as with muscle structure, the assaysrange from the simple to the more complex. Perhaps the sim-plest and most important assessment of muscle function isanimal survival, which Kawahara et al. used to show improve-ment of zebrafish dmd mutants following treatment withselected chemicals from their large-scale drug screens [54,55].Spontaneous embryo coiling and larval touch-evoked escaperesponse are two straightforward motor assays that havebeen used to characterize zebrafish myopathy models [47],and several studies have used these assays to demonstratemotor function rescue of zebrafish myopathies after drugtreatments [6,68,73]. Dowling et al. assessed motor function inN-acetylcysteine-treated ryr-mutant zebrafish larvae usingvideo monitoring to measure swim velocity and distance trav-elled (Figure 1; [71]). To further assess motor function, theymeasured multiple parameters of skeletal muscle contractileproperties in whole larvae by mounting larvae to a force trans-ducer [71]. Electrophysiological recordings from muscle inwhole larvae can assess muscle function in mutant and drug-treated animals [30,47]. Another option for muscle functionalanalysis in zebrafish is the study of cultured isolated myofib-ers, in which analyses such as electrophysiological measure-ments can be performed [82,83]. To assess locomotor abilitiesof swimming adult zebrafish under the demanding conditionsof a water current, Blazina et al. developed a Spinning Task ina beaker using a magnetic stirrer and were able to quantita-tively assess motor impairments induced by drug treat-ments [84]. This variety of approaches thus allows for theexquisite analysis of many aspects of muscle function andfor the identification of key pathological properties ofdifferent myopathies.

6. Using zebrafish to study pharmacologicalapproaches to muscle disease genemodification

Exon-skipping agents and stop-codon read-through arepotential therapeutic approaches for muscle diseases that

have been tested in zebrafish models. Table 1 includes theseapproaches as examples of drug studies in zebrafish dmdmodels.

6.1 Exon skippingOne promising therapeutic approach for DMD is exon skip-ping, in which antisense oligonucleotides are administered tosterically block splicing of the DMD pre-mRNA transcript,leading to exclusion of targeted exons and restoration oftruncated, but partially functional, dystrophin protein [85]. Itis estimated that about 83% of all DMD mutations couldpotentially be corrected by the skipping of specific exons [86].Although mouse and dog animal models for DMD exon skip-ping have already provided valuable guides for clinical trials[12,15,87], one question that has been tested in the zebrafishmodel is the level of dystrophin needed for functional res-cue [88]. The Currie laboratory used a forward genetic screento identify new zebrafish dmd alleles that are amenable to test-ing exon skipping strategies [88]. They injected dmd splice-sitetargeted antisense morpholino oligos into these dmd mutantembryos and then assayed dmd phenotype and dystrophinexpression. Their study showed that robust morpholino activ-ity, in which exon-skipped dmd transcript is induced to levelsof at least 30 -- 40% of total dmd transcript, is needed to signif-icantly improve the zebrafish dmdmuscle phenotype [88]. Theirstudy was also able to identify a conserved cryptic splice site inthe dmd gene [88]. Thus, this analysis in zebrafish could helpinform antisense oligo design and strategies for humanDMD exon-skipping therapies. Modulation of pre-mRNAsplicing has been investigated as a therapeutic option for severalmuscle diseases [85]. Because of the ease of testing antisense oli-gos in zebrafish, and because of the ability to engineer zebrafishcarrying specific disease gene alleles found in humans (seebelow 8. Expert opinion), zebrafish should serve as an excellentpreclinical animal model for testing exon-skipping strategies.

6.2 Stop-codon read-throughAbout 10 -- 15% of DMD patients have a mutation thatgenerates a premature stop codon [89]. Drugs that enable ribo-somal read-through of stop codons have thus been investi-gated as DMD therapies [14]. PTC Therapeutics identifiedAtaluren (PTC124) as a drug that promotes read-through ofpremature stop codons in the DMD gene and improves mus-cle function in the mouse mdx model [90], although the read-through effectiveness of Ataluren has been questioned [91,92].A Phase IIb clinical trial has suggested some success inpatients’ physical functioning [93]. Li et al. have used zebrafishto further investigate the ability of Ataluren to ameliorateDMD [94]. They find a significant improvement of musclestructure and function in dmd larvae after two days ofAtaluren treatment, accompanied by increased expression ofDystrophin protein [94]. Importantly, Ataluren shows dose-dependent effects in zebrafish, in which increasing doses canbegin to show reduced effects or even negative effects on mus-cle force production [94]. These findings may provide insight

L. Maves

1040 Expert Opin. Drug Discov. (2014) 9(9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 9: Recent advances using zebrafish animal models for muscle disease drug discovery

into the results from the Ataluren Phase IIb clinical trials,where patients’ walking improvement was observed at lowerdoses but not at higher doses [93].

Thus, even for drug therapies whose mechanism of action isrelatively well understood, zebrafish studies can providefurther insight into their design and use. Although the impor-tance of human-specific issues such as target-sequence speci-ficity and cellular delivery need to be considered, zebrafish isa valuable model for rapidly testing the efficacy and reproduc-ibility of these gene-product modification approaches.

7. Conclusion

The zebrafish system has a long history of providing insightinto skeletal muscle development and function. The lastcouple years have generated an exciting flurry of activity usingthe zebrafish model for the discovery and analysis of pharma-cological approaches to human muscle diseases. In particular,the large-scale drug screens using the zebrafish DMD model[54,55] have not only provided new insight into pharmacologi-cal therapies for DMD but also, for the future, provided amodel with which to approach large-scale drug screens withzebrafish models for additional myopathies. The ability totake a variety of simple as well as sophisticated approachesto assess muscle structure and function in zebrafish has alsorecently allowed for novel insights into the pathological mech-anisms of many muscle diseases. This insight gained in zebra-fish can provide clues to novel, rational pharmacologicalapproaches for muscle disease therapies, which, likewise, canthen be directly tested in zebrafish.

8. Expert opinion

A major advantage of the zebrafish system for future pharma-cological approaches for muscle diseases is in the study of com-binatorial drug treatments. Because corticosteroids are nowwidely used for treating DMD patients, any new potentialpharmacological therapies for DMD should be tested for effi-cacy with or interactions with corticosteroids [15]. The ability toeasily obtain many animals and rapidly assess the effects ofdrug treatments makes zebrafish an ideal model for optimizingthe timing and dosage of drugs alone and in combinations.

Because of the significance of addressing cardiac musclefunction in DMD patients [13], it will be important to deter-mine whether dmd zebrafish can serve as a model for cardiacmuscle defects in addition to the skeletal muscle defects.There are not yet any reports of dystrophin expression inthe zebrafish heart, nor have there been any reports of heartdefects in dmd mutant zebrafish [95]. This lack of data maybe due to zebrafish dystrophin being expressed or requiredin the heart at later stages than are typically examined.

As discussed earlier, most zebrafish models of humanmusclediseases have come from forward genetic screens and throughthe use of morpholinos. Genome editing technology, in partic-ular the CRISPR/Cas system [96], now opens up the possibility

of engineering zebrafish models for human muscle diseasesthat have not been identified through forward genetics, suchas mtm1 for myotubular myopathy [95,97]. Another use ofgenome editing approaches could be to engineer zebrafishstrains with muscle disease gene alleles that mimic the specificmutations found in humans [96]. With genome editing, zebra-fish strains can be created that carry a spectrum of mutationswithin an individual gene, to reflect the diversity of humanmutations that have been identified within certain muscle dis-ease genes [1,2]. Such engineered zebrafish could be useful fortesting muscle disease gene modification therapies such asexon skipping (see Section 6 Using zebrafish to study pharma-cological approaches to muscle disease gene modification) andalso would be valuable for in-depth cellular, structural andmuscle functional analyses to better understand disease mech-anisms. Engineered zebrafish strains can also be used in drugscreens to more directly screen for drugs that ameliorate thephenotypes caused by specific muscle disease gene mutations.

Even with the increasing development of genome editingtechnology for reverse genetics, antisense morpholinos cancontinue to provide important tools for zebrafish models ofmuscle disease. Morpholino knock-downs in zebrafishembryos can provide a rapid means to test new candidategenes for human muscle diseases or new modifier loci forexisting muscle disease genes. Two of the main potentialdisadvantages of morpholinos, incomplete knock-down andoff-target effects, can be alleviated through careful validationcontrols or by comparing the morpholino phenotype to thatof a mutant allele, identified through forward genetics, TILL-ING [98], or genome editing. As described above (Section3.2 Targeted drug studies), zebrafish muscle disease modelsgenerated using morpholinos can provide advantages over amutant allele for drug screening because about 100% ofmorpholino-injected embryos should show a muscle diseasephenotype, as opposed to only 25% for a genetic recessivemutant cross. Because the efficacy of morpholinos, typicallyinjected at the one-cell stage, lessens as larvae get older, it isbetter to perform long-term (> 4 -- 5 days) drug studies usingmutant strains instead of morpholinos.

One outstanding opportunity for use of the zebrafishmodel is in the study of inflammation and the immuneresponse in muscle disease. The inflammatory response playsan important role in normal muscle repair, but chronicinflammation contributes to the progressive pathology ofmuscle diseases such as DMD [99]. Understanding how, andwhen, different cell types of the immune system respond to,and influence, muscle degeneration and regeneration wouldprovide insight for improved pharmacological treatment ofmuscle diseases with chronic inflammation. Zebrafish are apowerful system with which to address these issues. Zebrafishdmd larvae show signs of acute inflammatory response in theirdystrophic muscle [50], and it would be informative to addresswhether and how drug treatments that ameliorate the zebra-fish dmd phenotype affect this inflammatory response. Theavailability of many zebrafish transgenic strains for labeling

Recent advances using zebrafish animal models for muscle disease drug discovery

Expert Opin. Drug Discov. (2014) 9(9) 1041

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 10: Recent advances using zebrafish animal models for muscle disease drug discovery

immune cell types [100], combined with live imaging and drugtreatment of muscle disease models, should allow elegantstudies of how pharmacological approaches might influenceinflammation and the immune response in muscle disease.

Acknowledgements

I would like to thank H Farr and N Johnson for their work ininitiating pharmacological studies of the zebrafish dmd modelin my laboratory.

Declaration of interest

The muscle disease research undertaken in L Maves’laboratory was funded by the Seattle Children’s MyocardialRegeneration Initiative and by the National Institutes ofHealth R03AR065760. The author has no other relevantaffiliations or financial involvement with any organization orentity with a financial interest in or financial conflict withthe subject matter or materials discussed in the manuscriptapart from those disclosed.

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Kaplan JC, Hamroun D. The

2014 version of the gene table of

monogenic neuromuscular disorders

(nuclear genome). Neuromuscul Disord

2013;23:1081-111

2. Rahimov F, Kunkel LM. The cell

biology of disease: cellular and molecular

mechanisms underlying muscular

dystrophy. J Cell Biol 2013;201:499-510

. This work provides an excellent recent

review of the molecular and cellular

mechanisms of muscle disease based on

combined insights from animal model

and human genetics studies.

3. Manring H, Abreu E, Brotto L, et al.

Novel excitation-contraction coupling

related genes reveal aspects of muscle

weakness beyond atrophy-new hopes for

treatment of musculoskeletal diseases.

Front Physiol 2014;5:37

. This work provides an excellent recent

review of the molecular and cellular

mechanisms of muscle disease based on

combined insights from animal model

and human genetics studies.

4. Ng R, Banks GB, Hall JK, et al. Animal

models of muscular dystrophy. Prog Mol

Biol Transl Sci 2012;105:83-111

5. Nance JR, Dowling JJ, Gibbs EM,

B€onnemann CG. Congenital myopathies:

an update. Curr Neurol Neurosci Rep

2012;12:165-74

6. Gibbs EM, Horstick EJ, Dowling JJ.

Swimming into prominence: the

zebrafish as a valuable tool for studying

human myopathies and muscular

dystrophies. FEBS J 2013;280:4187-97

7. Chartier A, Simonelig M. Animal models

in therapeutic drug discovery for

oculopharyngeal muscular dystrophy.

Drug Discov Today Technol

2013;10:e103-8

8. Lloyd TE, Taylor JP. Flightless flies:

drosophila models of neuromuscular

disease. Ann N Y Acad Sci

2010;1184:e1-20

9. Flisikowska T, Kind A, Schnieke A.

Genetically modified pigs to model

human diseases. J Appl Genet

2014;55:53-64

10. Monaco AP, Neve RL,

Colletti-Feener C, et al. Isolation of

candidate cDNAs for portions of the

Duchenne muscular dystrophy gene.

Nature 1986;323:646-50

11. Wallace GQ, McNally EM. Mechanisms

of muscle degeneration, regeneration, and

repair in the muscular dystrophies.

Annu Rev Physiol 2009;71:37-57

12. Fairclough RJ, Bareja A, Davies KE.

Progress in therapy for Duchenne

muscular dystrophy. Exp Physiol

2011;96:1101-13

13. Konieczny P, Swiderski K,

Chamberlain JS. Gene and cell-mediated

therapies for muscular dystrophy.

Muscle Nerve 2013;47:649-63

14. Pichavant C, Aartsma-Rus A,

Clemens PR, et al. Current status of

pharmaceutical and genetic therapeutic

approaches to treat DMD. Mol Ther

2011;19:830-40

15. Wilton SD, Fletcher S. Novel

compounds for the treatment of

Duchenne muscular dystrophy: emerging

therapeutic agents. Appl Clin Genet

2011;4:29-44

16. Abdel-Hamid H, Clemens PR.

Pharmacological therapies for muscular

dystrophies. Curr Opin Neurol

2012;25:604-8

17. Consalvi S, Saccone V, Giordani L, et al.

Histone deacetylase inhibitors in the

treatment of muscular dystrophies:

epigenetic drugs for genetic diseases.

Mol Med 2011;17:457-65

18. Pantoja M, Fischer KA, Ieronimakis N,

et al. Genetic elevation of sphingosine

1-phosphate suppresses dystrophic muscle

phenotypes in Drosophila. Development

2013;140:136-46

19. Kornegay JN, Bogan JR, Bogan DJ,

et al. Canine models of Duchenne

muscular dystrophy and their use in

therapeutic strategies. Mamm Genome

2012;23:85-108

20. Santoriello C, Zon LI. Hooked!

Modeling human disease in zebrafish.

J Clin Invest 2012;122:2337-43

21. Zon LI, Peterson RT. In vivo drug

discovery in the zebrafish. Nat Rev

Drug Discov 2005;4:35-44

22. North TE, Goessling W, Walkley CR,

et al. Prostaglandin E2 regulates

vertebrate haematopoietic stem cell

homeostasis. Nature 2007;447:1007-11

. One of the first examples of a chemical

screen in zebrafish that has directly led

to drug tests in human clinical trials.

23. White RM, Cech J,

Ratanasirintrawoot S, et al. DHODH

modulates transcriptional elongation in

the neural crest and melanoma. Nature

2011;471:518-22

. One of the first examples of a chemical

screen in zebrafish that has directly led

to drug tests in human clinical trials.

24. Hollway G, Currie P. Vertebrate

myotome development. Birth Defects Res

C Embryo Today 2005;75:172-9

25. Buckingham M, Vincent SD. Distinct

and dynamic myogenic populations in

the vertebrate embryo. Curr Opin

Genet Dev 2009;19:444-53

26. Jackson HE, Ingham PW. Control of

muscle fibre-type diversity during

embryonic development: the zebrafish

paradigm. Mech Dev 2013;130:447-57

L. Maves

1042 Expert Opin. Drug Discov. (2014) 9(9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 11: Recent advances using zebrafish animal models for muscle disease drug discovery

27. Parsons MJ, Campos I, Hirst EM,

Stemple DL. Removal of dystroglycan

causes severe muscular dystrophy in

zebrafish embryos. Development

2002;129:3505-12

28. Bassett DI, Bryson-Richardson RJ,

Daggett DF, et al. Dystrophin is

required for the formation of stable

muscle attachments in the zebrafish

embryo. Development 2003;130:5851-60

29. Guyon JR, Mosley AN, Zhou Y, et al.

The dystrophin associated protein

complex in zebrafish. Hum Mol Genet

2003;12:601-15

30. Hirata H, Watanabe T, Hatakeyama J,

et al. Zebrafish relatively relaxed mutants

have a ryanodine receptor defect, show

slow swimming and provide a model of

multi-minicore disease. Development

2007;134:2771-81

31. Steffen LS, Guyon JR, Vogel ED, et al.

Zebrafish orthologs of human muscular

dystrophy genes. BMC Genomics

2007;8:79

32. Berger J, Currie PD. Zebrafish models

flex their muscles to shed light on

muscular dystrophies. Dis Model Mech

2012;5:726-32

33. Kobayashi K, Izawa T, Kuwamura M,

Yamate J. Dysferlin and animal models

for dysferlinopathy. J Toxicol Pathol

2012;25:135-47

34. Roostalu U, Strahle U. In vivo imaging

of molecular interactions at damaged

sarcolemma. Dev Cell 2012;22:515-29

35. Montarras D, L’honore A,

Buckingham M. Lying low but ready for

action: the quiescent muscle satellite cell.

FEBS J 2013;280:4036-50

36. Seger C, Hargrave M, Wang X, et al.

Analysis of Pax7 expressing myogenic

cells in zebrafish muscle development,

injury, and models of disease. Dev Dyn

2011;240:2440-51

37. Siegel AL, Gurevich DB, Currie PD.

A myogenic precursor cell that could

contribute to regeneration in zebrafish

and its similarity to the satellite cell.

FEBS J 2013;280:4074-88

38. Kimmel CB, Patterson J, Kimmel RO.

The development and behavioral

characteristics of the startle response in

the zebra fish. Dev Psychobiol

1974;7:47-60

39. Felsenfeld AL, Walker C, Westerfield M,

et al. Mutations affecting skeletal muscle

myofibril structure in the zebrafish.

Development 1990;108:443-59

40. Granato M, van Eeden FJ, Schach U,

et al. Genes controlling and mediating

locomotion behavior of the zebrafish

embryo and larva. Development

1996;123:399-413

. This work, part of the first large-scale

forward genetic screen in zebrafish,

provided many of the current zebrafish

muscle disease models.

41. Saint-Amant L, Drapeau P. Time course

of the development of motor behaviors

in the zebrafish embryo. J Neurobiol

1998;37:622-32

42. Westerfield M, Liu DW, Kimmel CB,

Walker C. Pathfinding and synapse

formation in a zebrafish mutant lacking

functional acetylcholine receptors.

Neuron 1990;4:867-74

43. Behra M, Cousin X, Bertrand C, et al.

Acetylcholinesterase is required for

neuronal and muscular development in

the zebrafish embryo. Nat Neurosci

2002;5:111-18

44. Saint-Amant L, Sprague SM, Hirata H,

et al. The zebrafish ennui behavioral

mutation disrupts acetylcholine receptor

localization and motor axon stability.

Dev Neurobiol 2008;68:45-61

45. Gupta V, Kawahara G, Gundry SR,

et al. The zebrafish dag1 mutant: a novel

genetic model for dystroglycanopathies.

Hum Mol Genet 2011;20:1712-25

46. Gleason MR, Armisen R, Verdecia MA,

et al. A mutation in serca underlies

motility dysfunction in accordion

zebrafish. Dev Biol 2004;276:441-51

47. Hirata H, Saint-Amant L, Waterbury J,

et al. accordion, a zebrafish behavioral

mutant, has a muscle relaxation defect

due to a mutation in the ATPase Ca2+

pump SERCA1. Development

2004;131:5457-68

48. Hall TE, Bryson-Richardson RJ,

Berger S, et al. The zebrafish candyfloss

mutant implicates extracellular matrix

adhesion failure in laminin alpha2-

deficient congenital muscular dystrophy.

Proc Natl Acad Sci USA

2007;104:7092-7

49. Lin YY. Muscle diseases in the zebrafish.

Neuromuscul Disord 2012;22:673-84

50. Berger J, Berger S, Hall TE, et al.

Dystrophin-deficient zebrafish feature

aspects of the Duchenne muscular

dystrophy pathology.

Neuromuscul Disord 2010;20:826-32

51. Hoffman EP, Brown RH Jr, Kunkel LM.

Dystrophin: the protein product of the

Duchenne muscular dystrophy locus.

Cell 1987;51:919-28

52. Behra M, Etard C, Cousin X, Strahle U.

The use of zebrafish mutants to identify

secondary target effects of acetylcholine

esterase inhibitors. Toxicol Sci

2004;77:325-33

53. Etard C, Behra M, Ertzer R, et al.

Mutation in the delta-subunit of the

nAChR suppresses the muscle defects

caused by lack of Dystrophin. Dev Dyn

2005;234:1016-25

54. Kawahara G, Karpf JA, Myers JA, et al.

Drug screening in a zebrafish model of

Duchenne muscular dystrophy. Proc Natl

Acad Sci USA 2011;108:5331-6

.. This work was the first large-scale drug

screen in a zebrafish muscle

disease model.

55. Kawahara G, Gasperini MJ, Myers JA,

et al. Dystrophic muscle improvement in

zebrafish via increased heme oxygenase

signaling. Hum Mol Genet

2014;23:1869-78

56. Asai A, Sahani N, Kaneki M, et al.

Primary role of functional ischemia,

quantitative evidence for the two-hit

mechanism, and phosphodiesterase-5

inhibitor therapy in mouse muscular

dystrophy. PLoS One 2007;2:e806

57. Adamo CM, Dai DF, Percival JM, et al.

Sildenafil reverses cardiac dysfunction in

the mdx mouse model of Duchenne

muscular dystrophy. Proc Natl Acad

Sci USA 2010;107:19079-83

58. Percival JM, Whitehead NP, Adams ME,

et al. Sildenafil reduces respiratory

muscle weakness and fibrosis in the mdx

mouse model of Duchenne muscular

dystrophy. J Pathol 2012;228:77-87

59. Winder SJ, Lipscomb L,

Angela Parkin C, Juusola M. The

proteasomal inhibitor MG132 prevents

muscular dystrophy in zebrafish.

PLoS Curr 2011;3:RRN1286

60. Bonuccelli G, Sotgia F, Schubert W,

et al. Proteasome inhibitor (MG-132)

treatment of mdx mice rescues the

expression and membrane localization of

dystrophin and dystrophin-associated

proteins. Am J Pathol 2003;163:1663-75

61. Assereto S, Stringara S, Sotgia F, et al.

Pharmacological rescue of the

Recent advances using zebrafish animal models for muscle disease drug discovery

Expert Opin. Drug Discov. (2014) 9(9) 1043

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 12: Recent advances using zebrafish animal models for muscle disease drug discovery

dystrophin-glycoprotein complex in

Duchenne and Becker skeletal muscle

explants by proteasome inhibitor

treatment. Am J Physiol Cell Physiol

2006;290:C577-82

62. Johnson NM, Farr GH III, Maves L.

The HDAC inhibitor TSA ameliorates a

zebrafish model of Duchenne Muscular

Dystrophy. PLoS Curr

2013;5; Epub ahead of print

63. Mozzetta C, Consalvi S, Saccone V,

et al. Fibroadipogenic progenitors

mediate the ability of HDAC inhibitors

to promote regeneration in dystrophic

muscles of young, but not old Mdx

mice. EMBO Mol Med 2013;5:1-14

64. Minetti GC, Colussi C, Adami R, et al.

Functional and morphological recovery

of dystrophic muscles in mice treated

with deacetylase inhibitors. Nat Med

2006;12:1147-50

65. Lampe AK, Bushby KM. Collagen VI

related muscle disorders. J Med Genet

2005;42:673-85

66. Irwin WA, Bergamin N, Sabatelli P,

et al. Mitochondrial dysfunction and

apoptosis in myopathic mice with

collagen VI deficiency. Nat Genet

2003;35:367-71

67. Angelin A, Tiepolo T, Sabatelli P, et al.

Mitochondrial dysfunction in the

pathogenesis of Ullrich congenital

muscular dystrophy and prospective

therapy with cyclosporins. Proc Natl

Acad Sci USA 2007;104:991-6

68. Telfer WR, Busta AS, Bonnemann CG,

et al. Zebrafish models of collagen VI-

related myopathies. Hum Mol Genet

2010;19:2433-44

.. This work provides an excellent

example of taking advantage of many

strengths of the zebrafish model to

discover new pharmacological

approaches for muscle disease.

69. Bonaldo P, Braghetta P, Zanetti M, et al.

Collagen VI deficiency induces early

onset myopathy in the mouse: an animal

model for Bethlem myopathy.

Hum Mol Genet 1998;7:2135-40

70. Amburgey K, Bailey A, Hwang JH, et al.

Genotype-phenotype correlations in

recessive RYR1-related myopathies.

Orphanet J Rare Dis 2013;8:117

71. Dowling JJ, Arbogast S, Hur J, et al.

Oxidative stress and successful

antioxidant treatment in models of

RYR1-related myopathy. Brain

2012;135:1115-27

.. This work provides an excellent

example of taking advantage of many

strengths of the zebrafish model to

discover new pharmacological

approaches for muscle disease.

72. Holmberg J, Durbeej M. Laminin-211 in

skeletal muscle function. Cell Adh Migr

2013;7:111-21

73. Goody MF, Kelly MW, Reynolds CJ,

et al. NAD+ biosynthesis ameliorates a

zebrafish model of muscular dystrophy.

PLoS Biol 2012;10:e1001409

.. This work provides an excellent

example of taking advantage of many

strengths of the zebrafish model to

discover new pharmacological

approaches for muscle disease.

74. Goody MF, Kelly MW, Lessard KN,

et al. Nrk2b-mediated NAD+ production

regulates cell adhesion and is required for

muscle morphogenesis in vivo: nrk2b and

NAD+ in muscle morphogenesis.

Dev Biol 2010;344:809-26

75. Romero NB. Centronuclear myopathies:

a widening concept.

Neuromuscul Disord 2010;20:223-8

76. Gibbs EM, Clarke NF, Rose K, et al.

Neuromuscular junction abnormalities in

DNM2-related centronuclear myopathy.

J Mol Med (Berl) 2013;91:727-37

.. This work provides an excellent

example of taking advantage of many

strengths of the zebrafish model to

discover new pharmacological

approaches for muscle disease.

77. B€ohm J, Biancalana V, Dechene ET,

et al. Mutation spectrum in the large

GTPase dynamin 2, and genotype-

phenotype correlation in autosomal

dominant centronuclear myopathy.

Hum Mutat 2012;33:949-59

78. Gibbs EM, Davidson AE, Telfer WR,

et al. The myopathy-causing mutation

DNM2-S619L leads to defective

tubulation in vitro and in developing

zebrafish. Dis Model Mech

2014;7:157-61

79. Smith LL, Beggs AH, Gupta VA.

Analysis of skeletal muscle defects in

larval zebrafish by birefringence and

touch-evoke escape response assays.

J Vis Exp 2013;82:e50925

80. Berger J, Sztal T, Currie PD.

Quantification of birefringence readily

measures the level of muscle damage in

zebrafish. Biochem Biophys

Res Commun 2012;423:785-8

81. Snow CJ, Goody M, Kelly MW, et al.

Time-lapse analysis and mathematical

characterization elucidate novel

mechanisms underlying muscle

morphogenesis. PLoS Genet

2008;4:e1000219

82. Horstick EJ, Gibbs EM, Li X, et al.

Analysis of embryonic and larval

zebrafish skeletal myofibers from

dissociated preparations. J Vis Exp

2013;81:e50259

83. Schredelseker J, Shrivastav M, Dayal A,

Grabner M. Non-Ca2+-conducting Ca2+

channels in fish skeletal muscle

excitation-contraction coupling.

Proc Natl Acad Sci USA

2010;107:5658-63

84. Blazina AR, Vianna MR, Lara DR. The

spinning task: a new protocol to easily

assess motor coordination and resistance

in zebrafish. Zebrafish 2013;10:480-5

85. Spitali P, Aartsma-Rus A. Splice

modulating therapies for human disease.

Cell 2012;148:1085-8

86. Aartsma-Rus A, Fokkema I,

Verschuuren J, et al. Theoretic

applicability of antisense-mediated exon

skipping for Duchenne muscular

dystrophy mutations. Hum Mutat

2009;30:293-9

87. Lu QL, Cirak S, Partridge T. What Can

We Learn From Clinical Trials of Exon

Skipping for DMD? Mol Ther

Nucleic Acids 2014;3:e152

88. Berger J, Berger S, Jacoby AS, et al.

Evaluation of exon-skipping strategies for

Duchenne muscular dystrophy utilizing

dystrophin-deficient zebrafish. J Cell

Mol Med 2011;15:2643-51

89. Aartsma-Rus A, Van Deutekom JC,

Fokkema IF, et al. Entries in the Leiden

Duchenne muscular dystrophy mutation

database: an overview of mutation types

and paradoxical cases that confirm the

reading-frame rule. Muscle Nerve

2006;34:135-44

90. Welch EM, Barton ER, Zhuo J, et al.

PTC124 targets genetic disorders caused

by nonsense mutations. Nature

2007;447:87-91

91. Auld DS, Lovell S, Thorne N, et al.

Molecular basis for the high-affinity

binding and stabilization of firefly

luciferase by PTC124. Proc Natl Acad

Sci USA 2010;107:4878-83

L. Maves

1044 Expert Opin. Drug Discov. (2014) 9(9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.

Page 13: Recent advances using zebrafish animal models for muscle disease drug discovery

92. McElroy SP, Nomura T, Torrie LS,

et al. A lack of premature termination

codon read-through efficacy of PTC124

(Ataluren) in a diverse array of reporter

assays. PLoS Biol 2013;11:e1001593

93. Peltz SW, Morsy M, Welch EM,

Jacobson A. Ataluren as an agent for

therapeutic nonsense suppression.

Annu Rev Med 2013;64:407-25

94. Li M, Andersson-Lendahl M, Sejersen T,

Arner A. Muscle dysfunction and

structural defects of dystrophin-null sapje

mutant zebrafish larvae are rescued by

ataluren treatment. FASEB J

2013;28(4):1593-9

95. Bradford Y, Conlin T, Dunn N, et al.

ZFIN: enhancements and updates to the

Zebrafish Model Organism Database.

Nucleic Acids Res 2011;39:D822-9

96. Blackburn PR, Campbell JM, Clark KJ,

Ekker SC. The CRISPR system--keeping

zebrafish gene targeting fresh. Zebrafish

2013;10:116-18

97. Dowling JJ, Vreede AP, Low SE, et al.

Loss of myotubularin function results in

T-tubule disorganization in zebrafish and

human myotubular myopathy.

PLoS Genet 2009;5:e1000372

98. Kettleborough RN, de Bruijn E,

van Eeden F, et al. High-throughput

target-selected gene inactivation in

zebrafish. Methods Cell Biol

2011;104:121-7

99. Kharraz Y, Guerra J, Mann CJ, et al.

Macrophage plasticity and the role of

inflammation in skeletal muscle repair.

Mediators Inflamm 2013;2013:491497

100. Renshaw SA, Trede NS. A model

450 million years in the making:

zebrafish and vertebrate immunity.

Dis Model Mech 2012;5:38-47

101. Koshimizu E, Imamura S, Qi J, et al.

Embryonic senescence and laminopathies

in a progeroid zebrafish model.

PLoS One 2011;6:e17688

AffiliationLisa Maves1,2 PhD1Acting Assistant Professor,

University of Washington School of Medicine,

Department of Pediatrics, Division of

Cardiology, Seattle, WA, USA2Seattle Children’s Research Institute, Center for

Developmental Biology and Regenerative

Medicine, 1900 Ninth Avenue, Seattle,

WA 98101, USA

Tel: +1 206 884 1052;

E-mail: [email protected]

Recent advances using zebrafish animal models for muscle disease drug discovery

Expert Opin. Drug Discov. (2014) 9(9) 1045

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y Q

UT

Que

ensl

and

Uni

vers

ity o

f T

ech

on 1

0/13

/14

For

pers

onal

use

onl

y.


Recommended