Abstract
Rhabdomyosarcomas (RMS) are aggressive childhood soft-tissue malignancies deriving from mesenchymal progenitors that are committed to muscle-specific lineages. Despite the histopathological signatures associated with three main histological variants, termed embryonal, alveolar and pleomorphic, a plethora of genetic and molecular changes are recognized in RMS. Over the years, exposure to carcinogens or ionizing radiations and gene-targeting approaches in vivo have greatly contributed to disclose some of the mechanisms underlying RMS onset. In this review, we describe the principal distinct features associated with RMS variants and focus on the current available experimental animal models to point out the molecular determinants cooperating with RMS development and progression.
Keywords: rhabdomyosarcomas, animal models, skeletal muscle
Introduction
Rhabdomyosarcomas have an incidence of about 50% of all soft-tissue sarcomas and 10% of all malignant solid tumours in children [1,2]. They derive from mesenchimal progenitors committed to myogenic lineages and so may arise in almost any body district [3,4,5,6], thus exhibiting a muscle-specific expression pattern that makes this malignancy rather unique [7]. Diagnosis of RMS, indeed, is predicted by the immunohistochemical or molecular detection of Myogenic Regulatory Factors, such as MyoD and myogenin [8,9], whereas the expression of contractile proteins, like myosin, is indicative of differentiated tumour phenotypes [10]. Basically, several different genomic imbalances and translocations have been recognized in RMS, leading to identification of a rather complex number of deregulated pathways and targets [[3],[4],,[5]]. Recently, much attention has been devoted to the tumour-initiating cells involved in RMS development, suggesting that differences in tumour histology may be dependent on the presence of specific genetic changes in different mesenchymal cell progenitors [11,12,13,14,15]. In this article, we present an overview of the chemical, physical and genetic approaches employed to trigger RMS formation in different mammalian and non-mammalian models.
Histological, genetic and molecular characteristics of RMS
RMS have been classified on the basis of histopathological criteria and genetic signatures. They include two major histological variants, termed embryonal (ERMS) and alveolar (ARMS), and a less common pleomorphic (PRMS) variant. ERMS are more responsive to treatments and make up to 80% of RMS in children of less than 10 yrs of age. ERMS may occur in any body district and are heterogeneous in terms of histological appearance, ranging from poorly to highly differentiated lesions, virtually resembling the multi-step process of embryonic muscle differentiation. ARMS, instead, are characterized by poorer prognosis and are mainly detected in the trunk and body extremities in adolescents and adults [16]. ARMS cells resemble lung alveoli, with clusters of eosinophilic tumour cells arranged loosely and disposed in an alveolar pattern. PRMS are rare and mainly found in adults, and typically have a poor clinical outcome [17,18].
Rhabdomyosarcomas histotype is preferentially correlated with particular genomic aberrations (Table); in particular, ERMS are characterized by a severe genomic instability primarily due to losses or gains on different chromosomes [19,20,21,22]. The most frequent signatures characterizing ERMS are the loss of heterozygosis (LOH) and imprinting (LOI) on chromosome region 11p15.5 [23,24,25,26,27,28,29]. These genetic alterations trigger the impaired expression of different putative tumour suppressor genes like H19 [30], CDKN1C (p57/KIP2) [31,32], and SLC22AIL (BWR1A) [33], but also the overexpression of IGF-2, a tumour-promoting gene imprinted in the opposite direction [24,34]. In addition, frequently associated with ERMS are the deficiency in Patched (PTCH) gene due to LOH on chromosome 9q22 [21,35,,36] and activating mutations in RAS gene [37,38,,39]. Alveolar, instead, are predominantly characterized by the presence of non-random chromosomal translocations [40,41,,42], as well as by other less frequent genetic changes [43,44,45,46,47,48,49]. In particular, the t(2;13)(q35;q14) and t(1;13)(p36;q14) translocations account for about 70% and 10% of ARMS, respectively, giving rise to chimeric proteins that are formed by the fusion of the paired and homeo-DNA binding domain of Pax3 or Pax7 factors with the transactivation domain of Fkhr (FoxO1a) [40,41,,42]. The so-originated Pax3-Fkhr and Pax7-Fkhr transcription factors enable an aberrant transcriptional programme, contributing to RMS progression through multiple mechanisms [19,50,51,52]. Finally, in PRMS, a miscellaneous of several different genetic aberrations has been detected [53,54].
Table 1.
Genetic signatures associated with RMS histotypes
ERMS | ARMS | PRMS | |
---|---|---|---|
LOH and/or LOI | 11p15.5 [23,24,25,26,27,28,29] 9q22 [21,35,,36] | ||
Translocations | 11p1-q11 [22] | t(2;13)(q35;q14) t(1;13)(p36;q14) [40,41,,42] | |
Amplifications | 12q13-15 [20] | 12q13-15 [43,44,,45] 2p24 [43,46,47,48] 2q34-qter 15q24-26 1p36 13q31-32 1q21 8q13-21 [43] | 1q25-q31 11q13.5-q14 8p11.2-p11.1 [53] |
Gains of chromosomes | 2, 7, 8, 11, 12, 13q21, 17, 18, 19, 20 [20,21] | 13 [43] | 1p22-p23 7p 20/20p 1q21-q25 3p12 3q26-pter 4q28-q31 [54] 18/18p 8q21-q23/8q 22q [53,54] 5, 6q [53] |
Losses of chromosomes | 3, 6, 10, 14, 15, 16, 17 [20,21] 9q22, 1p35-36, 14q21-q32 [21] | 16q 9q32-34 13q14-qter [43] 17p 9p21 [49] | 1q, 14, 17p 12q13.2-q13.3 [53] 10q23 15q21-q22 2q21-q35 [54] 3p 5q32-qter 13 [53,54] |
Among the several molecular alterations found in RMS, some are frequent and others are rare, unveiling heterogeneous aetiologies under the convergent phenotype. As summarized in Table, the network of these alterations encompasses the expression of the chimeric Pax3- and Pax7-Fkhr factors [40,41,,42] and the loss or gain of activity of different players, including members of the p53 [55,56,57,58,59,60,61], Rb [62] and CDKs families [31,32,44,49], tumour-suppressor genes [30,33], autocrine/paracrine growth factors [24,34,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85], chemokines [86,87], immunoglobulin superfamily members [88], myogenic proteins [50,52,89,90,91,92], and components of the Akt [93], n- and c-Myc [46,47,48,94], Ras/Erk [37,38,,39] and Sonic hedgehog [21,35] pathways. Moreover, the involvement of certain gene aberrations has been inferred from the study of different human syndromes particularly correlated with RMS [59] (Table), including the Li-Fraumeni syndrome [55], Beckwith-Wiedemann syndrome [95], neurofibromatosis-1 [96], Costello syndrome [97], Gorlin syndrome 2004, retinoblastoma [99], mosaic variegated aneuploidy syndrome [100], mismatch repair deficiency syndrome [101] and Rubinstein-Taybi syndrome [102].
Table 2.
Principal molecular alterations detected in RMS
Gene family | Molecular target | Alteration in RMS | References |
---|---|---|---|
Chimeric fusion genes | PAX3-FKHR PAX7-FKHR | Product of translocation t(2;13)(q35;q14) Product of translocation t(1;13)(q36,q14) | [40,41,,42] |
P53, RB and CDKs | P53 | Loss of activity due to several different gene mutations and/or deletions | [55,56,57,58,59] |
MDM2 | Overexpression and/or gene amplification | [58,60] | |
P63, P73 | Transcript overexpression | [61] | |
RB | Homozygous deletion on the protein-binding pocket domain | [62] | |
CDKN2A, ARF | Gene deletion | [49] | |
CDKN2B | Gene deletion | [49] | |
CDK4 | Overexpression due to locus amplification in 12q13-15 | [44] | |
CDKN1C (p57/KIP2) | Loss of expression due to LOH in 11p15.5 locus | [31,32] | |
Tumour-suppressor genes | H19 | Loss of expression due to LOH in 11p15.5 locus | [30] |
SLC22AIL (BWR1A) | Loss of expression due to LOH in 11p15.15 locus | [33] | |
Autocrine/paracrine growth factors | HGF/c-MET | c-MET activating mutations c-MET overexpression and/or amplification Pax-Fkhr-dependent c-MET overexpression | [63,64,65,66,67,68] |
IGF1R | Pax-Fkhr-dependent IGF1R up-regulation | [69,70,,71] | |
IGF-2 | Overexpression due to LOI, LOH and paternal disomy Pax3-Fkhr-dependent overexpression | [24,34] | |
IGFBP5 | Overexpression | [72,73] | |
HER-1/EGFR | Overexpression | [74] | |
HER-2 | Overexpression | [74] | |
PDGFR-A, PDGF-A and C | Overexpression | [75] | |
VEGF | Overexpression of both short and long isoforms | [76] | |
VEGFR1 | Overexpression at mRNA and protein levels Pax3-Fkhr-dependent overexpression | [76,77,,78] | |
FGFR4 | High expression at mRNA and protein levels Activating mutations in the tyrosine-kinase domain | [79] | |
FGF, glypican-5 | Gene amplification | [80] | |
NGF pathway | Anti-apoptotic autocrine loop | [81] | |
TGF-β/myostatin | Increased expression | [82,83,84,85] | |
Chemokines | MMP2, CXCR4 | Pax3-Fkhr-dependent overexpression | [86] |
IL-4R | Pax3-Fkhr-dependent overexpression | [87] | |
Immunoglobulin superfamily | RAGE | Reduced gene expression | [88] |
Myogenic proteins | MyoD | Frequent expression in inactivated form | [50,52,89,90,91,92] |
AKT | Akt | High levels of phosphorylated Akt (Thr308 and Ser473) | [93] |
RAS/ERK and MYC | KRAS-2, NRAS | Activating point mutations | [37] |
HRAS-1 | Activating point mutations | [38,39] | |
n-MYC | Gene amplification | [46,47,,48] | |
c-MYC | Overexpression | [94] | |
Sonic hedgehog | PTCH1 | Gene deletion | [21,35] |
Table 3.
Human syndromes associated with RMS
Human cancer syndromes | Locus | Genetic mutation | MIM ID | References |
---|---|---|---|---|
Li-Fraumeni syndrome | 17p13.1, 9p21 | Germline transmission of a mutated P53 allele | 151623 | [55] |
Beckwith-Wiedemann syndrome | 11p15.5 | Mutation or deletion of imprinted genes within the 11p15.5 locus | 130650 | [95] |
Neurofibromatosis-1 | 17q11.2 | Mutation in NF1 | 162200 | [96] |
Costello syndrome | 11p15.5 | Germline mutation in HRAS | 218040 | [97] |
Gorlin syndrome | 9q22.3 | Germline mutations in PTCH1 or PTCH2 | 109400 | [98] |
Retinoblastoma | 13q14.1-q14.2 | Germline mutation in RB1 | 180200 | [99] |
Mosaic variegated aneuploidy syndrome | 15q15 | Constitutional biallelic truncating and missense mutations in BUB1B | 257300 | [100] |
Mismatch repair deficiency syndrome | 7p22, 3p21.3, 2p16, 2p22-p21 | Biallelic germline mutations of MLH1, MSH2, MSH6 or PMS2 | 276300 | [101] |
Rubinstein-Taybi syndrome | Unknown | Mutations in CREBBP (>60%) orEP300 (about 3%) | 180849 | [102] |
MIM (Mendelian Inheritance in Man) identification numbers referred to each syndrome can be used to retrieve further information at the following site: http://www.ncbi.nlm.nih.gov/omim.
Collectively, the large body of experimental evidence indicates that RMS development frequently requires the suppression of the p53 pathway in conjunction with secondary cooperating events, including the aberrant activity of different tyrosine-kinases receptors along the Ras axis, or the occurrence of Pax3/7-Fkhr chimeric products in the case of ARMS [3,4,5,6].
Experimental animal models of RMS
Rhabdomyosarcomas development has been detected upon exposure to carcinogen agents or ionizing radiations (Table 4), as well as in several different genetically engineered animal models (Table 5). An overview of these models, together with their main pathological features, is provided below.
Carcinogen agents and ionizing radiations-exposed animal models
Heavy metals
Heavy metals (such as As, Cd, Cr, Ni, Co, Cu, Fe, Hg, Pb, Pt) represent an important family of highly toxic environmental pollutants arising as industrial by-products and displaying mutagenic and carcinogenic potential. Indeed, metal cations can catalyse the production of reactive oxygen species which, in turn, elicit a variety of macromolecular alterations by impairing cellular functions. About 50 years ago, RMS formation was first observed upon intramuscular injection of nickel and cobalt compounds into adult rats 103,104]. Since then, other works have shown that nickel compounds are efficient triggers of RMS formation in rat and rabbit models 105,106,107]. More recently, an in vitro study revealed that cancerous cells derived from the nickel compounds-treated rats were characterized by a mature phenotype [108].
Table 4.
Experimental animal models of RMS – chemical and physical triggers
Triggers | Modality of administration and animal models | Tumour analysis | References |
---|---|---|---|
Heavy metals: nickel and cobalt compounds | Intramuscular single injection in thigh of Wistar rats, C3H or Swiss mice | PRMS, highly anaplastic RMS, fibrosarcoma and myoma | 103,104,105] |
Testis injection in Fisher rats | RMS, fibrous histiocytomas and fibromas | [106] | |
Intramuscular implant in rabbits | RMS with small polygonal or elongated cells, mature myofibres | [107] | |
Pyrrolizidine alkaloids: dehydroretronecine or monocrotaline | Subcutaneous injection in male Sprague-Dawley rats | RMS | [111] |
Benzenediazonium sulphate and derivates | Subcutaneous injection in Swiss mice | RMS, fibrosarcomas, osteosarcoma, fibromas and myxosarcomas | 112,113,114] |
N-methyl-N′-nitro-N-nitrosoguanidine | Microinjection, water or dietary exposure in Medaka or Zebrafish | RMS and other mesenchymal-derived sarcomas | 116,117] |
Azoxymethane and methylazoxymethanol acetate | Water exposure in Medaka and Guppy fish | RMS | [122] |
Polycyclic aromatic hydrocarbons: dimethylbenz[a]anthracene and benzo[a]pyrene | Microinjection, water or dietary exposure in Zebrafish | RMS in embryos and juveniles | [124] |
Subcutaneous injection in neonatal male Sprague-Dawley rats | PRMS and ERMS | [125] | |
Subcutaneous implantation of filters overlaid with gelatine containing benzo[a]pyrene in mice | Foreign-body-induced sarcoma and RMS | [126] | |
Tungsten alloy-based munitions embedded with uranium and lead | Intramuscular leg implantation of nickel- and tantalum-pellets in male F344 rats | Polycythemia and PRMS with lung metastases | [128] |
Ionizing radiations | Repeated doses of β-radiation in CD-1 mice | RMS, squamous-cell carcinoma and malignant fibrous histiocytoma | [129] |
Pyrrolizidine alkaloids
The pyrrolizidine alkaloid monocrotaline and its major metabolite dehydroretronecine are naturally occurring toxins widely distributed in the world. These alkaloids are among the most common poisonous plants affecting livestock, wildlife and humans, as they cause liver toxicity and cancer 109,110]. Administration of dehydroretronecine produced RMS in over 50% of the treated rats, in addition to other neoplasms occurring at lower percentage such as myelogenous leukaemias, hepatocellular carcinomas, and pulmonary adenomas [111].
Benzenediazonium sulphate and relative compounds
Benzenediazonium sulphate (BD) is formed during the cytochrome P-450 catalysed metabolism of the carcinogenic 1-(phenylazo)-2-hydroxynaphthalene (Sudan I, Solvent Yellow 14), which was used as a colouring agent for food and other materials in several countries. Furthermore, BD is a metabolic breakdown product of different classes of nitrogen-nitrogen bond-containing chemicals. Subcutaneous injections of BD in Swiss mice gave rise to different neoplasms that were classified as RMS, fibrosarcomas and osteosarcomas [112]. Other variants of this molecule, namely 4-hydroxy- and 4-methyl-BD, are two ingredients derived from the non-cultivable unedible Agaricus xanthodermus and the cultivable Agaricus bisporus mushrooms, respectively. These molecules promoted RMS development in mice, in addition to fibromas, fibrosarcomas and myxosarcomas 113,114].
N-methyl-N′-nitro-N-nitrosoguanidine
N-nitroso compounds, such as N-methyl-N′-nitro-N-nitrosoguanidine (MNNG), are chemical carcinogens detectable in preserved foodstuffs and cigarettes and represent important risk factors contributing to development of nasopharyngeal carcinoma [115]. It has been shown that fish models, such as Medaka (Oryzias latipes) and Zebrafish (Danio rerio), develop a broad range of neoplasms of mesenchymal derivation, including RMS, when exposed to MNNG 116,117].
Azoxymethane and methylazoxymethanol
Azoxymethane (AOM) is a potent carcinogen causing a high incidence of colon cancer in rodents [118]. Once administered, AOM is metabolized into methylazoxymethanol [119], a mitotoxic molecule that spontaneously decomposes to a reactive alkylating agent with tumourigenic [120] and neurotoxic properties [121]. Different fish species exposed to methylazoxymethanol-acetate exhibited RMS formation [122].
Polycyclic aromatic hydrocarbons
Polycyclic aromatic hydrocarbons are implicated in the aetiology of human cancer due to exposure to cigarette smoke, urban air, pollution, coal combustion and certain occupational situations [123]. For example, exposure to 7,12-dimethylbenz[a]anthracene elicited RMS formation in Zebrafish, although at low incidence [124], whereas the same molecule was recently shown more effective in causing RMS in male Sprague–Dawley rats [125]. Also, RMS was detected in mice treated with benzo[a]pyrene [126], even in association with nickel compounds [105]. Interestingly, benzo[a]pyrene carcinogenicity was lost in mice lacking the aryl hydrocarbon receptor [127].
Depleted uranium and lead
An interesting study was recently performed to assess the health risk effects of soldiers exposed to depleted uranium and lead employed to build tungsten alloy-based munitions [128]. For this purpose, male F344 rats were implanted intramuscularly with pellets of weapons-grade alloy to simulate shrapnel wounds. Within 4–5 months from the time of implantation, animals exhibited serious haematological changes indicative of polycythemia and aggressive PRMS in the surrounding of the pellets, with rapid formation of lung metastases.
Ionizing radiations
Epidemiological evidence identifies ionizing radiations as causative agents contributing to the stepwise process of carcinogenesis. Indeed, repeated doses of β-radiation on the backs of CD-1 mice triggered a subset of neoplasms, with RMS being the most frequently observed [129]. RMS cell lines derived from these tumours all exhibited p53 inactivating mutations, suggesting that excessive exposure to radiation contributes to RMS development through the loss of p53 tumour-suppressor activity.
Virus infection and transgenic expression of viral proteins
Results from a study performed in the 1968s have shown that the infection of newborn rats with Moloney-murine sarcoma virus (MoMSV) predisposes to RMS formation [130]. After virus inoculation in the inguinal area, the solid tumour grown rapidly and expanded toward the leg muscles and the dorsal musculature. Metastases occurred regularly in the lungs and draining lymph nodes, and the metastatic cells in the lung closely resembled the cells of the primary tumour. A subsequent in vitro analysis demonstrated that RMS cells derived from the MoMSV-infected rats had an immature phenotype, displaying staining for desmin, but lack of myosin expression [108].
More recently, transgenic mice harbouring the Simian Virus T Antigene (SV40 TAg) gene under the control of the beta-globin control region were generated, attempting the possibility to observe haematopoietic malignancies as a consequence of the erythroid-specific expression [131]. Unlikely, these mice developed PRMS in different anatomic sites and showed hyperplasia of the pancreatic islet cells, which progressed to pancreatic islet tumour.
In another model, transgenic mice expressing the 2.7-kb SV40 TAg early region under the control of the 5′ region of the SM22alpha gene (expressed in embryonic cardiac muscle) developed a rare form of cardiac RMS at the age of approximately 8–12 weeks [132]. Authors reasoned that the SV40 TAg protein and/or its upstream regulatory region may be implicated in the binding and sequestration of specific protein partners whose identity was yet unknown and whose loss of function may be involved in RMS formation. To reconcile these data with some recent findings, it is now accepted that SV40 TAg, like other viruses products, has the ability to bind p53 and determine its loss of function, as recently demonstrated in patients affected by Li-Fraumeni syndrome with one P53 allele active [133].
Table 5.
Experimental animal models of RMS – biological triggers
Triggers | Treatment or genetic approach and animal model | Tumour analysis | References |
---|---|---|---|
Virus and viral proteins | MoMSV inoculation in newborn Wistar rats | Undifferentiated RMS with lung and limph node metastases | [130] |
Erythroid-specific transgenic expression of SV40 Tag in SJL mice | PRMS and pancreatic islet tumour | [131] | |
Cardiac-specific transgenic expression of SV40 TAg in C57BL/6 mice | Cardiac RMS | [132] | |
P53 | Knock-out P53 in C57BL/6 × CBA mice | Undifferentiated RMS and other neoplasms | [135] |
Double knock-out P53 and FOS in 129Sv X C57BL/6 mice | ERMS | [139] | |
Transgenic HER-2/neu expression in Balb/c mice with P53+/− background | ERMS | [143] | |
KRASG12V conditional expression in adult Balb/c mice with P53+/− or P53−/− background | PRMS | [144] | |
Transgenic KRASG12V expression and simultaneous loss of P53 or gain of P53R172H mutant in C57B16J/S129 mice | PRMS with lung metastases | [145] | |
PAX3-FKHR | PAX3-FKHR knock-in expression and P53 or INK4a/ARF knock-out expression in conditional mice | ARMS | [149] |
RAS | KRASG12D expression alone or combined with a P53+/− or P53−/− background in Zebrafish | Highly invasive ERMS | [158] |
HGF | Transgenic broad overexpression of HGF in FVB/N mice | RMS, amelanotic melanoma, hepatic and mammary tumours | [64] |
Transgenic HGF expression in FVB/C57BL/6 mice with a background INK4a deficient | Lymphomas, fibrosarcomas and multifocal ERMS | [68] | |
Sonic hedgehog | PTCH+/− knock-out in CD-1 mice | RMS | [36] |
Conditional PTCH+/− knock-out in C57BL/6 × CD-1 mice | RMS | [174] | |
Conditional P53−/− in Balb/c mice with PTCH+/− background | ERMS | [175] | |
Gene-trap mediated SUFU+/− in C57BL/6 mice with P53−/−background | RMS | [176] | |
Muscular disorders-associated proteins | Nonsense mutation in dystrophin in non-transgenic mdx mice (model of DMD) | ARMS and ERMS | 185,186] |
Mdx mice interbreeded with P53-deficient mice | ERMS | [191] | |
Knock-out αSGCA in C57BL/10ScSn/J mice (model of LGMD-2D) | ERMS | [186] | |
Deficiency of dysferlin in A/J mice (model of LGMD-2B) | PRMS at high frequency | [192] |
Thus, these findings suggest that viral proteins such as SV40 TAg, having the ability to bind proteins like p53 and pRb, may contribute to RMS pathogenesis.
Gene-targeted animal models
P53 pathway
P53 tumour-suppressor activity promotes apoptosis, senescence or reversible protective cell cycle arrest upon a variety of cellular damage signals [134]. In this sense, cells harbouring inactivating P53 gene mutations are predisposed to cancer, as they escape self-protecting cell death and acquire a long-lived resistant condition. Patients affected by the Li-Fraumeni syndrome, harbouring germline P53 mutations, develop soft-tissue sarcomas [55], including a significant percentage of RMS [13,56]. In addition, P53 mutations and/or overexpression of its negative regulator MDM2 are frequently recognized in RMS [58,60]. So far, P53 null mice were generated through homologous recombination strategy [135]. These mice had normal development, but were susceptible to spontaneous formation of different cancers, including RMS at low incidence 135,136,137,138]. Subsequent works have firmly supported that RMS incidence is greatly increased when loss of p53 activity occurs in association with other deregulated pathways. As such, RMS tumour frequency was increased in mice upon concomitant loss of P53 and FOS [139], the latter being a major component of the AP-1 transcription factor, which regulates various biological processes by converting extracellular signals into changes in the expression of specific target genes [140]. Also, the activity of the tyrosine-kinase HER-2/neu receptor, which is expressed in approximately one-half of human RMS [141] and is involved in the transformation of many cell types [142], promoted ERMS formation in transgenic mice when coupled to loss of P53 [143].
A synergism between p53 and Ras pathways has been frequently observed in RMS. For instance, the conditional expression of the cancer-related activating KRASG12V mutation in adult muscles of P53 null mice triggered formation of PRMS [144], whereas the same KRASG12V form in the presence of the P53R172H mutant triggered PRMS with more aggressive metastases [145], suggesting that p53 mutants, due to gain of toxicity, can have more deleterious effects on tumour development compared with the sole p53 loss [146].
In summary, loss of p53 activity and/or gain of p53 cytotoxic function play(s) a central role in RMS development, especially when coupled to the aberrant activity of additional pathways.
Pax3-Fkhr transcription factor
Pax3-Fkhr chimeric factor strongly cooperates together with the loss of P53 or INK4a/ARF locus in ARMS onset. Transgenic mice carrying PAX3-FKHR exhibited defects in muscle development, including ectopic skeletal myogenesis in the developing neural tube, although they did not exhibit spontaneous tumour formation 147,148], supporting the idea that Pax3-Fkhr expression was not sufficient per se to cause RMS. However, targeting a conditional PAX3-FKHR knock-in allele in terminally differentiating Myf6-expressing myofibres promoted ARMS formation [149]. Strikingly, ARMS frequency was increased in these conditional mice by the simultaneous disruption of either p53 pathway or INK4a/ARF locus [14,149], the latter containing two overlapping tumour suppressor genes, p16INK4a and p14ARF [68], involved in the regulation of cell cycle, senescence and apoptosis 150,151]. These data suggest that expression of Pax3-Fkhr in differentiating Myf6-myofibres seems to be a sine qua non condition predisposing to ARMS, particularly when coupled to disruption of gene targets controlling cell cycle, such as P53 and those included in the INK4a/ARF locus.
Ras/Erk pathway
Activating RAS mutations have been primarily associated with ERMS [13,37,38,39,152,153,154,155]. Indeed, mutations in components of the Ras pathway are responsible of clinically overlapping dominant disorders that are characterized by RMS development, including the Noonan syndrome, Costello syndrome, cardiofaciocutaneous syndrome and LEOPARD syndrome [156]. Costello syndrome, in particular, is characterized by short stature, facial dysmorphism, cardiac defects and predisposition to cancers, including ERMS, because of germline activating mutations in the HRAS gene on chromosome 11p15.5 [97, 157]. Indeed, an elegant model of ERMS has been established by the delivery of a transgenic construct harbouring an activated RAS form (KRASG12D) in muscle-associated cells of Zebrafish [158]. Injected embryos developed highly invasive tumours composed of heterogeneous cell populations, comprising undifferentiated muscle cells, multi-nucleated striated muscle fibres, and infiltrating blood cells. In addition, tumour incidence markedly increased when the KRASG12D transgene was injected into mutant fish with a P53+/− and −/− background, confirming the cooperation between Ras and p53 pathways in RMS development. On the basis of microarray analysis on sorted cell populations of Zebrafish, authors postulated that the tumour-initiating cells were reasonably similar to muscle satellite cells. In summary, oncogenic RAS may itself play a primary role in ERMS development, especially when the gain of activity occurs in muscle satellite cells. Moreover, RMS frequency is greatly increased upon the simultaneous loss of p53 function, as observed in Zebrafish [158] and adult mice 144,145].
Hgf/c-Met pathway
C-MET proto-oncogene encodes a tyrosine-kinase receptor that, upon binding with the hepatocyte growth factor (Hgf), promotes cellular growth, motility and survival, extracellular matrix degradation and angiogenesis 159,160]. Excessive activation of this pathway has been implicated in a subset of human cancers, including RMS 161,162]. In transgenic mice, inappropriate Hgf expression gave rise to distinct tumours of both mesenchymal and epithelial origin [64,163,164], with a prevalence of malignant mammary tumours, melanomas, RMS, fibrosarcomas, squamous papillomas, basal cell and hair follicle tumours. Later, it has been demonstrated that aberrant c-Met signalling and simultaneous INK4a/ARF locus inactivation are critical for RMS genesis [68]. Indeed, a consistent percentage of INK4a/ARF−/− mice developed lymphomas and fibrosarcomas [165], whereas almost all INK4a/ARF−/− mice overexpressing Hgf exhibited highly invasive RMS at 3 months of age [68]. These data suggest that constitutive activation of c-Met and simultaneous absence of p16INK4a and p19ARF may give rise to a pre-malignant population of myogenic precursors, which cannot withdraw from the cell cycle and are resistant to p53-mediated apoptosis 166,167]. Unlike embryogenesis, during which a Pax3-dependent expression of c-MET in the lateral dermomyotome is required for the appropriate migration of myogenic precursors to the limb [168], an aberrant Pax3-Fkhr-dependent c-MET transcription takes place in RMS. As the Hgf/c-Met pathway is only transiently required for the activation of satellite cells following skeletal muscle injury [169], its persistent gain of function in RMS cells has been supposed to allow invasiveness through continuous proliferation and migration [63,65,66,67], ideally resembling a regenerating muscle that fails to repair [68].
Sonic hedgehog pathway
Inappropriate activation of the Sonic hedgehog (Shh) pathway, due to Ptch1 receptor inactivation, has been associated with familial cancer. In particular, germline mutations in the PTCH1 gene lead to Gorlin syndrome, also termed Nevoid Basal Cell Carcinoma Syndrome, characterized by a variety of clinical problems such as increased body size, developmental abnormalities of the skeleton, and increased incidence of sporadic Basal Cell Carcinomas (BCC), Medulloblastoma (MB) and RMS [98]. In addition, a deficiency in PTCH1 gene due to LOH on chromosome 9q22 has been further implicated in RMS [21]. Ptch1 basally suppresses the activity of the seven-pass membrane protein Smoothened (Smo), while the binding to the Sonic hedgehog (Shh) relieves the inhibition of Smo, culminating in the activation of the downstream Gli transcription factors. The latter regulate a variety of processes in invertebrate and vertebrate embryonic development 170,171]. So far, PTCH1+/− mice have been considered as a model of multi-organ tumourigenesis [172], as they were characterized by the development of many characteristic features of Gorlin syndrome, including a predisposition to radiation-induced teratogenesis and RMS formation [36]. Importantly, PTCH1+/−mice displayed elevated Igf-2 levels [35,36], suggesting that Ptch1 acts as a negative regulator of Igf-2, which is in turn required for the formation of MB and RMS. In comparison with P53+/− mice, PTCH1+/− mice predominantly showed less aggressive RMS due to a greater degree of differentiation [173], clearly highlighting how different mutations can have a different impact on tumour behaviour. Later on, the development of an elegant conditional mouse model allowed to demonstrate that the time-point and the gene dose of PTCH1 inactivation predispose to development of certain tumours rather than others [174]. In particular, RMS was observed when PTCH1 heterozygosity was induced prenatally [174], especially in the presence of simultaneous loss of p53 [175]. On the other hand, mono- or bi-allelic postnatal deletion of PTCH1 respectively lead to hamartomatous gastrointestinal cystic tumours, BCC precancerous lesions of the gastrointestinal epithelium and mesenteric tumours [174].
Recently, mice deficient for another regulator of the Shh pathway, termed Suppressor of Fused (SUFU), have been generated [176]. SUFU is a negative modulator of Shh signalling [170] and its gene ablation results in embryonic lethality [177], suggesting a critical role in higher organisms. Like PTCH1, SUFU is believed to be a tumour-suppressor gene [178], as a subset of MB patients carry germline and somatic SUFU mutations. In comparison with PTCH1+/− mice, SUFU+/− mice were not tumour prone; however, simultaneous loss of SUFU and P53 triggered MB and RMS in mice [176].
Overall, these data suggest that inappropriate activation of the Shh pathway contributes to RMS development, especially in association with p53 loss of function.
Muscular disorders associated-proteins
Four different animal models of neuromuscular disorders have been associated with RMS development. Among them, the non-transgenic mdx mice ideally represent the animal phenocopy of the X-linked Duchenne Muscular Dystrophy (DMD) [179], as they lack dystrophin due to a premature stop codon in exon 23 of dystrophin gene 180,181,182]. Dystrophin confers resistance to skeletal, cardiac and smooth muscle cells [183] by connecting F-actin in the subsarcolemmal cytoskeleton to the Dystrophin-Glycoprotein Complex (DGC) that spans the sarcolemma and attaches to laminin-2 (merosin) in the extracellular matrix [184]. In particular, it has been shown that old mdx mice (between 16.5 and 24 months of age) develop ARMS [185], although another research group has described development of ERMS in the same model [186]. Spontaneous formation of ERMS has been also detected in alpha sarcoglycan (αSGCA) deficient mice [186], which represent the animal phenocopy of the autosomal recessive Limb-Girdle Muscular Dystrophy-2D form (LGMD-2D) 187,188]. The αSGCA gene encodes a transmembrane glycoprotein that stabilizes the DGC complex [184] and protects muscle cells from contraction-induced damage 189,190]. It is worth noting that all mdx and αSGCA−/− mice developing RMS were characterized by harbouring cancer-related mutated p53 forms or overexpressing mutated or deleted Mdm2 forms lacking the p53-binding domain (in the case of mdx model) [186], confirming that disruption of the p53 pathway cooperates with RMS formation. To further corroborate this evidence, P53-deficient mdx mice have been recently generated, thus demonstrating that the regenerative microenvironment in skeletal muscle of mdx mice, when coupled to P53 deficiency, is sufficient to robustly induce ERMS in young mice [191]. Finally, development of PRMS at a high frequency has been detected in the A/J mouse strain [192], characterized by a progressive muscular dystrophy homologous to LGMD-2B due to lack of dysferlin, a protein involved in muscle repair 193,194].
Results from studies on these animal models suggest that the continuous activation and proliferation of satellite cells, characterizing the lifelong myofibre degeneration and regeneration in muscular disorders, predispose a local environment that may greatly increase the chance of developing RMS, particularly in the presence of Mdm2/p53 cancer-associated alterations.
Conclusions
Generation of animal models has provided a powerful tool for understanding the molecular determinants cooperating with RMS formation. Due to technology limitations, until few years ago, experimental induction of RMS was merely obtained by exposure to disparate classes of chemicals and ionizing radiations as well. Over the last two decades, the growing availability of genetic models has clearly outlined that the onset of RMS, as commonly seen in different cancers, requires the simultaneous occurrence of multiple aberrant molecular events, such as the loss of P53, RB and INK4a/ARF function along with the gained activity of Hgf/c-Met, Ras, Shh pathways and Pax3/7-Fkhr chimeric factors. Recent data further confirmed that loss of skeletal muscle integrity, as observed in some neuromuscular disorders, may supply a local tissue environment predisposing to RMS development. Apart from these different cues, RMS tumour histotype seems to be developmentally stage-dependent, being dictated from the timing and cell host in which specific molecular alterations arise.
In perspective, it will be attractive to bring together old and new evidence by coupling chemical and physical exposure in gene-targeted animal models. This combined approach could help to unravel how a specific genetic background may predispose to or protect from cancer formation in the presence of environmental risk factors.
Acknowledgments
We are grateful to Dr. Arianna Bellucci for kindly revising the manuscript. This work was supported by the Associazione Amici per il Cuore-ONLUS, Chiari (Brescia)-Italy to A.F. and E.M., by the Fondazione Cariplo grant to E.M. and by the University of Brescia research fund (ex 60%) to A.F.
Conflict of interest
The authors confirm that there are no conflicts of interest.
References
- Ognjanovic S, Linabery AM, Charbonneau B, et al. Trends in childhood rhabdomyosarcoma incidence and survival in the United States, 1975-2005. Cancer. 2009;115:4218–26. doi: 10.1002/cncr.24465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer WH, Spunt SL. Soft tissue sarcomas of childhood. Cancer Treat Rev. 2004;30:269–80. doi: 10.1016/j.ctrv.2003.11.001. [DOI] [PubMed] [Google Scholar]
- Barr FG. The role of chimeric paired box transcription factors in the pathogenesis of pediatric rhabdomysarcoma. Cancer Res. 1999;59:1711s–5s. [PubMed] [Google Scholar]
- Merlino G, Helman LJ. Rhabdomyosarcoma–working out the pathways. Oncogene. 1999;18:5340–8. doi: 10.1038/sj.onc.1203038. [DOI] [PubMed] [Google Scholar]
- De Giovanni C, Landuzzi L, Nicoletti G, et al. Molecular and cellular biology of rhabdomyosarcoma. Future Oncol. 2009;5:1449–75. doi: 10.2217/fon.09.97. [DOI] [PubMed] [Google Scholar]
- Xia SJ, Pressey JG, Barr FG. Molecular pathogenesis of rhabdomyosarcoma. Cancer Biol Ther. 2002;1:97–104. doi: 10.4161/cbt.51. [DOI] [PubMed] [Google Scholar]
- Tonin PN, Scrable H, Shimada H, et al. Muscle-specific gene expression in rhabdomyosarcomas and stages of human foetal skeletal muscle development. Cancer Res. 1991;51:5100–6. [PubMed] [Google Scholar]
- Berkes CA, Tapscott SJ. MyoD and the transcriptional control of myogenesis. Semin Cell Dev Biol. 2005;16:585–95. doi: 10.1016/j.semcdb.2005.07.006. [DOI] [PubMed] [Google Scholar]
- Bergstrom DA, Tapscott SJ. Molecular distinction between specification and differentiation in the myogenic basic helix-loop-helix transcription factor family. Mol Cell Biol. 2001;21:2404–12. doi: 10.1128/MCB.21.7.2404-2412.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morotti RA, Nicol KK, Parham DM, et al. An immunohistochemical algorithm to facilitate diagnosis and subtyping of rhabdomyosarcoma: the Children's Oncology Group experience. Am J Surg Pathol. 2006;30:962–8. doi: 10.1097/00000478-200608000-00005. [DOI] [PubMed] [Google Scholar]
- Charytonowicz E, Cordon-Cardo C, Matushansky I, et al. Alveolar rhabdomyosarcoma: is the cell of origin a mesenchymal stem cell? Cancer Lett. 2009;279:126–36. doi: 10.1016/j.canlet.2008.09.039. [DOI] [PubMed] [Google Scholar]
- Merlino G, Khanna C. Fishing for the origins of cancer. Genes Dev. 2007;21:1275–9. doi: 10.1101/gad.1563707. [DOI] [PubMed] [Google Scholar]
- Linardic CM, Downie DL, Qualman S, et al. Genetic modeling of human rhabdomyosarcoma. Cancer Res. 2005;65:4490–5. doi: 10.1158/0008-5472.CAN-04-3194. [DOI] [PubMed] [Google Scholar]
- Keller C, Capecchi MR. New genetic tactics to model alveolar rhabdomyosarcoma in the mouse. Cancer Res. 2005;65:7530–2. doi: 10.1158/0008-5472.CAN-05-0477. [DOI] [PubMed] [Google Scholar]
- Ren YX, Finckenstein FG, Abdueva DA, et al. Mouse mesenchymal stem cells expressing PAX-FKHR form alveolar rhabdomyosarcomas by cooperating with secondary mutations. Cancer Res. 2008;68:6587–97. doi: 10.1158/0008-5472.CAN-08-0859. [DOI] [PubMed] [Google Scholar]
- Tsokos M, Webber BL, Parham DM, et al. Rhabdomyosarcoma. A new classification scheme related to prognosis. Arch Pathol Lab Med. 1992;116:847–55. [PubMed] [Google Scholar]
- Sultan I, Qaddoumi I, Yaser S, et al. Comparing adult and pediatric rhabdomyosarcoma in the surveillance, epidemiology and end results program, 1973 to 2005: an analysis of 2,600 patients. J Clin Oncol. 2009;27:3391–7. doi: 10.1200/JCO.2008.19.7483. [DOI] [PubMed] [Google Scholar]
- Wolden SL, Alektiar KM. Sarcomas across the age spectrum. Semin Radiat Oncol. 2010;20:45–51. doi: 10.1016/j.semradonc.2009.09.003. [DOI] [PubMed] [Google Scholar]
- Barr FG. Molecular genetics and pathogenesis of rhabdomyosarcoma. J Pediatr Hematol Oncol. 1997;19:483–91. doi: 10.1097/00043426-199711000-00001. [DOI] [PubMed] [Google Scholar]
- Weber-Hall S, Anderson J, McManus A, et al. Gains, losses, and amplification of genomic material in rhabdomyosarcoma analyzed by comparative genomic hybridization. Cancer Res. 1996;56:3220–4. [PubMed] [Google Scholar]
- Bridge JA, Liu J, Weibolt V, et al. Novel genomic imbalances in embryonal rhabdomyosarcoma revealed by comparative genomic hybridization and fluorescence in situ hybridization: an intergroup rhabdomyosarcoma study. Genes Chromosomes Cancer. 2000;27:337–44. doi: 10.1002/(sici)1098-2264(200004)27:4<337::aid-gcc1>3.0.co;2-1. [DOI] [PubMed] [Google Scholar]
- Gordon T, McManus A, Anderson J, et al. Cytogenetic abnormalities in 42 rhabdomyosarcoma: a United Kingdom Cancer Cytogenetics Group Study. Med Pediatr Oncol. 2001;36:259–67. doi: 10.1002/1096-911X(20010201)36:2<259::AID-MPO1063>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
- Koufos A, Hansen MF, Copeland NG, et al. Loss of heterozygosity in three embryonal tumours suggests a common pathogenetic mechanism. Nature. 1985;316:330–4. doi: 10.1038/316330a0. [DOI] [PubMed] [Google Scholar]
- Scrable H, Cavenee W, Ghavimi F, et al. A model for embryonal rhabdomyosarcoma tumorigenesis that involves genome imprinting. Proc Natl Acad Sci USA. 1989;86:7480–4. doi: 10.1073/pnas.86.19.7480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scrable HJ, Witte DP, Lampkin BC, et al. Chromosomal localization of the human rhabdomyosarcoma locus by mitotic recombination mapping. Nature. 1987;329:645–7. doi: 10.1038/329645a0. [DOI] [PubMed] [Google Scholar]
- Loh WE, Scrable HJ, Livanos E, et al. Human chromosome 11 contains two different growth suppressor genes for embryonal rhabdomyosarcoma. Proc Natl Acad Sci USA. 1992;89:1755–9. doi: 10.1073/pnas.89.5.1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anderson J, Gordon A, McManus A, et al. Disruption of imprinted genes at chromosome region 11p15.5 in paediatric rhabdomyosarcoma. Neoplasia. 1999;1:340–8. doi: 10.1038/sj.neo.7900052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Besnard-Guérin C, Newsham I, Winqvist R, et al. A common region of loss of heterozygosity in Wilms' tumour and embryonal rhabdomyosarcoma distal to the D11S988 locus on chromosome 11p15.5. Hum Genet. 1996;97:163–70. doi: 10.1007/BF02265259. [DOI] [PubMed] [Google Scholar]
- Davicioni E, Anderson MJ, Finckenstein FG, et al. Molecular classification of rhabdomyosarcoma–genotypic and phenotypic determinants of diagnosis: a report from the Children's Oncology Group. Am J Pathol. 2009;174:550–64. doi: 10.2353/ajpath.2009.080631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hao Y, Crenshaw T, Moulton T, et al. Tumour-suppressor activity of H19 RNA. Nature. 1993;365:764–7. doi: 10.1038/365764a0. [DOI] [PubMed] [Google Scholar]
- Matsuoka S, Thompson JS, Edwards MC, et al. Imprinting of the gene encoding a human cyclin-dependent kinase inhibitor, p57KIP2, on chromosome 11p15. Proc Natl Acad Sci USA. 1996;93:3026–30. doi: 10.1073/pnas.93.7.3026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roeb W, Boyer A, Cavenee WK, et al. PAX3-FOXO1 controls expression of the p57Kip2 cell-cycle regulator through degradation of EGR1. Proc Natl Acad Sci USA. 2007;104:18085–90. doi: 10.1073/pnas.0708910104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schwienbacher C, Sabbioni S, Campi M, et al. Transcriptional map of 170-kb region at chromosome 11p15.5: identification and mutational analysis of the BWR1A gene reveals the presence of mutations in tumor samples. Proc Natl Acad Sci USA. 1998;95:3873–8. doi: 10.1073/pnas.95.7.3873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhan S, Shapiro DN, Helman LJ. Activation of an imprinted allele of the insulin-like growth factor II gene implicated in rhabdomyosarcoma. J Clin Invest. 1994;94:445–8. doi: 10.1172/JCI117344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hahn H, Wojnowski L, Specht K, et al. Patched target Igf2 is indispensable for the formation of medulloblastoma and rhabdomyosarcoma. J Biol Chem. 2000;275:28341–4. doi: 10.1074/jbc.C000352200. [DOI] [PubMed] [Google Scholar]
- Hahn H, Wojnowski L, Zimmer AM, et al. Rhabdomyosarcomas and radiation hypersensitivity in a mouse model of Gorlin syndrome. Nat Med. 1998;4:619–22. doi: 10.1038/nm0598-619. [DOI] [PubMed] [Google Scholar]
- Stratton MR, Fisher C, Gusterson BA, et al. Detection of point mutations in N-ras and K-ras genes of human embryonal rhabdomyosarcomas using oligonucleotide probes and the polymerase chain reaction. Cancer Res. 1989;49:6324–7. [PubMed] [Google Scholar]
- Wilke W, Maillet M, Robinson R. H-ras-1 point mutations in soft tissue sarcomas. Mod Pathol. 1993;6:129–32. [PubMed] [Google Scholar]
- Yoo J, Robinson RA. H-ras and K-ras mutations in soft tissue sarcoma: comparative studies of sarcomas from Korean and American patients. Cancer. 1999;86:58–63. [PubMed] [Google Scholar]
- Galili N, Davis RJ, Fredericks WJ, et al. Fusion of a fork head domain gene to PAX3 in the solid tumour alveolar rhabdomyosarcoma. Nat Genet. 1993;5:230–5. doi: 10.1038/ng1193-230. [DOI] [PubMed] [Google Scholar]
- Barr FG, Galili N, Holick J, et al. Rearrangement of the PAX3 paired box gene in the paediatric solid tumour alveolar rhabdomyosarcoma. Nat Genet. 1993;3:113–7. doi: 10.1038/ng0293-113. [DOI] [PubMed] [Google Scholar]
- Davis RJ, D'Cruz CM, Lovell MA, et al. Fusion of PAX7 to FKHR by the variant t(1;13)(p36;q14) translocation in alveolar rhabdomyosarcoma. Cancer Res. 1994;54:2869–72. [PubMed] [Google Scholar]
- Gordon AT, Brinkschmidt C, Anderson J, et al. A novel and consistent amplicon at 13q31 associated with alveolar rhabdomyosarcoma. Genes Chromosomes Cancer. 2000;28:220–6. [PubMed] [Google Scholar]
- Berner JM, Forus A, Elkahloun A, et al. Separate amplified regions encompassing CDK4 and MDM2 in human sarcomas. Genes Chromosomes Cancer. 1996;17:254–9. doi: 10.1002/(SICI)1098-2264(199612)17:4<254::AID-GCC7>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
- Forus A, Flørenes VA, Maelandsmo GM, et al. Mapping of amplification units in the q13-14 region of chromosome 12 in human sarcomas: some amplica do not include MDM2. Cell Growth Differ. 1993;4:1065–70. [PubMed] [Google Scholar]
- Dias P, Kumar P, Marsden HB, et al. N-myc gene is amplified in alveolar rhabdomyosarcomas (RMS) but not in embryonal RMS. Int J Cancer. 1990;45:593–6. doi: 10.1002/ijc.2910450403. [DOI] [PubMed] [Google Scholar]
- Driman D, Thorner PS, Greenberg ML, et al. MYCN gene amplification in rhabdomyosarcoma. Cancer. 1994;73:2231–7. doi: 10.1002/1097-0142(19940415)73:8<2231::aid-cncr2820730832>3.0.co;2-e. [DOI] [PubMed] [Google Scholar]
- Hachitanda Y, Toyoshima S, Akazawa K, et al. N-myc gene amplification in rhabdomyosarcoma detected by fluorescence in situ hybridization: its correlation with histologic features. Mod Pathol. 1998;11:1222–7. [PubMed] [Google Scholar]
- Iolascon A, Faienza MF, Coppola B, et al. Analysis of cyclin-dependent kinase inhibitor genes (CDKN2A, CDKN2B, and CDKN2C) in childhood rhabdomyosarcoma. Genes Chromosomes Cancer. 1996;15:217–22. doi: 10.1002/(SICI)1098-2264(199604)15:4<217::AID-GCC3>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
- Graf Finckenstein F, Shahbazian V, Davicioni E, et al. PAX-FKHR function as pangenes by simultaneously inducing and inhibiting myogenesis. Oncogene. 2008;27:2004–14. doi: 10.1038/sj.onc.1210835. [DOI] [PubMed] [Google Scholar]
- Anderson J, Ramsay A, Gould S, et al. PAX3-FKHR induces morphological change and enhances cellular proliferation and invasion in rhabdomyosarcoma. Am J Pathol. 2001;159:1089–96. doi: 10.1016/S0002-9440(10)61784-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Relaix F, Polimeni M, Rocancourt D, et al. The transcriptional activator PAX3-FKHR rescues the defects of Pax3 mutant mice but induces a myogenic gain-of-function phenotype with ligand-independent activation of Met signaling in vivo. Genes Dev. 2003;17:2950–65. doi: 10.1101/gad.281203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goldstein M, Meller I, Issakov J, et al. Novel genes implicated in embryonal, alveolar, and pleomorphic rhabdomyosarcoma: a cytogenetic and molecular analysis of primary tumors. Neoplasia. 2006;8:332–43. doi: 10.1593/neo.05829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gordon A, McManus A, Anderson J, et al. Chromosomal imbalances in pleomorphic rhabdomyosarcomas and identification of the alveolar rhabdomyosarcoma-associated PAX3-FOXO1A fusion gene in one case. Cancer Genet Cytogenet. 2003;140:73–7. doi: 10.1016/s0165-4608(02)00631-3. [DOI] [PubMed] [Google Scholar]
- Malkin D, Li FP, Strong LC, et al. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science. 1990;250:1233–8. doi: 10.1126/science.1978757. [DOI] [PubMed] [Google Scholar]
- Felix CA, Kappel CC, Mitsudomi T, et al. Frequency and diversity of p53 mutations in childhood rhabdomyosarcoma. Cancer Res. 1992;52:2243–7. [PubMed] [Google Scholar]
- Diller L, Sexsmith E, Gottlieb A, et al. Germline p53 mutations are frequently detected in young children with rhabdomyosarcoma. J Clin Invest. 1995;95:1606–11. doi: 10.1172/JCI117834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor AC, Shu L, Danks MK, et al. P53 mutation and MDM2 amplification frequency in pediatric rhabdomyosarcoma tumors and cell lines. Med Pediatr Oncol. 2000;35:96–103. doi: 10.1002/1096-911x(200008)35:2<96::aid-mpo2>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
- Lindor NM, McMaster ML, Lindor CJ, et al. Concise handbook of familial cancer susceptibility syndromes — second edition. J Natl Cancer Inst Monogr. 2008;38:1–93. doi: 10.1093/jncimonographs/lgn001. [DOI] [PubMed] [Google Scholar]
- Keleti J, Quezado MM, Abaza MM, et al. The MDM2 oncoprotein is overexpressed in rhabdomyosarcoma cell lines and stabilizes wild-type p53 protein. Am J Pathol. 1996;149:143–51. [PMC free article] [PubMed] [Google Scholar]
- Cam H, Griesmann H, Beitzinger M, et al. p53 family members in myogenic differentiation and rhabdomyosarcoma development. Cancer Cell. 2006;10:281–93. doi: 10.1016/j.ccr.2006.08.024. [DOI] [PubMed] [Google Scholar]
- Kohashi K, Oda Y, Yamamoto H, et al. Alterations of RB1 gene in embryonal and alveolar rhabdomyosarcoma: special reference to utility of pRB immunoreactivity in differential diagnosis of rhabdomyosarcoma subtype. J Cancer Res Clin Oncol. 2008;134:1097–103. doi: 10.1007/s00432-008-0385-3. [DOI] [PubMed] [Google Scholar]
- Taulli R, Scuoppo C, Bersani F, et al. Validation of met as a therapeutic target in alveolar and embryonal rhabdomyosarcoma. Cancer Res. 2006;66:4742–9. doi: 10.1158/0008-5472.CAN-05-4292. [DOI] [PubMed] [Google Scholar]
- Takayama H, LaRochelle WJ, Sharp R, et al. Diverse tumorigenesis associated with aberrant development in mice overexpressing hepatocyte growth factor/scatter factor. Proc Natl Acad Sci USA. 1997;94:701–6. doi: 10.1073/pnas.94.2.701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferracini R, Olivero M, Di Renzo MF, et al. Retrogenic expression of the MET proto-oncogene correlates with the invasive phenotype of human rhabdomyosarcomas. Oncogene. 1996;12:1697–705. [PubMed] [Google Scholar]
- Ginsberg JP, Davis RJ, Bennicelli JL, et al. Up-regulation of MET but not neural cell adhesion molecule expression by the PAX3-FKHR fusion protein in alveolar rhabdomyosarcoma. Cancer Res. 1998;58:3542–6. [PubMed] [Google Scholar]
- Rees H, Williamson D, Papanastasiou A, et al. The MET receptor tyrosine kinase contributes to invasive tumour growth in rhabdomyosarcomas. Growth Factors. 2006;24:197–208. doi: 10.1080/08977190600759923. [DOI] [PubMed] [Google Scholar]
- Sharp R, Recio JA, Jhappan C, et al. Synergism between INK4a/ARF inactivation and aberrant HGF/SF signaling in rhabdomyosarcomagenesis. Nat Med. 2002;8:1276–80. doi: 10.1038/nm787. [DOI] [PubMed] [Google Scholar]
- Kim SY, Toretsky JA, Scher D, et al. The role of IGF-1R in pediatric malignancies. Oncologist. 2009;14:83–91. doi: 10.1634/theoncologist.2008-0189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ayalon D, Glaser T, Werner H. Transcriptional regulation of IGF-I receptor gene expression by the PAX3-FKHR oncoprotein. Growth Horm IGF Res. 2001;11:289–97. doi: 10.1054/ghir.2001.0244. [DOI] [PubMed] [Google Scholar]
- Blandford MC, Barr FG, Lynch JC, et al. Rhabdomyosarcomas utilize developmental, myogenic growth factors for disease advantage: a report from the Children's Oncology Group. Pediatr Blood Cancer. 2006;46:329–38. doi: 10.1002/pbc.20466. [DOI] [PubMed] [Google Scholar]
- Khan J, Bittner ML, Saal LH, et al. cDNA microarrays detect activation of a myogenic transcription program by the PAX3-FKHR fusion oncogene. Proc Natl Acad Sci USA. 1999;96:13264–9. doi: 10.1073/pnas.96.23.13264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Astolfi A, De Giovanni C, Landuzzi L, et al. Identification of new genes related to the myogenic differentiation arrest of human rhabdomyosarcoma cells. Gene. 2001;274:139–49. doi: 10.1016/s0378-1119(01)00619-9. [DOI] [PubMed] [Google Scholar]
- Ganti R, Skapek SX, Zhang J, et al. Expression and genomic status of EGFR and ErbB-2 in alveolar and embryonal rhabdomyosarcoma. Mod Pathol. 2006;19:1213–20. doi: 10.1038/modpathol.3800636. [DOI] [PubMed] [Google Scholar]
- Taniguchi E, Nishijo K, McCleish AT, et al. PDGFR-A is a therapeutic target in alveolar rhabdomyosarcoma. Oncogene. 2008;27:6550–60. doi: 10.1038/onc.2008.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Onisto M, Slongo ML, Gregnanin L, et al. Expression and activity of vascular endothelial growth factor and metalloproteinases in alveolar and embryonal rhabdomyosarcoma cell lines. Int J Oncol. 2005;27:791–8. [PubMed] [Google Scholar]
- Gee MF, Tsuchida R, Eichler-Jonsson C, et al. Vascular endothelial growth factor acts in an autocrine manner in rhabdomyosarcoma cell lines and can be inhibited with all-trans-retinoic acid. Oncogene. 2005;24:8025–37. doi: 10.1038/sj.onc.1208939. [DOI] [PubMed] [Google Scholar]
- Wang W, Slevin M, Kumar S, et al. The cooperative transforming effects of PAX3-FKHR and IGF-II on mouse myoblasts. Int J Oncol. 2005;27:1087–96. doi: 10.3892/ijo.27.4.1087. [DOI] [PubMed] [Google Scholar]
- Taylor JG, Cheuk AT, Tsang PS, et al. Identification of FGFR4-activating mutations in human rhabdomyosarcomas that promote metastasis in xenotransplanted models. J Clin Invest. 2009;119:3395–407. doi: 10.1172/JCI39703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williamson D, Selfe J, Gordon T, et al. Role for amplification and expression of glypican-5 in rhabdomyosarcoma. Cancer Res. 2007;67:57–65. doi: 10.1158/0008-5472.CAN-06-1650. [DOI] [PubMed] [Google Scholar]
- Astolfi A, Nanni P, Landuzzi L, et al. An anti-apoptotic role for NGF receptors in human rhabdomyosarcoma. Eur J Cancer. 2001;37:1719–25. doi: 10.1016/s0959-8049(01)00190-3. [DOI] [PubMed] [Google Scholar]
- Bouché M, Canipari R, Melchionna R, et al. TGF-beta autocrine loop regulates cell growth and myogenic differentiation in human rhabdomyosarcoma cells. FASEB J. 2000;14:1147–58. doi: 10.1096/fasebj.14.9.1147. [DOI] [PubMed] [Google Scholar]
- Ricaud S, Vernus B, Duclos M, et al. Inhibition of autocrine secretion of myostatin enhances terminal differentiation in human rhabdomyosarcoma cells. Oncogene. 2003;22:8221–32. doi: 10.1038/sj.onc.1207177. [DOI] [PubMed] [Google Scholar]
- Langley B, Thomas M, McFarlane C, et al. Myostatin inhibits rhabdomyosarcoma cell proliferation through an Rb-independent pathway. Oncogene. 2004;23:524–34. doi: 10.1038/sj.onc.1207144. [DOI] [PubMed] [Google Scholar]
- Rossi S, Stoppani E, Puri PL, et al. Differentiation of human rhabdomyosarcoma RD cells is regulated by reciprocal, functional interactions between myostatin, p38 and extracellular regulated kinase signalling pathways. Eur J Cancer. 2011;47:1095–105. doi: 10.1016/j.ejca.2010.12.010. [DOI] [PubMed] [Google Scholar]
- Tomescu O, Xia SJ, Strezlecki D, et al. Inducible short-term and stable long-term cell culture systems reveal that the PAX3-FKHR fusion oncoprotein regulates CXCR4, PAX3, and PAX7 expression. Lab Invest. 2004;84:1060–70. doi: 10.1038/labinvest.3700125. [DOI] [PubMed] [Google Scholar]
- Ebauer M, Wachtel M, Niggli FK, et al. Comparative expression profiling identifies an in vivo target gene signature with TFAP2B as a mediator of the survival function of PAX3/FKHR. Oncogene. 2007;26:7267–81. doi: 10.1038/sj.onc.1210525. [DOI] [PubMed] [Google Scholar]
- Riuzzi F, Sorci G, Donato R. RAGE expression in rhabdomyosarcoma cells results in myogenic differentiation and reduced proliferation, migration, invasiveness, and tumor growth. Am J Pathol. 2007;171:947–61. doi: 10.2353/ajpath.2007.070049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davicioni E, Finckenstein FG, Shahbazian V, et al. Identification of a PAX-FKHR gene expression signature that defines molecular classes and determines the prognosis of alveolar rhabdomyosarcomas. Cancer Res. 2006;66:6936–46. doi: 10.1158/0008-5472.CAN-05-4578. [DOI] [PubMed] [Google Scholar]
- Puri PL, Wu Z, Zhang P, et al. Induction of terminal differentiation by constitutive activation of p38 MAP kinase in human rhabdomyosarcoma cells. Genes Dev. 2000;14:574–84. [PMC free article] [PubMed] [Google Scholar]
- Tapscott SJ, Thayer MJ, Weintraub H. Deficiency in rhabdomyosarcomas of a factor required for MyoD activity and myogenesis. Science. 1993;259:1450–3. doi: 10.1126/science.8383879. [DOI] [PubMed] [Google Scholar]
- Yang Z, MacQuarrie KL, Analau E, et al. MyoD and E-protein heterodimers switch rhabdomyosarcoma cells from an arrested myoblast phase to a differentiated state. Genes Dev. 2009;23:694–707. doi: 10.1101/gad.1765109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cen L, Hsieh FC, Lin HJ, et al. PDK-1/AKT pathway as a novel therapeutic target in rhabdomyosarcoma cells using OSU-03012 compound. Br J Cancer. 2007;97:785–91. doi: 10.1038/sj.bjc.6603952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marampon F, Ciccarelli C, Zani BM. Down-regulation of c-Myc following MEK/ERK inhibition halts the expression of malignant phenotype in rhabdomyosarcoma and in non muscle-derived human tumors. Mol Cancer. 2006;5:31. doi: 10.1186/1476-4598-5-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoovers JM, Kalikin LM, Johnson LA, et al. Multiple genetic loci within 11p15 defined by Beckwith-Wiedemann syndrome rearrangement breakpoints and subchromosomal transferable fragments. Proc Natl Acad Sci USA. 1995;92:12456–60. doi: 10.1073/pnas.92.26.12456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferrari A, Bisogno G, Macaluso A, et al. Soft-tissue sarcomas in children and adolescents with neurofibromatosis type 1. Cancer. 2007;109:1406–12. doi: 10.1002/cncr.22533. [DOI] [PubMed] [Google Scholar]
- Aoki Y, Niihori T, Kawame H, et al. Germline mutations in HRAS proto-oncogene cause Costello syndrome. Nat Genet. 2005;37:1038–40. doi: 10.1038/ng1641. [DOI] [PubMed] [Google Scholar]
- Gorlin RJ. Nevoid basal cell carcinoma (Gorlin) syndrome. Genet Med. 2004;6:530–9. doi: 10.1097/01.gim.0000144188.15902.c4. [DOI] [PubMed] [Google Scholar]
- Kleinerman RA, Tucker MA, Abramson DH, et al. Risk of soft tissue sarcomas by individual subtype in survivors of hereditary retinoblastoma. J Natl Cancer Inst. 2007;99:24–31. doi: 10.1093/jnci/djk002. [DOI] [PubMed] [Google Scholar]
- Hanks S, Coleman K, Summersgill B, et al. Comparative genomic hybridization and BUB1B mutation analyses in childhood cancers associated with mosaic variegated aneuploidy syndrome. Cancer Lett. 2006;239:234–8. doi: 10.1016/j.canlet.2005.08.006. [DOI] [PubMed] [Google Scholar]
- Kratz CP, Holter S, Etzler J, et al. Rhabdomyosarcoma in patients with constitutional mismatch-repair-deficiency syndrome. J Med Genet. 2009;46:418–20. doi: 10.1136/jmg.2008.064212. [DOI] [PubMed] [Google Scholar]
- Miller RW, Rubinstein JH. Tumors in Rubinstein-Taybi syndrome. Am J Med Genet. 1995;56:112–5. doi: 10.1002/ajmg.1320560125. [DOI] [PubMed] [Google Scholar]
- Gilman JP, Herchen H. The effect of physical form of implant on nickel sulphide tumourigenesis in the rat. Acta Unio Int Contra Cancrum. 1963;19:615–9. [PubMed] [Google Scholar]
- Gilman JP. Metal carcinogenesis. II. A study on the carcinogenic activity of cobalt, copper, iron, and nickel compounds. Cancer Res. 1962;22:158–62. [PubMed] [Google Scholar]
- Maenza RM, Pradhan AM, Sunderman FW. Rapid induction of sarcomas in rats by combination of nickel sulfide and 3,4-benzpyrene. Cancer Res. 1971;31:2067–71. [PubMed] [Google Scholar]
- Damjanov I, Sunderman FW, Mitchell JM, et al. Induction of testicular sarcomas in Fischer rats by intratesticular injection of nickel subsulfide. Cancer Res. 1978;38:268–76. [PubMed] [Google Scholar]
- Hildebrand HF, Biserte G. Cylindrical laminated bodies in nickel-subsulphide-induced rhabdomyosarcoma in rabbits. Eur J Cell Biol. 1979;19:276–80. [PubMed] [Google Scholar]
- Nanni P, Azzarello G, Tessarollo L, et al. In vitro differentiation of rhabdomyosarcomas induced by nickel or by Moloney murine sarcoma virus. Br J Cancer. 1991;63:736–42. doi: 10.1038/bjc.1991.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edgar JA, Colegate SM, Boppré M, et al. Pyrrolizidine alkaloids in food: a spectrum of potential health consequences. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 2011;28:308–24. doi: 10.1080/19440049.2010.547520. [DOI] [PubMed] [Google Scholar]
- Fu PP, Xia Q, Lin G, et al. Pyrrolizidine alkaloids–genotoxicity, metabolism enzymes, metabolic activation, and mechanisms. Drug Metab Rev. 2004;36:1–55. doi: 10.1081/dmr-120028426. [DOI] [PubMed] [Google Scholar]
- Allen JR, Hsu IC, Carstens LA. Dehydroretronecine-induced rhabdomyosarcomas in rats. Cancer Res. 1975;35:997–1002. [PubMed] [Google Scholar]
- Toth B, Patil K, Erickson J, et al. Carcinogenesis by benzenediazonium sulfate in mice. In Vivo. 1998;12:379–82. [PubMed] [Google Scholar]
- Toth B, Patil K, Taylor J, et al. Cancer induction in mice by 4-hydroxybenzenediazonium sulfate of the Agaricus xanthodermus mushroom. In Vivo. 1989;3:301–5. [PubMed] [Google Scholar]
- Toth B, Taylor J, Mattson B, et al. Tumor induction by 4-(methyl)benzenediazonium sulfate in mice. In Vivo. 1989;3:17–21. [PubMed] [Google Scholar]
- Bouvier G, Poirier S, Shao YM, et al. Epstein-Barr virus activators, mutagens and volatile nitrosamines in preserved food samples from high-risk areas for nasopharyngeal carcinoma. IARC Sci Publ. 1991;105:204–9. [PubMed] [Google Scholar]
- Bunton TE, Wolfe MJ. N-methyl-N'-nitro-N-nitrosoguanidine-induced neoplasms in medaka (Oryzias latipes) Toxicol Pathol. 1996;24:323–30. doi: 10.1177/019262339602400308. [DOI] [PubMed] [Google Scholar]
- Spitsbergen JM, Tsai HW, Reddy A, et al. Neoplasia in zebrafish (Danio rerio) treated with N-methyl-N'-nitro-N-nitrosoguanidine by three exposure routes at different developmental stages. Toxicol Pathol. 2000;28:716–25. doi: 10.1177/019262330002800512. [DOI] [PubMed] [Google Scholar]
- Lijinsky W, Saavedra JE, Reuber MD. Organ-specific carcinogenesis in rats by methyl- and ethylazoxyalkanes. Cancer Res. 1985;45:76–9. [PubMed] [Google Scholar]
- Sohn OS, Fiala ES, Requeijo SP, et al. Differential effects of CYP2E1 status on the metabolic activation of the colon carcinogens azoxymethane and methylazoxymethanol. Cancer Res. 2001;61:8435–40. [PubMed] [Google Scholar]
- Chen J, Huang XF. The signal pathways in azoxymethane-induced colon cancer and preventive implications. Cancer Biol Ther. 2009;8:1313–7. doi: 10.4161/cbt.8.14.8983. [DOI] [PubMed] [Google Scholar]
- Cattabeni F, Di Luca M. Developmental models of brain dysfunctions induced by targeted cellular ablations with methylazoxymethanol. Physiol Rev. 1997;77:199–215. doi: 10.1152/physrev.1997.77.1.199. [DOI] [PubMed] [Google Scholar]
- Hawkins W, Fournie J, Overstreet R, et al. Rhabdomyosarcoma in the Japanese medaka, oryzias-latipes (temminck and schlegel) and guppy, poecilia-reticulata peters. J Fish Dis. 1988;11:259–66. [Google Scholar]
- Warshawsky D. Polycyclic aromatic hydrocarbons in carcinogenesis. Environ Health Perspect. 1999;107:317–9. doi: 10.1289/ehp.99107317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spitsbergen JM, Tsai HW, Reddy A, et al. Neoplasia in zebrafish (Danio rerio) treated with 7,12-dimethylbenz[a]anthracene by two exposure routes at different developmental stages. Toxicol Pathol. 2000;28:705–15. doi: 10.1177/019262330002800511. [DOI] [PubMed] [Google Scholar]
- Taguchi S, Kuriwaki K, Souda M, et al. Induction of sarcomas by a single subcutaneous injection of 7,12-dimethylbenz[a]anthracene into neonatal male Sprague-Dawley rats: histopathological and immunohistochemical analyses. Toxicol Pathol. 2006;34:336–47. doi: 10.1080/01926230600773966. [DOI] [PubMed] [Google Scholar]
- Westwood FR, Longstaff E, Butler WH. Cellular progression of neoplasia in the subcutis of mice after implantation of 3,4-benzpyrene. Br J Cancer. 1979;39:761–72. doi: 10.1038/bjc.1979.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shimizu Y, Nakatsuru Y, Ichinose M, et al. Benzo[a]pyrene carcinogenicity is lost in mice lacking the aryl hydrocarbon receptor. Proc Natl Acad Sci USA. 2000;97:779–82. doi: 10.1073/pnas.97.2.779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalinich JF, Emond CA, Dalton TK, et al. Embedded weapons-grade tungsten alloy shrapnel rapidly induces metastatic high-grade rhabdomyosarcomas in F344 rats. Environ Health Perspect. 2005;113:729–34. doi: 10.1289/ehp.7791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gupta A, Andrews KL, McDaniel KM, et al. Experimental induction of rhabdomyosarcoma in mice with fractionated doses of beta-irradiation. J Cancer Res Clin Oncol. 1999;125:257–67. doi: 10.1007/s004320050272. [DOI] [PubMed] [Google Scholar]
- Perk K, Shachat DA, Moloney JB. Pathogenesis of a rhabdomyosarcoma (undifferentiated type) in rats induced by a murine sarcoma virus (Moloney) Cancer Res. 1968;28:1197–206. [PubMed] [Google Scholar]
- Teitz T, Chang JC, Kitamura M, et al. Rhabdomyosarcoma arising in transgenic mice harboring the beta-globin locus control region fused with simian virus 40 large T antigen gene. Proc Natl Acad Sci USA. 1993;90:2910–4. doi: 10.1073/pnas.90.7.2910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Köbbert C, Möllmann C, Schäfers M, et al. Transgenic model of cardiac rhabdomyosarcoma formation. J Thorac Cardiovasc Surg. 2008;136:1178–86. doi: 10.1016/j.jtcvs.2008.04.022. [DOI] [PubMed] [Google Scholar]
- Malkin D, Chilton-MacNeill S, Meister LA, et al. Tissue-specific expression of SV40 in tumors associated with the Li-Fraumeni syndrome. Oncogene. 2001;20:4441–9. doi: 10.1038/sj.onc.1204583. [DOI] [PubMed] [Google Scholar]
- Levine AJ, Oren M. The first 30 years of p53: growing ever more complex. Nat Rev Cancer. 2009;9:749–58. doi: 10.1038/nrc2723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donehower LA, Harvey M, Slagle BL, et al. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature. 1992;356:215–21. doi: 10.1038/356215a0. [DOI] [PubMed] [Google Scholar]
- Jacks T, Remington L, Williams BO, et al. Tumor spectrum analysis in p53-mutant mice. Curr Biol. 1994;4:1–7. doi: 10.1016/s0960-9822(00)00002-6. [DOI] [PubMed] [Google Scholar]
- Harvey M, McArthur MJ, Montgomery CA, et al. Spontaneous and carcinogen-induced tumorigenesis in p53-deficient mice. Nat Genet. 1993;5:225–9. doi: 10.1038/ng1193-225. [DOI] [PubMed] [Google Scholar]
- Harvey M, McArthur MJ, Montgomery CA, et al. Genetic background alters the spectrum of tumors that develop in p53-deficient mice. FASEB J. 1993;7:938–43. doi: 10.1096/fasebj.7.10.8344491. [DOI] [PubMed] [Google Scholar]
- Fleischmann A, Jochum W, Eferl R, et al. Rhabdomyosarcoma development in mice lacking Trp53 and Fos: tumor suppression by the Fos protooncogene. Cancer Cell. 2003;4:477–82. doi: 10.1016/s1535-6108(03)00280-0. [DOI] [PubMed] [Google Scholar]
- Jochum W, Passegué E, Wagner EF. AP-1 in mouse development and tumorigenesis. Oncogene. 2001;20:2401–12. doi: 10.1038/sj.onc.1204389. [DOI] [PubMed] [Google Scholar]
- Ricci C, Landuzzi L, Rossi I, et al. Expression of HER/erbB family of receptor tyrosine kinases and induction of differentiation by glial growth factor 2 in human rhabdomyosarcoma cells. Int J Cancer. 2000;87:29–36. [PubMed] [Google Scholar]
- Andrechek ER, Hardy WR, Girgis-Gabardo AA, et al. ErbB2 is required for muscle spindle and myoblast cell survival. Mol Cell Biol. 2002;22:4714–22. doi: 10.1128/MCB.22.13.4714-4722.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nanni P, Nicoletti G, De Giovanni C, et al. Development of rhabdomyosarcoma in HER-2/neu transgenic p53 mutant mice. Cancer Res. 2003;63:2728–32. [PubMed] [Google Scholar]
- Tsumura H, Yoshida T, Saito H, et al. Cooperation of oncogenic K-ras and p53 deficiency in pleomorphic rhabdomyosarcoma development in adult mice. Oncogene. 2006;25:7673–9. doi: 10.1038/sj.onc.1209749. [DOI] [PubMed] [Google Scholar]
- Doyle B, Morton JP, Delaney DW, et al. p53 mutation and loss have different effects on tumourigenesis in a novel mouse model of pleomorphic rhabdomyosarcoma. J Pathol. 2010;222:129–37. doi: 10.1002/path.2748. [DOI] [PubMed] [Google Scholar]
- Post SM, Lozano G. You can win by losing: p53 mutations in rhabdomyosarcomas. J Pathol. 2010;222:124–8. doi: 10.1002/path.2757. [DOI] [PubMed] [Google Scholar]
- Anderson MJ, Shelton GD, Cavenee WK, et al. Embryonic expression of the tumor-associated PAX3-FKHR fusion protein interferes with the developmental functions of Pax3. Proc Natl Acad Sci USA. 2001;98:1589–94. doi: 10.1073/pnas.98.4.1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Finckenstein FG, Davicioni E, Osborn KG, et al. Transgenic mice expressing PAX3-FKHR have multiple defects in muscle development, including ectopic skeletal myogenesis in the developing neural tube. Transgenic Res. 2006;15:595–614. doi: 10.1007/s11248-006-9011-9. [DOI] [PubMed] [Google Scholar]
- Keller C, Arenkiel BR, Coffin CM, et al. Alveolar rhabdomyosarcomas in conditional Pax3:Fkhr mice: cooperativity of Ink4a/ARF and Trp53 loss of function. Genes Dev. 2004;18:2614–26. doi: 10.1101/gad.1244004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chin L, Pomerantz J, DePinho RA. The INK4a/ARF tumor suppressor: one gene–two products–two pathways. Trends Biochem Sci. 1998;23:291–6. doi: 10.1016/s0968-0004(98)01236-5. [DOI] [PubMed] [Google Scholar]
- Sherr CJ. The INK4a/ARF network in tumour suppression. Nat Rev Mol Cell Biol. 2001;2:731–7. doi: 10.1038/35096061. [DOI] [PubMed] [Google Scholar]
- Chardin P, Yeramian P, Madaule P, et al. N-ras gene activation in the RD human rhabdomyosarcoma cell line. Int J Cancer. 1985;35:647–52. doi: 10.1002/ijc.2910350513. [DOI] [PubMed] [Google Scholar]
- Bos JL, Verlaan-de Vries M, Jansen AM, et al. Three different mutations in codon 61 of the human N-ras gene detected by synthetic oligonucleotide hybridization. Nucleic Acids Res. 1984;12:9155–63. doi: 10.1093/nar/12.23.9155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinelli S, McDowell HP, Vigne SD, et al. RAS signaling dysregulation in human embryonal Rhabdomyosarcoma. Genes Chromosomes Cancer. 2009;48:975–82. doi: 10.1002/gcc.20702. [DOI] [PubMed] [Google Scholar]
- Chen Y, Takita J, Hiwatari M, et al. Mutations of the PTPN11 and RAS genes in rhabdomyosarcoma and pediatric hematological malignancies. Genes Chromosomes Cancer. 2006;45:583–91. doi: 10.1002/gcc.20322. [DOI] [PubMed] [Google Scholar]
- Kratz CP, Rapisuwon S, Reed H, et al. Cancer in Noonan, Costello, cardiofaciocutaneous and LEOPARD syndromes. Am J Med Genet C Semin Med Genet. 2011;157:83–9. doi: 10.1002/ajmg.c.30300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kratz CP, Steinemann D, Niemeyer CM, et al. Uniparental disomy at chromosome 11p15.5 followed by HRAS mutations in embryonal rhabdomyosarcoma: lessons from Costello syndrome. Hum Mol Genet. 2007;16:374–9. doi: 10.1093/hmg/ddl458. [DOI] [PubMed] [Google Scholar]
- Langenau DM, Keefe MD, Storer NY, et al. Effects of RAS on the genesis of embryonal rhabdomyosarcoma. Genes Dev. 2007;21:1382–95. doi: 10.1101/gad.1545007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeffers M, Rong S, Woude GF. Hepatocyte growth factor/scatter factor-Met signaling in tumorigenicity and invasion/metastasis. J Mol Med (Berl) 1996;74:505–13. doi: 10.1007/BF00204976. [DOI] [PubMed] [Google Scholar]
- Matsumoto K, Nakamura T. Emerging multipotent aspects of hepatocyte growth factor. J Biochem. 1996;119:591–600. doi: 10.1093/oxfordjournals.jbchem.a021283. [DOI] [PubMed] [Google Scholar]
- Trusolino L, Comoglio PM. Scatter-factor and semaphorin receptors: cell signalling for invasive growth. Nat Rev Cancer. 2002;2:289–300. doi: 10.1038/nrc779. [DOI] [PubMed] [Google Scholar]
- Danilkovitch-Miagkova A, Zbar B. Dysregulation of Met receptor tyrosine kinase activity in invasive tumors. J Clin Invest. 2002;109:863–7. doi: 10.1172/JCI15418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sakata H, Takayama H, Sharp R, et al. Hepatocyte growth factor/scatter factor overexpression induces growth, abnormal development, and tumor formation in transgenic mouse livers. Cell Growth Differ. 1996;7:1513–23. [PubMed] [Google Scholar]
- Otsuka T, Takayama H, Sharp R, et al. c-Met autocrine activation induces development of malignant melanoma and acquisition of the metastatic phenotype. Cancer Res. 1998;58:5157–67. [PubMed] [Google Scholar]
- Serrano M, Lee H, Chin L, et al. Role of the INK4a locus in tumor suppression and cell mortality. Cell. 1996;85:27–37. doi: 10.1016/s0092-8674(00)81079-x. [DOI] [PubMed] [Google Scholar]
- Zacksenhaus E, Jiang Z, Chung D, et al. pRb controls proliferation, differentiation, and death of skeletal muscle cells and other lineages during embryogenesis. Genes Dev. 1996;10:3051–64. doi: 10.1101/gad.10.23.3051. [DOI] [PubMed] [Google Scholar]
- Pomerantz J, Schreiber-Agus N, Liégeois NJ, et al. The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2's inhibition of p53. Cell. 1998;92:713–23. doi: 10.1016/s0092-8674(00)81400-2. [DOI] [PubMed] [Google Scholar]
- Epstein JA, Shapiro DN, Cheng J, et al. Pax3 modulates expression of the c-Met receptor during limb muscle development. Proc Natl Acad Sci USA. 1996;93:4213–8. doi: 10.1073/pnas.93.9.4213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tatsumi R, Anderson JE, Nevoret CJ, et al. HGF/SF is present in normal adult skeletal muscle and is capable of activating satellite cells. Dev Biol. 1998;194:114–28. doi: 10.1006/dbio.1997.8803. [DOI] [PubMed] [Google Scholar]
- Lum L, Beachy PA. The Hedgehog response network: sensors, switches, and routers. Science. 2004;304:1755–9. doi: 10.1126/science.1098020. [DOI] [PubMed] [Google Scholar]
- Ingham PW, McMahon AP. Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 2001;15:3059–87. doi: 10.1101/gad.938601. [DOI] [PubMed] [Google Scholar]
- Pazzaglia S. Ptc1 heterozygous knockout mice as a model of multi-organ tumorigenesis. Cancer Lett. 2006;234:124–34. doi: 10.1016/j.canlet.2005.03.047. [DOI] [PubMed] [Google Scholar]
- Kappler R, Bauer R, Calzada-Wack J, et al. Profiling the molecular difference between patched- and p53-dependent rhabdomyosarcoma. Oncogene. 2004;23:8785–95. doi: 10.1038/sj.onc.1208133. [DOI] [PubMed] [Google Scholar]
- Zibat A, Uhmann A, Nitzki F, et al. Time-point and dosage of gene inactivation determine the tumor spectrum in conditional Ptch knockouts. Carcinogenesis. 2009;30:918–26. doi: 10.1093/carcin/bgp068. [DOI] [PubMed] [Google Scholar]
- Rubin BP, Nishijo K, Chen HI, et al. Evidence for an unanticipated relationship between undifferentiated pleomorphic sarcoma and embryonal rhabdomyosarcoma. Cancer Cell. 2011;19:177–91. doi: 10.1016/j.ccr.2010.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee Y, Kawagoe R, Sasai K, et al. Loss of suppressor-of-fused function promotes tumorigenesis. Oncogene. 2007;26:6442–7. doi: 10.1038/sj.onc.1210467. [DOI] [PubMed] [Google Scholar]
- Svärd J, Heby-Henricson K, Henricson KH, et al. Genetic elimination of Suppressor of fused reveals an essential repressor function in the mammalian Hedgehog signaling pathway. Dev Cell. 2006;10:187–97. doi: 10.1016/j.devcel.2005.12.013. [DOI] [PubMed] [Google Scholar]
- Taylor MD, Liu L, Raffel C, et al. Mutations in SUFU predispose to medulloblastoma. Nat Genet. 2002;31:306–10. doi: 10.1038/ng916. [DOI] [PubMed] [Google Scholar]
- Partridge T. Animal models of muscular dystrophy–what can they teach us? Neuropathol Appl Neurobiol. 1991;17:353–63. doi: 10.1111/j.1365-2990.1991.tb00735.x. [DOI] [PubMed] [Google Scholar]
- Bulfield G, Siller WG, Wight PA, et al. X chromosome-linked muscular dystrophy (mdx) in the mouse. Proc Natl Acad Sci USA. 1984;81:1189–92. doi: 10.1073/pnas.81.4.1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sicinski P, Geng Y, Ryder-Cook AS, et al. The molecular basis of muscular dystrophy in the mdx mouse: a point mutation. Science. 1989;244:1578–80. doi: 10.1126/science.2662404. [DOI] [PubMed] [Google Scholar]
- Hoffman EP, Morgan JE, Watkins SC, et al. Somatic reversion/suppression of the mouse mdx phenotype in vivo. J Neurol Sci. 1990;99:9–25. doi: 10.1016/0022-510x(90)90195-s. [DOI] [PubMed] [Google Scholar]
- Hoffman EP, Brown RH, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell. 1987;51:919–28. doi: 10.1016/0092-8674(87)90579-4. [DOI] [PubMed] [Google Scholar]
- Ervasti JM, Campbell KP. A role for the dystrophin-glycoprotein complex as a transmembrane linker between laminin and actin. J Cell Biol. 1993;122:809–23. doi: 10.1083/jcb.122.4.809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chamberlain JS, Metzger J, Reyes M, et al. Dystrophin-deficient mdx mice display a reduced life span and are susceptible to spontaneous rhabdomyosarcoma. FASEB J. 2007;21:2195–204. doi: 10.1096/fj.06-7353com. [DOI] [PubMed] [Google Scholar]
- Fernandez K, Serinagaoglu Y, Hammond S, et al. Mice lacking dystrophin or alpha sarcoglycan spontaneously develop embryonal rhabdomyosarcoma with cancer-associated p53 mutations and alternatively spliced or mutant Mdm2 transcripts. Am J Pathol. 2010;176:416–34. doi: 10.2353/ajpath.2010.090405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allamand V, Campbell KP. Animal models for muscular dystrophy: valuable tools for the development of therapies. Hum Mol Genet. 2000;9:2459–67. doi: 10.1093/hmg/9.16.2459. [DOI] [PubMed] [Google Scholar]
- Duclos F, Straub V, Moore SA, et al. Progressive muscular dystrophy in alpha-sarcoglycan-deficient mice. J Cell Biol. 1998;142:1461–71. doi: 10.1083/jcb.142.6.1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weller B, Karpati G, Carpenter S. Dystrophin-deficient mdx muscle fibers are preferentially vulnerable to necrosis induced by experimental lengthening contractions. J Neurol Sci. 1990;100:9–13. doi: 10.1016/0022-510x(90)90005-8. [DOI] [PubMed] [Google Scholar]
- Petrof BJ, Shrager JB, Stedman HH, et al. Dystrophin protects the sarcolemma from stresses developed during muscle contraction. Proc Natl Acad Sci USA. 1993;90:3710–4. doi: 10.1073/pnas.90.8.3710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Camboni M, Hammond S, Martin LT, et al. Induction of a regenerative microenvironment in skeletal muscle is sufficient to induce embryonal rhabdomyosarcoma in p53-deficient mice. J Pathol. 2011;226:40–9. doi: 10.1002/path.2996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sher RB, Cox GA, Mills KD, et al. Rhabdomyosarcomas in aging a/j mice. PLoS ONE. 2011;6:e23498. doi: 10.1371/journal.pone.0023498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ho M, Post CM, Donahue LR, et al. Disruption of muscle membrane and phenotype divergence in two novel mouse models of dysferlin deficiency. Hum Mol Genet. 2004;13:1999–2010. doi: 10.1093/hmg/ddh212. [DOI] [PubMed] [Google Scholar]
- Bittner RE, Anderson LV, Burkhardt E, et al. Dysferlin deletion in SJL mice (SJL-Dysf) defines a natural model for limb girdle muscular dystrophy 2B. Nat Genet. 1999;23:141–2. doi: 10.1038/13770. [DOI] [PubMed] [Google Scholar]