severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular...

11
2185 Research Article Introduction Duchenne muscular dystrophy (DMD) is the most common X- chromosome-linked human disease that affects approximately 1 in 3,500 males. DMD patients suffer from severe, progressive muscle wasting with clinical symptoms first detected between 2 and 5 years of age. As the disease progresses, patients are confined to a wheelchair in their early teens and die in their early twenties from cardiopulmonary failure (Emery, 1993; Moser, 1984). DMD patients and mdx mice have dystrophin gene mutations that result in an absence of the dystrophin protein (Bulfield et al., 1984; Campbell, 1995; Matsumura et al., 1992; Monaco et al., 1986; Sicinski et al., 1989). Dystrophin is localized to the cytoplasmic face of the plasma membrane and mediates the interaction between the cell cytoskeleton and the extracellular matrix (ECM) through a complex of associated glycoproteins called the dystrophin-glycoprotein complex (DGC). In skeletal muscle, the DGC is localized along muscle fibers and at myotendinous and neuromuscular junctions (MTJs and NMJs, respectively). This complex includes dystroglycans, sarcoglycans and syntrophins (Suzuki et al., 1994). Dystrophin interacts with F-actin through its N-terminus (Way et al., 1992). The C-terminus interacts with syntrophins and with the cytoplasmic tail of -dystroglycan (Henry and Campbell, 1996). -dystroglycan binds to laminin in the extracellular matrix. Overall, the DGC links the muscle cell cytoskeleton to the extracellular matrix and maintains sarcolemmal integrity. In DMD patients, loss of dystrophin results in a disruption of the DGC-extracellular matrix linkage. This leads to the loss of sarcolemmal integrity, unregulated influx of Ca 2+ into muscle cells, activation of Ca 2+ -dependent proteases and muscle degeneration (Gillis, 1996). Muscle degeneration is accompanied by regeneration through the activation of muscle satellite cells; however, regenerative capacity in DMD patients is exceeded by progressive muscle degeneration. This results in the replacement of muscle fibers with adipose and connective tissue (O’Brien and Kunkel, 2001). The mdx mouse is a valuable model for DMD and has been used extensively to analyze the progression of muscle disease. Although DMD patients (Monaco et al., 1986) and mdx mice (Bulfield et al., 1984; Sicinski et al., 1989) both lack dystrophin, the mdx mouse develops less severe muscle pathology compared with DMD patients, and has a normal life span. Several factors may explain the reduced pathology in mdx mice including differences in muscle use between humans and captive mice. In addition, mdx mice might compensate for the loss of dystrophin, possibly by overexpressing one or more proteins with functional similarity to dystrophin. One candidate protein is utrophin, an autosomal homolog of dystrophin that is expressed at the post-synaptic membrane of the NMJ in adult skeletal muscle. Utrophin is upregulated in the muscle of both DMD patients and mdx mice and is localized at extrajunctional sites (Ohlendieck et al., 1991; Pons et al., 1991). The dystrophin glycoprotein complex links laminin in the extracellular matrix to the cell cytoskeleton. Loss of dystrophin causes Duchenne muscular dystrophy, the most common human X-chromosome-linked genetic disease. The 71 integrin is a second transmembrane laminin receptor expressed in skeletal muscle. Mutations in the 7 integrin gene cause congenital myopathy in humans and mice. The 71 integrin is increased in the skeletal muscle of Duchenne muscular dystrophy patients and mdx mice. This observation has led to the suggestion that dystrophin and 71 integrin have complementary functional and structural roles. To test this hypothesis, we generated mice lacking both dystrophin and 7 integrin (mdx/7 –/– ). The mdx/7 –/– mice developed early-onset muscular dystrophy and died at 2-4 weeks of age. Muscle fibers from mdx/7 –/– mice exhibited extensive loss of membrane integrity, increased centrally located nuclei and inflammatory cell infiltrate, greater necrosis and increased muscle degeneration compared to mdx or 7-integrin null animals. In addition, loss of dystrophin and/or 7 integrin resulted in altered expression of laminin-2 chain. These results point to complementary roles for dystrophin and 71 integrin in maintaining the functional integrity of skeletal muscle. Supplementary material available online at http://jcs.biologists.org/cgi/content/full/119/11/2185/DC1 Key words: 7 integrin, Dystrophin, Transgenic mice, Double knockout, Skeletal muscle, Muscular dystrophy Summary Severe muscular dystrophy in mice that lack dystrophin and 7 integrin Jachinta E. Rooney 1, *, Jennifer V. Welser 1, *, Melissa A. Dechert 1 , Nichole L. Flintoff-Dye 1 , Stephen J. Kaufman 2 and Dean J. Burkin 1,3,‡ 1 Department of Pharmacology, University of Nevada, Reno, NV 89557, USA 2 Department of Cell and Developmental Biology, University of Illinois, Urbana, IL 61801, USA 3 Nevada Transgenic Center, University of Nevada, Reno, NV 89557, USA *These authors contributed equally to this work Author for correspondence (e-mail: [email protected]) Accepted 21 February 2006 Journal of Cell Science 119, 2185-2195 Published by The Company of Biologists 2006 doi:10.1242/jcs.02952 Journal of Cell Science

Upload: others

Post on 05-Aug-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2185Research Article

IntroductionDuchenne muscular dystrophy (DMD) is the most common X-chromosome-linked human disease that affects approximately1 in 3,500 males. DMD patients suffer from severe, progressivemuscle wasting with clinical symptoms first detected between2 and 5 years of age. As the disease progresses, patients areconfined to a wheelchair in their early teens and die in theirearly twenties from cardiopulmonary failure (Emery, 1993;Moser, 1984). DMD patients and mdx mice have dystrophingene mutations that result in an absence of the dystrophinprotein (Bulfield et al., 1984; Campbell, 1995; Matsumura etal., 1992; Monaco et al., 1986; Sicinski et al., 1989). Dystrophinis localized to the cytoplasmic face of the plasma membraneand mediates the interaction between the cell cytoskeleton andthe extracellular matrix (ECM) through a complex of associatedglycoproteins called the dystrophin-glycoprotein complex(DGC). In skeletal muscle, the DGC is localized along musclefibers and at myotendinous and neuromuscular junctions (MTJsand NMJs, respectively). This complex includes dystroglycans,sarcoglycans and syntrophins (Suzuki et al., 1994). Dystrophininteracts with F-actin through its N-terminus (Way et al., 1992).The C-terminus interacts with syntrophins and with thecytoplasmic tail of �-dystroglycan (Henry and Campbell,1996). �-dystroglycan binds to laminin in the extracellularmatrix. Overall, the DGC links the muscle cell cytoskeleton tothe extracellular matrix and maintains sarcolemmal integrity.

In DMD patients, loss of dystrophin results in a disruptionof the DGC-extracellular matrix linkage. This leads to the lossof sarcolemmal integrity, unregulated influx of Ca2+ intomuscle cells, activation of Ca2+-dependent proteases andmuscle degeneration (Gillis, 1996). Muscle degeneration isaccompanied by regeneration through the activation of musclesatellite cells; however, regenerative capacity in DMD patientsis exceeded by progressive muscle degeneration. This resultsin the replacement of muscle fibers with adipose andconnective tissue (O’Brien and Kunkel, 2001).

The mdx mouse is a valuable model for DMD and has beenused extensively to analyze the progression of muscle disease.Although DMD patients (Monaco et al., 1986) and mdx mice(Bulfield et al., 1984; Sicinski et al., 1989) both lack dystrophin,the mdx mouse develops less severe muscle pathologycompared with DMD patients, and has a normal life span.Several factors may explain the reduced pathology in mdx miceincluding differences in muscle use between humans andcaptive mice. In addition, mdx mice might compensate for theloss of dystrophin, possibly by overexpressing one or moreproteins with functional similarity to dystrophin. One candidateprotein is utrophin, an autosomal homolog of dystrophin that isexpressed at the post-synaptic membrane of the NMJ in adultskeletal muscle. Utrophin is upregulated in the muscle of bothDMD patients and mdx mice and is localized at extrajunctionalsites (Ohlendieck et al., 1991; Pons et al., 1991).

The dystrophin glycoprotein complex links laminin in theextracellular matrix to the cell cytoskeleton. Loss ofdystrophin causes Duchenne muscular dystrophy, the mostcommon human X-chromosome-linked genetic disease.The ��7��1 integrin is a second transmembrane lamininreceptor expressed in skeletal muscle. Mutations in the ��7integrin gene cause congenital myopathy in humans andmice. The ��7��1 integrin is increased in the skeletal muscleof Duchenne muscular dystrophy patients and mdx mice.This observation has led to the suggestion that dystrophinand ��7��1 integrin have complementary functional andstructural roles. To test this hypothesis, we generated micelacking both dystrophin and ��7 integrin (mdx/��7–/–). Themdx/��7–/– mice developed early-onset muscular dystrophyand died at 2-4 weeks of age. Muscle fibers from mdx/��7–/–

mice exhibited extensive loss of membrane integrity,increased centrally located nuclei and inflammatory cellinfiltrate, greater necrosis and increased muscledegeneration compared to mdx or ��7-integrin null animals.In addition, loss of dystrophin and/or ��7 integrin resultedin altered expression of laminin-��2 chain. These resultspoint to complementary roles for dystrophin and ��7��1integrin in maintaining the functional integrity of skeletalmuscle.

Supplementary material available online athttp://jcs.biologists.org/cgi/content/full/119/11/2185/DC1

Key words: �7 integrin, Dystrophin, Transgenic mice, Doubleknockout, Skeletal muscle, Muscular dystrophy

Summary

Severe muscular dystrophy in mice that lackdystrophin and ��7 integrinJachinta E. Rooney1,*, Jennifer V. Welser1,*, Melissa A. Dechert1, Nichole L. Flintoff-Dye1,Stephen J. Kaufman2 and Dean J. Burkin1,3,‡

1Department of Pharmacology, University of Nevada, Reno, NV 89557, USA2Department of Cell and Developmental Biology, University of Illinois, Urbana, IL 61801, USA3Nevada Transgenic Center, University of Nevada, Reno, NV 89557, USA*These authors contributed equally to this work‡Author for correspondence (e-mail: [email protected])

Accepted 21 February 2006Journal of Cell Science 119, 2185-2195 Published by The Company of Biologists 2006doi:10.1242/jcs.02952

Jour

nal o

f Cel

l Sci

ence

Page 2: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2186

Overexpression of utrophin can rescue thedystrophic phenotype in mdx mice(Deconinck et al., 1997b; Rafael et al., 1998;Squire et al., 2002; Tinsley et al., 1998;Tinsley et al., 1996). Mice that lack utrophinshow mild neuromuscular defects (Deconincket al., 1997a; Grady et al., 1997a). Bycontrast, mice lacking both utrophin anddystrophin (mdx/utr–/– mice) develop severemuscular dystrophy and cardiomyopathysimilar to that of DMD patients, and diebetween 4-14 weeks of age (Deconinck et al.,1997b; Grady et al., 1997b).

The �7�1 integrin is a second lamininreceptor in skeletal muscle (Burkin andKaufman, 1999). Like the DGC, �7�1 integrinserves as a structural link between the cellcytoskeleton and laminin in the basal laminaand, therefore, contributes to the overallintegrity of the sarcolemma. Mutations in the�7-integrin gene are responsible for humancongenital myopathy, in which patientsexhibit delayed motor milestones (Hayashiet al., 1998). Mice that lack �7 integrin developmyopathy and demonstrate altered forcetransmission, compliance and viscoelasticity indiaphragm muscle (Lopez et al., 2005; Mayeret al., 1997). In addition, �7�1 integrin playsan important role in the development of NMJs(Burkin et al., 1998; Burkin et al., 2000) andMTJs (Mayer et al., 1997; Nawrotzki et al.,2003). These junctional sites are oftendamaged in diseased muscle (Lyons and Slater,1991; Nagel et al., 1990; Nawrotzki et al.,2003; Rafael et al., 2000).

Increased amounts of �7 integrin are foundin DMD patients and mdx mice (Hodges et al.,1997). These observations suggest thatenhanced �7 integrin expression is amechanism by which muscle can compensatefor the loss of dystrophin. This hypothesiswas supported by transgenic overexpressionof the �7-integrin chain which alleviated thesevere muscular dystrophy and extended thelife-span of mice that lacked dystrophin andutrophin (Burkin et al., 2001; Burkin et al.,2005). These observations suggest potentialstructural and functional overlap between the�7�1 integrin and DGC.

To explore the structural and functional overlap betweendystrophin and �7�1 integrin, we generated mice that lack bothlaminin-binding complexes (mdx/�7–/– mice). These miceexhibited more severe muscular dystrophy than either mdx- or�7-integrin-deficient animals and died at 2-4 weeks of age. Inaddition, the muscle of 3-week-old mdx/�7–/– mice showedinflammation and centrally located nuclei, indicative of severemuscle damage and increased requirement for muscle repair.Finally, loss of dystrophin and/or �7 integrin resulted inreduced laminin-�2-chain expression in skeletal muscle,demonstrating regulation of laminin expression by �7�1integrin. Collectively, these studies support functional and

Journal of Cell Science 119 (11)

structural overlap between DGC and �7�1 integrin inmaintaining the integrity of skeletal muscle.

Resultsmdx/�7–/– mice exhibit muscle wasting and reducedviabilityTo determine whether functional overlap exists betweendystrophin and �7�1 integrin, mdx mice were bred with micethat lack the �7-integrin gene (Flintoff-Dye et al., 2005) toproduce mice that lack both dystrophin and �7 integrin(mdx/�7–/–). Mutations in the dystrophin and �7 integrin geneswere confirmed by genotyping (Fig. 1A). Multiplex PCR

Fig. 1. mdx/�7–/– mice lack dystrophin and �7 integrin. (A) Multiplex PCR and ARMSassays were performed on genomic tail DNA to detect wild-type versus �7-integrin-targeted allele and the wild-type versus mdx mutation, respectively. The wild-type �7-integrin allele (727 bp) was detected in wild-type and mdx mice, whereas the targeted�7-integrin allele (482 bp) was detected in the �7–/– and double-knockout mice. Wild-type dystrophin (275 bp) was detected in wild-type and �7–/– mice, whereas the mdxmutation (275 bp) was present in the mdx and double-knockout mice. Neg, negativecontrol lacking DNA. (B) Western blotting with anti-�7A-integrin and �7B-integrinantibodies in gastrocnemius muscle of 3-week-old mice confirmed the absence of �7-integrin protein in �7–/– and mdx/�7–/– mice. (C) Immunofluorescence with anti-CA5.5antibody, which recognizes �7 integrin, demonstrated loss of the �7 integrin in tricepsmuscle from 3-week-old �7–/– and mdx/�7–/– mice. The absence of dystrophin in mdxand mdx/�7–/– mice was verified by immunofluorescence with an anti-dystrophinantibody. Bar, 10 �m.

Jour

nal o

f Cel

l Sci

ence

Page 3: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2187Dystrophin and �7-integrin null mice

detected a 482 bp product representing the targeted �7-integrin allele in both the �7–/– and mdx/�7–/– mice,whereas the wild-type allele was 727 bp. Amplification-resistant mutation system (ARMS) PCR yielded a 275 bpproduct and confirmed the presence of the mdx mutationin mdx and mdx/�7–/– mice (Fig. 1A). Loss of �7 proteinwas confirmed by immunoblotting. Western analysisdemonstrated that both �7A-integrin and �7B-integrinisoforms were missing in mdx/�7–/– mice (Fig. 1B). Theabsence of both dystrophin and �7 integrin in the skeletalmuscle of mdx/�7–/– mice was confirmed byimmunofluorescence (Fig. 1C).

At 10 days of age, the average weight of male wild-type, mdx, �7–/– or mdx/�7–/– pups was 7.9±0.2 g,6.3±0.9 g, 7.8±0.17 g or 5.6±0.45 g, respectively (Fig.2B). The weight of mdx and mdx/�7–/– littermates wasstatistically different from wild-type and �7–/– mice(P<0.05), but was not different from each other. By day18, mdx/�7–/– mice exhibited a failure to thrive andappeared smaller compared with mdx littermates (Fig.2A). By day 21, male mdx/�7–/– mice weighed only5.05±0.51 g (n=8), less than half the weight of wild-type(11.89±0.83 g, n=5), mdx (9.86±0.15 g, n=7) or �7–/–

(13.04±0.54 g, n=9) animals, indicating that double-knockout mice failed to gain weight (Fig. 2C). mdx/�7–/–

animals displayed kyphosis, joint contractures of thelimbs, an abnormal waddling gate, tremors and reducedmobility (a movie that contrasts mobility, jointcontractures, tremor and gait in mdx and mdx/�7–/– micecan be viewed as supplementary material, Movie 1). Thisdebilitating phenotype progressed rapidly, resulting inthe death of these mice at day 16-26 (Fig. 2D).

Muscle membrane integrity is compromised inmdx/�7–/– miceTo evaluate skeletal muscle integrity, mice were injectedwith Evan’s Blue dye (EBD). EBD binds albumin,fluoresces red and can enter cells with compromisedplasma membranes. At 3 weeks, wild-type and �7–/– miceshowed no evidence of EBD uptake, signifying that thesarcolemma was undamaged in these animals (Fig. 3A).By contrast, myofibers from both mdx and dystrophin/�7double-knockout mice showed considerable EBD uptake(Fig. 3A). Muscle from mdx and mdx/�7–/– mice had14.6% and 18.9% EBD-positive fibers, respectively;however, these were not significantly different from eachother (Fig. 3B). These data indicate that the loss of dystrophinbut not �7 integrin, in the skeletal muscle of 3-week-old micedecreases sarcolemmal integrity.

mdx/�7–/– mice exhibit severe muscle pathologyTo investigate whether the skeletal muscle from mdx/�7–/–

mice exhibited greater pathology compared with controls,muscle sections were stained with hematoxylin and eosin.Skeletal muscle from 3-week-old wild-type and �7–/– miceshowed no pathology (Fig. 4A). By comparison, skeletalmuscle from 3-week-old mdx mice exhibited small necroticlesions (Fig. 4A). By contrast, skeletal muscle from mdx/�7–/–

mice exhibited severe pathology including variation inmyofiber size, large necrotic regions and areas of mononuclearcell infiltration (Fig. 4A). These data demonstrate that the

skeletal muscle of 3-week-old mdx/�7–/– mice has undergoneincreased muscle degeneration compared with wild-type, mdxor �7-integrin null animals.

Earlier studies have implicated inflammation in thepathogenesis of muscular dystrophy (McDouall et al., 1990;Tews and Goebel, 1996). To assess the inflammatory responsein mice lacking dystrophin and �7 integrin, tissue cryosectionsfrom the triceps muscle of 3-week-old wild-type, mdx, �7–/–

and mdx/�7–/– mice were incubated with anti-CD4 antibody.CD4 is a type-I membrane glycoprotein expressed onthymocytes, and to some extent, monocytes and macrophage-related cells. There was on average less than 1 CD4-positivecell per field of view from each of the control genotypes (Fig.4B). By contrast, an average of 44 CD4-positive cells per fieldwas observed in the muscle of mdx/�7–/– mice. These results

Fig. 2. Muscle-wasting and reduced viability in mdx/�7–/– mice.(A) Photograph of mdx and mdx/�7–/– littermates aged 18 days. Double-knockout mice appear smaller than mdx littermates and show outwardsigns of kyphosis. These mice also exhibit joint contractures, reducedmobility and tremors (see supplementary material, Movie 1). (B) Theaverage weight of 10-day-old male mdx/�7–/– mice was indistinguishablefrom that of mdx littermates. The weight of both mdx and mdx/�7–/– micewas significantly different from wild-type and �7–/– animals (*P<0.05).(C) The average weight of 21-day-old male mdx/�7–/– mice wassignificantly different from wild-type, mdx, and �7–/– animals (*P<0.05),indicating that double-knockout mice failed to gain weight. (D) Theviability of mdx/�7–/– mice (n=21) is severely reduced compared withwild-type (n=12), �7–/– (n=148) and mdx (n=19) mice. All mdx/�7–/– micedied at 16-26 days.

Jour

nal o

f Cel

l Sci

ence

Page 4: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2188

demonstrate that mdx/�7–/– mice experienced an increasedinflammatory response in skeletal muscle compared with wild-type, mdx or �7-integrin null animals at 3 weeks of age.

Increased muscle regeneration in mdx/�7–/– miceTo examine the extent of skeletal muscle regeneration inmdx/�7–/– mice, fibers containing centrally located nuclei intriceps and tibialis anterior muscles were counted andcompared with control genotypes. As expected, skeletal musclefrom wild-type mice had few centrally located nuclei (Fig. 5A-B). By comparison, the tibialis anterior muscle from 3-week-old mdx and �7–/– mice showed 4.1% and 3.2% fibers withcentrally located nuclei, respectively (Fig. 5B). Their triceps

muscle showed 2.8% and 0.3% of muscle fibers with centrallylocated nuclei, respectively, which was not statisticallysignificant from the wild-type controls (Fig. 5B). By contrast,17.8% of muscle fibers in the tibialis anterior muscle and40.1% of muscle fibers from the triceps muscle from mdx/�7–/–

contained centrally located nuclei (Fig. 5B). These resultssuggest an earlier onset of regeneration in response to musclefiber damage in mdx/�7–/– mice.

Embryonic myosin heavy chain (eMyHC) expression wasexamined as a second measure of muscle regeneration.Consistent with data obtained from the analysis of centrallylocated nuclei in muscle fibers, a significant increase in thenumber of eMyHC-positive fibers was observed in 3-week-old

Journal of Cell Science 119 (11)

Fig. 3. Reduced sarcolemmal integrity inmuscle from mdx/�7–/– mice. (A) Evan’sBlue dye (EBD) uptake was used to assessmembrane integrity of the tibialis anteriorskeletal muscle. Myofibers were outlinedwith Oregon Green-488-conjugated WGA(green). EBD uptake (red) was notdetected in wild-type and �7–/– myofibers.By contrast, significant EBD uptake wasobserved in muscle from mdx andmdx/�7–/– animals. Bar, 10 �m. (B) Thenumber of EBD-positive muscle fibers wasassayed. Wild-type and �7–/– muscle hadless than 1% EBD-positive fibers. Bycontrast, 18.9% of fibers were positive forEBD in mdx/�7–/– mice, but this was notsignificantly different from the number ofEBD-positive fibers seen in mdx animals.

Fig. 4. Severe muscle pathology in mdx/�7–/–

mice. (A) Hematoxylin and eosin staining oftibialis anterior muscle from 3-week-old miceshows the characteristically uniform fiberdiameters in wild-type mice. Small necroticregions and variations in myofiber size wereobserved in muscle of 3-week-old mdx mice.At this age, muscle from �7–/– mice appearedto be similar to wild-type. By contrast, musclefrom mdx/�7–/– mice exhibited extremevariations myofiber size and large areas ofmononuclear cell infiltration (arrow). Bar,10 �m. (B) Inflammation in the muscle ofmdx/�7–/– mice was quantified by counting thenumber of CD4-positive immune cells per fieldof view in triceps muscle of 3-week-old mice.Muscle of wild-type, mdx and �7–/– animalsshowed less than 1% CD4-positive cells. Bycontrast, an average of 43.88 CD4-positiveimmune cells per field was observed in thetriceps muscle of mdx/�7–/– mice, which wassignificantly higher when compared withcontrol genotypes (*P<0.05).

Jour

nal o

f Cel

l Sci

ence

Page 5: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2189Dystrophin and �7-integrin null mice

mdx/�7–/– muscle. As expected, wild-type muscle showed only0.3 eMyHC-positive fibers per field. Muscle from 3-week-oldmdx and �7–/– mice showed 0.2 and 0.1 eMyHC-positive fibersper field, respectively. By contrast, 7.5 eMyHC-positive fibersper field of view were counted in muscle from mdx/�7–/– mice(Fig. 6B). Collectively, these data indicate that loss ofdystrophin and �7 integrin results in more muscle regenerationin mdx/�7–/– mice at the age of 3 weeks compared with wild-type, mdx or �7 integrin deficient animals.

Altered expression of laminin-�2-chain expression in�7–/– and mdx/�7–/– miceIn mature skeletal muscle, the �7-integrin chain preferentiallyassociates with the �1D subunit and binds to laminin-211 andlaminin-221 in the extracellular matrix. The �7�1 integrin has

also been shown to regulate laminin deposition (Li et al., 2003).Immunoblotting and immunofluorescence were undertaken todetermine whether the absence of �7, coupled with the loss ofdystrophin, affected the expression and localization of �1Dintegrin, utrophin and/or laminin. Wild-type, mdx, �7–/– andmdx/�7–/– mice exhibited similar amounts of �1D integrin inskeletal muscle (Fig. 7A), which was confirmed by westernanalysis (data not shown).

Immunofluorescence revealed an increase in laminin-�2-chain expression in mdx mice and downregulation in themuscle of �7–/– and mdx/�7–/– mice (Fig. 7A). This wasconfirmed by quantitative western analysis with laminin-�2bands normalized for protein loading to the constitutively andubiquitously expressed protein cyclooxygenase-1 (Cox-1).Expression of Cox-1 did not appear to change in wild-type or

Fig. 5. Enhanced myofiber regeneration inmdx/�7–/– mice. (A) Hematoxylin and eosinstaining of tibialis anterior muscle of 3-week-old mice to analyze the localization of nucleiwithin myofibers. Centrally localized nucleiwere observed in the muscle of mdx/�7–/– mice.Compared with control genotypes they indicateextensive muscle regeneration, which followsdegeneration of diseased myofibers. Centralnuclei are indicated by arrows. Bar, 10 �m.(B) Quantitation of centrally-located nuclei intriceps and tibialis anterior muscles. Asexpected, wild-type mice exhibited peripherallylocated nuclei in both muscle groups. Centrallylocated nuclei were observed in muscle fibersfrom mdx and �7–/– mice compared with wild-type (2.8% and 0.32%, respectively, for tricepsmuscle; 4.1% and 3.2%, respectively, fortibialis anterior muscle). By contrast, mdx/�7–/–

mice showed 40.1% and 17.5% centrallylocated nuclei in triceps and tibialis anteriormuscle, respectively, which are significantlyhigher than controls (*P<0.05).

Fig. 6. Increased expression of eMyHC inmdx/�7–/– mice. (A) Immunofluorescencewas used to detect eMyHC expression intibialis anterior muscle fibers from 3-week-old mice. Muscle from wild-type, mdx and�7–/– mice showed few eMyHC-positivefibers (green). Many muscle fibers frommdx/�7–/– mice were positive for eMyHC.Rhodamine-labeled WGA (red) was used todefine muscle fibers. Bar, 10 �m.(B) Quantitation of eMyHC-positive fibers.Few eMyHC-positive muscle fibers werecounted in wild-type, mdx or �7–/– mice. Bycontrast, an average of 7.46 fibers per fieldwere positive for eMyHC in mdx/�7–/– mice,which is significantly different from thecontrols (*P<0.05).

Jour

nal o

f Cel

l Sci

ence

Page 6: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2190

diseased states. In mdx mice an increase in laminin-�2 chainwas detected compared with wild-type muscle. By contrast, a7.4-fold decrease in laminin-�2 protein was observed in themuscle of �7-integrin-deficient mice and a 3.3-fold decreasewas observed in mdx/�7–/– mice compared with wild-typeanimals (Fig. 7B). These results indicate that loss of �7 integrinled to reduced deposition of laminin-211 or laminin-221 in thebasal lamina of muscle.

Utrophin expression was assayed with two independent anti-utrophin antibodies (MANCHO3 and MANCHO7). Utrophinis normally localized at MTJs and NMJs in adult mice (Law etal., 1994). Utrophin was localized at the NMJs of 3-week-oldwild-type mice. As previously reported, utrophin expressionwas increased and localized at neuromuscular andextrajunctional sites in 3-week-old mdx mice (Law et al.,1994). In �7–/– and mdx/�7–/– mice, utrophin appearedlocalized at junctional and extrajunctional sites (Fig. 8A).Western blot analysis revealed a significant increase in utrophinexpression in mdx mice (P<0.05, n=3 mice per genotype)compared with wild-type animals (Fig. 8B), which confirmsprevious reports. Utrophin levels in the gastrocnemius musclefrom �7 and mdx/�7–/– mice were similar to those observed inwild-type animals (Fig. 8). These results indicate that the lossof dystrophin alone resulted in increased utrophin. However,the additional loss of the �7 integrin did not result in anincrease in utrophin expression in the skeletal muscle ofmdx/�7–/– mice.

Structural changes in the MTJs of �7 and mdx/�7–/–

miceSince the �7�1 integrin and dystrophin are localized at MTJs

and loss of �7 integrin has been shown to result in defectivematrix deposition at this site (Mayer et al., 1997; Nawrotzki etal., 2003), we compared the ultrastructure of the MTJs from 3-week-old wild-type, mdx, �7–/– and mdx/�7–/– mice (Fig. 9).Longitudinal sections were prepared from the gastrocemiusmuscle and examined by electron microscopy. The MTJs ofwild-type and mdx mice showed extensive sarcolemmal folding(Fig. 9, arrows) increasing the contact area between muscle andtendon; also, a highly organized, continuous and denseextracellular matrix was observed. By contrast, the MTJs of�7–/– mice showed few sarcolemmal folds (Fig. 9, arrows)indicating loss of structural integrity at this site. Theextracellular matrix appeared less organized and was not aselectron-dense as wild-type or mdx tissue confirming previousobservations (Fig. 9, arrowheads) (Mayer et al., 1997;Nawrotzki et al., 2003). Like those of �7-integrin-deficientmice, the MTJs of mdx/�7–/– mice showed few sarcolemmalfolds (arrow) and altered deposition of extracellular matrix(Fig. 9, arrowheads). In addition to these structural changes,mdx/�7–/– mice also showed mononuclear cell infiltrate at thisjunctional site.

DiscussionThe dystrophin glycoprotein complex and �7�1 integrin aretransmembrane receptors in skeletal muscle that providemolecular continuity between laminin in the extracellularmatrix and the cell cytoskeleton. Disruption of either receptorleads to neuromuscular disease in both humans and micereflecting the importance of both complexes in maintainingmuscle integrity (Emery, 1993; Hayashi et al., 1998; Mayer etal., 1997; Monaco et al., 1986; Moser, 1984; Sicinski et al.,

Journal of Cell Science 119 (11)

Fig. 7. Altered expression of laminin-�2chain in the skeletal muscle of mdx/�7–/–

mice. (A) Immunofluorescence was usedto detect the localization of �1D andlaminin-�2 chain in triceps muscle of 3-week-old mice. A similar pattern of �1Dlocalization at the myofiber sarcolemmawas observed in wild-type, �7–/–, mdxand mdx/�7–/– mice.Immunofluorescence revealed a decreasein laminin-�2 chain in the muscle of�7–/– and mdx/�7–/– mice compared withboth wild-type and mdx mice. Bar,10 �m. (B) Laminin-�2 expression in thegastrocnemius muscle was quantitated bywestern analysis. mdx mice showed asignificant increase in laminin-�2expression compared with wild-type.�7–/– and mdx/�7–/– mice showed adecrease in laminin-�2-chain expressioncompared with wild-type and mdxanimals (*P<0.05).

Jour

nal o

f Cel

l Sci

ence

Page 7: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2191Dystrophin and �7-integrin null mice

1989). Levels of the �7-integrin chain are increased twofold inthe muscle of DMD patients and mdx mice (Hodges et al.,1997). Transgenic overexpression of �7 integrin in the skeletalmuscle of severely dystrophic mdx/utr–/– mice can partiallyrescue the diseased phenotype and extend the lifespan of theseanimals without restoring the expression of DGC components(Burkin et al., 2001; Burkin et al., 2005). Together, theseresults suggest the DGC and �7�1 integrin have overlappingand compensatory functions. The severe muscle phenotype andpremature death of the mdx/�7–/– mice reported in this studyfurther extends these studies, and supports the hypothesis thatdystrophin and the �7�1 integrin have overlapping roles inmaintaining the structural and functional integrity of skeletalmuscle. We observed only mild muscle pathology in 3-week-old �7–/– and mdx mice, which is consistent with previouslyreported studies (Grady et al., 1997b; Mayer et al., 1997). Bycontrast, the absence of both proteins in mdx/�7–/– mice resultsin severe muscle degeneration, variation in myofiber size andinflammatory cell infiltration.

The muscle pathology of mice that lack dystrophin andutrophin is similar to that seen in DMD patients (Deconinck etal., 1997b; Grady et al., 1997b). Utrophin is an autosomalhomolog of dystrophin localized to junctional sites in adultskeletal muscle (Blake et al., 1996). In mdx mice, utrophin isincreased and is localized at extrajunctional sites were itreplaces dystrophin and associates with an otherwise normaldystrophin-associated protein complex (Matsumura et al.,1992). Mice that lack dystrophin and utrophin have a mean lifespan of 14 weeks (Burkin et al., 2001; Deconinck et al., 1997b;Grady et al., 1997b), whereas mice that lack dystrophin and �7integrin all die before the age of 4 weeks. These results indicate

Fig. 8. Increased utrophin expression in mdxbut not mdx/ �7–/– mice. (A) Utrophinlocalization was examined byimmunofluorescence in the gastrocnemiusmuscle of 3-week-old wild-type, mdx, �7–/–

and mdx/ �7–/– mice. As expected, utrophinwas localized at the NMJs of muscle fromwild-type mice and at neuromuscular andextrajunctional sites of muscle from mdxanimals, confirming previous studies.Utrophin localization appeared at bothjunctional and extrajunctional sites in �7–/– ormdx/�7–/– mice. Bar, 10 �m. (B) Utrophinexpression in the gastrocnemius muscle wasquantitated by western analysis. mdx miceshowed a statistically significant increase inutrophin expression compared with wild-type(*P<0.05). �7–/– and mdx/�7–/– mice showedno increase in utrophin compared with wild-type animals.

Fig. 9. Ultrastructure of MTJs in muscle from 3-week-old wild-type, mdx, �7–/– and mdx/�7–/– mice. The sarcolemma of the MTJsof wild-type and mdx mice is highly folded, increasing the surfacearea between muscle and tendon (arrows); the extracellular matrixat this site is highly organized and electron-dense. By contrast,MTJs of �7–/– and mdx/�7–/– mice show little or no sarcolemmalfolding (arrows) and the extracellular matrix appeared less denseand organized compared with those of wild-type and mdx mice(arrowheads). Bar, 1 �m.

Jour

nal o

f Cel

l Sci

ence

Page 8: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2192

that compensation by �7 integrin allows mdx/utr–/– mice to livelonger. This hypothesis is supported by the partial rescue of thedystrophic phenotype in mdx/utr–/– mice by transgenicexpression of �7 integrin, which extends the life span of theseanimals approximately threefold (Burkin et al., 2001; Burkinet al., 2005). By contrast, endogenous expression of utrophinin mdx/�7–/– mice appears to be unable to compensate to thesame extent for the loss of �7 integrin and dystrophin.Interestingly, the loss of dystrophin and �7 integrin does notresult in increased utrophin expression as seen in the skeletalmuscle of mdx mice.

The mdx/�7–/– mice analyzed in this study exhibit aphenotype similar to mice deficient in �7-integrin and �-sarcoglycan (Allikian et al., 2004). The phenotypic similaritybetween these mice suggests that functional overlap existsbetween �-sarcoglycan in the dystrophin complex and �7�1integrin. Alternatively, loss of �-sarcoglycan might affect thestability of the dystrophin-glycoprotein complex, resulting inthe observed phenotypic similarity (Hack et al., 2000b; Hacket al., 2000a; Zhu et al., 2001). The production and analysis ofdouble-knockout mice lacking �7 integrin and othersarcoglycan subunits may elucidate further the functionaloverlap between members of the sarcoglycan complex and �7integrin.

EBD uptake in the muscle of mdx/�7–/– mice was similar tothat observed in mdx animals, implying that loss of dystrophinis responsible for the observed membrane damage in bothgroups of animals. Prior investigations have shown that theheight of muscle degeneration and its ensuing regeneration inmdx mice occurs at 4-6 weeks of age (Grady et al., 1997b;McGeachie et al., 1993). By conducting the present studies in3-week-old mice, only the onset of changes in membranepermeability in mdx muscle fibers was observed. This conceptof age-dependency was further supported by the extensivemuscle damage and centrally located nuclei observed inmdx/�7–/– mice compared with the single-knockoutphenotypes. The absence of both dystrophin and �7 integrinresulted in significantly more centrally located nuclei andeMyHC expression in skeletal muscle fibers compared withcontrol genotypes. Therefore, our data indicate that mdx/�7–/–

mice have a requirement for increased muscle regenerationcompared with mdx or �7-integrin null animals. Compromisedcontractile function or too little muscle regeneration might bemechanisms underlying the severe muscle damage observed inthe mdx/�7–/– mice.

The severe muscle pathology observed in mdx/�7–/– mice issimilar to that reported in laminin-�2-deficient mice (Miyagoe-Suzuki et al., 2000). The skeletal muscle of laminin-�2 nullmice show reduced expression of �7 integrin (Hodges et al.,1997). Our data showed increased endogenous expression ofboth �7-integrin and laminin-�2 proteins in the muscle of mdxmice. Although �7 deficient mice exhibit decreased levels oflaminin-�2 in skeletal muscle, these mice live beyond 20weeks of age. These observations suggest that levels oflaminin, dystrophin and utrophin in �7-integrin-deficient miceare sufficient to maintain muscle integrity. A further reductionin laminin-�2-chain expression was not observed in mdx/�7–/–

mice compared with �7–/– animals, indicating that the loss ofthe laminin-�2 chain is primarily due to the loss of �7 integrin.Together, these data support the idea that, in skeletal muscle,a regulatory mechanism exists between the expression of �7

integrin and the laminin-�2 chain. This mechanism might alsobe involved in the rescue of severely dystrophic mice by thetransgenic overexpression of �7 integrin (Burkin et al., 2001).Interestingly, utrophin and laminin expression are upregulatedin mdx mice, but not mdx/�7–/– mice, suggesting the possibilitythat utrophin expression is regulated by laminin.

The inflammatory response to chronic muscle-fiber damageis believed to be a major factor contributing to the progressionof pathology observed in DMD patients (Gorospe et al., 1994;Gosselin and McCormick, 2004). Muscle of mdx mice showschanges in the expression of genes involved in theinflammatory response (Porter et al., 2002). A significantinflammatory response was observed in the skeletal muscle andMTJs of mdx/�7–/– mice compared with mdx or �7-integrin-deficient animals at 3 weeks of age. These observations suggestthat loss of dystrophin and integrin protein complexes lead todecreased extracellular matrix-to-muscle-cell contacts withinmuscle and at critical junctional sites, resulting in increasedmuscle-fiber damage and inflammation.

Although our data support the model of overlappingstructural roles for dystrophin and �7 integrin in maintainingthe integrity of the sarcolemma, it is also possible that theseproteins have functionally intersecting cell-signaling pathwaysthat regulate muscle survival because both mdx and laminin-deficient mice show increased apoptosis (Ruegg et al., 2002;Tidball et al., 1995; Tombes et al., 1995). Integrins exhibitmechanotransduction ability, and the binding of extracellularligands or integrin-clustering can initiate cell signaling throughproteins such as focal adhesion kinase or phosphoinisitol-3-kinase (Schlaepfer et al., 1999; Schwartz, 2001). Interestingly,signaling through FAK and the adaptor molecule growth-factor-receptor-binding protein 2 (Grb2) can lead to activationof the mitogen-activated protein kinase (MAPK) pathway(Schlaepfer et al., 1994), which promotes cell survival (Bonniet al., 1999).

Evidence that dystrophin and other members of the DCGhave cell-signaling capacities that regulate cell survival andgrowth is beginning to emerge. Dystrophin itself can becomephosphorylated, which can alter its affinity for actin (Senter etal., 1993). Calmodulin binding sites are present on bothdystrophin and syntrophin (Madhavan et al., 1992), which linkthe DGC to Ca2+/calmodulin-dependent protein kinase II(CaMKII)-mediated signaling (Rando, 2001). Neuronal nitricoxide synthase (nNOS), which regulates many signalingpathways, is also associated with DGC (Abdelmoity et al.,2000). Finally, Grb2 can bind to �-dystroglycan (Yang et al.,1995) and may therefore be a point of convergence betweendystrophin and integrins.

Recently, several genes have been identified that appear tocompensate for the loss of dystrophin. Using a complementarygene therapy approach, these ‘booster’ genes have beenoverexpressed in skeletal muscle of dystrophic mouse modelsand were shown to rescue the diseased phenotype (Engvall andWewer, 2003). These complementary proteins includeutrophin, �7 integrin, GalNac, nNOS and Adam12 (Burkin etal., 2001; Moghadaszadeh et al., 2003; Nguyen et al., 2002;Tidball and Wehling-Henricks, 2004; Tinsley et al., 1998). Bytaking advantage of single- and double-knockout mice, thisstudy has identified areas of structural and functionalconvergence and independence between the �7�1 integrin anddystrophin protein complexes. Understanding the functional

Journal of Cell Science 119 (11)

Jour

nal o

f Cel

l Sci

ence

Page 9: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2193Dystrophin and �7-integrin null mice

overlap between each of the complementary proteins anddystrophin might identify new pathways that can be targetedto treat muscular dystrophy.

Materials and MethodsGeneration of mdx/�7–/– mice�7–/– mice (C57BL6-�7�gal strain) produced in the Nevada Transgenic Center(Flintoff-Dye et al., 2005) were bred with mdx (C57BL10ScSn-Dmd strain) femaleanimals. The resulting F1 males, which were heterozygous at the �7 locus, werebackcrossed with mdx (C57BL10ScSn-Dmd strain) females. Males and femalesproduced in the F2 generation that were heterozygous at the �7 locus were bred togenerate mdx/� double-knockout mice. The wild-type control strain used in thisstudy was C57BL10ScSn. Male littermates or age-matched mice were used foranalysis.

To genotype mice, genomic DNA was isolated from tail clips using a Wizard SVDNA purification system (Promega, Madison, WI) following the manufacturer’sinstructions. Mice were genotyped by multiplex PCR to detect the wild-type andtargeted �7-integrin alleles with the following primers: �7PF10 (5�-TGAAG-GAATGAGTGCACAGTGC-3�), �7exon1R1 (5�-AGATGCCTGTGGGCAGAG-TAC-3�) and �galR2 (5�-GACCTGCAGGCATGCAAGC-3�). PCR conditions wereas follows: 95°C for 4 minutes then 34 cycles at 95°C for 1 minute, 62°C for 1minute and 72°C for 1 minute. A wild-type band was 727 bp, whereas the �7-integrin-targeted allele produced a 482 bp band.

The mutation in the dystrophin gene was detected by a modified ARMS assay(Amalfitano and Chamberlain, 1996; Burkin et al., 2001). The following primerswere used: p259E (5�-GTCACTCAGATAGTTGAAGCCATTTAA-3�), p260E (5�-GTCACTCAGATAGTTGAAGCCATTTAG-3�) and p306F (5�-CATAGTTATT-AATGCATAGATATTCAG-3�). PCR conditions were as follows: 95°C for 4minutes then 34 cycles at 95°C for 1 minute, 55°C for 1 minute and 72°C for 1minute. Primer set p259 and p306 produced a 275 bp wild-type dystrophin allele.Primer set p260 and p306 detects the mdx point mutation and produced a 275 bpproduct. To genotype the dystrophin gene, separate PCR reactions were performedbecause the product sizes are identical in wild-type and mdx mice.

Isolation of skeletal muscleThree-week-old wild-type, mdx, �7–/– and mdx/�7–/– male mice were euthanized byCO2 inhalation in accordance with a protocol approved by the University of Nevada,Reno Animal Care and Use Committee. The gluteus, gastrocnemius, triceps andtibialis anterior muscles from these mice were dissected, flash-frozen in liquidnitrogen and stored at –80°C.

ImmunoblottingThe gastrocnemius muscle from 3-week-old male mice was ground in liquidnitrogen. Protein was extracted in 200 mM octyl-�-D-glucopyranoside (SigmaAldrich, St Louis, MO), 50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM CaCl2, 1mM MgCl2, 2 mM PMSF and a 1:200 dilution of Protease Inhibitor Cocktail SetIII (Calbiochem, EMD Biosciences, San Diego, CA) for the detection of the �7 and�1 integrins. Protein was quantified by Bradford assay and 80 �g of total proteinwere separated on 8% SDS-PAGE gels under non-reduced conditions, andtransferred to nitrocellulose membranes. Membranes were blocked in OdysseyBlocking Buffer (LiCor Biosciences, Lincoln, NE) that was diluted 1:1 inphosphate-buffered saline (PBS). The �7 integrin was detected with a 1:500 dilutionof rabbit anti-�7A (A2 345) and rabbit anti-�7B (B2 347) polyclonal antibodies.�1D integrin was detected with a 1:500 dilution of rabbit anti-�1D polyclonalantibody (a kind gift of Woo Keun Song, Gwanju Institute for Science andTechnology, South Korea). Blots were incubated with a 1:5000 dilution of AlexaFluor 680-coupled goat anti-rabbit IgG (Molecular Probes, Eugene, OR) to detectprimary antibodies.

To analyze laminin-�2, protein was extracted from gastrocnemius muscle with abuffer containing 2% NP40 and 2% Triton X-100. Protein was quantified by aBradford assay. 60 �g of protein were loaded per well, separated on a 7.5%polyacrylamide gel and transferred for 1 hour to nitrocellulose. Laminin-�2 wasdetected with an anti-laminin-�2 antibody (sc-20142, Santa Cruz Biotechnology,Santa Cruz, CA) at 1:200 followed by a 1:5000 dilution of Alexa Fluor 680-labeledgoat anti-rabbit IgG. The 300 kDa laminin-�2 band was normalized for proteinloading by re-probing the blot with goat anti-Cox-1 polyclonal antibody (Santa CruzBiotechnology, Santa Cruz, CA). Blots were visualized with the Odyssey ImagingSystem and band intensities determined by using Odyssey Imaging software.

To examine utrophin expression, protein was extracted from the gastrocnemiusmuscle with RIPA buffer (50 mM Hepes pH 7.4, 150 mM NaCl, 1 mM Na3VO4,10 mM NaF, 0.5% Triton X-100, 0.5% NP40, 10% glycerol, 2 mM PMSF and a1:200 dilution of Protease Inhibitor Cocktail Set III) and quantified by a BradfordAssay (BioRad Laboratories Inc., Hercules, CA). 80 �g of total protein wereseparated on a 7.5% SDS-PAGE gel and transferred to nitrocellulose. The blot wasincubated with a 1:200 dilution of anti-utrophin mouse monoclonal antibody(MANCHO3 8A4, a kind gift of Glenn Morris, Center for Inherited NeuromuscularDisease, Shropshire, UK) followed by a 1:50,000 dilution of horseradish peroxidase

(HRP)-labeled goat anti-mouse secondary antibody. The 395 kDa utrophin band wasdetected by chemiluminescence and normalized for protein loading by probing thesame blot with anti-Cox-1 antibody (Santa Cruz Biotechnology, Santa Cruz, CA).Band intensities were quantified by using ImageQuant TL software (AmershamBiosciences, Piscataway, NJ).

ImmunofluorescenceTriceps muscles were embedded in Tissue-TEK Optimal Cutting Temperaturecompound (Sakura Finetek USA Inc., Torrance, CA). Ten-�m sections were cutusing a Leica CM1850 cryostat and placed onto Surgipath microscope slides(Surgipath Medical Industries, Richmond, IL). The �7 integrin was detected witha 1:1000 dilution of anti-CA5.5 rat monoclonal antibody (Sierra Biosource, MorganHill, CA) followed by a 1:1000 dilution of FITC-conjugated anti-rat secondaryantibody. The �1D integrin was detected with a 1:500 dilution of rabbit polyclonalantibody followed by a 1:500 dilution of FITC-conjugated anti-rabbit antibody.Laminin-�2 chain was detected with a 1:500 dilution of rabbit anti-�2G polyclonalantibody (a kind gift from Peter Yurchenco, Robert Wood Johnson Medical School,Department of Pathology, Piscataway, NJ). Dystrophin was detected with the mousemonoclonal anti-Dys2 antibody (Novacastra Laboratories, Ltd, Newcastle uponTyne, UK) and utrophin was detected with MANCHO7 7F3 monoclonal antibodyagainst utrophin (a kind gift from Glenn Morris, Center for Inherited NeuromuscularDisease, Shropshire, UK) at a dilution of 1:200. Anti-CD4 monoclonal antibody (akind gift from Dorothy Hudig, University of Nevada, Reno) was used to assaymuscle inflammation at a dilution of 1:200. The mouse monoclonal antibodies wereused in conjunction with a mouse-on-mouse (MOM) immunodetection kit (VectorLaboratories, Burlingame, CA) to block mouse immunoglobulin and a 1:500dilution of FITC-conjugated anti-mouse secondary antibody. Acetylcholinereceptors at NMJs were detected with Rhodamine-labeled �-bungarotoxin at 1:1000(Molecular Probes). Fluorescence was observed with a Zeiss Axioskop 2 Plusfluorescent microscope and images were captured with a Zeiss AxioCam HRcdigital camera and Axiovision 4.1 software.

Embryonic myosin heavy chainEmbryonic myosin heavy chain (eMyHC) was used in immunofluorescenceexperiments to measure muscle regeneration. Immunofluorescence was performedon 10-�m sections from tibialis anterior muscle from 3-week-old wild-type, mdx,�7–/– and mdx/�7–/– mice with a 1:500 dilution of anti-eMyHC antibody (F1.652,Developmental Studies Hybridoma Bank, University of Iowa, IA). The primaryantibody was detected with a 1:500 dilution of FITC-conjugated anti-mousesecondary antibody. A 1 �g/ml concentration of tetramethylrhodamine-conjugatedwheat-germ agglutinin (WGA) (Molecular Probes, Eugene, OR) was used to definemuscle fibers. Multiple adjacent sections were analyzed with ten random, non-overlapping microscopic fields that were counted per animal at 630� magnification.Data was reported as the number of eMyHC-positive fibers per field.

Hematoxylin and eosin stainingTibialis anterior and triceps muscles were cryosectioned and 10-�m sections wereplaced on Surgipath microscope slides. Tissue sections were fixed in ice-cold 95%ethanol for 2 minutes followed by 70% ethanol for 2 minutes and then re-hydratedin running water for 5 minutes. The tissue sections were then stained with Gill’shematoxylin (Fisher Scientific, Fair Lawn, NJ) and rinsed in water for 5 minutes.Tissue sections were placed in Scott’s solution (0.024 M NaHCO3, 0.17 M MgSO4)for 3 minutes and rinsed in water for 5 minutes. Tissue sections were stained ineosin solution (Sigma-Aldrich, St Louis, MO) for 2 minutes. Tissue sections werethen dehydrated in ice-cold 70% and 95% ethanol for 30 seconds each, followed by100% ethanol for 2 minutes. Tissue sections were then cleared in xylene for 5minutes prior to mounting with DePeX mounting medium (Electron MicroscopySciences, Washington, PA). Centrally located nuclei were counted at 630�magnification by bright-field microscopy. The number of central nuclei per musclefiber was determined by counting 1000 muscle fibers for triceps muscle and 700muscle fibers for tibialis anterior muscle per animal. At least five animals from eachgenotype (wild-type, mdx, �7–/– and mdx/�7–/–) were analyzed.

Evan’s Blue dye uptakeMice were injected intraperitoneally with 50 �l per 10 g of body weight sterileEvans Blue dye solution (10 mg/ml). After 3 hours, the gastrocnemius muscle washarvested and flash-frozen in liquid nitrogen. 10-�m cryosections were placed onmicroscope slides and fixed in 4% paraformaldehyde. To outline muscle fibers,tissue sections were incubated with 2 �g/ml Oregon Green-488-conjugated WGA(Molecular Probes, Eugene, OR). Muscle fibers positive for Evans Blue dye werecounted in a minimum of 1000 fibers per animal, and at least three animals fromeach phenotype were analyzed. Counting was conducted and images captured at630� magnification.

Electron microscopyThe gastrocnemius muscle and tendon were dissected from 3-week-old animals andfixed in 2% glutaraldehyde and 2.5% paraformaldehyde in 0.2% Sorenson’sphosphate buffer. Tissue was embedded in LX-112 epoxy (Ladd Research

Jour

nal o

f Cel

l Sci

ence

Page 10: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2194

Industries), sectioned at 0.1 �m wirth a Reichert Ultracut E Ultramicrotome, stainedwith uranyl actetate and lead citrate and viewed with a Hitachi H-600 transmissionelectron microscope at 5000� magnification.

Statistical analysisAll averaged data are reported as the mean ± s.d. Comparisons between multiplegroups were performed by one-way-analysis of variance (ANOVA) for parametricdata or by Kruskal-Wallis one-way-analysis of variance on ranks for non-parametricdata using SigmaStat 1.0 software (Jandel Corporation, San Rafael, CA). P<0.05was considered statistically significant.

The authors thank Glenn Morris (Center for InheritedNeuromuscular Disease, Shropshire, UK) for the MANCHO3 andMANCHO7 anti-utrophin antibodies, Peter Yurchenco (Robert WoodJohnson Medical School, Piscataway, NJ) for the anti-laminin-�2Gantibody, Woo Keun Song (Gwanju Institute for Science andTechnology, South Korea) for the anti-�1D antibody and DorothyHudig (University of Nevada, Reno) for the anti-CD4 antibody. Theauthors also thank Eric Chaney and Paul Scowen for technicalassistance and Dr Heather Burkin for critically reading themanuscript. This work was supported by grants from the NIH/NCRRP20 RR018751-01 and P20 RR15581-04 to D.J.B.

ReferencesAbdelmoity, A., Padre, R. C., Burzynski, K. E., Stull, J. T. and Lau, K. S. (2000).

Neuronal nitric oxide synthase localizes through multiple structural motifs to thesarcolemma in mouse myotubes. FEBS Lett. 482, 65-70.

Allikian, M. J., Hack, A. A., Mewborn, S., Mayer, U. and McNally, E. M. (2004).Genetic compensation for sarcoglycan loss by integrin alpha7beta1 in muscle. J. CellSci. 117, 3821-3830.

Amalfitano, A. and Chamberlain, J. S. (1996). The mdx-amplification-resistantmutation system assay, a simple and rapid polymerase chain reaction-based detectionof the mdx allele. Muscle Nerve 19, 1549-1553.

Blake, D. J., Tinsley, J. M. and Davies, K. E. (1996). Utrophin: a structural andfunctional comparison to dystrophin. Brain Pathol. 6, 37-47.

Bonni, A., Brunet, A., West, A. E., Datta, S. R., Takasu, M. A. and Greenberg, M.E. (1999). Cell survival promoted by the Ras-MAPK signaling pathway bytranscription-dependent and –independent mechanisms. Science 286, 1358-1362.

Bulfield, G., Siller, W. G., Wight, P. A. and Moore, K. J. (1984). X chromosome-linked muscular dystrophy (mdx) in the mouse. Proc. Natl. Acad. Sci. USA 81, 1189-1192.

Burkin, D. J. and Kaufman, S. J. (1999). The alpha7beta1 integrin in muscledevelopment and disease. Cell Tissue Res. 296, 183-190.

Burkin, D. J., Gu, M., Hodges, B. L., Campanelli, J. T. and Kaufman, S. J. (1998).A functional role for specific spliced variants of the alpha7beta1 integrin inacetylcholine receptor clustering. J. Cell Biol. 143, 1067-1075.

Burkin, D. J., Kim, J. E., Gu, M. and Kaufman, S. J. (2000). Laminin and alpha7beta1integrin regulate agrin-induced clustering of acetylcholine receptors. J. Cell Sci. 113,2877-2886.

Burkin, D. J., Wallace, G. Q., Nicol, K. J., Kaufman, D. J. and Kaufman, S. J. (2001).Enhanced expression of the alpha 7 beta 1 integrin reduces muscular dystrophy andrestores viability in dystrophic mice. J. Cell Biol. 152, 1207-1218.

Burkin, D. J., Wallace, G. Q., Milner, D. J., Chaney, E. J., Mulligan, J. A. andKaufman, S. J. (2005). Transgenic expression of {alpha}7{beta}1 integrin maintainsmuscle integrity, increases regenerative capacity, promotes hypertrophy, and reducescardiomyopathy in dystrophic mice. Am. J. Pathol. 166, 253-263.

Campbell, K. P. (1995). Three muscular dystrophies: loss of cytoskeleton-extracellularmatrix linkage. Cell 80, 675-679.

Deconinck, A. E., Potter, A. C., Tinsley, J. M., Wood, S. J., Vater, R., Young, C.,Metzinger, L., Vincent, A., Slater, C. R. and Davies, K. E. (1997a). Postsynapticabnormalities at the neuromuscular junctions of utrophin-deficient mice. J. Cell Biol.136, 883-894.

Deconinck, A. E., Rafael, J. A., Skinner, J. A., Brown, S. C., Potter, A. C., Metzinger,L., Watt, D. J., Dickson, J. G., Tinsley, J. M. and Davies, K. E. (1997b). Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy. Cell 90, 717-727.

Emery, A. E. (1993). Duchenne muscular dystrophy – Meryon’s disease. Neuromuscul.Disord. 3, 263-266.

Engvall, E. and Wewer, U. M. (2003). The new frontier in muscular dystrophy research:booster genes. FASEB J. 17, 1579-1584.

Flintoff-Dye, N. L., Welser, J., Rooney, J., Scowen, P., Tamowski, S., Hatton, W. andBurkin, D. J. (2005). Role for the alpha7beta1 integrin in vascular development andintegrity. Dev. Dyn. 234, 11-21.

Gillis, J. M. (1996). Membrane abnormalities and Ca homeostasis in muscles of the mdxmouse, an animal model of the Duchenne muscular dystrophy: a review. Acta Physiol.Scand. 156, 397-406.

Gorospe, J. R., Tharp, M. D., Hinckley, J., Kornegay, J. N. and Hoffman, E. P. (1994).A role for mast cells in the progression of Duchenne muscular dystrophy? Correlationsin dystrophin-deficient humans, dogs, and mice. J. Neurol. Sci. 122, 44-56.

Gosselin, L. E. and McCormick, K. M. (2004). Targeting the immune system to improveventilatory function in muscular dystrophy. Med. Sci. Sports Exerc. 36, 44-51.

Grady, R. M., Merlie, J. P. and Sanes, J. R. (1997a). Subtle neuromuscular defects inutrophin-deficient mice. J. Cell Biol. 136, 871-882.

Grady, R. M., Teng, H., Nichol, M. C., Cunningham, J. C., Wilkinson, R. S. andSanes, J. R. (1997b). Skeletal and cardiac myopathies in mice lacking utrophin anddystrophin: a model for Duchenne muscular dystrophy. Cell 90, 729-738.

Hack, A. A., Groh, M. E. and McNally, E. M. (2000a). Sarcoglycans in musculardystrophy. Microsc. Res. Tech. 48, 167-180.

Hack, A. A., Lam, M. Y., Cordier, L., Shoturma, D. I., Ly, C. T., Hadhazy, M. A.,Hadhazy, M. R., Sweeney, H. L. and McNally, E. M. (2000b). Differentialrequirement for individual sarcoglycans and dystrophin in the assembly and functionof the dystrophin-glycoprotein complex. J. Cell Sci. 113, 2535-2544.

Hayashi, Y. K., Chou, F. L., Engvall, E., Ogawa, M., Matsuda, C., Hirabayashi, S.,Yokochi, K., Ziober, B. L., Kramer, R. H., Kaufman, S. J. et al. (1998). Mutationsin the integrin alpha7 gene cause congenital myopathy. Nat. Genet. 19, 94-97.

Henry, M. D. and Campbell, K. P. (1996). Dystroglycan: an extracellular matrix receptorlinked to the cytoskeleton. Curr. Opin. Cell Biol. 8, 625-631.

Hodges, B. L., Hayashi, Y. K., Nonaka, I., Wang, W., Arahata, K. and Kaufman, S.J. (1997). Altered expression of the alpha7beta1 integrin in human and murinemuscular dystrophies. J. Cell Sci. 110, 2873-2881.

Law, D. J., Allen, D. L. and Tidball, J. G. (1994). Talin, vinculin and DRP (utrophin)concentrations are increased at mdx myotendinous junctions following onset ofnecrosis. J. Cell Sci. 107, 1477-1483.

Li, J., Rao, H., Burkin, D., Kaufman, S. J. and Wu, C. (2003). The muscle integrinbinding protein (MIBP) interacts with alpha7beta1 integrin and regulates cell adhesionand laminin matrix deposition. Dev. Biol. 261, 209-219.

Lopez, M. A., Mayer, U., Hwang, W., Taylor, T., Hashmi, M. A., Jannapureddy, S.R. and Boriek, A. M. (2005). Force transmission, compliance, and viscoelasticity arealtered in the alpha7-integrin-null mouse diaphragm. Am. J. Physiol. Cell Physiol. 288,C282-C289.

Lyons, P. R. and Slater, C. R. (1991). Structure and function of the neuromuscularjunction in young adult mdx mice. J. Neurocytol. 20, 969-981.

Madhavan, R., Massom, L. R. and Jarrett, H. W. (1992). Calmodulin specifically bindsthree proteins of the dystrophin-glycoprotein complex. Biochem. Biophys. Res.Commun. 185, 753-759.

Matsumura, K., Ervasti, J. M., Ohlendieck, K., Kahl, S. D. and Campbell, K. P.(1992). Association of dystrophin-related protein with dystrophin-associated proteinsin mdx mouse muscle. Nature 360, 588-591.

Mayer, U., Saher, G., Fassler, R., Bornemann, A., Echtermeyer, F., von der Mark, H.,Miosge, N., Poschl, E. and von der Mark, K. (1997). Absence of integrin alpha 7causes a novel form of muscular dystrophy. Nat. Genet. 17, 318-323.

McDouall, R. M., Dunn, M. J. and Dubowitz, V. (1990). Nature of the mononuclearinfiltrate and the mechanism of muscle damage in juvenile dermatomyositis andDuchenne muscular dystrophy. J. Neurol. Sci. 99, 199-217.

McGeachie, J. K., Grounds, M. D., Partridge, T. A. and Morgan, J. E. (1993). Age-related changes in replication of myogenic cells in mdx mice: quantitativeautoradiographic studies. J. Neurol. Sci. 119, 169-179.

Miyagoe-Suzuki, Y., Nakagawa, M. and Takeda, S. (2000). Merosin and congenitalmuscular dystrophy. Microsc. Res. Tech. 48, 181-191.

Moghadaszadeh, B., Albrechtsen, R., Guo, L. T., Zaik, M., Kawaguchi, N., Borup,R. H., Kronqvist, P., Schroder, H. D., Davies, K. E., Voit, T. et al. (2003).Compensation for dystrophin-deficiency: ADAM12 overexpression in skeletal muscleresults in increased alpha 7 integrin, utrophin and associated glycoproteins. Hum. Mol.Genet. 12, 2467-2479.

Monaco, A. P., Neve, R. L., Colletti-Feener, C., Bertelson, C. J., Kurnit, D. M. andKunkel, L. M. (1986). Isolation of candidate cDNAs for portions of the Duchennemuscular dystrophy gene. Nature 323, 646-650.

Moser, H. (1984). Duchenne muscular dystrophy: pathogenetic aspects and geneticprevention. Hum. Genet. 66, 17-40.

Nagel, A., Lehmann-Horn, F. and Engel, A. G. (1990). Neuromuscular transmission inthe mdx mouse. Muscle Nerve 13, 742-749.

Nawrotzki, R., Willem, M., Miosge, N., Brinkmeier, H. and Mayer, U. (2003).Defective integrin switch and matrix composition at alpha 7-deficient myotendinousjunctions precede the onset of muscular dystrophy in mice. Hum. Mol. Genet. 12, 483-495.

Nguyen, H. H., Jayasinha, V., Xia, B., Hoyte, K. and Martin, P. T. (2002).Overexpression of the cytotoxic T cell GalNAc transferase in skeletal muscle inhibitsmuscular dystrophy in mdx mice. Proc. Natl. Acad. Sci. USA 99, 5616-5621.

O’Brien, K. F. and Kunkel, L. M. (2001). Dystrophin and muscular dystrophy: past,present, and future. Mol. Genet. Metab. 74, 75-88.

Ohlendieck, K., Ervasti, J. M., Matsumura, K., Kahl, S. D., Leveille, C. J. andCampbell, K. P. (1991). Dystrophin-related protein is localized to neuromuscularjunctions of adult skeletal muscle. Neuron 7, 499-508.

Pons, F., Augier, N., Leger, J. O., Robert, A., Tome, F. M., Fardeau, M., Voit, T.,Nicholson, L. V., Mornet, D. and Leger, J. J. (1991). A homologue of dystrophin isexpressed at the neuromuscular junctions of normal individuals and DMD patients, andof normal and mdx mice. Immunological evidence. FEBS Lett. 282, 161-165.

Porter, J. D., Khanna, S., Kaminski, H. J., Rao, J. S., Merriam, A. P., Richmonds,C. R., Leahy, P., Li, J., Guo, W. and Andrade, F. H. (2002). A chronic inflammatoryresponse dominates the skeletal muscle molecular signature in dystrophin-deficientmdx mice. Hum. Mol. Genet. 11, 263-272.

Rafael, J. A., Tinsley, J. M., Potter, A. C., Deconinck, A. E. and Davies, K. E. (1998).

Journal of Cell Science 119 (11)

Jour

nal o

f Cel

l Sci

ence

Page 11: Severe muscular dystrophy in mice that lack dystrophin and ... · exhibited more severe muscular dystrophy than either mdx- or 7-integrin-deficient animals and died at 2-4 weeks

2195Dystrophin and �7-integrin null mice

Skeletal muscle-specific expression of a utrophin transgene rescues utrophin-dystrophin deficient mice. Nat. Genet. 19, 79-82.

Rafael, J. A., Townsend, E. R., Squire, S. E., Potter, A. C., Chamberlain, J. S. andDavies, K. E. (2000). Dystrophin and utrophin influence fiber type composition andpost-synaptic membrane structure. Hum. Mol. Genet. 9, 1357-1367.

Rando, T. A. (2001). The dystrophin-glycoprotein complex, cellular signaling, and theregulation of cell survival in the muscular dystrophies. Muscle Nerve 24, 1575-1594.

Ruegg, U. T., Nicolas-Metral, V., Challet, C., Bernard-Helary, K., Dorchies, O. M.,Wagner, S. and Buetler, T. M. (2002). Pharmacological control of cellular calciumhandling in dystrophic skeletal muscle. Neuromuscul. Disord. 12, S155-S161.

Schlaepfer, D. D., Hanks, S. K., Hunter, T. and van der Geer, P. (1994). Integrin-mediated signal transduction linked to Ras pathway by GRB2 binding to focal adhesionkinase. Nature 372, 786-791.

Schlaepfer, D. D., Hauck, C. R. and Sieg, D. J. (1999). Signaling through focal adhesionkinase. Prog. Biophys. Mol. Biol. 71, 435-478.

Schwartz, M. A. (2001). Integrin signaling revisited. Trends Cell Biol. 11, 466-470.Senter, L., Luise, M., Presotto, C., Betto, R., Teresi, A., Ceoldo, S. and Salviati, G.

(1993). Interaction of dystrophin with cytoskeletal proteins: binding to talin and actin.Biochem. Biophys. Res. Commun. 192, 899-904.

Sicinski, P., Geng, Y., Ryder-Cook, A. S., Barnard, E. A., Darlison, M. G. andBarnard, P. J. (1989). The molecular basis of muscular dystrophy in the mdx mouse:a point mutation. Science 244, 1578-1580.

Squire, S., Raymackers, J. M., Vandebrouck, C., Potter, A., Tinsley, J., Fisher, R.,Gillis, J. M. and Davies, K. E. (2002). Prevention of pathology in mdx mice byexpression of utrophin: analysis using an inducible transgenic expression system. Hum.Mol. Genet. 11, 3333-3344.

Suzuki, A., Yoshida, M., Hayashi, K., Mizuno, Y., Hagiwara, Y. and Ozawa, E.(1994). Molecular organization at the glycoprotein-complex-binding site of dystrophin.

Three dystrophin-associated proteins bind directly to the carboxy-terminal portion ofdystrophin. Eur. J. Biochem. 220, 283-292.

Tews, D. S. and Goebel, H. H. (1996). DNA fragmentation and BCL-2 expression ininfantile spinal muscular atrophy. Neuromuscul. Disord. 6, 265-273.

Tidball, J. G., Albrecht, D. E., Lokensgard, B. E. and Spencer, M. J. (1995). Apoptosisprecedes necrosis of dystrophin-deficient muscle. J. Cell Sci. 108, 2197-2204.

Tidball, J. G. and Wehling-Henricks, M. (2004). Expression of a NOS transgene indystrophin-deficient muscle reduces muscle membrane damage without increasing theexpression of membrane-associated cytoskeletal proteins. Mol. Genet. Metab. 82, 312-320.

Tinsley, J. M., Potter, A. C., Phelps, S. R., Fisher, R., Trickett, J. I. and Davies, K.E. (1996). Amelioration of the dystrophic phenotype of mdx mice using a truncatedutrophin transgene. Nature 384, 349-353.

Tinsley, J., Deconinck, N., Fisher, R., Kahn, D., Phelps, S., Gillis, J. M. and Davies,K. (1998). Expression of full-length utrophin prevents muscular dystrophy in mdxmice. Nat. Med. 4, 1441-1444.

Tombes, R. M., Grant, S., Westin, E. H. and Krystal, G. (1995). G1 cell cycle arrestand apoptosis are induced in NIH 3T3 cells by KN-93, an inhibitor of CaMK-II (themultifunctional Ca2+/CaM kinase). Cell Growth Differ. 6, 1063-1070.

Way, M., Pope, B., Cross, R. A., Kendrick-Jones, J. and Weeds, A. G. (1992).Expression of the N-terminal domain of dystrophin in E. coli and demonstration ofbinding to F-actin. FEBS Lett. 301, 243-245.

Yang, S. S., Van Aelst, L. and Bar-Sagi, D. (1995). Differential interactions of humanSos1 and Sos2 with Grb2. J. Biol. Chem. 270, 18212-18215.

Zhu, X., Hadhazy, M., Groh, M. E., Wheeler, M. T., Wollmann, R. and McNally, E.M. (2001). Overexpression of gamma-sarcoglycan induces severe muscular dystrophy.Implications for the regulation of Sarcoglycan assembly. J. Biol. Chem. 276, 21785-21790.

Jour

nal o

f Cel

l Sci

ence