Sabourin

Embed Size (px)

Citation preview

  • 7/29/2019 Sabourin

    1/10

    Clin Genet 2000: 57: 1625Printed in Ireland. All rights reser6ed

    Developmental Biology: Frontiers for Clinical Genetics

    Section Editor: Roderick R McInnes

    e-mail: [email protected]

    The molecular regulation of myogenesis

    Sabourin LA, Rudnicki MA. The molecular regulation of myogenesis.Clin Genet 2000: 57: 1625. Munksgaard, 1999

    Over the past years, several studies have unraveled important mecha-nisms by which the four myogenic regulatory factors (MRFs: MyoD,Myf-5, myogenin, and MRF4) control the specification and the differ-entiation of the muscle lineage. Early experiments led to the hypothesisthat these factors were redundant and could functionally replace oneanother. However, recent experiments using in 6i6o and in 6itro modelshave demonstrated that in fact different aspects of the myogenic pro-gram are controlled by different factors in 6i6o, suggesting that these

    factors play distinct roles during myogenesis. The activity of the MRFsduring proliferation and differentiation of muscle precursor cells hasclearly been demonstrated to be dependent on specific cell-cycle controlmechanisms as well as distinct interactions with other regulatorymolecules, such as the ubiquitously expressed E proteins and severalother transcription factors. Furthermore, the observation that theMRFs can recruit chromatin remodeling proteins has shed some lighton the mechanisms by which the MRFs activate gene expression. Re-cently, a functional role for MyoD during satellite cell activation andmuscle repair has been identified in 6i6o, which cannot be substitutedfor by the other MRFs. This has put forward the hypothesis that thesefactors also play specific biological roles following muscle injury andrepair.

    Luc A Sabourin and MichaelA Rudnicki

    Institute for Molecular Biology and

    Biotechnology, MOBIX, McMaster

    University, Hamilton, Ontario, Canada

    Corresponding author: Michael A Rudnicki,

    McMaster University, 1280 Main St. West,

    Life Sciences Rm 437, Hamilton, Ontario

    L8S 4K1, Canada. Tel: +1 905 5259140

    (ext: 27424); e-mail: [email protected]

    Received 8 October 1999, revised and ac-

    cepted for publication 14 October 1999

    The myogenic regulatory factors (MRFs) are partof a superfamily of basic helix-loop-helix (bHLH)transcription factors including c-myc and acheate-scute (13). The MRF subfamily consists ofMyoD (Myf-3) (4), Myf-5 (5), myogenin (Myf-1)(6), and MRF4 (Myf-6/Herculin) (7 9). Twelveyears ago, the MyoD gene was first isolated fromsubtractive hybridization procedures using my-oblast-specific cDNA libraries (4). The MyoDcDNA was identified by virtue of its ability toconvert fibroblasts into myogenic cells. Subse-quently, low stringency library screens uncovered

    three more MRFs all capable of inducing myo-genic conversion when overexpressed in a vastnumber of nonmuscle cell lines. The MRF proteinscontain a conserved basic DNA-binding domainessential for sequence-specific DNA binding and ahelix-loop-helix motif required for heterodimeriza-tion. Each of the MRFs has been shown to het-erodimerize in 6itro and in 6i6o with E proteins andto bind DNA in a sequence-specific manner at sitesknown as E-boxes (CANNTG). This DNA motif

    is present in the promoters of many skeletal mus-cle-specific genes and mediates gene activation inan MRF-dependent manner (13, 1012).

    Expression of the myogenic factors during

    embryogenesis

    The genes encoding the four MRFs have beenshown to be expressed in a temporally distinctpattern. As determined by in situ hybridization,activation of Myf-5 occurs first in the rostralsomites of the mouse around 8 days postcoitum

    (dpc) and is down-regulated after day 14 (13). Theactivation of myogenin is observed at 8.5 dpcfollowed by MyoD at about 10.5 dpc along withmarkers of terminal differentiation (14). MRF4 isexpressed transiently between days 9 and 12 andrepressed until after birth (15). In the limb bud,Myf-5 is expressed transiently between days 10 and12, followed by co-expression of myogenin andMyoD after day 10.5 and MRF4 after day 16(1315). The specific promoter elements that gov-

    16

  • 7/29/2019 Sabourin

    2/10

    The molecular regulation of myogenesis

    ern the temporo-spatial expression of the MRFshave yet to be fully defined. However, several studieshave identified specific enhancer elements for MyoDand myogenin (1623). Induction of the myogeningene has been shown to be dependent on an E-boxand a myocyte enhancer factor-2 element for properexpression in the somites and limbs of the develop-ing mouse, suggesting that its expression is MRF-dependent, in part (17, 23). The expression of the

    MyoD gene during embryogenesis has been foundto be regulated by at least two distinct enhancers. Afirst element, located 5 kb upstream of the corepromoter region, directs MyoD expression duringthe terminal differentiation of myogenic precursorcells into myotubes and myofibers (18, 22). A secondenhancer, found 20 kb upstream of the MyoD startsite, has been demonstrated to direct the expressionof a LacZ reporter gene during embryogenesis inspecific somitic subdomains (19 21). The superposi-tion of the expression patterns generated by bothenhancers results in a pattern that is indistinguish-

    able from the endogenous MyoD gene. The controlof MyoD expression through the 20-kb elementappears to be E-box-independent (24). In addition,demethylation of the MyoD locus has been demon-strated to play an important role during its activa-tion de no6o (25). Interestingly, Tajbakhsh andco-workers have shown that expression of MyoD isdependent on either Myf-5 or Pax-3 (a paired-boxdomain protein), demonstrating that MyoD actsdownstream of these genes during myogenesis (26)(and reviewed in (27)). In an independent study,ectopic expression of the Pax-3 gene in embryonictissues was shown to induce the expression of MyoD

    and Myf-5 (28). In contrast, overexpression of Pax-3in cultured myoblasts inhibits terminal differentia-tion in 6itro and this phenomenon appears to bedependent on Pax-3 DNA-binding activity (29).Further dissection of the MyoD promoter will likelyuncover other enhancer elements important for theMyoD gene activation.

    Targeted inactivation of the MRF genes

    Gene targeting experiments have provided muchinsight into the functions of the MRFs in 6i6o. The

    introduction of null mutations in the four MRFsinto the germline of mice has demonstrated theexistence of a hierarchical relationship among theMRFs. Inactivation of MyoD in mice results in anapparently normal muscle phenotype with a four-fold increase in Myf-5 expression (30). Similarly,Myf-5-deficient mice display normal skeletal mus-cles but die perinatally because of severe rib defects(31). Interestingly, introduction of the myogenincDNA into the Myf-5 locus is able to rescue the rib

    defect and results in viable and fertile mice but isunable to fully compensate for the absence of Myf-5during myogenic determination (32, 33). However,mice deficient for both MyoD and Myf-5 die at birthowing to a complete absence of skeletal myoblastsand muscle (34). Mice lacking myogenin display anormal number of myoblasts but die at birth be-cause of an absence of myofibers (35, 36). In con-trast, inactivation of MRF4 results in viable mice

    with apparently normal muscles but with a fourfoldincrease in myogenin expression (37 39). Takentogether, these experiments have then defined twogroups of MRFs. The primary MRFs, MyoD andMyf-5, appear to be required for myogenic determi-nation, whereas the secondary MRFs, myogeninand MRF4, are required downstream of MyoD andMyf-5 as differentiation factors (Fig. 1). In addition,these studies have demonstrated that some MRFscan substitute for one another without affectingoverall muscle development, suggesting the exis-tence of potential redundancy among the MRFs(40).

    Recently, the use of MyoD-lacZ transgenic micebred into the MyoD- or Myf-5-deficient back-grounds has been used to address potential redun-dancy (41). MyoD-/- mice display normal epaxial(paraspinal and intercostal) muscle development,whereas hypaxial (limb and abdominal wall) devel-opment is delayed by 2.5 days (Fig. 2). In contrast,Myf-5-/- embryos exhibit normal muscle develop-ment in the limb buds and branchial arches, andmarkedly delayed development of epaxial muscles(41). Furthermore, normal migration of Pax-3-ex-pressing cells into the limb buds and subsequentinduction of Myf-5 in myogenic precursors areobserved, suggesting that Myf-5 expression in thelimb is insufficient for the normal progression ofmyogenic development (41). These observationssuggest that MyoD and Myf-5 play distinct rolesduring the formation of epaxial and hypaxial mus-cles and argue against the existence of redundancyamong the MRFs.

    Fig. 1. Targeted inactivation of the MRFs has defined two

    groups of factors. The primary MRFs, MyoD and Myf-5, are

    required at the determination step for commitment of the

    proliferating somitic cells to the myogenic lineage. The com-

    mitted cells (myoblasts) can proliferate and further differenti-

    ate into myocytes and mature into myofibers under the action

    of the secondary MRFs, myogenin and MRF4.

    17

  • 7/29/2019 Sabourin

    3/10

    Sabourin and Rudnicki

    Fig. 2. Myogenic cell lineages. The temporal expression pattern of a MyoD-LacZ transgene in different null background shows that

    the 5-kb enhancer of MyoD, expressed in differentiated myocytes, is activated in the epaxial lineage (blue) in the absence of MyoD

    and in the hypaxial domain (red) in the absence of Myf-5. MyoD-/- mice display normal epaxial muscle development, whereas

    hypaxial development is delayed by 2.5 days, suggesting a specific role for Myf-5 in the establishment of the epaxial musculature.

    In contrast, Myf-5-/- embryos exhibit normal muscle development in the limb buds and branchial arches, and markedly delayed

    development of epaxial muscles, demonstrating a functional role for MyoD in the establishment of the hypaxial musculature.

    Regulatory mechanisms of MRF activity

    In 6itro differentiation systems have providedfurther into the regulation of MRFs activity andthe relationships between growth and differentia-tion. Several studies have demonstrated that theMRFs can efficiently heterodimerize with productsof the E2-2 (ITF2) and E2-5 genes (E12, E47, andITF1) (42 44) (see Fig. 3 and reviewed in (2)).These heterodimers activate muscle-specific tran-scription of E-box-containing muscle gene pro-moters (44). However, it is not clear whetherspecific heterodimers play distinct biological roles.

    The MRFs have been shown to be negatively regu-lated by the HLH protein Id, which lacks a basicDNA-binding domain (45, 46). The Id factors,encoded by at least four different genes (Id1, Id2,Id3, and Id4), act in a dominant negative mannerby heterodimerizing with E proteins preventingtheir association with the MRFs and subsequentmuscle-specific gene activation (46) (Fig. 3). Simi-larly, MyoD activity has been shown to be inhib-ited in 6itro by the murine twist (mTwist) protein(47). The mTwist protein is also thought to se-quester E proteins, preventing MRF-E protein het-

    erodimer formation. In addition to Id and mTwistproteins, MyoD has recently been shown to benegatively regulated by dimerization with Mist1, anovel bHLH factor that lacks a transactivationdomain (48). The resulting heterodimer does notbind E-box-containing promoters.

    The activity of the MRFs is also tightly coupledto the cell cycle (reviewed in (49, 50)). For exam-ple, the hypophosphorylated form of theretinoblastoma protein (Rb) has been demon-

    strated to associate with MyoD and to be requiredfor efficient transactivation of E-box-containingmuscle-specific promoters (51). The Rb protein isalso necessary to maintain the differentiated phe-notype of cultured myotubes. In addition, induc-tion of differentiation in cultured myoblasts resultsin up-regulation of the cell-cycle inhibitors p21(WAF-1, Cip1) and p16 (5254). The p21 gene hasbeen demonstrated to bear E-box in its upstreamregulatory regions and is activated by MyoD over-expression in transient assays (53). Supportingthese observations, high expression of p21 has been

    correlated with the activation of the myogeningene during embryogenesis (55). The cell-cycleMRF connection is further substantiated by theobservation that overexpression of cyclin D1, aG1-S cyclin and a Cdk4 activator, inhibits MyoDactivity and subsequent transactivation of E-box-containing reporter genes (56, 57). Furthermore,MyoD activity has been demonstrated to be down-regulated by direct interaction with the C-terminaldomain of Cdk4 (58) (Fig. 3). This interactionappears to require the cyclin D1-dependent nucleartargeting of Cdk4. The activity of the MRFs is

    further coupled to cellular proliferation by thenegative regulatory effect of the AP1 heterodimerFos/c-Jun. The proto-oncogene, c-Jun, directlybinds MyoD and inhibits its activity (59, 60). Fur-thermore, upon differentiation of cultured my-oblasts in 6itro, the c-Fos promoter isdown-regulated by MyoD through an E-boxwithin the c-Fos promoter (61). Various otheroncogenes such as c-myc, N-ras, and Ha-ras alsoinhibit muscle differentiation in 6itro, suggesting

    18

  • 7/29/2019 Sabourin

    4/10

    The molecular regulation of myogenesis

    that growth-promoting factors negatively regulatethe MRFs (62 66). In addition, viral oncogenessuch as adenovirus E1a inhibit differentiation andMRF activity by direct physical interaction (67, 68).

    MRF activity is also potentially negatively regu-lated through phosphorylation. For example, myo-genin is phosphorylated directly by protein kinase C(PKC) in 6itro and in 6i6o (69). Fibroblast growthfactor (FGF) treatment or PKC overexpression

    results in threonine-87 phosphorylation in theDNA-binding domain and a loss of DNA-bindingactivity. Similarly, protein kinase A negatively reg-ulates myogenin through an indirect mechanism(70). Recently, the mitogen-activated protein kinase(MAPK) pathway has been shown to be activatedin differentiating muscle cells and to positivelyregulate the expression and activity of the MyoDprotein (71). However, in contrast, Bennet andco-workers have demonstrated that upon mitogenwithdrawal from C2C12 myoblasts, the MAPKp42Erk2 is inactivated concomitant with up-regula-

    tion of muscle-specific genes (72). Supporting this,they showed that overexpression of MAPK phos-phatase-1 inhibited p42Erk2 activity and was suffi-cient to relieve the inhibitory effects of mitogens onmuscle-specific gene expression. Similarly, continu-ous activation of MEK, a MAP kinase kinase, isdetrimental to insulin-like growth factor-1- (IGF-1)or FGF-2-induced myogenesis (73). Recently, theactivity of stress-activated protein kinase 2(SAPK2/p38) has also been demonstrated to beimportant for the terminal differentiation of C2C12myoblasts (74). Whether different components ofthe MAPK pathway play distinct biological roles

    during growth or differentiation remains to beelucidated.

    Regulation of transcription through MRF/co-factor

    interactions

    In recent years, attempts at identifying the mecha-nisms underlying MRF functions have uncovered anumber of MRF-binding proteins and co-factors.

    Interestingly, these co-activators are known to playimportant roles in chromatin remodeling, RNApolymerase II functions, or are transcription fac-tors themselves. Among the co-activators, MyoDhas been shown to interact directly with p300/CBP(75 78). This interaction was demonstrated bothin 6i6o and in 6itro and appeared to be required forthe terminal differentiation of cultured myoblasts.This interaction occurs through the carboxyl cys-teine/histidine-rich (C/H3) domain of p300 andincreases the ability of MyoD to transactivate anE-box-containing reporter construct (76, 78).

    Active gene expression is associated with histoneacetylation and loss of histone/DNA interaction.Interestingly, in addition to p300/CBP, MyoD in-teracts with the histone acetyltransferase PCAF ina multiprotein complex also containing p300/CBP(78). Disruption of this complex by anti-PCAFantibody microinjection inhibits muscle differentia-tion, indicating that recruitment of histone acetyl-transferase activity of PCAF by MyoD, throughp300/CBP, is crucial for activation of the myogenicprogram. In a separate study, Gerber and co-work-ers have demonstrated that a cysteine-histidine-richregion of MyoD, upstream of the basic DNA-

    Fig. 3. The activity of the MyoD family is coupled to cell-cycle control. In proliferating myoblasts, activated cyclin-dependent

    kinases (Cdk4) inhibit MyoD activity through direct interaction. Expression of Id proteins precludes the formation of E

    protein-MRFs heterodimers. Upon differentiation, withdrawal from the cell cycle is maintained by a positive feedback loop in

    which high p21 and Rb expression prevents re-entry into the cell cycle and the MRF-E protein complex is activated. Green arrows

    denote positive, whereas blunt red arrows denote negative regulatory relationships.

    19

  • 7/29/2019 Sabourin

    5/10

    Sabourin and Rudnicki

    binding domain, is necessary for chromatin remod-eling and gene activation by MyoD (79).

    Although no known muscle diseases have beenassociated with genetic alterations in any of theMRFs (80), there is evidence that mutations inco-factors for the MRFs are contributing to thepathogenesis of rhabdomyosarcomas. Such geneticmodifications include the amplification of MDM2,for which overexpression has been shown to inhibit

    myoblast differentiation (81). In addition, het-erokaryon formation studies have revealed thatrhabdomyosarcomas are deficient for an unknownfactor required for MyoD activity (82).

    In addition to these factors, MyoD also interactswith components of the transcriptional machinery.Recently, the TATA-binding protein TFIID hasbeen identified as a novel MyoD-binding proteinand found to stabilize the binding of MyoD to itsconsensus binding site (83). Furthermore, MyoDhas been observed to facilitate the association ofTFIIB with the preinitiation complex subsequent

    to DNA binding. Interaction of the MRF-E12dimers with muscle LIM protein also results inincreased DNA-binding activity and stimulation ofmyogenesis (84). Transcription factors such asMEF2-C, a member of the MEF2 family of tran-scription factors, has also been demonstrated tointeract directly and synergize with the MyoD-E12heterodimer but not with either protein alone (re-viewed in (8587)). Similarly, serum response fac-tor, a MEF2-related protein, has been shown tobind and enhance the activity of MRFs-E12 het-erodimers (88).

    Muscle regeneration and satellite cell function: role

    of MyoD

    Satellite cells, the stem cells of adult skeletal mus-cles, reside beneath the basal lamina of adult skele-tal muscle closely juxtaposed against the musclefibers (89). Satellite cells arise around 17 dpc dur-ing mouse embryogenesis and are believed to rep-resent a unique myoblast lineage. Satellite cellsmediate the postnatal growth of muscle and con-tribute for the most part to the formation of theadult muscle mass (89). Satellite cells make up

    2 7% of the nuclei associated with a particularmyofiber. This proportion varies with age and aparticular muscle group.

    Satellite cells are normally mitotically quiescentbut are activated and re-enter the cell cycle inresponse to stress induced by weight-bearing exer-cise or trauma, including injury (89 91). Thedaughter cells of the activated satellite cells, calledmyogenic precursor cells (mpcs), undergo multiplerounds of division prior to fusion with the existing

    or new myofibers. Satellite cells appear to form apopulation of stem cells that are biologically andbiochemically distinct from their descendant mpcs(89, 92). The total number of quiescent satellitecells in adult muscle remains relatively constantover multiple cycles of degeneration and regenera-tion, suggesting that self-renewal in the satellite cellcompartment maintains a population of quiescentcells (89). However, the numbers and proliferative

    potential of satellite cells become progressively re-duced in muscle diseases presenting with muscularatrophy, such as Duchenne muscular dystrophy(DMD), which is likely due to high levels of ongo-ing regeneration (93, 94).

    The essential role played by satellite cells inmuscle regeneration, muscle hypertrophy, andpostnatal muscle growth has been demonstratedextensively (89, 92, 95). However, the molecularmechanisms underlying the activation and functionof myogenic stem cells are still unclear. As deter-mined by polymerase chain reaction analysis, qui-

    escent satellite cells display no detectable levels ofeither of the four MRFs (96). Upon injury andactivation, MyoD is rapidly up-regulated within 12h. This up-regulation of MyoD occurs prior to theexpression of proliferating cell nuclear antigen(PCNA), a marker for cell proliferation. The ex-pression of myogenin occurs last during the timeassociated with fusion and differentiation (97, 98).Analysis of gene expression by reverse transcrip-tion-polymerase chain reaction of individual satel-lite cells following their activation in intact musclefibers (96) substantiates that quiescent satellite cellsexpress no detectable MRFs but do express the

    c-met receptor tyrosine kinase (the receptor forhepatocyte growth factor). Activated satellite cellsfirst express either Myf-5 or MyoD. Subsequently,both factors are co-expressed during the prolifera-tive phase. Following proliferation, myogenin andMRF4 are expressed in cells entering the terminaldifferentiation program. The absence of MRFmRNA in satellite cells prior to activation suggeststhat satellite cells represent a stem cell lineage thatis distinct from myoblasts. Furthermore, the deno6o induction of Myf-5 and MyoD transcriptionimplies that inductive signals are involved,

    analogous to those that occur during embryogene-sis (27, 99).

    The role of MyoD in satellite cell function hasbeen investigated by interbreeding MyoD-/- mice(30) with mdx mice. The mdx mouse carries a lossof function point mutation in the X-linked dys-trophin gene, and thus is an animal model forhuman Duchenne and Becker muscular dystrophy(93). The mdx mice display a high regenerativecapacity leading to muscle hypertrophy, making it

    20

  • 7/29/2019 Sabourin

    6/10

    The molecular regulation of myogenesis

    an attractive model to investigate the role of theMRFs during muscle regeneration. Studies haveshown that the compound mutant mice(mdx:MyoD-/-) exhibit markedly increased pene-trance of the mdx phenotype characterized by mus-cle atrophy and increased myopathy leading topremature death (100). By 3 5 months of age,mdx:MyoD -/- mice develop a profound dorsal ventral curvature of the spine similar to the lordosis

    and kyphosis of patients with DMD. Interestingly,unlike mdx mice, mdx:MyoD-/- animals also dis-play severe cardiomyopathy, a hallmark of DMDpatients (101).

    Muscle regeneration is severely impaired inMyoD-/- mice and is characterized by an almostcomplete absence of proliferative myogenic precur-sor cells as determined by 3H-thymidine incorpora-tion or immunohistochemistry with antibodyreactive to PCNA (100). However, electron micro-scopic examination of MyoD-deficient muscle re-veals the presence of morphologically normal

    satellite cells. However, cell counts show that theirrelative abundance is increased by 1.8-fold inMyoD-/- muscle and 13-fold in mdx:MyoD-/- mus-cle. These data suggest that up-regulation of MyoDis required for satellite cells to enter the mpc prolif-erative phase prior to terminal differentiation. Inthe absence of MyoD, myogenic stem cells undergoseveral rounds of division and return to a quiescentstate rather than progressing through the develop-mental program. Taken together, these experimentsstrongly support the hypothesis that satellite cellsform a stem cell compartment that is the source ofmyogenic precursor cells (100).

    To gain insight into the regeneration deficit ofMyoD-/- muscle, satellite cell-derived primary cul-tures from adult MyoD-/- hind limb muscle weregenerated and analyzed for proliferative and differ-entiation potential. Low passage MyoD-/- myo-genic cells exhibit a fibroblast-like morphologydistinct from the bipolar morphology of wildtypemyoblasts (102). Myogenic cells lacking MyoDexpress c-met (96, 103), but do not express desmin,an intermediate filament protein typically expressedin myoblasts in 6itro and in 6i6o (104). Following theinduction of differentiation in 6itro, wildtype my-

    oblasts undergo cell-cycle arrest and fuse into mult-inucleated myotubes, whereas MyoD-/- cellscontinue to proliferate and yield reduced numbersof predominantly mononuclear myocytes after sev-eral days in differentiation medium (102, 105). Inaddition, the expression of differentiation-specificmarkers is drastically reduced or absent in MyoD-/-cells. As for MyoD-/- muscle tissue, MyoD-/- my-oblast cultures display a fourfold increase in Myf-5mRNA expression, suggesting that overexpression

    Fig. 4. Role of MyoD in satellite cell function. Upon activa-

    tion, quiescent satellite cells, expressing c-met, first express

    Myf-5 or MyoD before co-expressing both and progressing

    through the developmental program. In the absence of MyoD,

    satellite cells appear to exhibit a propensity for self-renewal

    rather than progression through the differentiation program.

    Expression of Myf-5 alone may allow self-renewal either before

    returning to quiescence (yellow arrow) or up-regulating MyoD

    and formation of proliferative mpcs (white arrows).

    of Myf-5 cannot alleviate the differentiation defectimparted by the inactivation of MyoD in these cells.

    Furthermore, culture mixing experiments usingLacZ-marked MyoD-/- cells has demonstrated thatthe MyoD-/- cellular phenotype is cell autonomous.Interestingly, expression of IGF-1 is markedly in-creased in MyoD-/- myogenic cells cultured underdifferentiation conditions, suggesting that MyoDnormally negatively regulates IGF-1 expression inprimary myogenic cells. One possibility is that IGF-1 promotes proliferation and inhibits differentiationof MyoD-/- myoblasts via an autocrine loop. Inaddition, the expression of M-cadherin is markedlyreduced in MyoD-/- myogenic cells and a require-

    ment for M-cadherin has been reported for cell-cy-cle withdrawal and myoblast fusion (106, 107).Taken together, these results suggest that MyoD-/-myogenic cells represent an intermediate stage inthe satellite cell activation pathway downstream ofthe quiescent state but upstream of the mpc com-partment (102) (see Fig. 4). Since Myf-5-/- mice dieperinatally (31), a definitive role for Myf-5 in satel-lite cell activation has yet to be determined.

    Future work

    It is now well established that the MRFs constitutea group of four bHLH transcription factors thatplay a pivotal role during the specification anddifferentiation of muscle cells. To date, severalsignaling pathways that regulate MRF activityhave been identified. However, it will be of interestto identify additional regulatory components suchas kinases, phosphatases, and other transducersthat are directly controlling the activity of theMRFs under growth and differentiation condi-

    21

  • 7/29/2019 Sabourin

    7/10

    Sabourin and Rudnicki

    tions. Of great interest will be the identificationof the regulatory elements and the factors in-volved in the control of the de no6o MRF geneexpression during embryogenesis and satellite cellactivation. This is especially important for the in-duction of MyoD and Myf-5 as these determina-tion factors are likely to be subject to differentregulatory mechanisms, whether the cells are in acontext of embryogenesis or muscle regeneration.

    The activity of the MyoD protein has beendemonstrated to be modulated by its interactionwith various nuclear co-factors and transactiva-tors. The identification of additional interactingco-factors or RNA polymerase II-associated com-ponents will provide further insights into our un-derstanding of the molecular mechanismsunderlying tissue-specific gene expression. In ad-dition, the identification of specific target genesfor which the expression is regulated by one ormore MRFs will be valuable for understandingthe distinct biological roles played by each of the

    MRFs.Finally, the specific functions of each of theMRFs during satellite cell activation and muscleregeneration remain to be determined. The use ofMyoD-deficient and MRF4-/- mice will proveuseful in elucidating their role during satellite cellactivation but a major difficulty lies in the gener-ation of viable animals bearing mutations for theother two MRFs. The establishment of condi-tional mutant lines may circumvent the viabilityproblems encountered with the Myf-5-/- or myo-genin-null mice. Alternatively, the use of knock-ins, as demonstrated by Wang and co-workers

    (33), may be helpful in this regard.

    References

    1. Olson EN, Klein WH. bHLH factors in muscle develop-

    ment: dead lines and commitments, what to leave in and

    what to leave out. Genes Dev 1994: 8: 18.

    2. Rudnicki MA, Jaenisch R. The MyoD family of tran-

    scription factors and skeletal myogenesis. Bioessays 1995:

    17: 203209.

    3. Weintraub H, Davis R, Tapscott S et al. The myoD gene

    family: nodal point during specification of the muscle cell

    lineage. Science 1991: 251: 761766.4. Davis RL, Weintraub H, Lassar AB. Expression of a

    single transfected cDNA converts fibroblasts to my-

    oblasts. Cell 1987: 51: 9871000.

    5. Braun T, Buschhausen-Denker G, Bober E, Tannich E,

    Arnold HH. A novel human muscle factor related to but

    distinct from MyoD1 induces myogenic conversion in

    10T1/2 fibroblasts. Embo J 1989: 8: 701709.

    6. Edmondson DG, Olson EN. A gene with homology to

    the myc similarity region of MyoD1 is expressed during

    myogenesis and is sufficient to activate the muscle differ-

    entiation program. Genes Dev 1989: 3: 628640.

    7. Braun T, Bober E, Winter B, Rosenthal N, Arnold HH.Myf-6, a new member of the human gene family of

    myogenic determination factors: evidence for a gene clus-ter on chromosome 12. Embo J 1990: 9: 821831.

    8. Miner JH, Wold B. Herculin, a fourth member of the

    MyoD family of myogenic regulatory genes. Proc NatlAcad Sci USA 1990: 87: 10891093.

    9. Rhodes SJ, Konieczny SF. Identification of MRF4: a new

    member of the muscle regulatory factor gene family.Genes Dev 1989: 3: 20502061.

    10. Emerson CP Jr. Skeletal myogenesis: genetics and embry-

    ology to the fore. Curr Opin Genet Dev 1993: 3: 265274.11. Buckingham M. Skeletal muscle development and the

    role of the myogenic regulatory factors. Biochem SocTrans 1996: 24: 506509.

    12. Buckingham M, Cossu G. Myogenesis in the mouse em-

    bryo. Methods Cell Biol 1997: 52: 2952.13. Ott MO, Bober E, Lyons G, Arnold H, Buckingham M.

    Early expression of the myogenic regulatory gene, myf-5,

    in precursor cells of skeletal muscle in the mouse embryo.Development 1991: 111: 10971107.

    14. Sassoon D, Lyons G, Wright WE et al. Expression of twomyogenic regulatory factors myogenin and MyoD1 dur-ing mouse embryogenesis. Nature 1989: 341: 303307.

    15. Bober E, Lyons GE, Braun T et al. The muscle regulatorygene, Myf-6, has a biphasic pattern of expression during

    early mouse development. J Cell Biol 1991: 113: 12551265.

    16. Cheng TC, Tseng BS, Merlie JP, Klein WH, Olson EN.

    Activation of the myogenin promoter during mouse em-bryogenesis in the absence of positive autoregulation.Proc Natl Acad Sci USA 1995: 92: 561565.

    17. Cheng TC, Wallace MC, Merlie JP, Olson EN. Separableregulatory elements governing myogenin transcription inmouse embryogenesis. Science 1993: 261: 215218.

    18. Asakura A, Lyons GE, Tapscott SJ. The regulation ofMyoD gene expression: conserved elements mediate ex-pression in embryonic axial muscle. Dev Biol 1995: 171:

    386398.19. Faerman A, Goldhamer DJ, Puzis R, Emerson CP Jr,

    Shani M. The distal human myoD enhancer sequences

    direct unique muscle-specific patterns of lacZ expressionduring mouse development. Dev Biol 1995: 171: 2738.

    20. Goldhamer DJ, Faerman A, Shani M, Emerson CP Jr.Regulatory elements that control the lineage-specific ex-pression of myoD. Science 1992: 256: 538542.

    21. Goldhamer DJ, Brunk BP, Faerman A et al. Embryonicactivation of the myoD gene is regulated by a highly

    conserved distal control element. Development 1995: 121:637649.

    22. Tapscott SJ, Lassar AB, Weintraub H. A novel myoblast

    enhancer element mediates MyoD transcription. Mol CellBiol 1992: 12: 4994 5003.

    23. Yee SP, Rigby PW. The regulation of myogenin gene

    expression during the embryonic development of themouse. Genes Dev 1993: 7: 12771289.

    24. Kucharczuk KL, Love CM, Dougherty NM, GoldhamerDJ. Fine-scale transgenic mapping of the MyoD coreenhancer: MyoD is regulated by distinct but overlapping

    mechanisms in myotomal and non-myotomal muscle lin-eages. Development 1999: 126: 19571965.

    25. Brunk BP, Goldhamer DJ, Emerson CP Jr. Regulated

    demethylation of the myoD distal enhancer during skele-tal myogenesis. Dev Biol 1996: 177: 490503.

    26. Tajbakhsh S, Rocancourt D, Cossu G, Buckingham M.

    Redefining the genetic hierarchies controlling skeletalmyogenesis: Pax-3 and Myf-5 act upstream of MyoD.Cell 1997: 89: 127138.

    22

  • 7/29/2019 Sabourin

    8/10

    The molecular regulation of myogenesis

    27. Tajbakhsh S, Cossu G. Establishing myogenic identity

    during somitogenesis. Curr Opin Genet Dev 1997: 7:

    634641.

    28. Maroto M, Reshef R, Munsterberg AE et al. Ectopic Pax-3

    activates MyoD and Myf-5 expression in embryonic meso-

    derm and neural tissue. Cell 1997: 89: 139148.

    29. EpsteinJA, Lam P, Jepeal L, Maas RL, Shapiro DN. Pax3

    inhibits myogenic differentiation of cultured myoblast

    cells. J Biol Chem 1995: 270: 1171911722.

    30. Rudnicki MA, Braun T, Hinuma S, Jaenisch R. Inactiva-

    tion of MyoD in mice leads to up-regulation of the

    myogenic HLH gene Myf-5 and results in apparently

    normal muscle development. Cell 1992: 71: 383390.

    31. Braun T, Rudnicki MA, Arnold HH, Jaenisch R. Targeted

    inactivation of the muscle regulatory gene Myf-5 results in

    abnormal rib development and perinatal death. Cell 1992:

    71: 369382.

    32. Wang Y, Jaenisch R. Myogenin can substitute for Myf5 in

    promoting myogenesis but less efficiently. Development

    1997: 124: 25072513.

    33. Wang Y, Schnegelsberg PN, Dausman J, Jaenisch R.

    Functional redundancy of the muscle-specific transcription

    factors Myf5 and myogenin. Nature 1996: 379: 823825.

    34. Rudnicki MA, Schnegelsberg PN, Stead RH et al. MyoD

    or Myf-5 is required for the formation of skeletal muscle.

    Cell 1993: 75: 13511359.35. Hasty P, Bradley A, Morris JH et al. Muscle deficiency and

    neonatal death in mice with a targeted mutation in the

    myogenin gene. Nature 1993: 364: 501506.

    36. Nabeshima Y, Hanaoka K, Hayasaka M et al. Myogenin

    gene disruption results in perinatal lethality because of

    severe muscle defect. Nature 1993: 364: 532535.

    37. Braun T, Arnold HH. Inactivation of Myf-6 and Myf-5

    genes in mice leads to alterations in skeletal muscle devel-

    opment. Embo J 1995: 14: 11761186.

    38. Patapoutian A, Yoon JK, Miner JH et al. Disruption of

    the mouse MRF4 gene identifies multiple waves of myoge-

    nesis in the myotome. Development 1995: 121: 33473358.

    39. Zhang W, Behringer RR, Olson EN. Inactivation of the

    myogenic bHLH gene MRF4 results in up-regulation of

    myogenin and rib anomalies. Genes Dev 1995: 9: 13881399.

    40. Weintraub H. The MyoD family and myogenesis: redun-

    dancy, networks,and thresholds. Cell 1993: 75: 1241 1244.

    41. Kablar B, Krastel K, Ying C et al. MyoD and Myf-5

    differentially regulate the development of limbversus trunk

    skeletal muscle. Development 1997: 124: 47294738.

    42. Murre C, McCaw PS, Vaessin H et al. Interactionsbetween

    heterologous helix-loop-helix proteins generate complexes

    that bind specifically to a common DNA sequence. Cell

    1989: 58: 537544.

    43. Murre C, McCaw PS, Baltimore D. A new DNA binding

    and dimerization motif in immunoglobulin enhancer bind-

    ing, daughterless, MyoD, and myc proteins. Cell 1989: 56:

    777783.

    44. LassarAB, Davis RL, Wright WE et al. Functional activity

    of myogenic HLH proteins requires hetero-oligomeriza-

    tion with E12/E47-like proteins in 6i6o. Cell 1991: 66:

    305315.

    45. Benezra R, Davis RL, Lassar A et al. Id: a negative

    regulator of helix-loop-helix DNA binding proteins. Con-

    trol of terminal myogenic differentiation. Ann NY Acad

    Sci 1990: 599: 111.

    46. Benezra R, Davis RL, Lockshon D, TurnerDL, Weintraub

    H. The protein Id: a negative regulator of helix-loop-helix

    DNA binding proteins. Cell 1990: 61: 4959.

    47. Spicer DB, Rhee J, Cheung WL, Lassar AB. Inhibition ofmyogenic bHLH and MEF2 transcription factors by the

    bHLH protein Twist [see comments]. Science 1996: 272:

    14761480.48. Lemercier C, To RQ, Carrasco RA, Konieczny SF. The

    basic helix-loop-helix transcription factor Mist1 functionsas a transcriptional repressor of myoD. Embo J 1998: 17:

    14121422.

    49. Olson EN. Interplay between proliferation and differenti-ation within the myogenic lineage. Dev Biol 1992: 154:

    261272.

    50. Lassar AB, Skapek SX, Novitch B. Regulatory mecha-nisms that coordinate skeletal muscle differentiation and

    cell cycle withdrawal. Curr Opin Cell Biol 1994: 6: 788794.

    51. Gu W, Schneider JW, Condorelli G et al. Interaction ofmyogenic factors and the retinoblastoma protein mediates

    muscle cell commitment and differentiation. Cell 1993: 72:309324.

    52. Guo K, Wang J, Andres V, Smith RC, Walsh K. MyoD-in-

    duced expression of p21 inhibits cyclin-dependent kinaseactivity upon myocyte terminal differentiation. Mol Cell

    Biol 1995: 15: 3823 3829.53. Halevy O, Novitch BG, Spicer DB et al. Correlation of

    terminal cell cycle arrest of skeletal muscle with induction

    of p21 by MyoD. Science 1995: 267: 10181021.

    54. Schneider JW, Gu W, Zhu L, Mahdavi V, Nadal-GinardB. Reversal of terminal differentiation mediated by p107in Rb-/- muscle cells. Science 1994: 264: 14671471.

    55. Parker SB, Eichele G, Zhang P et al. p53-independent

    expression of p21Cip1 in muscle and other terminallydifferentiating cells. Science 1995: 267: 10241027.

    56. Skapek SX, Rhee J, Kim PS, Novitch BG, Lassar AB.Cyclin-mediated inhibition of muscle gene expression via

    a mechanism that is independent of pRB hyperphosphory-

    lation. Mol Cell Biol 1996: 16: 70437053.57. Skapek SX, Rhee J, Spicer DB, Lassar AB. Inhibition of

    myogenic differentiation in proliferating myoblasts bycyclin D1-dependent kinase. Science 1995: 267: 10221024.

    58. Zhang JM, Wei Q, Zhao X, Paterson BM. Coupling of the

    cell cycle and myogenesis through the cyclin D1-dependent

    interaction ofMyoDwithcdk4. EmboJ 1999: 18: 926 933.59. Bengal E, Ransone L, Scharfmann R et al. Functional

    antagonism between c-Jun and MyoD proteins: a direct

    physical association. Cell 1992: 68: 507519.60. Li L, Chambard JC, Karin M, Olson EN. Fos and Jun

    repress transcriptional activation by myogenin and MyoD:

    the amino terminus of Jun can mediate repression. GenesDev 1992: 6: 676689.

    61. Trouche D, Grigoriev M, Lenormand JL et al. Repressionof c-fos promoter by MyoD on muscle cell differentiation.

    Nature 1993: 363: 7982.

    62. La Rocca SA, Crouch DH, Gillespie DA. c-Myc inhibitsmyogenic differentiation and myoD expression by a mech-

    anism which can be dissociated from cell transformation.Oncogene 1994: 9: 34993508.

    63. Miner JH, Wold BJ. c-myc inhibition of MyoD andmyogenin-initiated myogenic differentiation. Mol Cell Biol1991: 11: 2842 2851.

    64. Kong Y, Johnson SE, Taparowsky EJ, Konieczny SF. Rasp21Val inhibits myogenesis without altering the DNA

    binding or transcriptional activities of the myogenic basic

    helix-loop-helix factors. Mol Cell Biol 1995: 15: 52055213.

    65. Konieczny SF, Drobes BL, Menke SL, Taparowsky EJ.Inhibition of myogenic differentiation by the H-ras onco-

    gene is associated with the down regulation of the MyoD1gene. Oncogene 1989: 4: 473481.

    23

  • 7/29/2019 Sabourin

    9/10

    Sabourin and Rudnicki

    66. Olson EN, Spizz G, Tainsky MA. The oncogenic forms

    of N-ras or H-ras prevent skeletal myoblast differentia-

    tion. Mol Cell Biol 1987: 7: 21042111.

    67. Taylor DA, Kraus VB, Schwarz JJ, Olson EN, Kraus

    WE. E1A-mediated inhibition of myogenesis correlates

    with a direct physical interaction of E1A12S and basic

    helix-loop-helix proteins. Mol Cell Biol 1993: 13: 4714

    4727.

    68. Braun T, Bober E, Arnold HH. Inhibition of muscle

    differentiation by the adenovirus E1a protein: repression

    of the transcriptional activating function of the HLH

    protein Myf-5. Genes Dev 1992: 6: 888902.69. Li L, Zhou J, James G et al. FGF inactivates myogenic

    helix-loop-helix proteins through phosphorylation of a

    conserved protein kinase C site in their DNA-binding

    domains. Cell 1992: 71: 11811194.

    70. Li L, Heller-Harrison R, Czech M, Olson EN. Cyclic

    AMP-dependent protein kinase inhibits the activity of

    myogenic helix-loop-helix proteins. Mol Cell Biol 1992:

    12: 4478 4485.

    71. Gredinger E, Gerber AN, Tamir Y, Tapscott SJ, Bengal

    E. Mitogen-activated protein kinase pathway is involved

    in the differentiation of muscle cells. J Biol Chem 1998:

    273: 1043610444.

    72. Bennett AM, Tonks NK. Regulation of distinct stages of

    skeletal muscle differentiation by mitogen-activated

    protein kinases. Science 1997: 278: 12881291.73. Weyman CM, Wolfman A. Mitogen-activated protein

    kinase kinase (MEK) activity is required for inhibition of

    skeletal muscle differentiation by insulin-like growth fac-

    tor 1 or fibroblast growth factor 2. Endocrinology 1998:

    139: 1794 1800.

    74. Cuenda A, Cohen P. Stress-activated protein kinase-2/

    p38 and a rapamycin-sensitive pathway are required for

    C2C12 myogenesis. J Biol Chem 1999: 274: 43414346.

    75. Sartorelli V, Huang J, Hamamori Y, Kedes L. Molecular

    mechanisms of myogenic coactivation by p300: direct

    interaction with the activation domain of MyoD and with

    the MADS box of MEF2C. Mol Cell Biol 1997: 17:

    10101026.

    76. Yuan W, Condorelli G, Caruso M, Felsani A, Giordano

    A. Human p300 protein is a coactivator for the transcrip-

    tion factor MyoD. J Biol Chem 1996: 271: 90099013.

    77. Eckner R, Yao T-P, Oldread E, Livingston DM. Interac-

    tion and functional collaboration of p300/CBP and

    bHLH proteins in muscle and B-cell differentiation.

    Genes Dev 1996: 10: 24782490.

    78. Puri PL, Sartorelli V, Yang XJ et al. Differential roles of

    p300 and PCAF acetyltransferases in muscle differentia-

    tion. Mol Cell 1997: 1: 3545.

    79. Gerber AN, Klesert TR, Bergstrom DA, Tapscott SJ.

    Two domains of MyoD mediate transcriptional activa-

    tion of genes in repressive chromatin: a mechanism for

    lineage determination in myogenesis. Genes Dev 1997: 11:

    436450.

    80. Anand G, Shapiro DN, Dickman PS, Prochownik EV.

    Rhabdomyosarcomas do not contain mutations in the

    DNA binding domains of myogenic transcription factors.

    J Clin Invest 1994: 93: 59.

    81. Fiddler TA, Smith L, Tapscott SJ, Thayer MJ. Amplifica-

    tion of MDM2 inhibits MyoD-mediated myogenesis. Mol

    Cell Biol 1996: 16: 50485057.

    82. Tapscott SJ, Thayer MJ, Weintraub H. Deficiency in

    rhabdomyosarcomas of a factor required for MyoD ac-

    tivity and myogenesis. Science 1993: 259: 14501453.

    83. Heller H, Bengal E. TFIID (TBP) stabilizes the binding

    of MyoD to its DNA site at the promoter and MyoD

    facilitates the association of TFIIB with the preinitiation

    complex. Nucleic Acids Res 1998: 26: 21122119.

    84. Kong Y, Flick MJ, Kudla AJ, Konieczny SF. Muscle

    LIM protein promotes myogenesis by enhancing the ac-

    tivity of MyoD. Mol Cell Biol 1997: 17: 47504760.

    85. Olson EN, Perry M, Schulz RA. Regulation of muscle

    differentiation by the MEF2 family of MADS box tran-

    scription factors. Dev Biol 1995: 172: 214.

    86. Molkentin JD, Black BL, Martin JF, Olson EN. Cooper-

    ative activation of muscle gene expression by MEF2 and

    myogenic bHLH proteins. Cell 1995: 83: 11251136.

    87. Molkentin JD, Olson EN. Combinatorial control of mus-cle development by basic helix-loop-helix and MADS-box

    transcription factors. Proc Natl Acad Sci USA 1996: 93:

    93669373.

    88. Groisman R, Masutani H, Leibovitch MP et al. Physical

    interaction between the mitogen-responsive serum re-

    sponse factor and myogenic basic-helix-loop-helix

    proteins. J Biol Chem 1996: 271: 52585264.

    89. Bischoff R. The satellite cell and muscle regeneration. In:

    Engel AG, Franszini-Armstrong C, eds. Myogenesis, vol.

    2. New York: McGraw-Hill, 1994: 97118.

    90. Grounds MD. Towards understanding skeletal muscle

    regeneration. Pathol Res Pract 1991: 187: 122.

    91. Miller JB, Schaefer L, Dominov JA. Seeking muscle stem

    cells. Curr Top Dev Biol 1999: 43: 191219.

    92. Grounds MD, Yablonka-Reuveni Z. Molecular and cellbiology of skeletal muscle regeneration. Mol Cell Biol

    Hum Dis Ser 1993: 3: 210256.

    93. Bulfield G, Siller WG, Wight PA, Moore KJ. X chromo-

    some-linked muscular dystrophy (mdx) in the mouse.

    Proc Natl Acad Sci USA 1984: 81: 11891192.

    94. Webster C, Blau HM. Accelerated age-related decline in

    replicative life-span of Duchenne muscular dystrophy my-

    oblasts: implications for cell and gene therapy. Somat

    Cell Mol Genet 1990: 16: 557565.

    95. Grounds DM. Age-associated changes in the response of

    skeletal muscle cells to exercise and regeneration. Ann

    NY Acad Sci 1998: 854: 7891.

    96. Cornelison DD, Wold BJ. Single-cell analysis of regula-

    tory gene expression in quiescent and activated mouse

    skeletal muscle satellite cells. Dev Biol 1997: 191: 270

    283.

    97. Yablonka-Reuveni Z, Rivera AJ. Temporal expression of

    regulatory and structural muscle proteins during myogen-

    esis of satellite cells on isolated adult rat fibers. Dev Biol

    1994: 164: 588603.

    98. Smith CK II, Janney MJ, Allen RE. Temporal expression

    of myogenic regulatory genes during activation, prolifera-

    tion, and differentiation of rat skeletal muscle satellite

    cells. J Cell Physiol 1994: 159: 379385.

    99. Cossu G, Tajbakhsh S, Buckingham M. How is myogene-

    sis initiated in the embryo? Trends Genet 1996: 12: 218

    223.

    100. Megeney LA, Kablar B, Garrett K, Anderson JE, Rud-

    nicki MA. MyoD is required for myogenic stem cell

    function in adult skeletal muscle. Genes Dev 1996: 10:

    11731183.

    101. Megeney LA, Kablar B, Perry RL et al. Severe cardiomy-

    opathy in mice lacking dystrophin and MyoD. Proc Natl

    Acad Sci USA 1999: 96: 220225.

    102. Sabourin LA, Girgis-Gabardo A, Seale P, Asakura A,

    Rudnicki MA. Reduced differentiation potential of pri-

    mary MyoD-/- myogenic cells derived from adult skeletal

    muscle. J Cell Biol 1999: 144: 631643.

    103. Allen RE, Sheehan SM, Taylor RG, Kendall TL, Rice

    GM. Hepatocyte growth factor activates quiescent skele-

    24

  • 7/29/2019 Sabourin

    10/10

    The molecular regulation of myogenesis

    tal muscle satellite cells in 6itro. J Cell Physiol 1995: 165:

    307312.

    104. George-Weinstein M, Foster RF, Gerhart JV, Kaufman

    SJ. In 6itro and in 6i6o expression of alpha 7 integrin and

    desmin define the primary and secondary myogenic lin-

    eages. Dev Biol 1993: 156: 209229.

    105. Yablonka-Reuveni Z, Rudnicki MA, Rivera AJ et al. The

    transition from proliferation to differentiation is delayed

    in satellite cells from mice lacking MyoD. Dev Biol 1999:

    210: 440455.

    106. Zeschnigk M, Kozian D, Kuch C, Schmoll M, Starzinski-

    Powitz A. Involvement of M-cadherin in terminal differ-

    entiation of skeletal muscle cells. J Cell Sci 1995: 108:

    29732981.

    107. Irintchev A, Zeschnigk M, Starzinski-Powitz A, Wernig

    A. Expression pattern of M-cadherin in normal, dener-

    vated, and regenerating mouse muscles. Dev Dyn 1994:

    199: 326337.

    25