25
Muscle Diseases: The Muscular Dystrophies Elizabeth M. McNally and Peter Pytel Department of Medicine, Section of Cardiology, University of Chicago, Chicago, Illinois 60637; email: [email protected] Department of Pathology, University of Chicago, Chicago, Illinois 60637; email: [email protected] Annu. Rev. Pathol. Mech. Dis. 2007. 2:87–109 The Annual Review of Pathology: Mechanisms of Disease is online at pathmechdis.annualreviews.org This article’s doi: 10.1146/annurev.pathol.2.010506.091936 Copyright c 2007 by Annual Reviews. All rights reserved 1553-4006/07/0228-0087$20.00 Key Words myotonia, sarcopenia, muscle regeneration, dystrophin, lamin A/C, nucleotide repeat expansion Abstract Dystrophic muscle disease can occur at any age. Early- or childhood- onset muscular dystrophies may be associated with profound loss of muscle function, affecting ambulation, posture, and cardiac and respiratory function. Late-onset muscular dystrophies or myopathies may be mild and associated with slight weakness and an inability to increase muscle mass. The phenotype of muscular dystrophy is an endpoint that arises from a diverse set of genetic pathways. Genes associated with muscular dystrophies encode proteins of the plasma membrane and extracellular matrix, and the sarcomere and Z band, as well as nuclear membrane components. Because muscle has such distinctive structural and regenerative properties, many of the genes implicated in these disorders target pathways unique to muscle or more highly expressed in muscle. This chapter reviews the basic structural properties of muscle and genetic mechanisms that lead to myopathy and muscular dystrophies that affect all age groups. 87 Annu. Rev. Pathol. Mech. Dis. 2007.2:87-109. Downloaded from arjournals.annualreviews.org by Stanford University Robert Crown Law Lib. on 10/27/08. For personal use only.

Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Muscle Diseases: TheMuscular DystrophiesElizabeth M. McNally and Peter PytelDepartment of Medicine, Section of Cardiology, University of Chicago, Chicago,Illinois 60637; email: [email protected]

Department of Pathology, University of Chicago, Chicago, Illinois 60637;email: [email protected]

Annu. Rev. Pathol. Mech. Dis. 2007.2:87–109

The Annual Review ofPathology: Mechanisms of Disease is online atpathmechdis.annualreviews.org

This article’s doi:10.1146/annurev.pathol.2.010506.091936

Copyright c© 2007 by Annual Reviews.All rights reserved

1553-4006/07/0228-0087$20.00

Key Words

myotonia, sarcopenia, muscle regeneration, dystrophin, lamin A/C,nucleotide repeat expansion

AbstractDystrophic muscle disease can occur at any age. Early- or childhood-onset muscular dystrophies may be associated with profound lossof muscle function, affecting ambulation, posture, and cardiac andrespiratory function. Late-onset muscular dystrophies or myopathiesmay be mild and associated with slight weakness and an inability toincrease muscle mass. The phenotype of muscular dystrophy is anendpoint that arises from a diverse set of genetic pathways. Genesassociated with muscular dystrophies encode proteins of the plasmamembrane and extracellular matrix, and the sarcomere and Z band,as well as nuclear membrane components. Because muscle has suchdistinctive structural and regenerative properties, many of the genesimplicated in these disorders target pathways unique to muscle ormore highly expressed in muscle. This chapter reviews the basicstructural properties of muscle and genetic mechanisms that lead tomyopathy and muscular dystrophies that affect all age groups.

87

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 2: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Musculardystrophy:genetically linkedmuscle diseasecharacterized byprogressivedestruction ofmuscle tissue

Sarcomere: basicfunctional unit ofmuscle contractionconsisting of actin,myosin, andassociated proteins

MUSCLE STRUCTURE

The basic cellular unit of voluntary mus-cle is the myofiber. Myofibers are elongated,multinucleate cells. During development andregeneration, myofibers arise from the fusionof singly nucleated myoblasts to form myo-fibers. Individual myofibers are encased in anendomysial sheath of connective tissue, andgroups of myofibers are encased in epimysialconnective tissue (Figure 1). The ends ofmyofibers form myotendinous junctions, aspecialized attachment for bony insertion thatcan withstand considerable force. The cyto-plasm of each individual myofiber is highly

Figure 1Normal muscle histology. (a) Low-power H&E-stained cross section of normal skeletal muscle showingthe organization of myofibers into fascicles separated by epimysial connective tissue. Overall, there islittle variation in myofiber size. (b) A high-power H&E-stained cross section of normal skeletal muscle.The myofiber nuclei are located peripherally, just below the sarcolemmal membrane. Only thin strandsof connective tissue separate individual myofibers within each of the three depicted fascicles. (c) Ahigh-power H&E-stained longitudinal section of normal muscle shows cross striation, which is evidenceof the highly organized cytoskeletal architecture of the myofiber. (d ) The ATPase reaction candistinguish fiber types. At the pH shown here (pH 9.4), type I fibers are light and type II fibers are dark.The checkerboard pattern reflects the normal fiber-type distribution.

organized, containing chains of sarcomeresthat run parallel to the length of the myofiber.Electron-dense Z bands define the bordersof each sarcomere, and individual sarcomeresare composed of actin-containing thin fila-ments and myosin-containing thick filaments(Figure 2). The heavy chain of myosin hy-drolyzes ATP to provide the energy for mus-cle contraction. The rate at which myosin hy-drolyzes ATP is proportional to the speed ofmuscle contraction. Slow type I and fast typeII fibers express different myosin isoforms andcan be distinguished by enzyme histochem-istry in the ATPase reaction (Figure 1d ). The

88 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 3: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

fiber type is determined by the innervatingmotor neuron in the spinal cord. The motorunit is the functional unit of one motor neu-ron and all the myofibers innervated by it.

Several groups of muscle diseases presentas weakness, cramping, or muscle pain. Theseinclude the congenital myopathies, the mus-cular dystrophies, myotonic disorders, storagediseases, mitochondrial diseases, and inflam-matory myopathies. These primary musclediseases are distinct from neuropathic diseasesand disorders of neuromuscular transmission.This review focuses on myopathies arisingfrom defects of cytoskeletal, sarcomeric, andmembrane-associated proteins of the plasmamembrane and the nucleus (Table 1). Histor-ically, the diseases that fall into this group wereclassified on the basis of features such as thepattern of inheritance, the age of disease on-set, and the primarily affected muscle groups.The increasing ability to link these entities tospecific genetic defects has greatly increasedour understanding of these diseases.

MUSCLE DEGENERATION ANDREGENERATION

Muscle damage typically takes the form of my-ofiber necrosis and regeneration (Figure 3).This process can be segmental, involving onlya part of an individual myofiber. The necrosisis associated with membrane damage and leak-age of cytoplasmic proteins, such as creatinekinase (CK) and lactate dehydrogenase, thatcan serve as serum markers of muscle dam-age. The basement membrane of the necroticfiber remains as a scaffold for the regener-ative process. Muscle is highly regenerative.The regenerative process occurs with the or-ganization of necrotic cytoplasmic debris byinflammatory cells. Depending on the degreeof damage, the time course of muscle regener-ation occurs over one to three weeks. Maturemuscle contains mononuclear cells that re-side between the basement membrane andthe plasma membrane or sarcolemma of eachmyofiber. The nuclei of these mononuclearcells selectively take up bromodeoxyuridine,

Figure 2Transmission electron micrograph and schematic representation of asarcomere, the basic functional contraction unit in skeletal and cardiacmuscle. The A band (anisotropic as seen under polarized light) marks theextent of the thick myosin filaments and is indicated by the green box in theschematic diagram. The thin actin filaments are anchored at the Z disc.The area where the thin filaments do not overlap with the thick filaments isthe I band (isotropic as seen under polarized light). The membraneoverlying the sarcomere is also shown.

indicating mitotic activity. These two fea-tures, the mitotic activity and the location be-tween the plasma membrane and the basallamina, were used to define muscle satellitecells (1). Experimental evidence supports thatthe origin of the satellite cell is the myotome(2), but additional evidence suggests that

www.annualreviews.org • The Muscular Dystrophies 89

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 4: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Sarcolemma: thespecialized plasmamembrane of theindividual myofibersin skeletal muscle

Satellite cell: poolof localized tissuestem cells in theskeletal muscleimportant for itsdevelopment andregenerativepotential

CMD: congenitalmuscular dystrophy

Table 1 Summary of different genes orgene products and the type of muscledisease with which they associate

Gene product (gene) DiseaseLaminin α2 (LAMA2) CMDIntegrin α7 (ITGA7 ) CMDFukutin (FCMD) CMDPOMGnT1 CMDPOMT1 WWSFKRP CMD, LGMDLARGEDystrophin (DYS ) DMDα-sarcoglycan (SGCA ) LGMDβ-sarcoglycan (SGCB ) LGMDγ-sarcoglycan (SGCG ) LGMDδ-sarcoglycan (SGCD) LGMDDysferlin (DYSF ) LGMDCalpain-3 (CAPN3 ) LGMDCaveolin-3 (CAV3) LGMD, RMDE3-ubiquitin ligase(TRIM 32)

LGMD

Telethonin LGMDTitin LGMDMyotilin LGMDLamin A/C (LMNA) LGMD, EDMDEmerin (EMD) EDMD

Color coding reveals those genetic defects that arefunctionally related (see text). POMT: proteinO-mannosyltransferase; DMD, Duchennemuscular dystrophy; LGMD, limb-girdle musculardystrophy; CMD, congenital muscular dystrophy;EDMD, Emery-Dreifuss muscular dystrophy;WWS, Walker-Warburg syndrome; RMD,rippling muscle disease.

bone marrow–derived cells may contribute tomuscle regeneration (3).

Satellite cells are activated by injury. Asquiescent cells, satellite cells divide and main-tain the satellite cell pool. With injury, satel-lite cells activate and then differentiate intomyoblasts (for a review, see Reference 4).Myoblasts represent a committed cell thatwill eventually withdraw from cell cycle ac-tivity and express genes found in myotubes.Myoblasts can fuse to each other or to exist-ing myotubes to generate new muscle. Thenuclei from myoblasts that have recently fusedto an existing myofiber are found in the cen-ter of myotubes. Within one to three months,

these nuclei will assume a peripheral positionnear the sarcolemma, as is characteristic of themature myofiber.

Because most muscle disease involvesmyofiber damage or degeneration, regenera-tion is a feature of all muscle disease. Althoughcentral nucleation is seen in normal musclefiber, an increase in the number of centrallynucleated myofibers may indicate an ongoingdegenerative or necrotic process. The musclebiopsy, viewed perpendicular to the long axisof the myofiber, remains the gold standard forevaluating muscle disease. The main musclebiopsy changes found in myopathic diseasesare the presence of necrotic myofibers andregenerative myofibers. Necrotic myofiberscontain variable amounts of amorphous cel-lular debris, inflammatory cells, and satellitecells. Regenerative myofibers are character-ized by their enlarged nucleus and basophilicRNA-rich cytoplasm. After the reconstitutionof the sarcomeric myofiber architecture, themyofiber nuclei maintain a centralized posi-tion away from their normal subsarcolemmallocation for several weeks. In addition, myo-pathic diseases also show an increased varia-tion in myofiber size with regenerating fibersthat tend to be smaller, and with unaffectedfibers that may show compensatory hypertro-phy. In diseases associated with prolonged,continued myofiber injury, the regenerativeprocess fails to maintain normal skeletal mus-cle architecture. In these cases, increased con-nective tissue in the form of interstitial fibro-sis and fatty replacement are evidence of thechronicity of the disease (Figure 3).

CONGENITAL MUSCULARDYSTROPHIES

The congenital muscular dystrophies(CMDs) are apparent at birth, manifestingfrequently as a “floppy” infant lacking muscletone. CMD is a feature of Walker-WarburgSyndrome, muscle-eye-brain disease, andFukuyama-type CMD. In these disorders,additional neurologic features such aslissencephaly and ocular and retinal defects

90 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 5: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Figure 3Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers. The outline of theoriginal fiber is still detectable. The fibers are filled with cellular debris and inflammatory cells. Satellitecells cannot be reliably distinguished from mononuclear inflammatory cells. (c, d ) Examples ofregenerating myofibers with more blue-purple, basophilic cytoplasm, and enlarged activated nucleus.Some of these myofiber nuclei are internalized and do not occupy the normal subsarcolemmal location.

may occur and may reflect a failure ofneuronal migration. The genes mutatedin the CMDs encode enzymes that con-tribute to O-linked glycosylation (5–9). Ingeneral, asparagine-linked glycosylationis common to more proteins, with only aminority of proteins undergoing O-linkedglycosylation—usually on serine residues. Amajor target protein affected by these diversegenetic disorders is α-dystroglycan, as it isknown to undergo O-linked glycosylation(10, 11). As discussed in more detail below,dystroglycan is a core component of the dys-trophin glycoprotein complex (DGC) (12).Dystroglycan is composed of two subunitsproduced from a single gene (Figure 4).Dystroglycan is broadly expressed, andα-dystroglycan is variably glycosylatedwith tissue specificity (13). β-dystroglycanis a transmembrane protein that anchorsα-dystroglycan.

Although extramuscular involvement iscommon in CMDs, not all genes in thisclass lead to extramuscular involvement. Forexample, mutations in the gene encodingthe fukutin-related protein (FKRP) associate

DGC: dystrophinglycoproteincomplex

more with muscle defects with less severe orabsent central nervous system findings (14,15). The degree to which there are central andperipheral nervous system findings can be par-tially explained by the specific responsible mu-tations. As such, genotype-phenotype corre-lation in these disorders may, to some degree,be predicted by the amount of glycosylated α-dystroglycan present. In CMDs, muscle hasa dystrophic appearance and, consistent withthis, patients may have an elevated serumCK, indicating disruption of the sarcolemma(16). Immunostaining of muscle biopsies withantibodies directed at core residues in α-dystroglycan may appear normal, as mayantibodies directed at β-dystroglycan. Im-munostaining with antibodies directed atthe glycosylated forms of α-dystroglycangenerally show a reduction proportional tothe severity of disease. The decrease in α-dystroglycan glycosylation may be nonuni-form along the length of the myofiber. Thegenes associated with CMDs are thought toencode glycosyltransferases (Figure 5) (17,18). Where antibodies have been generated,it appears these proteins are resident in the

www.annualreviews.org • The Muscular Dystrophies 91

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 6: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Mucin

TMD Dystrophinbinding

Figure 4Dystroglycan structure. The dystroglycan complex has a single transmembrane domain and a shortcytoplasmic tail that contains a dystrophin-binding domain. Through its extracellular domain, it can bindto laminin α2 in the basement membrane. Dystroglycan is posttranslationally cleaved at its aminoterminus. Dystroglycan contains residues that serve as anchors for O-linked as well as N-linkedglycosylation. TMD, transmembrane domain.

β

Dystroglycan

Actin

Dystrophin

Sarcoglycan

Sarcospan

Plasmamembrane

Golgiapparatus

Endoplasmicreticulum

POMGnT1 FKRP

FukutinLARGE

POMT1

POMT2

Nucleus

αDG

Figure 5Glycosylation defects associated with congenital muscular dystrophy. During posttranslationalprocessing in the rough endoplasmic reticulum and the Golgi apparatus, dystroglycan is glycosylated.Deficiency in any of these enzymes has been linked to diseases and muscle dysfunction. POMGnT1,protein O-mannose 1,2-N-acetylglucosaminyl transferase; FKRP, fukutin-related protein; POMT1/2,protein O-mannosyltransferase; LARGE, like acetylglucoseaminyltransferase; DG, dystroglycan.

92 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 7: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Golgi apparatus or in the endoplasmic orsarcoplasmic reticulum (19–22).

One form of CMD arises from mutationsin the gene encoding the α2 chain of laminin(23, 24). In muscle, the major basement pro-tein laminin is composed of the α1, α2, andγ chains to form merosin or laminin-2. Al-though the defect here is not one that af-fects glycosylation of dystroglycan directly,merosin is a major ligand for α-dystroglycan;thus, disruption between the extracellular ma-trix and the membrane occurs, albeit by amechanism distinct from that in the CMDslisted above.

THE MUSCULAR DYSTROPHIES

Duchenne Muscular Dystrophy

Muscular dystrophies in children or adults arecharacterized by a progressive muscle weak-ness that may have a predilection for cer-tain muscle groups. Childhood-onset mus-cular dystrophies are usually lethal, owingto associated cardiac muscle or respiratorymuscle weakness. Duchenne muscular dystro-phy (DMD) represents the most common X-linked inherited disorder. DMD is producedby mutations in the dystrophin gene on the Xchromosome, and most affected boys are diag-nosed in the first few years of life. DMD boysdisplay delayed walking, falling, a toe gait, andcalf hypertrophy. Commonly, serum CK lev-els are substantially elevated. Gross deletionsor duplications in the dystrophin gene accountfor 60% of mutations, and these mutations aredetected by a PCR-based blood test (25). Theefficacy of this test is such that many DMDboys are no longer subjected to muscle biopsy.

As much as 40% of DMD patients donot have a large gene deletion or duplication;instead, a point mutation is responsible fortheir disorder (26). A number of these pointmutations may insert novel stop codons, andthis is the mechanism for the dystrophic phe-notype in the mdx mouse model. The mdxmouse displays many of the histopathologicfeatures seen in DMD, but these mice—unlike

DMD: Duchennemuscular dystrophy

Stop codonread-through:continuedtranscription of aprotein that occursthrough suppressingthe effect of a stopcodon

BMD: Beckermuscular dystrophy

their human counterparts—remain ambula-tory and have only a mildly reduced lifespan.A point mutation in exon 23 creates a novelstop codon and truncation of the dystrophinprotein (27). Recent strategies for treatmentdesign target stop codon read-through as amechanism for treating these DMD patients(28). At least 10% of DMD patients may betreated with this approach. Novel agents withimproved read-through capabilities and a re-duced side-effect profile are being developedand tested. These compounds are associatedwith a systemic delivery that leads to an in-crease of 10–30% over normal dystrophinprotein levels (29).

Gene deletions that only partially disruptdystrophin protein expression are usually as-sociated with the milder phenotype of Beckermuscular dystrophy (BMD). Many of thesemutations produce internally deleted dys-trophin that lacks spectrin repeats but main-tains the core actin-binding and carboxy-terminal regions. For many BMD patients,a muscle biopsy may be helpful in pro-viding a diagnosis that may be essentialfor genetic counseling. Maternal carriers ofDMD-associated dystrophin gene mutationscan show symptoms of mild muscle weak-ness. Cardiomyopathy may manifest in fe-male dystrophin mutation carriers owing toX-inactivation, which affects the wild-typedystrophin locus that remains in these women(30).

Dystrophin is a large protein and a centralcomponent of the DGC that provides stabil-ity to the sarcolemma (Figure 6). Dystrophinhas a calponin-like actin-binding domain at itsamino terminus and 24 spectrin repeats inter-rupted by four hinge points (31). The carboxylterminus of dystrophin binds β-dystroglycandirectly (32). The amino terminus and regionsalong the spectrin-repeat rod domain bind tocytoplasmic γ-actin, forming a mechanicallystrong link (33). In the absence of dystrophin,muscle contraction enhances membrane dis-ruption and produces myofiber damage. Dis-ruption of the myofiber can be imaged byuptake of the vital tracer Evans blue dye

www.annualreviews.org • The Muscular Dystrophies 93

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 8: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Integrin Sarcoglycan

β

β1

β-synα-syn

NOS

β

Dystroglycan

αα7α

γ δ

ActinActin SSN

Filamin

Caveolin

Dystrophin

Dystrobrevin

Figure 6The dystrophin glycoprotein complex (DGC). The main components of the DGC are the sarcoglycancomplex, the dystroglycan complex, and dystrophin. Through binding to laminin in the basementmembrane on the extracellular site and binding to actin on the cytoplasmic site, this complex is thoughtto provide stability to the sarcolemma during the mechanical changes that accompany musclecontraction. Neuronal nitric oxide synthase (NOS), dystrobrevins, syntrophins (α-syn and β-syn),filamin, and sarcospan (SSN) are additional dystrophin-associated proteins.

nNOS: neuronalnitric oxide synthase

(34, 35). Gene replacement experiments haveidentified a minimal region of dystrophin re-quired to protect muscle against contraction-induced damage (36). These experiments in-dicate that an intact actin-binding domain,four spectrin repeats, and the carboxyl termi-nus are needed to replace dystrophin func-tion. Membrane disruption that arises fromthe loss of dystrophin leads to an increasein intracellular calcium content, which pro-motes a series of pathogenic events includingcalcium-activated proteolysis and sarcomeredysfunction (37).

The carboxyl terminus of dystrophinlinks it to the remainder of the membrane-associated DGC (38, 39). The DGC includescytoplasmic and transmembrane components,and the entire complex encompasses mechan-ical and signaling roles (40, 41). The cyto-plasmic components of this complex includethe dystrobrevins, small proteins that binddirectly to dystrophin, and have homologyto the carboxyl terminus of dystrophin. ThePDZ domain-containing syntrophins bind todystrobrevin and to neuronal nitric oxide syn-thase (also known as nNOS or NOS1). In theabsence of dystrophin, the cytoplasmic DGCcomponents, in addition to the transmem-brane components dystroglycan and sarco-

glycan, are destabilized at the sarcolemma.The loss of dystrophin leads to the displace-ment of nNOS from the sarcolemma, and thisdisplacement mediates abnormal contraction-induced vasorelaxation (42, 43). The relation-ship of this loss to disease pathogenesis is notentirely clear because the loss of nNOS fromthe plasma membrane is insufficient to pro-duce a dystrophic phenotype (44). It is morelikely that the loss of nNOS from the mem-brane plays a contributory role in that it mayaugment tissue damage in response to dys-trophin loss.

In DMD, the muscle biopsies show my-opathic changes with myofiber degenera-tion and regeneration (Figure 7). Withdisease progression, the muscle shows in-creasing fatty replacement and endomysial fi-brosis. Immunohistochemistry demonstratesthe absence of the normal sarcolemmal stain-ing for dystrophin. Immunostaining biopsiesof DMD patients for the remaining DGCcomponents reveal secondary deficiencies ofthe sarcoglycans, dystroglycan, and the syn-trophins as evidence of the incomplete assem-bly of the remaining DGC in the absence ofdystrophin. In BMD, the biopsy changes de-velop at a much more protracted pace, and im-munohistochemical staining may only show

94 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 9: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Figure 7Examples of skeletal muscle biopsies in muscular dystrophies. (a, b) Biopsies from two brothers, agedseven and nine years old, respectively. The progression in the morphologic changes is clearly visible withmore severe fibrosis and fatty replacement, as well as more variation in fiber size in the latter (b) biopsy.(c) By immunohistochemistry staining, the biopsies of the above patients showed absent staining fordystrophin. The inset illustrates the normal continuous membranous staining found in the control tissue.(d ) Biopsy of a 40-year-old female with limb-girdle muscular dystrophy. The biopsy shows variation infiber size, fibrosis, and focal fatty replacement. (e) Immunohistochemical studies confirm the absence ofdysferlin. The normal staining for dysferlin is shown in the inset. ( f ) A case of myotonic dystrophy thatshows variation in myofiber size and abundant internalized central nuclei. As illustrated by this biopsy,chronic changes such as fibrosis and fatty replacement are more variable in myotonic dystrophy.

www.annualreviews.org • The Muscular Dystrophies 95

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 10: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

LGMD: limb-girdlemuscular dystrophy

deficient staining for certain domains of theprotein.

Limb-Girdle Muscular Dystrophies:The Sarcoglycans

The sarcoglycans are transmembrane ele-ments that form a tight unit within the DGC(40, 45). There are at least six sarcogly-can proteins, although the major sarcogly-can complex found at the muscle membraneis composed of four sarcoglycan proteins,α, β, γ, and δ. Mutations in these genesare the underlying defects in a subset ofthe recessively inherited limb-girdle muscu-lar dystrophies (LGMDs). ε-sarcoglycan wasidentified on the basis of its high homol-ogy to α-sarcoglycan, and mutations in thegene encoding ε-sarcoglycan lead to my-oclonic dystonia (46). ζ -sarcoglycan was iden-tified on the basis of its homology to γ-and δ-sarcoglycan, and mutations in this genehave not been described (47). Patients withLGMD due to sarcoglycan gene mutationshave a presentation similar to the pheno-typic range seen in DMD and BMD. Mu-tations in the α-sarcoglycan gene are fre-quently point mutations that may associatewith milder phenotypes (48). Frameshiftingand select point mutations may lead to severephenotypes. There is a common frameshiftingdeletion in the gene encoding γ-sarcoglycanthat has been described in many distinct pop-ulations consistent with a common disease al-lele (49). The identical mutation in the γ-sarcoglycan gene can produce a phenotypeof varying disease severity in humans and inmouse when placed in different genetic back-grounds. These findings suggest that geneticmodifiers may influence the phenotypic out-come in the muscular dystrophies (50).

The basic pathogenic features in musclebiopsies from sarcoglycan mutant patients areindistinguishable from those found in DMDor BMD muscle. Antibody staining to thesarcoglycan subunits is often all depleted in re-sponse to mutations in any single sarcoglycangene (51). An exception to this is with muta-

tions in γ-sarcoglycan, where residual sarco-glycan staining can be present but the pheno-type may still be severe. α-sarcoglycan genemutations may also display residual sarco-glycan expression at the plasma membrane.Mutations in sarcoglycan subunits generallydo not affect the distribution of dystrophin.Thus, mutations in dystrophin lead to the lossof the sarcoglycan subunits from the plasmamembrane, but the reverse is not true as sarco-glycan gene mutations leave dystrophin in-tact. As the phenotypes are equally severefrom dystrophin and sarcoglycan gene mu-tations, it is therefore the disruption of thesarcoglycan complex, as the common molec-ular feature, that is critical for the dystrophicprocess. Mutations in γ- and δ-sarcoglycanproduce a similar phenotype in mouse mod-els. Despite phenotypic similarities, these twomutations do not cause similar disruptions inthe sarcoglycan complex, but result in mus-cle damage through different pathways. Lossof δ-sarcoglycan causes contraction-inducedmuscle damage similar to that seen in dys-trophin mutations (52). The γ-sarcoglycanmutation is not associated with the same typeof mechanical damage, implicating a separatemechanism for the myopathic changes. Theseresults indicate that the sarcoglycan complexmay serve important functions beyond main-taining mechanical strength as part of theDGC.

Smooth muscle dysfunction may bepresent in DMD patients. However, thereare differences between the DGC in striatedand smooth muscle. Notably, the sarcogly-can complex in vascular smooth muscle iscomposed of ε-, β-, δ-, and ζ -sarcoglycans(53). Interestingly, vascular spasm can occurin response to sarcoglycan gene mutations(54). Vascular spasm is thought to be mostpathogenic to the heart, affecting the coro-nary artery vasculature. Restoration of car-diomyocyte δ-sarcoglycan expression in thebackground of δ-sarcoglycan null animals wassufficient to eliminate coronary artery vascu-lar spasm. Therefore, vascular smooth muscledefects in DGC mutations can arise from

96 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 11: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

vascular smooth muscle cell extrinsicprocesses (55).

Limb-Girdle Muscular Dystrophies:Dysferlin

Dysferlin is the protein product of the LGMDtype 2B locus and is a membrane-associatedprotein with a long cytoplasmic domain (56).Dysferlin is not associated with the DGCand plays a distinct pathogenic role whendisrupted. Dysferlin is homologous to theCaenorhabditis elegans protein fer-1, named forits role in fertilization defects in C. elegansmutants (57). In fer-1 mutants, there is a de-fect in vesicle fusion to the plasma mem-brane of the maturing sperm, leading to fer-tilization defects (58). Specifically, in fer-1mutants, there is an accumulation of submem-branous vesicles. Similarly, in dysferlin mu-tant muscle, there is also an accumulation ofsubmembranous vesicles (59–61). In a man-ner analogous to fer-1, dysferlin mediatesmembrane-resealing events required for themuscle membrane repair in mature muscle(62). Laser-mediated sarcolemmal disruptionsare repaired much more slowly in dysfer-lin mutant muscle compared with normalmuscle (61). In these studies, it was con-firmed that sarcolemmal resealing in responseto damage is a calcium-sensitive event. In-terestingly, resealing in the absence of cal-cium occurred at the same slow pace as re-sealing in the absence of dysferlin. Thesefindings are consistent with a role for dys-ferlin as the calcium sensor for vesicle fu-sion that mediates sarcolemmal resealing(63).

The cytoplasmic domain of dysferlin con-tains six C2 domains, and C2 domains areimplicated in calcium and phospholipid bind-ing. The C2 domains of the synaptotagminsdisplay homology to those found in dysferlin,and synaptotagmins mediate the calcium sen-sitivity of membrane fusion events associatedwith the neurotransmitter release at nerveterminals (64). The first C2 domain of dys-ferlin binds a mixture of phosphotidylserine

and phosphotidylcholine only in the presenceof physiologically relevant calcium concen-trations (65). Calcium-dependent phospho-lipid binding is abolished by a point muta-tion in the first C2 domain of dysferlin, andthis mutation produces muscular dystrophy.The closely related protein myoferlin is highlyexpressed in myoblasts undergoing fusion tomyotubes, and its first C2 domain displayssimilar phospholipid-binding capacity to thatseen for dysferlin (66). Mice lacking myofer-lin display reduced muscle size, and in culturemyoferlin null myoblasts fuse less well and donot form large myotubes (67). These findingsimplicate myoferlin in the membrane fusionevents associated with myoblast fusion to ex-isting myotubes.

LGMD 2B muscle biopsies display find-ings similar to those associated with otherforms of muscular dystrophy, with the ex-ception that an inflammatory infiltrate maybe seen as a prominent feature with dysferlingene mutations (68, 69). This finding may beso great as to mimic inflammatory myopathiessuch as what is seen in polymyositis or inclu-sion body myositis (70). A subset of biopsiesmay show a decrease in dysferlin staining,yet have a normal dysferlin gene indicat-ing an alternative mechanism for reducingdysferlin and for producing muscle pathol-ogy. Miyoshi myopathy is a mild form ofmuscular dystrophy associated with dysferlingene mutations that selectively affects the gas-trocnemius muscle but spares other muscula-ture. The identical mutation can be associ-ated with the more severe LGMD or Miyoshimyopathy, indicating that additional genesand/or environmental factors may contributestrongly to modulate the dystrophic process(71).

Limb-Girdle Muscular Dystrophies:Calpain, Titin, Caveolin, TRIM32,Myotilin, and Telethonin

LGMD 2A is a recessive form of musculardystrophy associated with homozygous mu-tation in the gene encoding calpain-3 (also

www.annualreviews.org • The Muscular Dystrophies 97

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 12: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

EDMD:Emery-Dreifussmuscular dystrophy

Nuclear membraneproteins: proteinsthat may play a rolein mechanicalfunctions and generegulation

known as p94). These mutations are com-mon and lead to a progressive loss of musclefunction (72). Mice lacking calpain-3 havebeen generated and recapitulate aspects ofthe human phenotype of LGMD. Calpain-3likely has a number of proteolytic targets, butfilamin C, a muscle-specific filamin, is cleavedby calpain-3 in a manner that alters the bind-ing of filamin C to subunits of the sarcogly-can complex (73). Calpain-3 is important forsarcomere turnover, and its pathways are dis-tinct and upstream from ubiquitin. In addi-tion, calpain-3 binds directly to titin, the giantprotein that spans sarcomeres (74). Mutationsin titin lead to LGMD and the milder tibialmyopathy. The murine model muscular dys-trophy with myositis (mdm) is associated witha mutation in titin’s NB2 domain that inter-acts with calpain-3 (75).

Several other gene products have been im-plicated in the formation of muscular dystro-phy, including caveolin-3, TRIM32, and my-otilin. Caveolae are membrane invaginationsthat participate in localizing components andproteins in the membrane. Caveolin-3 isfound inserted in the membrane, and dom-inant and recessive mutations are associatedwith muscular dystrophy, as well as with moremild disorders (76). Caveolin-3 may be selec-tively reduced in response to dysferlin genemutations (77). TRIM32 is a ubiquitin ligasethat binds to myosin and ubiquitinates actinand thereby participates in sarcomere recy-cling (78). Myotilin is a Z band–associatedprotein that binds to other Z band pro-teins, and defects in myotilin lead to dom-inantly inherited muscular dystrophy (79).The full role of myotilin is not appreciated,but its pathogenicity may be related to whatis seen from mutations in other sarcomere-associated genes such as telethonin or, po-tentially, α-actinin (80). In addition, a subsetof these disorders is associated with nemalinemyopathy, where an accumulation of nema-line rods can be found in the cytoplasm asan indicator of a pathologic disease process(81).

MUTATIONS OF NUCLEARMEMBRANE–ASSOCIATEDGENES

Emery Dreifuss muscular dystrophy (EDMD)is an X-linked disorder associated with pro-gressive muscle weakness and contractures.With EDMD, cardiac involvement such asatrioventricular heart block is frequent. Mu-tations in the gene encoding the nuclearmembrane protein emerin produce EDMD(Figure 8). Emerin is a 34-kDa proteinthat embeds in the inner nuclear membrane.Emerin contains within its primary struc-ture an LEM domain, named for its presencein lamin-associated protein 2, emerin, andMAN-1. The LEM domain of emerin binds tobarrier-to-autointegration factor (BAF). BAFis small peptide that oligomerizes and directlybinds to DNA (82). Its position at the nu-clear membrane coupled with its partneringto BAF suggests that the inner nuclear mem-brane may play a role in scaffolding chro-matin. Emerin is a broadly expressed protein,yet mutations in this gene primarily affectmuscle (83).

Dominantly inherited forms of EDMD(AD-EDMD) are more common (84). Mu-tations in the gene encoding inner nuclearmembrane protein lamin A/C (LMNA gene)are responsible for AD-EDMD. Lamins Aand C are type V intermediate filament pro-teins that form much of the structural appa-ratus of the inner nuclear membrane of post-mitotic cells (85). Lamins A and C dimerizeand then form higher-order structures to pro-vide tensile strength to the nucleus. In mostpostmitotic cells, lamins A and C dominatethe composition of the inner nuclear mem-brane intermediate filaments. In cells under-going division, lamin B is more likely to bethe dominant intermediate filament proteinof the inner nuclear membrane.

Exactly how mutations in broadly ex-pressed genes lead to tissue-specific pheno-types that affect muscle is not known. Sev-eral nonexclusive hypotheses may explain thisphenomenon. LMNA gene mutations may

98 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 13: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Nuclearlamina

Outer nuclear membrane

Figure 8Nuclear membrane proteins. Lamin A/C forms a scaffold at the inner nuclear membrane linked to otherproteins, including emerin ( yellow), nesprin 1-α ( green), lamin-associated protein 2 (LAP2) (blue), andMAN-1 ( purple). These proteins are thought to be involved in providing mechanical strength to thenucleus. In addition, LAP2, emerin, and MAN-1 bind directly to barrier-to-autointegration factors thatcan bind heterochromatin.

render the nuclear membrane weakened suchthat abnormal function develops when thesenuclei are subjected to the force associatedwith muscle contraction. This mechanicalweakness hypothesis may explain some of thesusceptibility of striated muscle to mutationsand defects in the nuclear membrane. Evi-dence supporting the mechanical hypothesiswas found in murine cells engineered to lackLMNA (86). In these homozygous null cells,maximal normalized nuclear strain was in-creased in LMNA null fibroblasts. Cytoskele-tal stiffness was reduced and nuclear fragilitywas increased in LMNA fibroblasts. Interest-ingly, defective nuclear factor-kappa B (NF-κB) signaling was associated with the loss ofLMNA. These observations were made in fi-broblasts from LMNA null mice, and there-fore investigators argued that those cells mostsubject to stress and thus strain, such as mus-cle, would be the most adversely affected byloss of LMNA. A similar defect was associatedwith the loss of emerin (87).

Many mutations in LMNA are not loss-of-function mutations, but instead are domi-nant point mutations that may produce gain-of-function activity and affect other attributesof the nuclear membrane. A second hypothe-sis to explain how LMNA gene defects tar-get certain cells and tissues may relate to

DM: myotonicdystrophy

other nuclear functions perturbed by muta-tions in LMNA. The nuclear membrane reg-ulates intracellular transport, DNA synthesis,and gene transcription. The observation thatmany LMNA mutations preferentially targetpostmitotic cells, such as myofibers and car-diomyocytes, may indicate defects in nucleartransport or that gene expression may morelikely explain the underlying cellular defectassociated with LMNA gene mutations. Notall LMNA mutations may lead to phenotypethrough the same mechanisms. In support ofthis, recent data indicate that LMNA mutantsare associated with defects in skeletal muscleregeneration (88–90). The regenerative de-fect may explain aspects of the muscle phe-notype seen in autosomal-dominant EDMDpatients, but may not fully explain the accom-panying cardiac defects.

MYOTONIC DYSTROPHY

Myotonic dystrophy type 1 (DM1, orSteinert’s disease) is one of the most commongenetic disorders, affecting 1 in 8000 individ-uals. DM1 is associated with a trinucleotideexpansion on chromosome 19. In subsequentgenerations within a family, increased ex-pansion of this repeat sequence produces anearlier age of onset consistent with classic

www.annualreviews.org • The Muscular Dystrophies 99

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 14: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Myotonia: delayedrelaxation aftervoluntary musclecontractionassociated withcharacteristicelectrophysiologicalchanges

Nucleotide repeatexpansion:expansion of normalrepeats that altersgene expression orcauses abnormalRNA or proteinaccumulation

genetic anticipation, similar to neurodegen-erative disorders such as Huntington’s or thespinocerebellar ataxias. The findings in DM1are those of progressive muscle weakness andmyotonia. Myotonia is characterized by a de-lay in muscle relaxation after normal contrac-tion and by characteristic changes on elec-tromyography. The typical electromyogra-phy manifestations are high-frequency musclefiber discharges of waxing and waning ampli-tude. Extramuscular findings include cardiacatrioventricular heart block and cardiomy-opathy, cataracts, testicular failure, disruptionof sleep, and profound fatigue (91). Neuropsy-chiatric abnormalities may also be present.The trinucleotide expansion on chromosome19 falls within the 3′ end of the myotonic dys-

trophy protein kinase (DMPK) gene, and as-pects of the cardiac dysfunction may relate tothe disruption of the function of DM proteinkinase. The trinucleotide expansion also en-compasses the promoter of the adjacent SIX5gene, and deletions of this gene recapitulateextramuscular aspects of the DM1-associatedphenotype.

Myotonic dystrophy is associated with anucleotide repeat expansion. The current fa-vored hypothesis is that myotonia relates toa gain of function associated with the pro-duction and accumulation of CUG repeat–containing RNA within the nuclei of DM1patients (Figure 9). Transgenic overexpres-sion of the trinucleotide repeat associatedwith an inconsequential gene (human skeletal

DNARepeat expansion

RNARetention in nuclear fociMuscleblind sequestration

Aberrant splicingChloride channel, insulin receptor, troponin T

DMPK SIX5(CTG35

)

(CTGexpansion)

(CUG expansion)

MBNLMBNL

MBNL

MBNL

Figure 9Effect of CTG repeat expansion in myotonic dystrophy type 1 (DM1). Expansion of the CTG tripletrepeat in DM1 is thought to result in the nuclear accumulation of RNA containing the abnormal CUGexpansions. By binding muscleblind-like (MBNL), these repeats are thought to sequester MBNL,creating a functional state of MBNL deficiency. This results in abnormal splicing in functionallyimportant proteins such as chloride channel, insulin receptor, and troponin T. DMPK, myotonicdystrophy protein kinase.

100 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 15: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

actin) was sufficient to produce myotonia andmuscle wasting (92). In this case, the trans-gene was expressed only in muscle, therebylimiting the pathology to muscle. This find-ing was extended further when it was observedthat expanded, expressed repeats sequester themuscleblind proteins, limiting their normalparticipation in RNA binding (93, 94). Theloss of muscleblind proteins results in abnor-mal splicing of a number of genes (95). Targetgenes whose RNA is improperly spliced in-clude the insulin receptor, the chloride chan-nel, myotubularin, tau, and troponin T, pro-teins that play roles in the variable aspectsof the phenotype of DM1. Mice with tar-geted gene disruption of muscleblind genesdevelop myotonia and have confirmed therole of muscleblind genes in specific dereg-ulation of splicing (96). As it is this my-otonia that defines the disorder, these an-imals display the typical, small, polyphasicshort-duration motor unit potentials as elec-tromyography findings. Particularly interest-ing is that misregulation of splicing is notuniform, but rather, affects a subset of geneproducts.

A clear genotype-phenotype correlationdoes not exist, except in the broadest sensefor myotonic dystrophy. For CTG nucleotiderepeat expansions less than 400, there may besome correlation with phenotype. For longerrepeats, there is less correlation. Individualswith profound expansions between two subse-quent generations with maternal inheritanceare more likely to suffer congenital myotonicdystrophy. Congenital myotonic dystrophy isa severe, neonatal-onset disorder that can belethal or lead to very early onset of diseasesassociated with cognitive impairment, in ad-dition to profound muscle weakness. In gen-eral, CTG trinucleotide repeat expansions canbe quite variable in their presentation withrespect to the degree of muscle weakness.The pattern of muscle weakness in DM1 fre-quently involves the facial muscles, with pto-sis, difficulty closing the eyes and mouth, anddifficulty chewing being among the earliersigns. Weakness can affect any muscle and

FSHD: fascioscapu-lohumeral musculardystrophy

usually progresses. The histological pheno-type of DM1 (Figure 7) on muscle biopsies ismuch more variable than that of DMD. Oftenthere is very little evidence of myofiber necro-sis. This is also reflected in often near-normalCK levels. Instead, the most prominent fea-tures are often internalized central nuclei andvariation in fiber size, with atrophic changesthat predominantly affect type I fibers andsome compensatory type II fiber hypertro-phy. Chronic changes in the form of fibrosisand fatty replacement develop at a much morevariable rate than in DMD.

Myotonic Dystrophy Type 2

Not all patients with a DM-like phenotypeshowed an expansion in the chromosome 19region, nor did these individuals link to thisregion genetically. A second locus on chromo-some 3 was mapped and subsequently identi-fied. DM2, like DM1, is associated with an ex-pansion in untranslated RNA (97). The DM2region arises from a CCTG tetranucleotiderepeat expansion in zinc finger protein 9. Thephenotype with DM2 repeat expansions maybe milder, especially with respect to the car-diac phenotype, although cardiac involvementcan occur (98, 99). Presently, evidence sup-ports that DM2, like DM1, is associated with asimilar RNA-mediated toxicity (100). Finally,there are clearly patients with a DM-like phe-notype who do not have expansions at eitherthe chromosome 19 or chromosome 3 locus,suggesting that other repeats can expand andbe expressed highly enough to participate insimilar processes.

FASCIOSCAPULOHUMERALMUSCULAR DYSTROPHY

Fascioscapulohumeral muscular dystrophy(FSHD) is the third most common form ofinherited myopathy and is a dominantly in-herited form of progressive muscle wasting(101, 102). FSHD is striking for its predilec-tion to involve discreet muscle groups. Mus-cles of the face are characteristically affected,

www.annualreviews.org • The Muscular Dystrophies 101

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 16: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

Sarcopenia:age-related loss ofmuscle mass thatmay interfere withdaily living

with ptosis being a prominent feature. Scapu-lar winging is common. Molecularly, FSHD isassociated with a deletion that affects the sub-telomeric region of chromosome 4q. A 3.3 kbrepeated DNA sequence termed D4Z4 is nor-mally found over a 50–300 kb region, with11 to 150 copies of the repeat. In FSHD pa-tients, there are typically fewer than 11 re-peats present in this region, and this effec-tive deletion is thought to have a positionaleffect on neighboring genes. Most patientswith an FSHD-like phenotype display a re-duced number of D4Z4 repeats, and there ap-pears to be little genetic heterogeneity whenstrict diagnostic criteria are used. Curiously,there is a similar repeat constellation at thesubtelomeric region of chromosome 10p, butin this case alteration of the repeats at this lo-cus is not associated with a muscle phenotypeor any other extraskeletal manifestation. Theprevailing theory is that the D4Z4 repeat pro-duces the phenotype through a positional ef-fect because D4Z4 contains a transcriptionalsilencer. A recent study directly examined thishypothesis through overexpression of threepotential target genes on 4q. FRG1 (FSHDregion gene 1) is located approximately 100kb centromeric to the D4Z4 region. Overex-pression using murine transgenesis of FRG1,but not FRG2 or ANT1, was sufficient toproduce a myopathic phenotype consistentwith the notion that overexpression of FRG1is toxic to muscle and, therefore, pathogenicin FSHD (103). These findings, although in-triguing, may not fully account for the patho-genesis of FSHD because whether FRG1 isabnormally overexpressed in FSHD muscleremains in debate (104).

In addition to these studies, the telomericregion of chromosome 4q is also found atthe nuclear periphery, unlike other telomericregions (105). Although this position is notthought to be different between FSHD mu-tant and normal cells, this nuclear position-ing may further influence gene expression ofneighboring genes and provide a distinct, po-tentially additional mechanism for position-effect variegation.

REGULATORS OF MUSCLEGROWTH: THERAPY BEYONDMUSCLE-WASTING DISORDERS

In addition to these disorders, there are othercauses of muscle degeneration, and the mostcommon form of muscle wasting, sarcope-nia, occurs with aging. The mechanisms thatunderlie sarcopenia are probably multifoldand likely include defects of muscle growthor regeneration. Several distinct and impor-tant pathways that mediate growth and re-generation during development and in adultlife, including mature or older animals, havebeen uncovered in recent years. There are atleast two pathways being pursued as treat-ment strategies in muscle-wasting disorders.In both cases, the effects of these pathwaysare not specific to diseased muscle and maybe expanded upon to treat sarcopenia.

Myostatin, also known as growth and dif-ferentiation factor 8, is a member of the trans-forming growth factor β family of proteins(106). Myostatin is a natural inhibitor of mus-cle growth and is highly expressed in mus-cle. Mice engineered to lack the myostatingene display a profound increase in musclemass with more than a 200% increase in mus-cle size. Myostatin is processed to form adimer and binds the activin type IIB recep-tor and subsequently activates SMADs and af-fects gene expression. Myostatin can bind tofollistatin in a latent complex. Mice overex-pressing follistatin similarly show an increasein muscle mass (107). A number of naturallyoccurring alleles of myostatin loss of functionhave been described in large animals, lead-ing to the double muscling phenotype in cat-tle. Myostatin shows promise as a therapeutictarget because an antibody that inhibits mus-cle growth can lead to an increase in mus-cle mass in normal muscle (108). In addition,myostatin inhibition in the background of themdx mouse that lacks dystrophin also pro-duced larger and stronger muscles (109). Mostrecently, myostatin inhibition has also beenachieved through peptide inhibition (110). Inaddition to its role in muscle hypertrophy,

102 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 17: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

myostatin exerts an effect on the proliferationand differentiation of muscle precursor cellsor myoblasts.

Insulin-like Growth Factor-1

Insulin-like growth factor-1 (IGF-1) medi-ates growth of a number of somatic tissues,including muscle. Specific forms of IGF-1mediate muscle growth in the setting of nor-mal and dystrophic muscle. Transgenic over-expression of IGF-1 leads to a profound in-crease in muscle mass, similar to what is seenwith the deletion of the myostatin locus (111).Interestingly, this result is highly dependenton the specific splice form of IGF-1 used.IGF-1 may act locally as well. Viral deliveryof IGF-1 is also effective at generating en-hanced muscle mass both normally and in thesetting of dystrophic muscle (112). A smallform of IGF-1, known as muscle growth fac-tor, is thought to be very effective at in-creasing muscle mass when delivered locally(113).

SUMMARY

Genetic studies have identified a large num-ber of distinct monogenic causes of mus-cular degeneration and muscular dystrophy.The phenotype associated with these disor-ders varies and includes the mildest weak-

ness to the most profound loss of skeletalmuscle function. Subsets of genes implicatedin the muscular dystrophies share specificpathologic defects. The DGC, including dys-trophin and the sarcoglycans, is essential forthe mechanosignaling maintenance of plasmamembrane stability in muscle. Dysferlin and,potentially, caveolin facilitate membrane re-pair. Calpain, potentially titin, and TRIM32may mediate protein turnover. Proteins ofthe Z band such as myotilin and telethoninmay have diverse roles related to sarcomerestability and signaling. Nuclear membraneproteins mediate dystrophic pathology byproviding structural integrity to the nuclearmembrane and potentially through epistaticmechanisms. Myotonic dystrophy is associ-ated with a toxic RNA pathology, whereasFSHD arises from a positional effect on geneexpression. These diverse mechanisms lead tothe common pathway of muscle degenerationand weakness, and therapy aimed at stimulat-ing muscle growth may be effective againstthese common pathways. Other approachesto therapy, such as stop codon read-through,require the detailed specific knowledge of thegene defect. Because skeletal muscle regener-ation is ongoing and mediated by the fusionof mononuclear stem cells, cell-based therapymay prove effective at introducing correctedgenes into the multinuclear syncytium that ismature muscle.

ACKNOWLEDGMENTS

EMM is supported by the Muscular Dystrophy Association, the Burroughs Wellcome Fund,the Heart Research Foundation, and the National Institutes of Health.

LITERATURE CITED

1. Mauro A. 1961. Satellite cell of skeletal muscle fibers. J. Biophys. Biochem. Cytol. 9:493–952. Relaix F, Rocancourt D, Mansouri A, Buckingham M. 2005. A Pax3/Pax7-dependent

population of skeletal muscle progenitor cells. Nature 435:948–533. LaBarge MA, Blau HM. 2002. Biological progression from adult bone marrow to

mononucleate muscle stem cell to multinucleate muscle fiber in response to injury. Cell111:589–601

4. Reviewedsatellite cells astissue-specific stemcells and providednew insights intothe heterogeneousnature of this cellpool.4. Morgan JE, Partridge TA. 2003. Muscle satellite cells. Int. J. Biochem. Cell Biol.

35:1151–56

IGF-1: insulin-likegrowth factor-1

www.annualreviews.org • The Muscular Dystrophies 103

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 18: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

5. Kobayashi K, Nakahori Y, Miyake M, Matsumura K, Kondo-Iida E, et al. 1998. Anancient retrotransposal insertion causes Fukuyama-type congenital muscular dystrophy.Nature 394:388–92

6. Yoshida A, Kobayashi K, Manya H, Taniguchi K, Kano H, et al. 2001. Muscular dys-trophy and neuronal migration disorder caused by mutations in a glycosyltransferase,POMGnT1. Dev. Cell 1:717–24

7. Beltran-Valero de Bernabe D, Currier S, Steinbrecher A, Celli J, van Beusekom E, et al.2002. Mutations in the O-mannosyltransferase gene POMT1 give rise to the severe neu-ronal migration disorder Walker-Warburg syndrome. Am. J. Hum. Genet. 71:1033–43

8. Beltran-Valero de Bernabe D, Voit T, Longman C, Steinbrecher A, Straub V, et al. 2004.Mutations in the FKRP gene can cause muscle-eye-brain disease and Walker-Warburgsyndrome. J. Med. Genet. 41:e61

9. Mercuri E, Brockington M, Straub V, Quijano-Roy S, Yuva Y, et al. 2003. Phenotypicspectrum associated with mutations in the fukutin-related protein gene. Ann. Neurol.53:537–42

10. Reviewed thenormal function ofthe dystroglycancomplex and thealterations causedby defectiveposttranslationalprocessing.

10. Barresi R, Campbell KP. 2006. Dystroglycan: from biosynthesis to pathogenesisof human disease. J. Cell Sci. 119:199–207

11. Brancaccio A. 2005. Alpha-dystroglycan, the usual suspect? Neuromuscul. Disord. 15:825–28

12. Durbeej M, Henry MD, Campbell KP. 1998. Dystroglycan in development and disease.Curr. Opin. Cell Biol. 10:594–601

13. Gee SH, Blacher RW, Douville PJ, Provost PR, Yurchenco PD, Carbonetto S. 1993.Laminin-binding protein 120 from brain is closely related to the dystrophin-associatedglycoprotein, dystroglycan, and binds with high affinity to the major heparin bindingdomain of laminin. J. Biol. Chem. 268:14972–80

14. Brockington M, Blake DJ, Prandini P, Brown SC, Torelli S, et al. 2001. Mutations in thefukutin-related protein gene (FKRP) cause a form of congenital muscular dystrophy withsecondary laminin alpha2 deficiency and abnormal glycosylation of alpha-dystroglycan.Am. J. Hum. Genet. 69:1198–209

15. Kondo-Iida E, Kobayashi K, Watanabe M, Sasaki J, Kumagai T, et al. 1999. Novelmutations and genotype-phenotype relationships in 107 families with Fukuyama-typecongenital muscular dystrophy (FCMD). Hum. Mol. Genet. 8:2303–9

16. Kirschner J, Bonnemann CG. 2004. The congenital and limb-girdle muscular dystro-phies: sharpening the focus, blurring the boundaries. Arch. Neurol. 61:189–99

17. Muntoni F. 2004. Journey into muscular dystrophies caused by abnormal glycosylation.Acta Myol. 23:79–84

18. Haliloglu G, Topaloglu H. 2004. Glycosylation defects in muscular dystrophies. Curr.Opin. Neurol. 17:521–27

19. Torelli S, Brown SC, Brockington M, Dolatshad NF, Jimenez C, et al. 2005. Sub-cellularlocalisation of fukutin related protein in different cell lines and in the muscle of patientswith MDC1C and LGMD2I. Neuromuscul. Disord. 15:836–43

20. Esapa CT, McIlhinney RA, Blake DJ. 2005. Fukutin-related protein mutations that causecongenital muscular dystrophy result in ER-retention of the mutant protein in culturedcells. Hum. Mol. Genet. 14:295–305

21. Grewal PK, McLaughlan JM, Moore CJ, Browning CA, Hewitt JE. 2005. Characteri-zation of the LARGE family of putative glycosyltransferases associated with dystrogly-canopathies. Glycobiology 15:912–23

22. Matsumoto H, Noguchi S, Sugie K, Ogawa M, Murayama K, et al. 2004. Subcellularlocalization of fukutin and fukutin-related protein in muscle cells. J. Biochem. 135:709–12

104 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 19: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

23. Guicheney P, Vignier N, Helbling-Leclerc A, Nissinen M, Zhang X, et al. 1997. Geneticsof laminin alpha 2 chain (or merosin) deficient congenital muscular dystrophy: fromidentification of mutations to prenatal diagnosis. Neuromuscul. Disord. 7:180–86

24. Pegoraro E, Marks H, Garcia CA, Crawford T, Mancias P, et al. 1998. Laminin alpha2muscular dystrophy: genotype/phenotype studies of 22 patients. Neurology 51:101–10

25. Miller RG, Hoffman EP. 1994. Molecular diagnosis and modern management ofDuchenne muscular dystrophy. Neurol. Clin. 12:699–725

26. Chaturvedi LS, Mukherjee M, Srivastava S, Mittal RD, Mittal B. 2001. Point mutationand polymorphism in Duchenne/Becker muscular dystrophy (D/BMD) patients. Exp.Mol. Med. 33:251–56

27. Sicinski P, Geng Y, Ryder-Cook AS, Barnard EA, Darlison MG, Barnard PJ. 1989.The molecular basis of muscular dystrophy in the mdx mouse: a point mutation. Science244:1578–80

28. Howard MT, Anderson CB, Fass U, Khatri S, Gesteland RF, et al. 2004. Readthrough ofdystrophin stop codon mutations induced by aminoglycosides. Ann. Neurol. 55:422–26

29. Aminoglycosideantibiotics produceread-throughexpression ofdystrophin bysuppressing theeffect of stop codonmutations.

29. Barton-Davis ER, Cordier L, Shoturma DI, Leland SE, Sweeney HL. 1999. Amino-glycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice.J. Clin. Invest. 104:375–81

30. Hoogerwaard EM, Bakker E, Ippel PF, Oosterwijk JC, Majoor-Krakauer DF, et al. 1999.Signs and symptoms of Duchenne muscular dystrophy and Becker muscular dystrophyamong carriers in The Netherlands: a cohort study. Lancet 353:2116–19

31. Koenig M, Kunkel LM. 1990. Detailed analysis of the repeat domain of dystrophin revealsfour potential hinge segments that may confer flexibility. J. Biol. Chem. 265:4560–66

32. Ibraghimov-Beskrovnaya O, Ervasti JM, Leveille CJ, Slaughter CA, Sernett SW,Campbell KP. 1992. Primary structure of dystrophin-associated glycoproteins linkingdystrophin to the extracellular matrix. Nature 355:696–702

33. Dystrophin andthe DGC form astrong mechanicalcoupling betweenthe sarcolemmaand the sarcomericactin.

33. Rybakova IN, Patel JR, Ervasti JM. 2000. The dystrophin complex forms a me-chanically strong link between the sarcolemma and costameric actin. J. Cell Biol.150:1209–14

34. Matsuda R, Nishikawa A, Tanaka H. 1995. Visualization of dystrophic muscle fibers inmdx mouse by vital staining with Evans blue: evidence of apoptosis in dystrophin-deficientmuscle. J. Biochem. 118:959–64

35. Straub V, Rafael JA, Chamberlain JS, Campbell KP. 1997. Animal models for musculardystrophy show different patterns of sarcolemmal disruption. J. Cell Biol. 139:375–85

36. Gregorevic P, Blankinship MJ, Allen JM, Crawford RW, Meuse L, et al. 2004. Sys-temic delivery of genes to striated muscles using adeno-associated viral vectors. Nat.Med. 10:828–34

37. Alderton JM, Steinhardt RA. 2000. How calcium influx through calcium leak channelsis responsible for the elevated levels of calcium-dependent proteolysis in dystrophic my-otubes. Trends Cardiovasc. Med. 10:268–72

38. Ervasti JM, Ohlendieck K, Kahl SD, Gaver MG, Campbell KP. 1990. Deficiency of a gly-coprotein component of the dystrophin complex in dystrophic muscle. Nature 345:315–19

39. Yoshida M, Ozawa E. 1990. Glycoprotein complex anchoring dystrophin to sarcolemma.J. Biochem. 108:748–52

40. Lapidos KA, Kakkar R, McNally EM. 2004. The dystrophin glycoprotein complex: sig-naling strength and integrity for the sarcolemma. Circ. Res. 94:1023–31

41. Lovering RM, Porter NC, Bloch RJ. 2005. The muscular dystrophies: from genes totherapies. Phys. Ther. 85:1372–88

www.annualreviews.org • The Muscular Dystrophies 105

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 20: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

42. Loufrani L, Levy BI, Henrion D. 2002. Defect in microvascular adaptation to chronicchanges in blood flow in mice lacking the gene encoding for dystrophin. Circ. Res.91:1183–89

43. Sander M, Chavoshan B, Harris SA, Iannaccone ST, Stull JT, et al. 2000. Functionalmuscle ischemia in neuronal nitric oxide synthase-deficient skeletal muscle of childrenwith Duchenne muscular dystrophy. Proc. Natl. Acad. Sci. USA 97:13818–23

44. Thomas GD, Shaul PW, Yuhanna IS, Froehner SC, Adams ME. 2003. Vasomodulationby skeletal muscle-derived nitric oxide requires alpha-syntrophin-mediated sarcolemmallocalization of neuronal Nitric oxide synthase. Circ. Res. 92:554–60

45. Ozawa E, Mizuno Y, Hagiwara Y, Sasaoka T, Yoshida M. 2005. Molecular and cell biologyof the sarcoglycan complex. Musc. Nerv. 32:563–76

46. Zimprich A, Grabowski M, Asmus F, Naumann M, Berg D, et al. 2001. Mutations inthe gene encoding epsilon-sarcoglycan cause myoclonus-dystonia syndrome. Nat. Genet.29:66–69

47. Wheeler MT, Zarnegar S, McNally EM. 2002. Zeta-sarcoglycan, a novel component ofthe sarcoglycan complex, is reduced in muscular dystrophy. Hum. Mol. Genet. 11:2147–54

48. Bueno MR, Moreira ES, Vainzof M, Chamberlain J, Marie SK, et al. 1995. A commonmissense mutation in the adhalin gene in three unrelated Brazilian families with a rela-tively mild form of autosomal recessive limb-girdle muscular dystrophy. Hum. Mol. Genet.4:1163–67

49. McNally EM, Passos-Bueno MR, Bonnemann CG, Vainzof M, de Sa Moreira E, et al.1996. Mild and severe muscular dystrophy caused by a single gamma-sarcoglycan muta-tion. Am. J. Hum. Genet. 59:1040–47

50. Heydemann A, Huber JM, Demonbreun A, Hadhazy M, McNally EM. 2005. Geneticbackground influences muscular dystrophy. Neuromuscul. Disord. 15:601–9

51. Vainzof M, Passos-Bueno MR, Canovas M, Moreira ES, Pavanello RC, et al. 1996. Thesarcoglycan complex in the six autosomal recessive limb-girdle muscular dystrophies.Hum. Mol. Genet. 5:1963–69

52. Hack AA, Lam MY, Cordier L, Shoturma DI, Ly CT, et al. 2000. Differential requirementfor individual sarcoglycans and dystrophin in the assembly and function of the dystrophin-glycoprotein complex. J. Cell Sci. 113(Pt 14):2535–44

53. Straub V, Ettinger AJ, Durbeej M, Venzke DP, Cutshall S, et al. 1999. ε-sarcoglycanreplaces α-sarcoglycan in smooth muscle to form a unique dystrophin-glycoprotein com-plex. J. Biol. Chem. 274:27989–96

54. Coral-Vazquez R, Cohn RD, Moore SA, Hill JA, Weiss RM, et al. 1999. Disruption ofthe sarcoglycan-sarcospan complex in vascular smooth muscle: a novel mechanism forcardiomyopathy and muscular dystrophy. Cell 98:465–74

55. Coronary arteryvasospasm insarcoglycan-deficient animals iscaused by extrinsicfactors, not primarysmooth muscledysfunction.

55. Wheeler MT, Allikian MJ, Heydemann A, Hadhazy M, Zarnegar S, McNally EM.2004. Smooth muscle cell-extrinsic vascular spasm arises from cardiomyocyte de-generation in sarcoglycan-deficient cardiomyopathy. J. Clin. Invest. 113:668–75

56. Anderson LV, Davison K, Moss JA, Young C, Cullen MJ, et al. 1999. Dysferlin is a plasmamembrane protein and is expressed early in human development. Hum. Mol. Genet. 8:855–61

57. Achanzar WE, Ward S. 1997. A nematode gene required for sperm vesicle fusion. J. CellSci. 110:1073–81

58. Ward S, Argon Y, Nelson GA. 1981. Sperm morphogenesis in wild-type and fertilization-defective mutants of Caenorhabditis elegans. J. Cell Biol. 91:26–44

106 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 21: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

59. Piccolo F, Moore SA, Ford GC, Campbell KP. 2000. Intracellular accumulation andreduced sarcolemmal expression of dysferlin in limb-girdle muscular dystrophies. Ann.Neurol. 48:902–12

60. Selcen D, Stilling G, Engel AG. 2001. The earliest pathologic alterations in dysferlinopa-thy. Neurology 56:1472–81

61. Dysferlin is atransmembraneprotein importantin calcium-dependent repair ofdefects in thesarcolemmalmembrane.

61. Bansal D, Miyake K, Vogel SS, Groh S, Chen CC, et al. 2003. Defective membranerepair in dysferlin-deficient muscular dystrophy. Nature 423:168–72

62. Cenacchi G, Fanin M, De Giorgi LB, Angelini C. 2005. Ultrastructural changes in dys-ferlinopathy support defective membrane repair mechanism. J. Clin. Pathol. 58:190–95

63. Doherty KR, McNally EM. 2003. Repairing the tears: dysferlin in muscle membranerepair. Trends Mol. Med. 9:327–30

64. Fernandez-Chacon R, Konigstorfer A, Gerber SH, Garcia J, Matos MF, et al. 2001.Synaptotagmin I functions as a calcium regulator of release probability. Nature 410:41–49

65. Davis DB, Doherty KR, Delmonte AJ, McNally EM. 2002. Calcium-sensitive phos-pholipid binding properties of normal and mutant ferlin C2 domains. J. Biol. Chem.277:22883–88

66. Davis DB, Delmonte AJ, Ly CT, McNally EM. 2000. Myoferlin, a candidate gene andpotential modifier of muscular dystrophy. Hum. Mol. Genet. 9:217–26

67. Doherty KR, Cave A, Davis DB, Delmonte AJ, Posey A, et al. 2005. Normal myoblastfusion requires myoferlin. Development 132:5565–75

68. McNally EM, Ly CT, Rosenmann H, Mitrani Rosenbaum S, Jiang W, et al. 2000. Splicingmutation in dysferlin produces limb-girdle muscular dystrophy with inflammation. Am.J. Med. Genet. 91:305–12

69. Gallardo E, Rojas-Garcia R, de Luna N, Pou A, Brown RHJ, Illa I. 2001. Inflammationin dysferlin myopathy: immunohistochemical characterization of 13 patients. Neurology57:2136–38

70. Hoffman EP, Rao D, Pachman LM. 2002. Clarifying the boundaries between the inflam-matory and dystrophic myopathies: insights from molecular diagnostics and microarrays.Rheum. Dis. Clin. North Am. 28:743–57

71. Weiler T, Bashir R, Anderson LV, Davison K, Moss JA, et al. 1999. Identical mutationin patients with limb girdle muscular dystrophy type 2B or Miyoshi myopathy suggests arole for modifier gene(s). Hum. Mol. Genet. 8:871–77

72. Zatz M, Starling A. 2005. Calpains and disease. N. Engl. J. Med. 352:2413–2373. Guyon JR, Kudryashova E, Potts A, Dalkilic I, Brosius MA, et al. 2003. Calpain 3 cleaves

filamin C and regulates its ability to interact with gamma- and delta-sarcoglycans. Musc.Nerv. 28:472–83

74. Sorimachi H, Kinbara K, Kimura S, Takahashi M, Ishiura S, et al. 1995. Muscle-specificcalpain, p94, responsible for limb girdle muscular dystrophy type 2A, associates withconnectin through IS2, a p94-specific sequence. J. Biol. Chem. 270:31158–62

75. Garvey SM, Rajan C, Lerner AP, Frankel WN, Cox GA. 2002. The muscular dystrophywith myositis (mdm) mouse mutation disrupts a skeletal muscle-specific domain of titin.Genomics 79:146–49

76. Woodman SE, Sotgia F, Galbiati F, Minetti C, Lisanti MP. 2004. Caveolinopathies:mutations in caveolin-3 cause four distinct autosomal dominant muscle diseases. Neurology62:538–43

77. Walter MC, Braun C, Vorgerd M, Poppe M, Thirion C, et al. 2003. Variable reductionof caveolin-3 in patients with LGMD2B/MM. J. Neurol. 250:1431–38

www.annualreviews.org • The Muscular Dystrophies 107

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 22: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

78. Kudryashova E, Kudryashov D, Kramerova I, Spencer MJ. 2005. Trim32 is a ubiquitinligase mutated in limb girdle muscular dystrophy type 2H that binds to skeletal musclemyosin and ubiquitinates actin. J. Mol. Biol. 354:413–24

79. Hauser MA, Horrigan SK, Salmikangas P, Torian UM, Viles KD, et al. 2000. Myotilinis mutated in limb girdle muscular dystrophy 1A. Hum. Mol. Genet. 9:2141–47

80. Z-disc proteinsmay play animportant rolebeyond themechanics of thesarcomeres bybeing involved incell signaling.

80. Pyle WG, Solaro RJ. 2004. At the crossroads of myocardial signaling: the role ofZ-discs in intracellular signaling and cardiac function. Circ. Res. 94:296–305

81. Wallgren-Pettersson C. 2002. Nemaline and myotubular myopathies. Semin. Pediatr.Neurol. 9:132–44

82. Segura-Totten M, Wilson KL. 2004. BAF: roles in chromatin, nuclear structure andretrovirus integration. Trends Cell Biol. 14:261–66

83. Ostlund C, Worman HJ. 2003. Nuclear envelope proteins and neuromuscular diseases.Musc. Nerv. 27:393–406

84. Helbling-Leclerc A, Bonne G, Schwartz K. 2002. Emery-Dreifuss muscular dystrophy.Eur. J. Hum. Genet. 10:157–61

85. Smith ED, Kudlow BA, Frock RL, Kennedy BK. 2005. A-type nuclear lamins, progeriasand other degenerative disorders. Mech. Ageing Dev. 126:447–60

86. Defects innuclear envelopeproteins impair themechanicalfunction of thenucleus and reducethe expression ofmechanosensitivegenes.

86. Lammerding J, Schulze PC, Takahashi T, Kozlov S, Sullivan T, et al. 2004. LaminA/C deficiency causes defective nuclear mechanics and mechanotransduction. J.

Clin. Invest. 113:370–7887. Lammerding J, Hsiao J, Schulze PC, Kozlov S, Stewart CL, Lee RT. 2005. Abnormal

nuclear shape and impaired mechanotransduction in emerin-deficient cells. J. Cell Biol.170:781–91

88. Bakay M, Wang Z, Melcon G, Schiltz L, Xuan J, et al. 2006. Nuclear envelope dystrophiesshow a transcriptional fingerprint suggesting disruption of Rb-MyoD pathways in muscleregeneration. Brain 129(4):996–1013

89. Melcon G, Kozlov S, Cutler DA, Sullivan T, Hernandez L, et al. 2006. Loss of emerin atthe nuclear envelope disrupts the Rb1/E2F and MyoD pathways during muscle regener-ation. Hum. Mol. Genet. 15:637–51

90. Frock RL, Kudlow BA, Evans AM, Jameson SA, Hauschka SD, Kennedy BK. 2006.Lamin A/C and emerin are critical for skeletal muscle satellite cell differentiation. GenesDev. 20:486–500

91. Machuca-Tzili L, Brook D, Hilton-Jones D. 2005. Clinical and molecular aspects of themyotonic dystrophies: a review. Musc. Nerv. 32:1–18

92. Mankodi A, Logigian E, Callahan L, McClain C, White R, et al. 2000. Myotonic dys-trophy in transgenic mice expressing an expanded CUG repeat. Science 289:1769–73

93. Miller JW, Urbinati CR, Teng-Umnuay P, Stenberg MG, Byrne BJ, et al. 2000. Recruit-ment of human muscleblind proteins to (CUG)(n) expansions associated with myotonicdystrophy. EMBO J. 19:4439–48

94. Mankodi A, Urbinati CR, Yuan QP, Moxley RT, Sansone V, et al. 2001. Muscleblindlocalizes to nuclear foci of aberrant RNA in myotonic dystrophy types 1 and 2. Hum.Mol. Genet. 10:2165–70

95. Ho TH, Charlet BN, Poulos MG, Singh G, Swanson MS, Cooper TA. 2004. Muscleblindproteins regulate alternative splicing. EMBO J. 23:3103–12

96. Muscleblind-like deleted miceshow the samesplicingabnormalitiesfound in DMpatients.Sequestration ofmuscleblind-likethrough RNAbinding can causeDM.

96. Kanadia RN, Johnstone KA, Mankodi A, Lungu C, Thornton CA, et al. 2003. Amuscleblind knockout model for myotonic dystrophy. Science 302:1978–80

97. Liquori CL, Ricker K, Moseley ML, Jacobsen JF, Kress W, et al. 2001. Myotonic dys-trophy type 2 caused by a CCTG expansion in intron 1 of ZNF9. Science 293:864–67

108 McNally · Pytel

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 23: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

ANRV295-PM02-04 ARI 13 December 2006 2:57

98. Ranum LP, Day JW. 2002. Myotonic dystrophy: clinical and molecular parallels betweenmyotonic dystrophy type 1 and type 2. Curr. Neurol. Neurosci. Rep. 2:465–70

99. Schoser BG, Ricker K, Schneider-Gold C, Hengstenberg C, Durre J, et al. 2004. Suddencardiac death in myotonic dystrophy type 2. Neurology 63:2402–4

100. Day JW, Ricker K, Jacobsen JF, Rasmussen LJ, Dick KA, et al. 2003. Myotonic dystrophytype 2: molecular, diagnostic and clinical spectrum. Neurology 60:657–64

101. Tupler R, Gabellini D. 2004. Molecular basis of facioscapulohumeral muscular dystrophy.Cell Mol. Life Sci. 61:557–66

102. Tawil R. 2004. Facioscapulohumeral muscular dystrophy. Curr. Neurol. Neurosci. Rep.4:51–54

103. Deletions maycause FSHD byinappropriateoverexpression ofFRG1 throughpositional changeson gene regulation.

103. Gabellini D, D’Antona G, Moggio M, Prelle A, Zecca C, et al. 2006. Facioscapu-lohumeral muscular dystrophy in mice overexpressing FRG1. Nature 439:973–77

104. van Deutekom JC, Lemmers RJ, Grewal PK, van Geel M, Romberg S, et al. 1996.Identification of the first gene (FRG1) from the FSHD region on human chromosome4q35. Hum. Mol. Genet. 5:581–90

105. Masny PS, Bengtsson U, Chung SA, Martin JH, van Engelen B, et al. 2004. Localizationof 4q35.2 to the nuclear periphery: Is FSHD a nuclear envelope disease? Hum. Mol. Genet.13:1857–71

106. Lee SJ, McPherron AC. 1999. Myostatin and the control of skeletal muscle mass. Curr.Opin. Genet. Dev. 9:604–7

107. Lee SJ, McPherron AC. 2001. Regulation of myostatin activity and muscle growth. Proc.Natl. Acad. Sci. USA 98:9306–11

108. Whittemore LA, Song K, Li X, Aghajanian J, Davies M, et al. 2003. Inhibition of myo-statin in adult mice increases skeletal muscle mass and strength. Biochem. Biophys. Res.Commun. 300:965–71

109. Bogdanovich S, Krag TO, Barton ER, Morris LD, Whittemore LA, et al. 2002. Func-tional improvement of dystrophic muscle by myostatin blockade. Nature 420:418–21

110. Bogdanovich S, Perkins KJ, Krag TO, Whittemore LA, Khurana TS. 2005. Myostatinpropeptide-mediated amelioration of dystrophic pathophysiology. FASEB J. 19:543–49

111. Musaro A, McCullagh KJ, Naya FJ, Olson EN, Rosenthal N. 1999. IGF-1 induces skeletalmyocyte hypertrophy through calcineurin in association with GATA-2 and NF-ATc1.Nature 400:581–85

112. Barton ER, Morris L, Musaro A, Rosenthal N, Sweeney HL. 2002. Muscle-specificexpression of insulin-like growth factor I counters muscle decline in mdx mice. J. CellBiol. 157:137–48

113. Yang SY, Goldspink G. 2002. Different roles of the IGF-I Ec peptide (MGF) and matureIGF-I in myoblast proliferation and differentiation. FEBS Lett. 522:156–60

www.annualreviews.org • The Muscular Dystrophies 109

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 24: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

Contents ARI 13 December 2006 2:13

Annual Reviewof Pathology:Mechanisms ofDisease

Volume 2, 2007Contents

FrontispieceHenry C. Pitot � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �xii

Adventures in HepatocarcinogenesisHenry C. Pitot � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 1

Endocrine Functions of Adipose TissueHironori Waki and Peter Tontonoz � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �31

Endometrial CarcinomaAntonio Di Cristofano and Lora Hedrick Ellenson � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �57

Muscle Diseases: The Muscular DystrophiesElizabeth M. McNally and Peter Pytel � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �87

Pathobiology of Neutrophil Transepithelial Migration: Implications inMediating Epithelial InjuryAlex C. Chin and Charles A. Parkos � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 111

von Hippel-Lindau DiseaseWilliam G. Kaelin, Jr. � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 145

Cancer Stem Cells: At the Headwaters of Tumor DevelopmentRyan J. Ward and Peter B. Dirks � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 175

NeurofibromatosisAndrea I. McClatchey � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 191

Malaria: Mechanisms of Erythrocytic Infection and PathologicalCorrelates of Severe DiseaseKasturi Haldar, Sean C. Murphy, Dan A. Milner, Jr., and Terrie E. Taylor � � � � � � � � � � � � 217

VEGF-A and the Induction of Pathological AngiogenesisJanice A. Nagy, Ann M. Dvorak, and Harold F. Dvorak � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 251

In Vivo Pathology: Seeing with Molecular Specificity and CellularResolution in the Living BodyChristopher H. Contag � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 277

vi

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.

Page 25: Muscle Diseases: The Muscular Dystrophies · ANRV295-PM02-04 ARI 13 December 2006 2:57 Figure 3 Myopathic muscle histology. (a, b) H&E staining of two examples of necrotic fibers.The

Contents ARI 13 December 2006 2:13

Cell-Based Therapy for Myocardial Ischemia and Infarction:Pathophysiological MechanismsMichael A. Laflamme, Stephan Zbinden, Stephen E. Epstein,

and Charles E. Murry � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 307

Cystic Disease of the KidneyPatricia D. Wilson and Beatrice Goilav � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 341

Pathobiology of Pulmonary HypertensionMarlene Rabinovitch � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 369

Body Traffic: Ecology, Genetics, and Immunity in InflammatoryBowel DiseaseJonathan Braun and Bo Wei � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 401

Contents vii

Ann

u. R

ev. P

atho

l. M

ech.

Dis

. 200

7.2:

87-1

09. D

ownl

oade

d fr

om a

rjou

rnal

s.an

nual

revi

ews.

org

by S

tanf

ord

Uni

vers

ity R

ober

t Cro

wn

Law

Lib

. on

10/2

7/08

. For

per

sona

l use

onl

y.