Oncogene (2014), 1–9 © 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc

REVIEW

PAX genes in childhood oncogenesis: developmental biology gone awry? P Mahajan1, PJ Leavey1 and RL Galindo1,2,3 Childhood solid tumors often arise from embryonal-like cells, which are distinct from the epithelial cancers observed in adults, and etiologically can be considered as ‘developmental patterning gone awry’. Paired-box (PAX) genes encode a family of evolutionarily conserved transcription factors that are important regulators of cell lineage specification, migration and tissue patterning. PAX loss-of-function mutations are well known to cause potent developmental phenotypes in animal models and underlie genetic disease in humans, whereas dysregulation and/or genetic modification of PAX genes have been shown to function as critical triggers for human tumorigenesis. Consequently, exploring PAX-related pathobiology generates insights into both normal developmental biology and key molecular mechanisms that underlie pediatric cancer, which are the topics of this review. Oncogene advance online publication, 21 July 2014; doi:10.1038/onc.2014.209

INTRODUCTION The developmental mechanisms necessary to generate a fully patterned, complex organism from a nascent embryo are precise. Undifferentiated primordia undergo a vast array of cell lineage specification, migration and patterning, and differentiate into an ensemble of interdependent connective, muscle, nervous and epithelial tissues. Dysregulation of these precise developmental programs cause various diseases/disorders, including—and relevant to this review—childhood cancer. Interestingly, the pathobiology of childhood cancer is different from adult neoplasia. Solid tumors in adults are most often epithelial in origin (for example, prostate and breast carcinoma), whereas pediatric solid tumors are typically comprised of histologically primitive blastemal/embryonal-type cells (for example, neuroblastoma, nephroblastoma, medulloblastoma and rhabdomyosarcoma (RMS)). Developmental dysregulation of precursor cell maturation and terminal differentiation, or ‘development gone awry,’ represents a seminal feature of childhood cancer. In this review, we discuss in particular the misregulation of Paired-Box (PAX) gene-mediated developmental programs as important underpinnings of childhood cancer. The evolutionarily conserved PAX transcription factors are master regulators of histogenesis and organogenesis. Spontaneous and targeted murine Pax gene mutations disrupt many aspects of tissue/organ patterning, as well as interfere with the maintenance of adult stem cells necessary for tissue-specific repair/regeneration. Here we profile how PAX genes normally direct precursor cell differentiation, whereas dysregulation and/or misexpression of PAX orthologs serve as critical participants in a broad spectrum of childhood malignancies (Table 1). Of note, we refer interested readers to Li et al.,1 Lang et al.2 and Robson et al.,3 for more detailed reviews of PAX

developmental mechanisms and PAX genes in medical (adult) oncology.

STRUCTURAL MOTIFS DEFINE THE PAX FAMILY SUBGROUPS The mammalian PAX family of transcription factors is comprised of nine members that function as ‘master regulators’ of organogenesis4 (Figure 1). The structural motif that defines this unique family of molecules is the evolutionarily conserved paired domain (PD). The PD, 128 amino acids in length, is a DNA-binding motif that recognizes highly related DNA sequences (TCACGC/G; minor variability can be seen for each PAX ortholog).5–7 Seven of the nine PAX molecules, PAX2 through -8, additionally possess a second DNA-binding domain, a homeodomain (HD) motif (consensus binding sequence TAAT–ATTA),7,8 which adds functional complexity to these PAX orthologs, as the PD and HD can operate cooperatively (consensus sequence AATTA–GTCACGC)7,9 or independently of one another. Unlike the PD, the HD varies structurally among the HD-containing PAX proteins, and is used to further organize the PAX orthologs into subfamilies (described in more detail below). Finally, except for PAX4 and -6, PAX molecules contain a conserved octapeptide motif (OP) positioned between the PD and HD. This eight-amino-acid element has been shown to function as a transcriptional inhibitory motif evolutionarily related to the Drosophila-engrailed family eh1 repression domain and Goosecoid protein, which behave as transcriptional repressors.10 Thus, PAX molecules, depending on context and cofactors, can function either as transcriptional activators or repressors, underscoring even more so their experimentally problematic functional complexity. Below, we discuss in more detail aspects of PAX-mediated development and pathobiology that pertain specifically to PAXrelated pediatric malignancies, organized into sections corresponding to the four PAX family subgroups.

1 Department of Pediatrics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA; 2Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA and 3Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA. Correspondence: Dr Galindo, Department of Pathology, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, TX 75390-9072, USA. E-mail: [email protected] Received 3 April 2014; revised 10 June 2014; accepted 11 June 2014

PAX genes in childhood cancers P Mahajan et al

2 Table 1. PAX genes

PAX genes in normal development and childhood cancers Subgroup Normal development

PAX3 PAX7

III

PAX2 PAX5 PAX8

II

PAX4 PAX6 PAX1 PAX9

IV I

Pediatric cancer

Skeletal muscle Rhabdomyosarcoma development and regeneration Kidney Wilms tumor Renal cell carcinoma Thyroid Thyroid carcinoma B cell B-cell acute lymphoblastic leukemia Acute myeloblastic leukemia B-cell lymphomas T-lymphoblastic leukemia Medulloblastoma Neuroblastoma Pancreas Retinoblastoma? Eye Musculoskeletal Pediatric bone-related malignancies?

PAX-MEDIATED DEVELOPMENT, PATHOBIOLOGY AND ONCOGENICITY PAX3 and PAX7 are canonical proto-oncogenes PAX3 and PAX7, members of subgroup III, are structurally the most complex PAX orthologs, being the only two family members containing a PD, a three-helix-length HD and an OP motif. In addition to being master regulators of neural tube, neural crest and somatic muscle development, PAX3 and -7 are also the clearest examples of how PAX can function as proto-oncogenes and drive human tumorigenesis, as clearly illustrated in the context of the skeletal muscle-lineage soft tissue sarcoma, rhabdomyosarcoma (RMS). PAX3 and PAX7 in skeletal muscle development and regeneration. Mutation of the Pax3 locus was identified as the molecular basis for the spontaneous-occurring semidominant lethal murine splotch mutation.11 Splotch homozygous mice were initially characterized for phenotypes related to neural tube- and neural crest-derived tissues. Further analysis showed that the splotch mouse also served as a model for skeletal muscle patterning (myogenesis). Developmental studies uncovered that Pax3 is expressed in the somites and mesenchymal limb buds,12 and that splotch mice, although exhibiting a mild reduction in body wall muscle, showed a striking failure to develop normal limb musculature.13 The observation that Pax3 mutation blocks the onset of myogenesis in a subset of somitic cells otherwise fated to become myoblasts (no expressions of the myogenic transcription factors Myogenin and MyoD, for example, were detected in these precursors) clearly pointed to Pax3 as a critical upstream myogenesis determinant.14 The importance of PAX3 in development, including myogenesis, is also seen in humans: mutations in the PAX3 gene have been identified in patients with Waardenburg Syndrome, a rare autosomal dominant disorder characterized by deafness, pigmentation anomalies and disruptions in myogenesis.15 Similar to its ‘sibling’ Pax3, Pax7 is also expressed in embryonic muscle progenitor cells, most notably those arising in the central dermomyotome.16 Pax7 homozygous mutant embryos, however, demonstrate no obvious muscle patterning defects, as mice with targeted null mutations for Pax7 express myogenesis markers (for example, MyoD, Myf5) in a normal spatiotemporal pattern.17 Pax7 loss-of-function muscle phenotypes, instead, are revealed postnatally, as Pax7 mutant mice die within few weeks after birth from failure to thrive due to severe muscular atrophy.17 Further probing of these animals revealed an absence of muscle-specific Oncogene (2014), 1 – 9

Figure 1. The mammalian family of PAX transcription factors. The PAX family of proteins is comprised of nine members, which are further classified into four distinct subgroups I–IV based on structural composition. All nine PAX proteins have a highly conserved PD. Group I includes PAX1 and 9; group II consists of PAX2, 5 and 8; group III includes PAX3 and 7; and PAX4 and 6 belong to group IV. PAX2, 3, 4, 5, 6, 7 and 8 contain a HD and all of the PAX proteins, with the exception of PAX4 and 6, contain an OP. TAD, transcription activation domain.

monopotential stem cells, termed satellite cells, which are critically required for skeletal muscle physiologic growth, maintenance and regeneration.17,18 Realix et al.19 further demonstrated that the loss of satellite cells is progressive in Pax7 mutants, pointing toward an antiapoptic role of Pax7 for which Pax3 is not compensatory. In addition, Pax7 deletion in primary murine myoblasts and satellite cells resulted in cell cycle arrest and early differentiation,20 highlighting that Pax7 is necessary for maintaining satellite cells in a stem cell-like state for ongoing muscle renewal. Of note, although numerous findings support the notion that Pax7 is indispensible for the proper establishment and function of perinatal satellite cells, Cre/loxP lineage tracing and tamoxifeninducible conditional gene inactivation interestingly reveal that loss of Pax7 in adult satellite cells does not compromise muscle regeneration. In these studies, Lepper et al.21 showed that adult satellite cells lacking Pax7 (as well as cells lacking both Pax7 and Pax3) proliferate and properly reoccupy the sublaminal niche during muscle repair from injury, and report that Pax7 is only required in satellite cells up to the juvenile period, at which point these progenitors make a transition into quiescence. The authors of these studies appropriately remind us that stem cell biology is not static, but is instead dynamic throughout an organism’s life cycle, and that developmental windows exist that are important to consider. © 2014 Macmillan Publishers Limited

PAX genes in childhood cancers P Mahajan et al

3 Translocation involving PAX3 and PAX7 drives RMS. Turning toward pediatric oncology, RMS is the most common pediatric soft tissue tumor, with an incidence rate of 4.5 per million US children.22,23 The estimated 5-year failure-free survival rate for lowrisk RMS patients is 90%.24 However, despite intensive therapies, the 3-year event-free survival for children with high-risk RMS is only 20%, with no significant improvement in the past 25 years.25 RMS is a malignancy comprised of skeletal muscle precursors that fail to exit the cell cycle and are blocked from differentiating into syncytial muscle. RMS is typically divided into two distinct histologic subgroups, each with differing clinical features: embryonal RMS, which is more common, and alveolar RMS (A-RMS), which is notoriously more aggressive. RMS cells are known to misexpress/overexpress PAX3 and/or PAX7, thus suggesting a potential pathogenetic role for PAX3/PAX7 in RMS.26–28 A critical breakthrough regarding PAX3’s potent myogenic influence on RMS occurred when a recurrent chromosomal translocation uniquely associating with A-RMS, t(2;13)(q35;q14), was molecularly characterized and found to fuse PAX3 on chromosome 2 with the FOXO1 forkhead transcription factor locus on chromosome 13 (Figure 2). The newly encoded PAX3– FOXO1 chimera, which fuses the PAX3 DNA-binding domains with the stronger FOXO1 transcriptional activation domain, is misexpressed from the endogenous PAX3 enhancer/promoter, which dysregulates myogenesis in vivo and drives RMS tumorigenesis. A similar finding occurred not long after, when a second (though less common) recurrent translocation, t(1;13)(p36;q14), was characterized and found to encode an equivalent PAX7–FOXO1 oncogenic fusion.29 Thus, A-RMS is a genetically distinct disease driven by the PAX–FOXO1 fusion oncoprotein, which is both unique to and diagnostic for ARMS.30 Clinically, expression of the PAX–FOXO1 fusion is now widely used as a diagnostic marker (identified by PCR/FISH technology), which harbors prognostic significance.30 PAX3–FOXO1, the more frequent chimera encountered, is associated with a worse clinical outcome among patients with metastatic disease,30 as PAX3– FOXO1-positive patients with metastatic disease have a significant increased risk for treatment failure. Fusion gene status has also been shown to be associated with distinct clinical phenotypes, where PAX7–FOXO1 tumors are more often localized and typically present in younger patients within the extremities.31 Of note, tumors that are classified as A-RMS based on histologic morphology but are otherwise PAX–FOXO1 fusion negative (~20–30% of all A-RMS cases) have been shown to behave similarly to E-RMS with regard to clinical outcome.32 As the PAX–FOXO1 fusions contain the native PAX3/7 DNAbinding domains, it is postulated that myogenic genes normally regulated by PAX3 and PAX7 underlie PAX–FOXO1 A-RMS pathogenesis. PAX3 has been shown to exert ongogenic potential by modulating the c-MET receptor in RMS cell lines with a t(2;13) translocation. In addition, genome-wide analysis of PAX3–FOXO1 RMS points toward ALK, FGFR4, MYCN and IGFIR as direct targets of the fusion gene.33 Interestingly, high levels of PAX7 expression are noted in RMS tumors lacking a PAX–FOXO1 gene fusion, suggesting that overexpression of PAX7 may facilitate fusionnegative RMS.26,27 Of note, molecular genetic studies have identified additional rare, novel fusion proteins in A-RMS, such as fusion of PAX3 to the nuclear receptor coactivator NCOA1, which appears to demonstrate similar transactivation properties as PAX3–FOXO1.34 Shern et al.35 have also reported a new PAX3NCOA1 fusion in RMS, as well as a PAX3 fusion involving the C terminus of INO80D. The influence that these rare fusions exert on the clinical behavior of these RMS variants at present remains unclear. As mentioned above, alterations of PAX genes in RMS presumably induce aberrant myogenesis, tumor growth and resistance to apoptosis. The observation that PAX3–FOXO1, in © 2014 Macmillan Publishers Limited

Figure 2. PAX–FOXO1 fusion gene is both unique and diagnostic for ARMS. The PAX–FOXO1 translocation gene involves FOXO1 on chromosome 13 and PAX3 or PAX7 on chromosomes 2 and 1, respectively. This translocation retains the wild-type PAX DNAbinding site with the transactivation domain from the FOXO1 gene fused to the 3′ end. FD, forkhead domain.

comparison with wild-type PAX3, potently inhibits the differentiation of the C2C12 murine myoblasts’ cell line6 suggests that dysregulated PAX3/7 activity contributes to the A-RMS phenotype by interfering with terminal differentiation in the myogenic pathway, as well as the discrete process of myoblast cell–cell fusion.36,37 It is important to note, however, that even though PAX–FOXO1 expression is viewed as the driver mutation in precursor cells that newly acquire a PAX–FOXO1-encoding translocation, PAX–FOXO1 alone is apparently a weak driver of RMS tumorigenesis. For example, expression of PAX3–FOXO1 or PAX7–FOXO1 in cultured mesenchymal cells generates A-RMS-like tumors in mice only in the presence of additional oncogenesispromoting mutations, such as p53, RB or Ras pathway disruptions.38,39 In addition, mutations in p53 or INK/ARF are also known to significantly increase A-RMS tumorigenesis in vivo in genetically engineered PAX3–FOXO1 mouse models.40 Interestingly, genomic analysis of a comprehensive collection of human RMS tumors,41 however, showed that—in comparison with the E-RMS, for example—A-RMS has far fewer genetic alterations overall, with no lesions isolated in well-known genes (for example, p53) recurrently mutated in cancer. Thus, the influence that cooperative genetic mutations might impart on PAX–FOXO1mediated tumorigenesis in humans remains an intriguing, openended question. In summary, together these studies clearly identify PAX dysregulation and overexpression as RMS underpinnings, and that PAX3 and -7 function as canonical proto-oncogenes. PAX2, PAX5 and PAX8 in development and childhood cancer Similar to PAX3/7, PAX 2, PAX5 and PAX8, which belong to subgroup II, contain a PD, HD and OP motif, but are distinguished structurally by a truncated, single-helix HD. The developmental functions of PAX2, -5 and -8 are diverse and impinge on renal, thyroid, eye and lymphocyte development.42–46 Here we will focus on three aspects of PAX2/5/8 in developmental programming and childhood cancer: (1) PAX2 and -8 in renal patterning and neoplasia; (2) PAX8 in thyroid development and follicular carcinoma; and (3) PAX5 in B-cell lineage specification and leukemia/lymphoma. PAX2 and PAX8 in kidney development. PAX2 is critical in excretory system development—in particular, the urogenital tract. Pax2 is normally expressed throughout the developing branching uteric bud and Wolffian/Müllerian ducts, which persists into the nephrogenic mesenchyme and the early epithelia of patterning Oncogene (2014), 1 – 9

PAX genes in childhood cancers P Mahajan et al

4 nephrons, with Pax2 expression downregulated as nephrogenesis concludes.43,47–49 Mutant mice further confirm the importance of Pax2 for proper nephrogenesis, as Pax2 heterozygotes demonstrate mild-to-moderate renal hypoplasia, whereas Pax2 homozygotes show renal agenesis, as well as the absence of additional genitourinary structures.48 Similar to these murine phenotypes, PAX2 heterozygous mutations in humans cause renal-coloboma syndrome, so named due to the presence of renal hypoplasia and optic nerve colobomas.46 Even though Pax8 is expressed in the developing renal vesicle and persists in conjunction with Pax2 during kidney development, Pax8 mutation itself does not disrupt nephrogenesis, suggesting a shared or compensatory role between Pax2 and -8.42,44,50,51 Compound Pax2/Pax8 mutant mouse models indeed demonstrate a cooperative Pax2/Pax8 relationship: mouse embryos heterozygous for Pax2 and Pax8 (Pax2+/ − /Pax8+/ − ) show enhanced growth impairment when compared with Pax2+/ − mice, with severe reduction in kidney size (~25–50% of normal size).51 These results illustrate the critical role of both PAX2 and PAX8 in renal cell specification, morphogenesis, growth and cell survival. Regulatory roles of PAX2 and PAX8 in cell survival and apoptosis. Consistent with the roles PAX2 and PAX8 provide in regulating the survival and propagation of nephrogenic-lineage precursors, PAX2/8 influence survival and proliferation in renal neoplasia. In renal cell carcinoma (RCC) cell lines, inhibition of PAX2 expression by RNA silencing results in growth inhibition.52 Similarly, silencing of PAX2 expression through RNA interference (RNAi) in additional genitourinary cancer cell lines (bladder, ovarian) induces apoptosis, despite the presence of cooperating p53 and/or HRAS mutations.53 In addition, PAX2 facilitates angiogenesis, tumor growth and proliferation, whereas downregulation of PAX2 expression in renal tumor-derived endothelial cells results in an increase in the PTEN tumor suppressor gene.54 In these same studies, normal human endothelial cells—when transfected with PAX2—induced a proangiogenic phenotype, further illustrating the pro-growth potential of PAX2. In invertebrate genetic models, two PAX2/5/8-related genes, egl-38 and pax-2, promote cell survival in Caenorhabditis elegans, thus showing evolutionary conservation of PAX2/5/8 anti-apoptotic activity.55 Finally, PAX8 has also been shown to promote tumor cell growth and cell cycling by transcriptionally regulating E2F1 and stabilizing the retinoblastoma protein (RB) protein.56 Aberrant PAX2 and PAX8 expressions associate with a variety of renal tumors, including Wilms tumor. PAX2 and PAX8 are implicated in both Wilms tumor and RCC pathobiology. Wilms tumor is the most common form of renal cancer in patients o 15 years of age, accounting for 95% of the childhood renal cancer diagnoses in this age group.57 The incidence rate is 8 cases per million children in the United States less than age 15 and ~ 500 new cases are diagnosed each year in the United States. It is most commonly diagnosed in children younger than 5 years of age and the 5-year survival rate is ~ 90%.58 In contrast, RCC rarely occurs in children, and occurrence is most frequently in the second decade of life, with the highest incidence among 15–19-year olds.59 Although Pax2 expression is necessary for nephrogenic precursor cell specification and survival, Pax2 must be downregulated for nephrogenesis to complete properly, as persistent transgenic expression of Pax2 results in nephric phenotypes similar to human nephrotic syndromes.60 Given that Wilms tumor (also referred to as nephroblastoma) is a blastemal neoplasm tumor, it is not surprizing that Pax2 expression persists in the epithelial blastema of these tumors. Further probing of Pax2 regulation in nephrogenesis revealed that downregulation of Pax2 expression coincided with the known tumor suppressor gene Wilms Tumor protein 1 (WT1), and that Pax2 expression is directly regulated by WT1,61 whereas additional studies have shown that Oncogene (2014), 1 – 9

in Wilms tumor sections where PAX2 is expressed, WT1 expression is absent.61 Interestingly, PAX2 and WT1 were subsequently shown to interact genetically and molecularly, as mice with heterozygous mutations in both genes harbor kidneys that are 50% smaller than wild type, whereas WT1 and PAX2 can function in a protein complex in vitro and in vivo.62 Thus, PAX2 and WT1 are critical components of nephrogenic pathobiology. PAX2 is also a specific and sensitive marker for tumors of renal origin, including RCC.63 Immunostaining in primary RCC tumors detects PAX2 expression in the malignant cells but not in the surrounding tissue.52 Furthermore, treatment of RCC cell lines with RNAi results in significant growth inhibition, supporting the role for PAX2 in proliferation.52 Thus, PAX2 function is postulated to facilitate RCC initiation and tumor maintenance. Similar to PAX2, PAX8 expression is observed in renal neoplasms.50,64 Analysis of normal human kidney and Wilms tumor showed a strong expression of PAX8 in Wilms tumors in comparison with the adult kidney.50 A study comparing PAX2 and PAX8 expressions in both primary renal tumors and metastatic tumors demonstrates that PAX2 and PAX8 may be useful diagnostic markers for both primary and metastatic tumors of the kidney.64 Regardless of histologic subtype, PAX8 immunostaining is observed in more cells with stronger intensity, and PAX8 expression is more frequent within metastatic RCC than PAX2. Therefore, PAX8 may be a more sensitive marker than PAX2 for both primary and metastatic tumors of renal histogenesis. These studies together suggest a role for PAX proteins as diagnostic and potentially important prognostic markers. PAX8 expressed in the thyroid and a role in cancer cell growth. In addition to the developing kidney, Pax8/PAX8 is also expressed in the developing and adult thyroid.44,50 Analysis of Pax8−/− mice illustrates the requirement of Pax8 for proper formation of the follicular cells of the thyroid gland:42 mice embryos with inactivated Pax8 revealed smaller thyroid glands with no detectable follicles. Interestingly, no other defects were observed in other Pax8-involved structures, likely given the redundancy of other Pax genes. These Pax8−/− mice demonstrated growth retardation and eventually demised, thus supporting a role for Pax8 in thyroid development and differentiation. PAX8 expression is observed in normal thyroids and differentiated thyroid cancers more frequently than undifferentiated thyroid cancers, demonstrating a potential role for PAX8 in differentiation.65 Furthermore, nuclear and cytoplasmic localization of PAX8 is seen in pediatric thyroid tissues, and cytoplasmic PAX8 is more frequently noted in thyroid cancers and is associated with more aggressive disease and increased risk of recurrence.66 Cytoplasmic PAX8 may lead to decreased transcription of thyroidspecific genes and therefore dysregulation of proper PAX8 function; however, the complex molecular mechanisms that underlie thyroid cancers are not clearly established and further work with larger sample sizes is needed to distinguish clinical significance. Thyroid carcinoma is overall the most common endocrine malignancy; however in pediatrics, thyroid cancer is rare with the highest incidence occurring in the 15–19 age group among children and adolescents.67 Of the pediatric thyroid carcinomas, papillary thyroid carcinoma is the most prevalent, representing 80% of differentiated thyroid carcinomas, whereas follicular thyroid carcinoma is uncommon.68,69 In children and adolescents, tumor recurrence has been shown to occur at higher rates in the presence of vascular invasion.68 Also noteworthy, PAX8 is involved in a recurrent chromosomal translocation observed in a subset of follicular thyroid cancers. The rearrangement, t(2;3)(q13;p25), involves PAX8 on chromosome 2 and the nuclear receptor, peroxisome proliferator-activated receptor gamma-1 (PPARγ) on chromosome 3. PAX8-PPARγ was initially proposed to be specific to follicular thyroid carcinomas, as it was not detected in follicular adenomas, papillary carcinomas or © 2014 Macmillan Publishers Limited

PAX genes in childhood cancers P Mahajan et al

5

Figure 3. PAX9 is misexpressed in bone/bone marrow-related malignancies. Shown are probes for PAX9 and PAX1 expressions in a battery of samples for Pre-B-cell ALL, T-cell ALL, embryonal rhabdomyosarcoma (E-RMS), A-RMS, Ewing sarcoma (EWS), and osteosarcoma (OS). Profiles are from cell lines (C), tumor xenografts (X) and primary human tumors (T). Also shown for comparison is a probe for MyoD, a gene typically expressed in E-RMS. Data are obtained from the Pediatric Tumor Affymetrix Database (Oncogenomics; http://home.ccr.cancer.gov/oncology/ oncogenomics/).

mutinodular hyperplasias.70 However, the presence of the PAX8PPARγ has since been noted in other thyroid neoplasms, including follicular adenomas.71–75 The fusion gene inhibits normal PPARγ function,70 thus highlighting the potential role of PPARγ as a tumor suppressor and possible therapeutic target. Functionally, PAX8-PPARγ expression increases cell cycle transit and decreases apoptosis,76 pointing toward an oncogenic potential for the fusion, whereas PAX8 immunostaining has been shown to be useful diagnostically.77–81 Further probing of how PAX8-PPARγ might underlie pediatric thyroid cancers remains to be determined, and the clinical/therapeutic significance of PAX8 and/or the PAX8-PPARγ fusion clarified. PAX5 in B-cell development, leukemias, lymphomas and other cancer types. Acute lymphoblastic leukemia (ALL) is the most common pediatric malignancy and represents ~ 25% of childhood cancers. B-cell precursor ALL accounts for a majority of childhood leukemias and, in recent years, great improvements in ALL outcomes have been achieved, with a present-day 5-year survival rate of ~90%.82 Disruption of the genes involved in normal B-cell development, including PAX5, has been associated with leukemogenesis. PAX5, a B-cell-specific transcription factor, has a key role in B-cell lineage commitment83,84 and aberrant expression of PAX5 is associated with B-cell cancers, such as lymphoma and B-cell ALL.85–87 Pax5-deficient pro-B cells differentiate along myeloid and non-B-lymphoid lineages, whereas B-cell development is arrested,84 thus underscoring the critical importance of Pax5 in B-cell differentiation. A diverse array of PAX5 alterations, including PAX5 deletions and structural rearrangements, has been observed in both children and adults with acute B-cell ALL88 and recently germline PAX5 mutations have been linked with a susceptibility to pre B-cell ALL,89 implicating PAX5 in familial or hereditary ALL. PAX5 is a commonly targeted gene in ALL, as somatic mutations in PAX5 have been observed in ~ 30% of pediatric ALL cases.90 In ALL, PAX5 has been identified in various fusion genes.91–96 The incidence of PAX5 rearrangements in childhood B-cell precursor ALL is ~ 2.5%.97 In addition, intragenic PAX5 amplifications have been detected in pediatric B-cell precursor ALL patient samples, and although the importance of this amplification is unclear, PAX5 amplification may be a potential prognostic factor and preferentially associated with relapsed disease.98 PAX5 has also been associated with acute myeloblastic leukemia and PAX5 overexpression has been implicated in driving transcription of the B-cell-specific surface marker CD-19 in t(8;21) acute myeloblastic leukemia.99 PAX5 is additionally involved in a chromosomal rearrangement with the immunoglobulin heavy-chain locus on chromosome 14 [t(9;14)(p13;q32)], which results in the aberrant expression of PAX5 in variety of B-cell lymphomas,100,101 including aggressive B-cell non-Hodgkin’s lymphomas.102 Pax5 has also been shown to be a © 2014 Macmillan Publishers Limited

potent oncogene in the tumorgenesis of T-lymphoblastic lymphomas,103 suggesting that normal T-lymphoid development is interrupted with aberrant Pax5 expression. Loss-of-function Pax5 mutations in mice result in aggressive lymphomas, illustrating a role for Pax5 as a tumor suppressor gene.104 Although the prognostic implications of PAX5 alterations is unclear, PAX5 immunostaining has a potentially valuable diagnostic role in distinguishing B-cell lymphoid cancers from other lymphoid neoplasms.105 Of note, PAX5 expression is also observed in non-B-cell cancers, including nonhematopoietic solid tumors. PAX5 has been shown to be dysregulated in medulloblastoma106 and neuroblastoma.107 Medulloblastoma is the most common malignant pediatric brain tumor and neuroblastoma is the most common extracranial solid tumor in childhood. PAX5 is expressed in medulloblastoma, even though Pax5 transcripts are not detected in normal neonatal or adult cerebellar tissue, suggesting that PAX5 misexpression might be influential in medulloblastoma.106 Furthermore, downregulation of PAX5 in neuroblastoma cell lines decreases proliferation,107 highlighting the oncogenic potential of PAX5 in nonhematological malignancies. Interestingly, PAX5 has been shown to interact with the underphosphorylated form of the tumor suppressor RB108 and although this interaction warrants further investigation, PAX5 may interfere with normal RB function, thereby contributing to oncogenesis. PAX4 and PAX6 in the pancreas, eye and retinoblastoma PAX4 and PAX6, which contain the PD and full-length HD, but lack the OP motif, belong to subgroup IV. Pax4 and Pax6 are predominantly expressed in the pancreatic islet cells,109 whereas Pax6 is additionally critical in eye patterning.110–112 Pax4 specifically has a role in the differentiation of insulin-secreting β-cells in the pancreas113–116 and mutations in PAX4 have been associated with diabetes.117–119 In contrast, Pax6 is involved in the differentiation of the α-cells in the pancreas, which are responsible for synthesizing and secreting glucagon.120 Pax6/PAX6 is also a critical master regulator of eye development110–112 and has significant homology in Drosophila, mice and humans.121 Loss-of-function mutations in the evolutionary conserved Pax6 locus result in eye defects in mammals and flies, including the eyeless phenotype in Drosophila. Provocatively, ectopic compound eyes are induced within Drosophila wings, legs and antennae tissue by targeted misexpression of the Drosophila eyeless (ey) gene,122 as well as ectopic expression of murine Pax6, further illustrating PAX6’s role as a master regulator of eye patterning. Of note, an additional Drosophila Pax6 gene, twin of ey (toy), has also been identified and is required for the expression of ey to initiate eye development.123 Similar to the ey mutation, the small eye (Sey) mutation in mice, characterized by lack of eyes and nasal cavity, Oncogene (2014), 1 – 9

PAX genes in childhood cancers P Mahajan et al

6 and human aniridia, in which the iris is absent, arises from mutations in the homologous Pax6/PAX6 gene.124–126 Consistent with PAX6’s intimate association with eye development, the hypothesis exists questioning whether PAX6 activity might be associated with pediatric ocular neoplasms—in particular, retinoblastoma. Retinoblastoma is the most common malignant ocular tumor in children and ~ 200 children are affected each year in the United States.127 The role of PAX6 in retinoblastoma has been investigated128,129 and PAX6 overexpression using recombinant lentiviral vectors induces retinoblastoma tumor cell proliferation with a reduction in G0/G1 arrest.129 Furthermore, silencing PAX6 with small interfering molecules inhibits proliferation and increases apoptosis in retinoblastoma cells, suggesting a role for PAX6 in retinoblastoma tumor cell survival.128 Investigation of a particular microRNA shown to be downregulated in retinoblastoma, MiR-365b-3b, demonstrated that expression of MiR-365b-3b promotes apoptosis by targeting PAX6 in retinoblastoma.130 Therefore, there is a potential role for PAX6 in the growth of retinoblastoma tumor cells. PAX1 and PAX9, a ‘benign’ subgroup? With regard to structure, subgroup I, comprised of PAX1 and PAX9, is the ‘minimalist’ subgroup, as these two PAX orthologs are the only members that contain no additional DNA-binding domains beyond the PD (Figure 1), and thus presumably activate (or repress via the OP motif) transcription from PD-specific DNA elements. On the basis of murine studies, Pax1 and Pax9 are best known for their critical roles in musculoskeletal development.131–133 Specifically, Pax1 is necessary for normal development of the vertebral column, sternum and scapula,132,134 whereas Pax9 homozygous loss-of-function mutations demonstrate craniofacial and hind-/forelimb abnormalities,133 showing slightly different expression profiles of Pax1 and -9. The importance of PAX1 and PAX9 in development is further revealed by their involvement in human disease. Consistent with Pax1 as integral for murine vertebral development, mutation of the PAX1 PD has been identified in spina bifida,135 while oligodontia has been associated with PAX9 mutations.136–139 These findings underscore the importance PAX1 and -9 for proper vertebral, craniofacial and tooth development.140 Keeping with the notion of subgroup 1 being the ‘minimalist’ subgroup, no definitive role for PAX1 or -9 has been uncovered with regard to human (or pediatric) tumorigenesis. We wish to introduce here, however, the observation that PAX9 is preferentially misexpressed in childhood tumors from the bone and bone marrow. For this review, we surveyed PAX1 and PAX9 expression levels in a large collection of pediatric cancer cell lines, xenograft tumors and primary tumors using the Pediatric Tumor Affymetrix Database (Oncogenomics; http://home.ccr.cancer.gov/oncology/ oncogenomics/).141 When compared with normal tissues and other pediatric malignancies, PAX9 showed a provocative and significant upregulation in three malignancies related to the bone or bone marrow: T-cell ALL, Ewing sarcoma and osteosarcoma (Figure 3). Given that PAX9 is intimately involved in skeletal formation, we propose the hypothesis that PAX9 misregulation might influence childhood malignancies (or a subset of these malignancies) originating from pediatric bone or bone marrow tissues. CONCLUDING REMARKS PAX genes encode evolutionary conserved transcription factors and PAX mutations have been shown to be associated with distinct phenotypes in species ranging from Drosophila to mice and humans. Although they have precise and specific roles in organogenesis and tissue development, PAX genes similarly are important in proliferation, growth, cell maintenance and apoptosis. When PAX gene function and behavior are disrupted, Oncogene (2014), 1 – 9

PAX genes can act as oncogenes, as we have discussed, for example, in RMS, Wilms tumor and B-cell cancers. Their importance as diagnostic and prognostic markers, and more importantly as putative therapeutic targets, needs to and will be subjects of further interrogation. CONFLICT OF INTEREST The authors declare no conflict of interest.

ACKNOWLEDGEMENTS This work was supported by funding to RLG by the Burroughs Wellcome Fund (Career Award for Medical Scientists), Alex's Lemonade Stand Foundation ("A" Award), American Cancer Society (Research Scholars Grant) and UTSW Department of Pathology (Research Grant). We thank Stephen X. Skapek for helpful suggestions regarding this manuscript.

REFERENCES 1 Li CG, Eccles MR. PAX genes in cancer; friends or foes? Front Genet 2012; 3: 6. 2 Lang D, Powell SK, Plummer RS, Young KP, Ruggeri BA. PAX genes: roles in development, pathophysiology, and cancer. Biochem Pharmacol 2007; 73: 1–14. 3 Robson EJ, He SJ, Eccles MR. A PANorama of PAX genes in cancer and development. Nat Rev Cancer 2006; 6: 52–62. 4 Dahl E, Koseki H, Balling R. Pax genes and organogenesis. Bioessays 1997; 19: 755–765. 5 Epstein J, Cai J, Glaser T, Jepeal L, Maas R. Identification of a Pax paired domain recognition sequence and evidence for DNA-dependent conformational changes. J Biol Chem 1994; 269: 8355–8361. 6 Epstein JA, Lam P, Jepeal L, Maas RL, Shapiro DN. Pax3 inhibits myogenic differentiation of cultured myoblast cells. J Biol Chem 1995; 270: 11719–11722. 7 Apuzzo S, Gros P. Cooperative interactions between the two DNA binding domains of Pax3: helix 2 of the paired domain is in the proximity of the amino terminus of the homeodomain. Biochemistry 2007; 46: 2984–2993. 8 Wilson D, Sheng G, Lecuit T, Dostatni N, Desplan C. Cooperative dimerization of paired class homeo domains on DNA. Genes Dev 1993; 7: 2120–2134. 9 Jun S, Desplan C. Cooperative interactions between paired domain and homeodomain. Development 1996; 122: 2639–2650. 10 Mailhos C, Andre S, Mollereau B, Goriely A, Hemmati-Brivanlou A, Desplan C. Drosophila Goosecoid requires a conserved heptapeptide for repression of paired-class homeoprotein activators. Development 1998; 125: 937–947. 11 Epstein DJ, Vekemans M, Gros P. Splotch (Sp2H), a mutation affecting development of the mouse neural tube, shows a deletion within the paired homeodomain of Pax-3. Cell 1991; 67: 767–774. 12 Goulding MD, Chalepakis G, Deutsch U, Erselius JR, Gruss P. Pax-3, a novel murine DNA binding protein expressed during early neurogenesis. EMBO J 1991; 10: 1135–1147. 13 Goulding M, Lumsden A, Paquette AJ. Regulation of Pax-3 expression in the dermomyotome and its role in muscle development. Development 1994; 120: 957–971. 14 Bober E, Franz T, Arnold HH, Gruss P, Tremblay P. Pax-3 is required for the development of limb muscles: a possible role for the migration of dermomyotomal muscle progenitor cells. Development 1994; 120: 603–612. 15 Tassabehji M, Read AP, Newton VE, Harris R, Balling R, Gruss P et al. Waardenburg's syndrome patients have mutations in the human homologue of the Pax-3 paired box gene. Nature 1992; 355: 635–636. 16 Kassar-Duchossoy L, Giacone E, Gayraud-Morel B, Jory A, Gomes D, Tajbakhsh S. Pax3/Pax7 mark a novel population of primitive myogenic cells during development. Genes Dev 2005; 19: 1426–1431. 17 Seale P, Sabourin LA, Girgis-Gabardo A, Mansouri A, Gruss P, Rudnicki MA. Pax7 is required for the specification of myogenic satellite cells. Cell 2000; 102: 777–786. 18 Oustanina S, Hause G, Braun T. Pax7 directs postnatal renewal and propagation of myogenic satellite cells but not their specification. EMBO J 2004; 23: 3430–3439. 19 Relaix F, Montarras D, Zaffran S, Gayraud-Morel B, Rocancourt D, Tajbakhsh S et al. Pax3 and Pax7 have distinct and overlapping functions in adult muscle progenitor cells. J Cell Biol 2006; 172: 91–102. 20 von Maltzahn J, Jones AE, Parks RJ, Rudnicki MA. Pax7 is critical for the normal function of satellite cells in adult skeletal muscle. Proc Natl Acad Sci USA 2013; 110: 16474–16479. 21 Lepper C, Conway SJ, Fan CM. Adult satellite cells and embryonic muscle progenitors have distinct genetic requirements. Nature 2009; 460: 627–631.

© 2014 Macmillan Publishers Limited

PAX genes in childhood cancers P Mahajan et al

7 22 Gurney J, Young J, Roffers S, Smith M, Bunin G. Cancer indicences and survival among children and adolescents: United States SEER Program 1975–1995. In: Ries L (ed.) National Cancer Institute, SEER Program. NIH: Bethesda, MD, USA, 1999, pp 111–124. 23 Ognjanovic S, Linabery AM, Charbonneau B, Ross JA. Trends in childhood rhabdomyosarcoma incidence and survival in the United States, 1975–2005. Cancer 2009; 115: 4218–4226. 24 Meza JL, Anderson J, Pappo AS, Meyer WH, Children's Oncology Group, Analysis of prognostic factors in patients with nonmetastatic rhabdomyosarcoma treated on intergroup rhabdomyosarcoma studies III and IV: the Children's Oncology Group. J Clin Oncol 2006; 24: 3844–3851. 25 Huh WW, Skapek SX. Childhood rhabdomyosarcoma: new insight on biology and treatment. Curr Oncol Rep 2010; 12: 402–410. 26 Tiffin N, Williams RD, Shipley J, Pritchard-Jones K. PAX7 expression in embryonal rhabdomyosarcoma suggests an origin in muscle satellite cells. Br J Cancer 2003; 89: 327–332. 27 Barr FG, Fitzgerald JC, Ginsberg JP, Vanella ML, Davis RJ, Bennicelli JL. Predominant expression of alternative PAX3 and PAX7 forms in myogenic and neural tumor cell lines. Cancer Res 1999; 59: 5443–5448. 28 Bernasconi M, Remppis A, Fredericks WJ, Rauscher FJ 3rd, Schafer BW. Induction of apoptosis in rhabdomyosarcoma cells through down-regulation of PAX proteins. Proc Natl Acad Sci USA 1996; 93: 13164–13169. 29 Davis RJ, D'Cruz CM, Lovell MA, Biegel JA, Barr FG. Fusion of PAX7 to FKHR by the variant t(1;13)(p36;q14) translocation in alveolar rhabdomyosarcoma. Cancer Res 1994; 54: 2869–2872. 30 Sorensen PH, Lynch JC, Qualman SJ, Tirabosco R, Lim JF, Maurer HM et al. PAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicators in alveolar rhabdomyosarcoma: a report from the children's oncology group. J Clin Oncol 2002; 20: 2672–2679. 31 Kelly KM, Womer RB, Sorensen PH, Xiong QB, Barr FG. Common and variant gene fusions predict distinct clinical phenotypes in rhabdomyosarcoma. J Clin Oncol 1997; 15: 1831–1836. 32 Williamson D, Missiaglia E, de Reynies A, Pierron G, Thuille B, Palenzuela G et al. Fusion gene-negative alveolar rhabdomyosarcoma is clinically and molecularly indistinguishable from embryonal rhabdomyosarcoma. J Clin Oncol 2010; 28: 2151–2158. 33 Cao L, Yu Y, Bilke S, Walker RL, Mayeenuddin LH, Azorsa DO et al. Genome-wide identification of PAX3-FKHR binding sites in rhabdomyosarcoma reveals candidate target genes important for development and cancer. Cancer Res 2010; 70: 6497–6508. 34 Wachtel M, Dettling M, Koscielniak E, Stegmaier S, Treuner J, Simon-Klingenstein K et al. Gene expression signatures identify rhabdomyosarcoma subtypes and detect a novel t(2;2)(q35;p23) translocation fusing PAX3 to NCOA1. Cancer Res 2004; 64: 5539–5545. 35 Shern JF, Chen L, Chmielecki J, Wei JS, Patidar R, Rosenberg M et al. Comprehensive genomic analysis of rhabdomyosarcoma reveals a landscape of alterations affecting a common genetic axis in fusion-positive and fusionnegative tumors. Cancer Discov 2014; 4: 216–231. 36 Galindo RL, Allport JA, Olson EN. A Drosophila model of the rhabdomyosarcoma initiator PAX7-FKHR. Proc Natl Acad Sci USA 2006; 103: 13439–13444. 37 Avirneni-Vadlamudi U, Galindo KA, Endicott TR, Paulson V, Cameron S, Galindo RL. Drosophila and mammalian models uncover a role for the myoblast fusion gene TANC1 in rhabdomyosarcoma. J Clin Invest 2012; 122: 403–407. 38 Ren YX, Finckenstein FG, Abdueva DA, Shahbazian V, Chung B, Weinberg KI et al. Mouse mesenchymal stem cells expressing PAX-FKHR form alveolar rhabdomyosarcomas by cooperating with secondary mutations. Cancer Res 2008; 68: 6587–6597. 39 Naini S, Etheridge KT, Adam SJ, Qualman SJ, Bentley RC, Counter CM et al. Defining the cooperative genetic changes that temporally drive alveolar rhabdomyosarcoma. Cancer Res 2008; 68: 9583–9588. 40 Keller C, Arenkiel BR, Coffin CM, El-Bardeesy N, DePinho RA, Capecchi MR. Alveolar rhabdomyosarcomas in conditional Pax3:Fkhr mice: cooperativity of Ink4a/ARF and Trp53 loss of function. Genes Dev 2004; 18: 2614–2626. 41 Chen X, Stewart E, Shelat AA, Qu C, Bahrami A, Hatley M et al. Targeting oxidative stress in embryonal rhabdomyosarcoma. Cancer Cell 2013; 24: 710–724. 42 Mansouri A, Chowdhury K, Gruss P. Follicular cells of the thyroid gland require Pax8 gene function. Nat Genet 1998; 19: 87–90. 43 Dressler GR, Deutsch U, Chowdhury K, Nornes HO, Gruss P. Pax2, a new murine paired-box-containing gene and its expression in the developing excretory system. Development 1990; 109: 787–795. 44 Plachov D, Chowdhury K, Walther C, Simon D, Guenet JL, Gruss P. Pax8, a murine paired box gene expressed in the developing excretory system and thyroid gland. Development 1990; 110: 643–651. 45 Dressler GR, Douglass EC. Pax-2 is a DNA-binding protein expressed in embryonic kidney and Wilms tumor. Proc Natl Acad Sci USA 1992; 89: 1179–1183.

© 2014 Macmillan Publishers Limited

46 Sanyanusin P, Schimmenti LA, McNoe LA, Ward TA, Pierpont ME, Sullivan MJ et al. Mutation of the PAX2 gene in a family with optic nerve colobomas, renal anomalies and vesicoureteral reflux. Nat Genet 1995; 9: 358–364. 47 Eccles MR, Wallis LJ, Fidler AE, Spurr NK, Goodfellow PJ, Reeve AE. Expression of the PAX2 gene in human fetal kidney and Wilms' tumor. Cell Growth Differ 1992; 3: 279–289. 48 Torres M, Gomez-Pardo E, Dressler GR, Gruss P. Pax-2 controls multiple steps of urogenital development. Development 1995; 121: 4057–4065. 49 Winyard PJ, Risdon RA, Sams VR, Dressler GR, Woolf AS. The PAX2 tanscription factor is expressed in cystic and hyperproliferative dysplastic epithelia in human kidney malformations. J Clin Invest 1996; 98: 451–459. 50 Poleev A, Fickenscher H, Mundlos S, Winterpacht A, Zabel B, Fidler A et al. PAX8, a human paired box gene: isolation and expression in developing thyroid, kidney and Wilms' tumors. Development 1992; 116: 611–623. 51 Narlis M, Grote D, Gaitan Y, Boualia SK, Bouchard M. Pax2 and pax8 regulate branching morphogenesis and nephron differentiation in the developing kidney. J Am Soc Nephrol 2007; 18: 1121–1129. 52 Gnarra JR, Dressler GR. Expression of Pax-2 in human renal cell carcinoma and growth inhibition by antisense oligonucleotides. Cancer Res 1995; 55: 4092–4098. 53 Muratovska A, Zhou C, He S, Goodyer P, Eccles MR. Paired-Box genes are frequently expressed in cancer and often required for cancer cell survival. Oncogene 2003; 22: 7989–7997. 54 Fonsato V, Buttiglieri S, Deregibus MC, Puntorieri V, Bussolati B, Camussi G. Expression of Pax2 in human renal tumor-derived endothelial cells sustains apoptosis resistance and angiogenesis. Am J Pathol 2006; 168: 706–713. 55 Park D, Jia H, Rajakumar V, Chamberlin HM. Pax2/5/8 proteins promote cell survival in C. elegans. Development 2006; 133: 4193–4202. 56 Li CG, Nyman JE, Braithwaite AW, Eccles MR. PAX8 promotes tumor cell growth by transcriptionally regulating E2F1 and stabilizing RB protein. Oncogene 2011; 30: 4824–4834. 57 Fernandez C, Geller J, Ehrlich P, Hill A, Kalapurakai J, Grundy P et al. Renal tumors. In: Pizzo P, Poplack D (eds) Principles and Practice of Pediatric Oncology, 6th edn, Lippincott WIlliams & Wilkins: Philadelphia, PA, USA, 2011, pp 861–885. 58 Ali AN, Diaz R, Shu HK, Paulino AC, Esiashvili N. A Surveillance, Epidemiology and End Results (SEER) program comparison of adult and pediatric Wilms' tumor. Cancer 2012; 118: 2541–2551. 59 Silberstein J, Grabowski J, Saltzstein SL, Kane CJ. Renal cell carcinoma in the pediatric population: Results from the California Cancer Registry. Pediatr Blood Cancer 2009; 52: 237–241. 60 Dressler GR, Wilkinson JE, Rothenpieler UW, Patterson LT, Williams-Simons L, Westphal H. Deregulation of Pax-2 expression in transgenic mice generates severe kidney abnormalities. Nature 1993; 362: 65–67. 61 Ryan G, Steele-Perkins V, Morris JF, Rauscher FJ 3rd, Dressler GR. Repression of Pax-2 by WT1 during normal kidney development. Development 1995; 121: 867–875. 62 Discenza MT, He S, Lee TH, Chu LL, Bolon B, Goodyer P et al. WT1 is a modifier of the Pax2 mutant phenotype: cooperation and interaction between WT1 and Pax2. Oncogene 2003; 22: 8145–8155. 63 Zhai QJ, Ozcan A, Hamilton C, Shen SS, Coffey D, Krishnan B et al. PAX-2 expression in non-neoplastic, primary neoplastic, and metastatic neoplastic tissue: a comprehensive immunohistochemical study. Appl Immunohistochem Mol Morphol 2010; 18: 323–332. 64 Ozcan A, de la Roza G, Ro JY, Shen SS, Truong LD. PAX2 and PAX8 expression in primary and metastatic renal tumors: a comprehensive comparison. Arch Pathol Lab Med 2012; 136: 1541–1551. 65 Puglisi F, Cesselli D, Damante G, Pellizzari L, Beltrami CA, Di Loreto C. Expression of Pax-8, p53 and bcl-2 in human benign and malignant thyroid diseases. Anticancer Res 2000; 20: 311–316. 66 Scouten WT, Patel A, Terrell R, Burch HB, Bernet VJ, Tuttle RM et al. Cytoplasmic localization of the paired box gene, Pax-8, is found in pediatric thyroid cancer and may be associated with a greater risk of recurrence. Thyroid 2004; 14: 1037–1046. 67 Holmes L Jr., Hossain J, Opara F. Pediatric thyroid carcinoma incidence and temporal trends in the USA (1973-2007): race or shifting diagnostic paradigm? ISRN Oncol 2012; 2012: 906197. 68 Enomoto K, Enomoto Y, Uchino S, Yamashita H, Noguchi S. Follicular thyroid cancer in children and adolescents: clinicopathologic features, long-term survival, and risk factors for recurrence. Endocr J 2013; 60: 629–635. 69 Hogan AR, Zhuge Y, Perez EA, Koniaris LG, Lew JI, Sola JE. Pediatric thyroid carcinoma: incidence and outcomes in 1753 patients. J Surg Res 2009; 156: 167–172. 70 Kroll TG, Sarraf P, Pecciarini L, Chen CJ, Mueller E, Spiegelman BM et al. PAX8-PPARgamma1 fusion oncogene in human thyroid carcinoma [corrected]. Science 2000; 289: 1357–1360.

Oncogene (2014), 1 – 9

PAX genes in childhood cancers P Mahajan et al

8 71 Marques AR, Espadinha C, Catarino AL, Moniz S, Pereira T, Sobrinho LG et al. Expression of PAX8-PPAR gamma 1 rearrangements in both follicular thyroid carcinomas and adenomas. J Clin Endocrinol Metab 2002; 87: 3947–3952. 72 Nikiforova MN, Biddinger PW, Caudill CM, Kroll TG, Nikiforov YE. PAX8PPARgamma rearrangement in thyroid tumors: RT-PCR and immunohistochemical analyses. Am J Surg Pathol 2002; 26: 1016–1023. 73 Cheung L, Messina M, Gill A, Clarkson A, Learoyd D, Delbridge L et al. Detection of the PAX8-PPAR gamma fusion oncogene in both follicular thyroid carcinomas and adenomas. J Clin Endocrinol Metab 2003; 88: 354–357. 74 Castro P, Rebocho AP, Soares RJ, Magalhaes J, Roque L, Trovisco V et al. PAX8-PPARgamma rearrangement is frequently detected in the follicular variant of papillary thyroid carcinoma. J Clin Endocrinol Metab 2006; 91: 213–220. 75 Lacroix L, Mian C, Barrier T, Talbot M, Caillou B, Schlumberger M et al. PAX8 and peroxisome proliferator-activated receptor gamma 1 gene expression status in benign and malignant thyroid tissues. Eu J Endocrinol 2004; 151: 367–374. 76 Gregory Powell J, Wang X, Allard BL, Sahin M, Wang XL, Hay ID et al. The PAX8/PPARgamma fusion oncoprotein transforms immortalized human thyrocytes through a mechanism probably involving wild-type PPARgamma inhibition. Oncogene 2004; 23: 3634–3641. 77 Ordonez NG. Value of PAX 8 immunostaining in tumor diagnosis: a review and update. Adv Anat Pathol 2012; 19: 140–151. 78 Nonaka D, Tang Y, Chiriboga L, Rivera M, Ghossein R. Diagnostic utility of thyroid transcription factors Pax8 and TTF-2 (FoxE1) in thyroid epithelial neoplasms. Mod Pathol 2008; 21: 192–200. 79 Ozcan A, Shen SS, Hamilton C, Anjana K, Coffey D, Krishnan B et al. PAX 8 expression in non-neoplastic tissues, primary tumors, and metastatic tumors: a comprehensive immunohistochemical study. Mod Pathol 2011; 24: 751–764. 80 Laury AR, Perets R, Piao H, Krane JF, Barletta JA, French C et al. A comprehensive analysis of PAX8 expression in human epithelial tumors. Am J Surg Pathol 2011; 35: 816–826. 81 Bishop JA, Sharma R, Westra WH. PAX8 immunostaining of anaplastic thyroid carcinoma: a reliable means of discerning thyroid origin for undifferentiated tumors of the head and neck. Hum Pathol 2011; 42: 1873–1877. 82 Hunger SP, Lu X, Devidas M, Camitta BM, Gaynon PS, Winick NJ et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the children's oncology group. J Clin Oncol 2012; 30: 1663–1669. 83 Holmes ML, Pridans C, Nutt SL. The regulation of the B-cell gene expression programme by Pax5. Immunol Cell Biol 2008; 86: 47–53. 84 Nutt SL, Heavey B, Rolink AG, Busslinger M. Commitment to the B-lymphoid lineage depends on the transcription factor Pax5. Nature 1999; 401: 556–562. 85 Familiades J, Bousquet M, Lafage-Pochitaloff M, Bene MC, Beldjord K, De Vos J et al. PAX5 mutations occur frequently in adult B-cell progenitor acute lymphoblastic leukemia and PAX5 haploinsufficiency is associated with BCR-ABL1 and TCF3-PBX1 fusion genes: a GRAALL study. Leukemia 2009; 23: 1989–1998. 86 Cobaleda C, Schebesta A, Delogu A, Busslinger M. Pax5: the guardian of B cell identity and function. Nat Immunol 2007; 8: 463–470. 87 O'Brien P, Morin P Jr, Ouellette RJ, Robichaud GA. The Pax-5 gene: a pluripotent regulator of B-cell differentiation and cancer disease. Cancer Res 2011; 71: 7345–7350. 88 Coyaud E, Struski S, Prade N, Familiades J, Eichner R, Quelen C et al. Wide diversity of PAX5 alterations in B-ALL: a Groupe Francophone de Cytogenetique Hematologique study. Blood 2010; 115: 3089–3097. 89 Shah S, Schrader KA, Waanders E, Timms AE, Vijai J, Miething C et al. A recurrent germline PAX5 mutation confers susceptibility to pre-B cell acute lymphoblastic leukemia. Nat Genet 2013; 45: 1226–1231. 90 Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan-Smith E, Dalton JD et al. Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature 2007; 446: 758–764. 91 Cazzaniga G, Daniotti M, Tosi S, Giudici G, Aloisi A, Pogliani E et al. The paired box domain gene PAX5 is fused to ETV6/TEL in an acute lymphoblastic leukemia case. Cancer Res 2001; 61: 4666–4670. 92 Bousquet M, Broccardo C, Quelen C, Meggetto F, Kuhlein E, Delsol G et al. A novel PAX5-ELN fusion protein identified in B-cell acute lymphoblastic leukemia acts as a dominant negative on wild-type PAX5. Blood 2007; 109: 3417–3423. 93 Denk D, Nebral K, Bradtke J, Pass G, Moricke A, Attarbaschi A et al. PAX5-AUTS2: a recurrent fusion gene in childhood B-cell precursor acute lymphoblastic leukemia. Leuk Res 2012; 36: e178–e181. 94 Strehl S, Konig M, Dworzak MN, Kalwak K, Haas OA. PAX5/ETV6 fusion defines cytogenetic entity dic(9;12)(p13;p13). Leukemia 2003; 17: 1121–1123. 95 Kawamata N, Pennella MA, Woo JL, Berk AJ, Koeffler HP. Dominant-negative mechanism of leukemogenic PAX5 fusions. Oncogene 2012; 31: 966–977. 96 Nebral K, Konig M, Harder L, Siebert R, Haas OA, Strehl S. Identification of PML as novel PAX5 fusion partner in childhood acute lymphoblastic leukaemia. Br J Haematol 2007; 139: 269–274.

Oncogene (2014), 1 – 9

97 Nebral K, Denk D, Attarbaschi A, Konig M, Mann G, Haas OA et al. Incidence and diversity of PAX5 fusion genes in childhood acute lymphoblastic leukemia. Leukemia 2009; 23: 134–143. 98 Ofverholm I, Tran AN, Heyman M, Zachariadis V, Nordenskjold M, Nordgren A et al. Impact of IKZF1 deletions and PAX5 amplifications in pediatric B-cell precursor ALL treated according to NOPHO protocols. Leukemia 2013; 27: 1936–1939. 99 Walter K, Cockerill PN, Barlow R, Clarke D, Hoogenkamp M, Follows GA et al. Aberrant expression of CD19 in AML with t(8;21) involves a poised chromatin structure and PAX5. Oncogene 2010; 29: 2927–2937. 100 Busslinger M, Klix N, Pfeffer P, Graninger PG, Kozmik Z. Deregulation of PAX-5 by translocation of the Emu enhancer of the IgH locus adjacent to two alternative PAX-5 promoters in a diffuse large-cell lymphoma. Proc Natl Acad Sci USA 1996; 93: 6129–6134. 101 Iida S, Rao PH, Nallasivam P, Hibshoosh H, Butler M, Louie DC et al. The t(9;14) (p13;q32) chromosomal translocation associated with lymphoplasmacytoid lymphoma involves the PAX-5 gene. Blood 1996; 88: 4110–4117. 102 Poppe B, De Paepe P, Michaux L, Dastugue N, Bastard C, Herens C et al. PAX5/IGH rearrangement is a recurrent finding in a subset of aggressive B-NHL with complex chromosomal rearrangements. Genes Chromosomes Cancer 2005; 44: 218–223. 103 Souabni A, Jochum W, Busslinger M. Oncogenic role of Pax5 in the T-lymphoid lineage upon ectopic expression from the immunoglobulin heavy-chain locus. Blood 2007; 109: 281–289. 104 Cobaleda C, Jochum W, Busslinger M. Conversion of mature B cells into T cells by dedifferentiation to uncommitted progenitors. Nature 2007; 449: 473–477. 105 Desouki MM, Post GR, Cherry D, Lazarchick J. PAX-5: a valuable immunohistochemical marker in the differential diagnosis of lymphoid neoplasms. Clin Med Res 2010; 8: 84–88. 106 Kozmik Z, Sure U, Ruedi D, Busslinger M, Aguzzi A. Deregulated expression of PAX5 in medulloblastoma. Proc Natl Acad Sci USA 1995; 92: 5709–5713. 107 Baumann Kubetzko FB, Di Paolo C, Maag C, Meier R, Schafer BW, Betts DR et al. The PAX5 oncogene is expressed in N-type neuroblastoma cells and increases tumorigenicity of a S-type cell line. Carcinogenesis 2004; 25: 1839–1846. 108 Eberhard D, Busslinger M. The partial homeodomain of the transcription factor Pax-5 (BSAP) is an interaction motif for the retinoblastoma and TATA-binding proteins. Cancer Res 1999; 59(7 Suppl): 1716s–1724s discussion 24s-25s. 109 Dohrmann C, Gruss P, Lemaire L. Pax genes and the differentiation of hormoneproducing endocrine cells in the pancreas. Mech Dev 2000; 92: 47–54. 110 Gehring WJ. The master control gene for morphogenesis and evolution of the eye. Genes Cells 1996; 1: 11–15. 111 Carbe C, Garg A, Cai Z, Li H, Powers A, Zhang X. An allelic series at the paired box gene 6 (Pax6) locus reveals the functional specificity of Pax genes. J Biol Chem 2013; 288: 12130–12141. 112 Walther C, Gruss P. Pax-6, a murine paired box gene, is expressed in the developing CNS. Development 1991; 113: 1435–1449. 113 Sosa-Pineda B. The gene Pax4 is an essential regulator of pancreatic beta-cell development. Mol Cells 2004; 18: 289–294. 114 Brun T, Gauthier BR. A focus on the role of Pax4 in mature pancreatic islet betacell expansion and survival in health and disease. J Mol Endocrinol 2008; 40: 37–45. 115 Smith SB, Ee HC, Conners JR, German MS. Paired-homeodomain transcription factor PAX4 acts as a transcriptional repressor in early pancreatic development. Mol Cell Biol 1999; 19: 8272–8280. 116 Sosa-Pineda B, Chowdhury K, Torres M, Oliver G, Gruss P. The Pax4 gene is essential for differentiation of insulin-producing beta cells in the mammalian pancreas. Nature 1997; 386: 399–402. 117 Jo W, Endo M, Ishizu K, Nakamura A, Tajima T. A novel PAX4 mutation in a Japanese patient with maturity-onset diabetes of the young. Tohoku J Exp Med 2011; 223: 113–118. 118 Shimajiri Y, Sanke T, Furuta H, Hanabusa T, Nakagawa T, Fujitani Y et al. A missense mutation of Pax4 gene (R121W) is associated with type 2 diabetes in Japanese. Diabetes 2001; 50: 2864–2869. 119 Plengvidhya N, Kooptiwut S, Songtawee N, Doi A, Furuta H, Nishi M et al. PAX4 mutations in Thais with maturity onset diabetes of the young. J Clin Endocrinol Metab 2007; 92: 2821–2826. 120 St-Onge L, Sosa-Pineda B, Chowdhury K, Mansouri A, Gruss P. Pax6 is required for differentiation of glucagon-producing alpha-cells in mouse pancreas. Nature 1997; 387: 406–409. 121 Quiring R, Walldorf U, Kloter U, Gehring WJ. Homology of the eyeless gene of Drosophila to the Small eye gene in mice and Aniridia in humans. Science 1994; 265: 785–789. 122 Halder G, Callaerts P, Gehring WJ. Induction of ectopic eyes by targeted expression of the eyeless gene in Drosophila. Science 1995; 267: 1788–1792.

© 2014 Macmillan Publishers Limited

PAX genes in childhood cancers P Mahajan et al

9 123 Czerny T, Halder G, Kloter U, Souabni A, Gehring WJ, Busslinger M. twin of eyeless, a second Pax-6 gene of Drosophila, acts upstream of eyeless in the control of eye development. Mol Cell 1999; 3: 297–307. 124 Hill RE, Favor J, Hogan BL, Ton CC, Saunders GF, Hanson IM et al. Mouse small eye results from mutations in a paired-like homeobox-containing gene. Nature 1991; 354: 522–525. 125 Glaser T, Walton DS, Maas RL. Genomic structure, evolutionary conservation and aniridia mutations in the human PAX6 gene. Nat Genet 1992; 2: 232–239. 126 Jordan T, Hanson I, Zaletayev D, Hodgson S, Prosser J, Seawright A et al. The human PAX6 gene is mutated in two patients with aniridia. Nat Genet 1992; 1: 328–332. 127 Hurwitz R, Shields C, Shields J, Chévez-Barrios P, Gombos D, Hurwitz M et al. Retinoblastoma. In: Pizzo P, Poplack D (eds) Principles and Practice of Pediatric Oncology, 6th edn, Lippincott WIlliams & Wilkins: Philadelphia, PA, USA, 2011, pp 809–837. 128 Bai SW, Li B, Zhang H, Jonas JB, Zhao BW, Shen L et al. Pax6 regulates proliferation and apoptosis of human retinoblastoma cells. Invest Ophthalmol Vis Sci 2011; 52: 4560–4570. 129 Li L, Li B, Zhang H, Bai S, Wang Y, Zhao B et al. Lentiviral vector-mediated PAX6 overexpression promotes growth and inhibits apoptosis of human retinoblastoma cells. Invest Ophthalmol Vis Sci 2011; 52: 8393–8400. 130 Wang J, Wang X, Wu G, Hou D, Hu Q. MiR-365b-3p, down-regulated in retinoblastoma, regulates cell cycle progression and apoptosis of human retinoblastoma cells by targeting PAX6. FEBS Lett 2013; 587: 1779–1786. 131 Su D, Ellis S, Napier A, Lee K, Manley NR. Hoxa3 and pax1 regulate epithelial cell death and proliferation during thymus and parathyroid organogenesis. Dev Biol 2001; 236: 316–329.

© 2014 Macmillan Publishers Limited

132 Dietrich S, Gruss P. undulated phenotypes suggest a role of Pax-1 for the development of vertebral and extravertebral structures. Dev Biol 1995; 167: 529–548. 133 Peters H, Neubuser A, Kratochwil K, Balling R. Pax9-deficient mice lack pharyngeal pouch derivatives and teeth and exhibit craniofacial and limb abnormalities. Genes Dev 1998; 12: 2735–2747. 134 Wilm B, Dahl E, Peters H, Balling R, Imai K. Targeted disruption of Pax1 defines its null phenotype and proves haploinsufficiency. Proc Natl Acad Sci USA 1998; 95: 8692–8697. 135 Hol FA, Geurds MP, Chatkupt S, Shugart YY, Balling R, Schrander-Stumpel CT et al. PAX genes and human neural tube defects: an amino acid substitution in PAX1 in a patient with spina bifida. J Med Genet 1996; 33: 655–660. 136 Jumlongras D, Lin JY, Chapra A, Seidman CE, Seidman JG, Maas RL et al. A novel missense mutation in the paired domain of PAX9 causes non-syndromic oligodontia. Hum Genet 2004; 114: 242–249. 137 Stockton DW, Das P, Goldenberg M, D'Souza RN, Patel PI. Mutation of PAX9 is associated with oligodontia. Nat Genet 2000; 24: 18–19. 138 Frazier-Bowers SA, Scott MR, Cavender A, Mensah J, D'Souza RN. Mutational analysis of families affected with molar oligodontia. Connect Tissue Res 2002; 43: 296–300. 139 Wang J, Jian F, Chen J, Wang H, Lin Y, Yang Z et al. Sequence analysis of PAX9, MSX1 and AXIN2 genes in a Chinese oligodontia family. Arch Oral Biol 2011; 56: 1027–1034. 140 Peters H, Neubuser A, Balling R. Pax genes and organogenesis: Pax9 meets tooth development. Eur J Oral Sci 1998; 106(Suppl 1): 38–43. 141 Khan J, Wei JS, Ringner M, Saal LH, Ladanyi M, Westermann F et al. Classification and diagnostic prediction of cancers using gene expression profiling and artificial neural networks. Nat Med 2001; 7: 673–679.

Oncogene (2014), 1 – 9

PAX genes in childhood oncogenesis: developmental biology gone awry?

Childhood solid tumors often arise from embryonal-like cells, which are distinct from the epithelial cancers observed in adults, and etiologically can...
1MB Sizes 2 Downloads 3 Views