<<

Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

Cancer Review Research

1p36 Tumor Suppression—A Matter of Dosage?

Kai-Oliver Henrich, Manfred Schwab, and Frank Westermann

Abstract A broad range of human malignancies is associated with nonrandom 1p36 deletions, suggesting the existence of tumor suppressors encoded in this region. Evidence for tumor-specific inactivation of 1p36 genes in the classic "two-hit" manner is scarce; however, many tumor suppressors do not require complete inactivation but contribute to tumorigenesis by partial impairment. We discuss recent data derived from both human tumors and functional models indicating that the 1p36 genes CHD5, CAMTA1, KIF1B, CASZ1, and miR-34a contribute to cancer development when reduced in dosage by genomic copy number loss or other mechanisms. We explore potential interactions among these candidates and propose a model where heterozygous 1p36 deletion impairs oncosuppressive pathways via simultaneous downregulation of several dosage-dependent tumor suppressor genes. Cancer Res; 72(23); 1–10. 2012 AACR.

Introduction (Fig. 1; refs. 1, 17–29). Despite extensive 1p36 candidate gene Deletions of the distal short arm of chromosome 1 (1p) are sequence analyses, success was limited for identifying tumor- fi frequently observed in a broad range of human , speci c in or other malignancies, including breast cancer, cervical cancer, pancreatic cancer, which led some to conclude that a deletion mapping approach pheochromocytoma, thyroid cancer, hepatocellular cancer, was unlikely to deliver tumor suppressor genes. Many tumor colorectal cancer, lung cancer, glioma, meningioma, neuro- suppressor genes, however, do not require inactivation in a blastoma, melanoma, Merkel cell carcinoma, rhabdomyosar- classic "two-hit" manner but contribute to tumor development coma, acute myeloid leukemia, chronic myeloid leukemia, and when their dosage is reduced, sometimes only subtly, by non-Hodgkin lymphoma (1, 2). These nonrandom aberrations mechanisms such as copy number change, transcriptional suggest that loss of genetic information mapping to this region repression, epigenetic downregulation, or aberrant miRNA contributes to cancer development. This is supported by regulation (30). Unlike in a classic "two-hit" mutational inac- fi constitutional 1p aberrations in patients (3, tivation scenario, de nite proof for dosage-sensitive tumor 4) and the association of 1p deletion with poor survival of suppressor gene involvement is not offered by a single straight neuroblastoma (5), breast cancer (6, 7), and colon cancer (8, 9) forward assay. Instead, evidence must be accumulated from patients. Deletion of 1p in premalignant lesions and/or early genetic, epigenetic, and transcriptional analyses of human tumor stages of colorectal, breast, and hepatocellular cancer tumors and functional in vitro and in vivo assays. This review fi (10–12) points to a role for 1p genes during the early steps of discusses ve 1p36 genes, CHD5, CAMTA1, KIF1B, CASZ1, and in these entities. This is supported by loss of 1p miR-34a, recently suggested as tumor suppressor candidates material during in vitro progression in a cell culture model of and likely to be impaired by partial reduction as suggested by colon carcinogenesis (13). Furthermore, transfer of 1p chro- both their status in human cancers and their activity in mosomal material suppresses tumorigenicity of both neuro- functional cancer models. blastoma and colon carcinoma cells (14, 15). Since the first report of 1p deletions in neuroblastomas in CHD5 1977 (16), smallest regions of overlapping heterozygous dele- Chromodomain helicase DNA binding (CHD) genes encode a tions (SRO) have been defined in various tumor entities in the class of ATPase-dependent DNA-binding proteins interacting pursuit of cancer-related genes. That 1p36 is a hot spot of with histones to modulate chromatin structure and transcrip- chromosomal aberrations became clear early on (1), with the tion. CHD5 resides in 1p36.31, is preferentially expressed in most detailed mapping picture appearing for neuroblastoma neuronal tissues, and its product regulates genes involved in neuronal function, cell-cycle control, and chromatin remodel- ing (31). Functional evidence for a tumor suppressive role of mouse Chd5 derives from an elegant approach using chromo- Authors' Affiliation: Division of Tumor Genetics B030, German Cancer Research Center, Heidelberg, Germany some engineering to generate mouse models with loss or gain of genomic regions corresponding to human 1p36 (22). Dele- Corresponding Author: Kai-Oliver Henrich, Division of Tumor Genetics B030, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 tion of a Chd5-containing 4.3 Mb genomic subinterval, corre- Heidelberg, Germany. Phone: 496-221-423-220; Fax: 496-221-423-277; sponding to 5.7 Mb of human 1p36, enhanced proliferation, loss E-mail: [email protected] of contact inhibition, spontaneous immortalization, and sen- doi: 10.1158/0008-5472.CAN-12-2230 sitivity to oncogenic transformation of cultured mouse embry- 2012 American Association for Cancer Research. onic fibroblasts. Mice with heterozygous deletion of this

www.aacrjournals.org OF1

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

Henrich et al.

0 Mb 5 Mb 10 Mb 15 Mb 20 Mb 25 Mb

1p36.33 1p36.32 1p36.31 1p36.23 1p36.22 1p36.21 1p36.13 1p36.12 1p36.11

p73 CAMTA1 CHD5 p73 CHD5 KIF1B CASZ1 miR-34a CAMTA1

Schwab et al., 1996 (review) (1) Neuronal D1S47 miR-34a D1S244 genes Caron et al., 2001 (17) D1S1615 MYCN / MYC CASZ1 Bauer et al., 2001 (18) D1S2731 D1S2666

White et al., 2005 (19) BMI1 EZH2 D1S214 D1S2660 Neuroblastoma Ohira et al., 2000 (homozygous, cell line) (20) CADM1 miR-101

HDNB1 KIF1B D1S2736 (11q23) (1p31) Ejeskär et al., 2001 (germ cell tumors + NB) (21) D1S508 D1S244 1p36-encoded Bagchi et al., 2007 (22)

Barbashina et al., 2005 (23) D1S2694 D1S2666 Glioma Felsberg et al., 2004 (24) D1S482 D1S489 D1S2633 D1S2642

Pheochromocytoma; Edström Elder et al., 2000 (25) D1S1612 Melanoma; Poetsch et al., 2003 (26) D1S214 D1S253 D1S243 D1S468

Small cell lung cancer; Girard et al., 2000 (27)

D1S214 Non-small cell lung cancer; Girard et al., 2000 (27) D1S199

Other entities Breast cancer; Bieche et al., 1999 (28) D1S243 D1S468 D1S160 D1S244

Colorectal cancer; Thorstensen et al., 2000 (29) D1S228 D1S2647

Figure 1. Localization of tumor suppressor candidates p73, CHD5, CAMTA1, miR-34a, KIF1b, and CASZ1 with respect to 1p36 alterations in human cancers. Horizontal bars illustrate the extension of commonly deleted regions; short vertical bars at their end represent the first nondeleted locus. Only size (5.4 Mb) and chromosomal extension (1p32.32–1p36.22) are available for the region identified by Bagchi et al. (22). Genomic positions correspond to the UCSC genome browser, assembly Feb. 2009 (GRCh37/hg19). Gray box, model illustrating potential interactions between 1p36 tumor suppressor candidates.

subinterval were prone to hyperplasia in a variety of tissues effects of the engineered deletion, including enhanced prolif- (22). Duplication of this subinterval in mouse embryonic eration, sensitivity to oncogenic transformation, and inhibition fibroblasts inhibited proliferation and increased the senescent of p19Arf/ (22). This suggests that Chd5 is a dose-dependent cell fraction. Mice with subinterval duplication had develop- gene within the identified 4.3 Mb genomic subinterval that mental abnormalities characterized by an increased apoptotic mediates the tumor suppressive mechanisms seen in mouse cell fraction in various tissues, including the neural tube (22). models. The functional role of human CHD5 in a cancer The identified subinterval includes 52 genes. Among 11 tested background was analyzed in neuroblastoma cells, where its candidate genes, knockdown of only Chd5 functionally rescued overexpression had no impact on proliferation, morphology, the proliferative defect of mouse embryonic fibroblasts with differentiation, or apoptosis but significantly inhibited clono- duplication of the subinterval. Chd5 knockdown in wild-type genic growth in soft agar and xenograft tumor growth in mice cells induced phenotypic changes closely resembling the (32). The absence of an impact on proliferation may indicate an

OF2 Cancer Res; 72(23) December 1, 2012 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

1p36 Tumor Suppression—A Matter of Dosage?

already impaired p14Arf (human p19Arf homolog)/p53 pathway nostic information to existing risk stratification (46). Conse- in these cells, a defect frequently seen in neuroblastomas (33). quently, CAMTA1 is included in most recent prognostic neu- CHD5 is one of 23 genes mapping to a 2 Mb SRO in neuro- roblastoma expression classifiers (51–54). Low CAMTA1 blastoma (34) and a 5.4 Mb SRO spanning 1p36.32 to 1p36.22 expression is also significantly associated with shorter survival in glioma (Fig. 1; refs. 22, 34). An SRO containing Chd5 was in patients (50), and, intriguingly, low CAMTA1 identified in a lymphoma mouse model with chromosomal expression emerged as a new independent predictor of poor instability, and syntenic CHD5-containing deletions were dis- outcome in patients with a tumor that is not of neural origin, covered in human T-cell acute lymphoblastic leukemia/lym- colorectal cancer (48). phomas (T-ALL; ref. 35). CHD5 mutations are rare in the Functional evidence for a tumor suppressive role of CAMTA1 entities analyzed so far. Heterozygous missense mutations comes from analyses in neuroblastoma and glioblastoma cells. were found in one of 30 neuroblastoma cell lines (34), 2 of In neuroblastoma cells with low endogenous CAMTA1 levels, 14 metastatic prostate tumors (36) and 3 of 123 primary ectopic CAMTA1 expression inhibits proliferation, induces ovarian cancers (37). Reports of CHD5 frequency accumulation of cells in the G1–G0 phase of the cell cycle and in breast cancer are controversial, ranging from 0% (0/60; inhibits anchorage-independent colony formation and xeno- ref. 37) to 8.5% (3/35; ref. 38). None of the studies have graft tumor growth (55). CAMTA1 induction shifts neuroblas- identified nonsense or frameshift mutations, and whether the toma cell morphology toward a more differentiated type, missense mutations impair CHD5 function, remains to be including induction of neuron-specific markers. The transcrip- investigated. Aberrant CHD5 promoter methylation in primary tome of CAMTA1-induced cells reflects their phenotype and is tumors was identified in 73% of gastric cancers, 17% of colon significantly enriched for genes that mediate cell-cycle inhibi- cancers, 10% of breast cancers, 10% of ovarian cancers, and 4% tion and neuronal function (55). CAMTA1 is upregulated in of gliomas (37, 39, 40), indicating that CHD5 downregulation neuroblastoma cells prompted to differentiate by retinoic acid via promoter methylation mediates a selective advantage in or other stimuli (55). In glioblastoma cell models, CAMTA1 the development of a subset of human tumors. In neuroblas- overexpression reduces both neurosphere formation and xeno- tomas, low CHD5 expression is associated with high-risk graft tumor growth, probably mediated by activation of natri- features such as 1p deletion, amplified MYCN , and uretic peptide A (NPPA), a secreted peptide with a strong advanced stage (41). Furthermore, low CHD5 mRNA and antiproliferative effect on glioblastoma cells (50). The mechan- protein expression in neuroblastomas are significantly asso- isms downregulating CAMTA1 in high-risk tumors are largely ciated with poor patient outcome, even when adjusted for unknown. CAMTA1 expression is significantly lower in neuro- established prognostic variables (32, 42). Together, this sug- blastomas, gliomas, and colorectal cancers with 1p deletion gests that CHD5 is a neuronal gene whose dose reduction compared with tumors retaining 1p (23, 46, 48). This conforms contributes to tumor development by inhibiting the p14Arf/p53 to a haploinsufficiency model where a single CAMTA1 copy pathway. This function is likely to be mediated by CHD5 acting would result in insufficient transcript levels. However, even in as a transcriptional regulator via chromatin remodeling, an neuroblastomas without 1p deletion, low CAMTA1 expression idea supported by the presence of CHD5 in a multiprotein predicts poor outcome (46), indicating additional CAMTA1- complex highly similar to NuRD chromatin remodeling com- repressive mechanisms. No evidence for aberrant methylation plexes (31). of CAMTA1-associated CpG islands was found in neuroblas- tomas or colorectal cancers (48, 55), but other epigenetic CAMTA1 mechanisms might be relevant, as indicated by upregulation CAMTA1 encodes a member of the calmodulin-binding of CAMTA1 in neuroblastoma cells treated with histone dea- transcription activator (CAMTA) protein family (43, 44), is cetylase inhibitors (55). Another mechanism of CAMTA1 localized in 1p36.31-p36.23 and predominantly expressed in downregulation was identified in glioblastoma cells, where it neural tissues, including brain and spinal chord (45). CAMTA1 is targeted by miR-9/9, a miRNA pair that is highly abundant þ maps to virtually all recently described 1p36 neuroblastoma in glioblastoma stem cell-enriched CD133 cell populations SROs (summarized in ref. 46 and Fig. 1), is the only gene (50). Summing up, nonrandom CAMTA1 deletions in tumors, mapping to a 150 Kb SRO in glioma (Fig. 1; ref. 23) and is the role of CAMTA1 in differentiation and growth suppression, homozygously deleted in a subgroup of (47). and the strong association between CAMTA1 downregulation In colorectal cancers, deletion of a small region at 1p36.31- and poor survival in neuroblastoma, glioma, and colorectal p36.23, including only the CAMTA1 gene, had the strongest cancer patients support its assignment as a dosage-dependent impact on survival among all identified genomic alterations . (48). A missense mutation was seen in one of 26 colorectal Further evidence for an involvement of CAMTA1 in cancer cancers (48), but somatic CAMTA1 mutations were not comes from epithelioid hemangioendothelioma (EHE), a rare observed in neuroblastomas or gliomas (23, 49); however, vascular sarcoma difficult to diagnose because of considerable CAMTA1 expression is significantly lower in high-risk tumors morphological overlap with other epithelioid vascular tumors. of both entities (46, 50). In neuroblastoma, low CAMTA1 mRNA Two independent studies identified an EHE-characteristic expression is significantly associated with prognostic markers translocation, t(1;3)(p36.3;q25), involving WWTR1 (WW of poor outcome, including amplified MYCN and advanced domain-containing transcription regulator 1) and CAMTA1 tumor stage (46). Low CAMTA1 expression was also identified (45, 56). The WWTR1/CAMTA1 translocation was found in as a new independent marker of poor outcome adding prog- 100% (56) and 87% to 89% (45) of 2 EHE cohorts but in no

www.aacrjournals.org Cancer Res; 72(23) December 1, 2012 OF3

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

Henrich et al.

other vascular analyzed. The translocation results in (70). Intriguingly, MYCN/MYC oncoproteins directly bind to a fusion gene encoding the N-terminus of WWTR1 fused in the BMI1 promoter and induce its transcription (70, 71), frame to the C-terminus of CAMTA1. This EHE-specific fusion suggesting a model where MYCN/MYC represses KIF1B via has the potential to (i) serve as a new marker for tumor BMI1-mediated epigenetic chromosome modification. Most detection, diagnosis, and monitoring; (ii) provide a highly MYCN-amplified neuroblastomas harbor 1p deletions indicat- specific therapeutic target; and (iii) act as a study model to ing that KIF1B expression may be inhibited by both reduced gather insights into the physiological functions of WWTR1 and copy number and amplified MYCN in this subgroup. A positive CAMTA1. The identification of a highly specific CAMTA1- regulator of KIF1Bb is the proapoptotic hydroxylase EGLN1, involving genomic rearrangement seen in virtually all tumors placing KIF1B in a pathway to eliminate excess neuroblasts of a single cancer entity further implicates CAMTA1 in cancer during embryonal development (64) that is likely to be impli- development. cated in the pathogenesis of neural crest-derived tumors such as neuroblastomas and pheochromocytomas (72). The iden- KIF1B tification of functionally impairing KIF1B mutations in neu- The KIF1B kinesin motor protein is involved in axon mye- roblastoma, pheochromocytoma, and medulloblastoma, lination and outgrowth as well as axonal transport of mito- together with KIF1Bb downregulation in advanced tumors chondria and synaptic vesicles (57–59). KIF1B maps to 1p36.22 and its inhibitory effect on cancer cells in vivo and in vitro, and encodes two alternatively spliced isoforms, KIF1Ba and support a tumor suppressive function for KIF1B that is linked KIF1Bb, conferring different axonal cargo specificity. KIF1B to its proapoptotic role. is one of 6 genes within a 500 Kb homozygous deletion found in a neuroblastoma cell line (Fig. 1; refs. 20, 60). Sequence CASZ1 analysis did not reveal mutations in oligodendrogliomas or a Castor zinc finger 1 (CASZ1), localized in 1p36.22, is the panel of pediatric solid tumor cell lines, including rhabdomyo- human homolog of the Drosophila zinc finger transcription sarcoma and Ewing sarcoma cells (61, 62). A missense variant factor Castor, which is expressed in a subset of central of unknown functional significance was detected in 6 of 100 nervous system neuroblasts and is involved in late stage neuroblastomas (63). Another mutation screen identified neurogenesis (73). CASZ1 maps near the border of a 3 Mb missense variants in three of 111 neuroblastomas, 2 of 52 SRO defined by integrating 1p deletions of neuroblastomas pheochromocytomas, and 1 of 14 medulloblastomas (64). and germ cell tumors (Fig. 1; ref. 21). Sequence analysis did Intriguingly, all variants identified in the latter study were not reveal evidence for tumor-specific CASZ1 mutations (74), shown to impair KIF1Bb function in vitro (64), and one of these but low CASZ1 expression is significantly correlated with loss-of-function variants was present in the germline of a three- unfavorable clinical and biologic features and poor overall generation cancer-prone family, segregating with predisposi- survival in neuroblastoma (75). Ectopic restoration of CASZ1 tion to pheochromocytoma, neuroblastoma, ganglioneuroma, enhanced cell adhesion, induced morphological differentia- and lung adenocarcinoma (65). This indicates that KIF1B tion, accompanied by expression of neuron-specificmarkers, sequence variants/mutations are infrequent but may be path- and inhibited migration, proliferation, and tumorigenicity ogenic in a subset of tumors, which is further supported by a (75). Furthermore, CASZ1 was increased in differentiating KIF1B single-nucleotide polymorphism (SNP) highly associat- neuroblastoma cells treated with retinoic acid or cAMP- ed with hepatitis virus B (HBV)-related hepatocellular carci- inducing agents (75, 76), which is in line with murine studies noma (66). KIF1B expression is significantly lower in advanced suggesting a developmental role for Casz1 in controlling neuroblastoma stages (63, 67, 68), as is KIF1B expression in neuronal subtype specification and differentiation (77). hepatocellular carcinomas from chronic HBV carriers com- Transcriptome analysis of CASZ1-overexpressing neuroblas- pared with tumor-adjacent tissue (66). toma cells revealed signatures consistent with CASZ1- Consistent with a tumor suppressive function, KIF1Bb induced phenotypes and a significant enrichment of genes induction triggers apoptosis in neuroblastoma cells, pheochro- involved in cell growth regulation and developmental pro- mocytoma cells, and rat sympathetic neurons (63, 64). KIF1Bb cesses (75). One means of CASZ1 downregulation in cancer knockdown in rat sympathetic neurons prevents apoptosis cells is genomic copy number loss, as indicated by lower following nerve growth factor (NGF) withdrawal, indicating CASZ1 expression in 1p-deleted tumors (75, 78). Evidence for that KIF1Bb plays a crucial role in neuronal apoptosis upon tumor-specific promoter CpG methylation has not been NGF limitation (64). KIF1Bb knockdown also enhances reported (74, 75), but CASZ1 induction by histone deacety- anchorage-independent colony formation and xenograft lase inhibitors in neuroblastoma cells (74, 75) suggests that tumor growth (63). Downregulation of KIF1B in advanced suppressive histone modifications inhibit CASZ1 expression. tumors can be mediated by heterozygous 1p loss, as indicated This idea is corroborated by the finding that the CASZ1 locus by significantly lower KIF1B levels in both gastrointestinal is repressed by the enhancer of zeste homolog 2 (EZH2) stromal tumors and neuroblastomas with heterozygous KIF1B Polycomb complex histone methyltransferase, an oncopro- deletion (63, 69). No evidence for methylation of KIF1B-asso- tein overexpressed in advanced tumors of various entities, ciated CpG islands was found in neuroblastomas (63, 67), but including neuroblastoma (79). Together, these data suggest chromatin remodeling mechanisms have been suggested to be that CASZ1 is a cell growth and differentiation regulator relevant as the BMI1 Polycomb group protein strongly that, when impaired by dosage reduction, contributes to the represses KIF1Bb by direct binding to the KIF1B promoter malignant phenotype of cancer cells.

OF4 Cancer Res; 72(23) December 1, 2012 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

1p36 Tumor Suppression—A Matter of Dosage?

miR-34a prostate carcinomas (80) and 74% of non–small cell lung MiRNAs are small noncoding RNAs involved in posttran- cancer samples (86). Moreover, miR-34a promoter methylation scriptional control of gene expression, and their deregulation was detected in melanoma (63%), colorectal cancer (74%), has been linked to a variety of diseases, including cancer. MiR- pancreatic cancer (64%), mammary cancer (60%), ovarian 34a, localized in 1p36.22 (Fig. 1), is ubiquitously expressed, with cancer (62%), urothelial cancer (71%), renal cell cancer highest levels in the brain (80). Aberrant miR-34a downregula- (58%), soft tissue sarcoma (68%), chronic lymphocytic leuke- tion has been detected in many cancer types, including breast mia (4%), multiple myeloma (6%), and non-Hodgkin lympho- cancer (81), epithelial ovarian cancer (82), prostate carcinoma ma (19%; refs. 80, 114, 115). An additional parameter affecting (80), pancreatic ductal adenocarcinoma (83), hepatocellular miR-34a expression is its genomic status, as indicated by carcinoma (84), colon cancer (85), non–small cell lung cancer significant association between lower miR-34a levels and (86), neuroblastoma (87), glioblastoma (88), malignant periph- 1p36 deletion in neuroblastomas (116, 117). In conclusion, eral nerve sheath tumors (89), melanoma (80), chronic lym- miR-34a acts as a pivotal element in the p53 tumor suppressive phocytic leukemia (90–92), and acute myeloid leukemia (93). In pathway, and its recurrent downregulation by a broad range of many cancer entities, low miR-34a expression is associated mechanisms in various malignancies together with its inhib- with advanced disease and/or poor patient survival, as seen in itory effect in cancer cell models suggests that aberrant reduc- neuroblastoma (87), epithelial ovarian cancer (82), peripheral tion of miR-34a levels contributes to cancer development. nerve sheath tumors (89), breast cancer (81), and pancreatic ductal adenocarcinoma (83). Ectopic miR-34a expression induces cell cycle arrest, apoptosis, and senescence, and inhi- Discussion bits migration and invasion of cancer cells (94). MiR-34a is It has been recognized early on that 1p36 harbors genetic directly induced by p53 (95–99) and plays a pivotal role within a information mediating tumor suppression. Here, we summa- p53-activating positive feedback loop where mirR-34a down- rize recent efforts substantiating the candidacy of CHD5, regulates the SIRT1 class III histone deacetylase, leading to CAMTA1, KIF1B, CASZ1, and miR-34a to be 1p36 genes con- accumulation of active acetylated p53 (100). Additional neg- tributing to tumor development when reduced in dosage. ative regulators of p53 (MTA2, HDAC1, and YY1) were iden- Cancer-specific mutations of these candidates are infrequent tified as mir-34a targets (101, 102), indicating the existence of but might play a role in a subset of tumors, as exemplified by several mir-34a-dependent mechanisms functioning within rare KIF1B mutations impairing its in vitro function and p53-activating feed back loops. Bioinformatic analyses and segregating with cancer predisposition (65). However, the global proteomic approaches indicate that regulation of hun- candidate genes seem to be impaired mainly on the transcrip- dreds of additional mir-34a targets contributes to mir-34a– tional rather than the genetic level. Each of them was reported associated cellular functions (101, 102). Validated direct targets to be aberrantly downregulated in one or more cancer entity, include factors involved in G1/S transition (E2F3, cyclin E2, and lower levels were associated with advanced disease and/or cyclin D1, CDK4, CDK6, MYC, MYCN), apoptosis (BCL2, Sur- poor patient survival. Compensation of their dosage in cancer vivin), metastatic potential (MET, AXL), Wnt signaling (WNT1, models via ectopic expression inhibited features of malignan- LEF1), and glycolysis (LDHA; discussed in refs. 94 and 102). cy, including tumorigenicity in xenografts, and low expression Other direct miR-34a targets play important roles in the Notch of all candidates was significantly associated with 1p deletion pathway (NOTCH1, NOTCH2, JAG1, DLL1; refs. 103–105), in at least one tumor type. This may indicate that their dose cancer stem cell functions (CD44; ref. 106), or growth factor reduction via single-copy loss compromises tumor suppressive signaling (ARAF, PIK3R2; ref. 107). Considering this broad functions and promotes cancer, as also seen for a growing spectrum of potentially oncogenic miR-34a targets, repression number of haploinsufficient tumor suppressor genes (30). of miR-34a expression is likely to create a selective advantage Multiple tumor suppressive genes simultaneously downregu- for cancer cells, also supported by miR-34a downregulation lated via genomic deletion might cooperate in an additive or during progressive carcinogenesis in a rat liver cancer model synergistic way, and targeting more than one component of a (108). Functional impairment of p53 downregulates miR-34a, pathway or regulatory loop could be a mechanism of circum- as seen upon p53 knockdown in vivo (99). In line with this, low venting redundant backup mechanisms that compensate for the miR-34a expression is significantly associated with either loss of single genes. Even within the limited set of 1p36 genes deleted or mutated p53 in various malignancies (88, 90– discussed here, potential interactions are found. It is tempting to 92, 109–111). In lymphocytic leukemia, a SNP within the speculate that a repressive effect of MYCN/MYC on KIF1B via promoter of the p53 inhibitor MDM2 is associated with higher BMI1 (70) may be inhibited by mir-34a impairing its direct target MDM2 levels and consequently lower miR-34a expression MYCN/MYC, thereby leading to an indirect activation of KIF1B (111). In HPV-induced cervical cancer, the E6 oncoprotein by miR-34a. In this scenario, 1p36 deletion would affect KIF1B destabilizes p53, resulting in miR-34a downregulation (112, both by direct copy number loss and downregulation of its 113). A p53-independent mechanism of miR-34a downregula- putative activator miR-34a. This cascade can be extended by tion is seen in acute myeloid leukemia, where the C/EBPa gene, adding p73, another 1p36 tumor suppressor candidate that has encoding a transcriptional activator of miR-34a, is mutated in been extensively reviewed elsewhere (118, 119). p73 drives 10% of the cases (93). Aberrant promoter CpG methylation is expression of miR-34a (120), so that 1p36 deletion would target another frequent mechanism of miR-34a inhibition, reported an p73/miR-34a/MYC(N)/BMI1/KIF1B axis at 3 levels (Fig. 1, with concomitant inhibition of expression in 79% of primary gray box). Another interaction level may be convergence of

www.aacrjournals.org Cancer Res; 72(23) December 1, 2012 OF5

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

Henrich et al.

signals downstream of 1p36 genes on identical pathways. In an inactivation by copy number-neutral gene-specific mutations, adequate cellular context, CHD5, CAMTA1, CASZ1,andmiR-34a copy number–dependent downregulation of 1p36 candidates all activate genetic programs implicated in neuronal function is likely to be accompanied by deregulation of a considerable and differentiation (31, 55, 75, 120). Expression of neuron- set of other cancer-related genes. These may be either targeted specific gene sets is associated with a markedly better prognosis by the same event (1p deletion) or genomic aberrations in both neuroblastomas and gliomas (68, 121, 122). A simulta- associated with 1p deletion. However, a large fraction of genes neous dosage-dependent impairment of the proneural regula- may passively reflect copy number change on the expression tors CHD5, CAMTA1, CASZ1,andmiR-34a via 1p36 deletion level without contributing to cancer development. Survival could shift the transcriptome toward dedifferentiation, thus, analyses adjusting for the respective copy number alteration contributing to neural tumor development. Taken together, may clarify whether the prognostic value of a gene's expression dosage-dependent 1p36 genes are likely to interact on more profile is independent of the underlying genomic alteration, as than one level to suppress malignancy. seen for CHD5 and CAMTA1 (32, 46). Such prognostic inde- Considering that most 1p deletions in human tumors extend pendency suggests that some tumors evolve additional, copy beyond 1p36 and that a certain fraction of genes reflects the number–independent mechanisms to regulate these genes, copy number loss on the expression level, genes from other 1p and identifying such mechanisms may strengthen the position regions may contribute to tumor suppression. Array-based of the respective candidates. expression analyses of 1p genes in oligodendrogliomas and A range of inhibitory mechanisms other than copy number neuroblastomas detected considerable copy number-depen- loss were identified for the 1p36 candidates discussed here. Mir- dent expression (123, 124). In neuroblastomas, 15% (124), 31% 34a is frequently downregulated by inactivation of upstream (125), and 61% (126) of all 1p genes were expressed significantly transcription factors, including p53 (88, 90–93, 109–111). Aber- lower in 1p-deleted tumors compared with tumors retaining rant promoter CpG methylation for miR-34a and CHD5 was 1p, being in favor of a strong impact of heterozygous loss on the observed in a broad range of tumors (37, 39, 40, 80, 86, 114, 115). 1p transcriptome. Thus, repression of CHD5, CAMTA1, KIF1B, Epigenetic mechanisms acting via histone modifications have CASZ1, and miR-34a via genomic loss in an individual tumor is also been linked to impairment of tumor suppressive activities likely to be accompanied by downregulation of a set of other, (134) and are likely to play a role in regulating CAMTA1 (55), potentially cancer-relevant genes, depending on extension and CASZ1 via EZH2 (79), KIF1B via BMI1 (70), and CHD5, acting nature of the 1p deletion. Other 1p genes could contribute to as a histone-interacting chromatin remodeler itself. tumor suppressive functions either by interaction with 1p36 In conclusion, the 1p36 genes CHD5, CAMTA1, KIF1B, genes or via independent routes. An example for an interre- CASZ1, and miR-34a may not necessarily require biallelic gional interaction of proximal 1p genes with 1p36 genes can be inactivation in a classic "two-hit" manner but contribute to proposed for CASZ1 regulation. EZH2, a potent suppressor of cancer development by partial dosage reduction via copy the CASZ1 locus (79) is a direct target of the 1p31.3-encoded number loss or other mechanisms, including epigenetic miR-101 (127), and a large terminal deletion including 1p31 inhibition. Functional studies indicate that these candidates would affect both CASZ1 and its indirect activator, miR-101 cooperate to suppress tumorigenesis. Their codeletion may (Fig. 1, gray box). A model where genes mapping to proximal 1p be one way for a developing cancer cell to acquire selective add to the tumor suppressive effect of 1p36 genes is further advantage by inhibiting an antioncogenic network at differ- supported by the observation that neuroblastomas with large ent positions in a single event.Aberrantexpressionofother terminal deletions are more aggressive than neuroblastomas dosage-dependent cancer-relevant genes on 1p or from with small deletions confined to 1p36 (128). Interaction of associated copy number alterations in other chromosomes cancer-relevant genes whose expression follows copy number is likely to further contribute to this selective advantage. In aberrations is certainly not limited to 1p, considering that 1p such a setting, deletion mapping and SRO identification will deletion is associated with other genomic aberrations in most not lead to identification of a single tumor suppressor gene tumors. Expression of a substantial fraction of genes is altered that is completely inactivated by a second hit, but may guide consistently with the underlying genomic changes in a variety the identification of a minimum gene set, whose reduction is of malignancies, including colon cancer (129), prostate cancer required for tumorigenesis in a certain cellular context. (130), glioblastoma (131), neuroblastoma (125, 126), and mul- Human genetics alone can deliver definitive proof of biallelic tiple myeloma (132). An interchromosomal interaction of inactivation of a classic tumor suppressor gene, but unequiv- dosage-dependent genes can also be illustrated by the p73/ ocal support for the dosage dependency of a tumor sup- miR-34a/MYC(N)/BMI1/KIF1B axis. Besides KIF1B, BMI1 sup- pressor requires other lines of evidence, including mouse presses CADM1 (70), a candidate for haploinsufficient tumor models. An elegant example of a tight correlation between suppression mapping to 11q23. This region is recurrently lost tumor suppressor expression level and its functionality was in a broad range of solid tumors and hematological malignan- shown by the generation of an allelic series of genetically cies (133). Thus, the putative interaction cascade can be engineered mice expressing varying levels of the Pten tumor extended to p73/miR-34a/MYC(N)/BMI1/KIF1B-CADM1 and, suppressor (135). Surprisingly, even in animals with a subtle accordingly, might be targeted by copy number–dependent 20% reduction of the normal Pten level, tumor incidence was gene deregulation via 1p36 deletion (p73, miR-34a, KIF1B), increased and survival was decreased, with the wild-type 11q23 deletion (CADM1), and/or MYC(N) amplification (Fig. allele remaining fully functional in all induced tumors 1, gray box). Collectively, in contrast to tumor suppressor (135). A similar mouse-based assay may clarify whether a

OF6 Cancer Res; 72(23) December 1, 2012 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

1p36 Tumor Suppression—A Matter of Dosage?

comparable dosage dependency can be modeled for the Authors' Contributions candidate genes discussed here. Conception and design: K.-O. Henrich, M. Schwab, F. Westermann Development of methodology: K.-O. Henrich, M. Schwab In contrast to genetic inactivation of tumor suppressor Acquisition of data (provided animals, acquired and managed patients, genes by mutation, inhibitory mechanisms acting on the provided facilities, etc.): K.-O. Henrich, M. Schwab, F. Westermann expression level are in principle reversible as long as an intact Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): K.-O. Henrich, M. Schwab, F. Westermann allele is present. This may pave the way for intervention Writing, review, and/or revision of the manuscript: K.-O. Henrich, strategies that restore expression of dosage-sensitive tumor M. Schwab, F. Westermann suppressor genes. The genes discussed in this review are Study supervision: K.-O. Henrich, M. Schwab, F. Westermann targeted by aberrant epigenetic mechanisms, at least in a subset of tumors, and recent advancements in depicting the Grant Support Plus enzymatic processes controlling the cancer epigenome should BMBF: NGFN #01GS0896 (K.-O. Henrich, M. Schwab, and F. Westermann), MYC-NET, CancerSys #0316076A (F. Westermann), EU (FP7): ASSET #259348 (F. open doors for developing new therapeutic approaches (134). Westermann).

Disclosure of Potential Conflicts of Interest Received June 5, 2012; revised July 6, 2012; accepted July 18, 2012; No potential conflicts of interest were disclosed. published OnlineFirst November 20, 2012.

References 1. Schwab M, Praml C, Amler LC. Genomic instability in 1p and human 15. Tanaka K, Yanoshita R, Konishi M, Oshimura M, Maeda Y, Mori T, malignancies. Genes Chromosomes Cancer 1996;16:211–29. et al. Suppression of tumourigenicity in human colon carcinoma cells 2. Bagchi A, Mills AA. The quest for the 1p36 tumor suppressor. Cancer by introduction of normal chromosome 1p36 region. Oncogene Res 2008;68:2551–6. 1993;8:2253–8. 3. Laureys G, Speleman F, Opdenakker G, Benoit Y, Leroy J. Consti- 16. Brodeur GM, Sekhon G, Goldstein MN. Chromosomal aberrations in tutional translocation t(1;17)(p36;q12–21) in a patient with neuroblas- human neuroblastomas. Cancer 1977;40:2256–63. toma. Genes Chromosomes Cancer 1990;2:252–4. 17. Caron H, Spieker N, Godfried M, Veenstra M, van Sluis P, de Kraker J, 4. Biegel JA, White PS, Marshall HN, Fujimori M, Zackai EH, Scher CD, et al. Chromosome bands 1p35–36 contain two distinct neuroblas- et al. Constitutional 1p36 deletion in a child with neuroblastoma. Am toma tumor suppressor loci, one of which is imprinted. Genes J Hum Genet 1993;52:176–82. Chromosomes Cancer 2001;30:168–74. 5. Maris JM, Weiss MJ, Guo C, Gerbing RB, Stram DO, White PS, et al. 18. Bauer A, Savelyeva L, Claas A, Praml C, Berthold F, Schwab M. Loss of heterozygosity at 1p36 independently predicts for disease Smallest region of overlapping deletion in 1p36 in human neuroblas- progression but not decreased overall survival probability in neuro- toma: a 1 Mbp cosmid and PAC contig. Genes Chromosomes Cancer blastoma patients: a Children's Cancer Group study. J Clin Oncol 2001;31:228–39. 2000;18:1888–99. 19. White PS, Thompson PM, Gotoh T, Okawa ER, Igarashi J, Kok M, 6. Ragnarsson G, Eiriksdottir G, Johannsdottir JT, Jonasson JG, Egils- et al. Definition and characterization of a region of 1p36.3 consistently son V, Ingvarsson S. Loss of heterozygosity at chromosome 1p in deleted in neuroblastoma. Oncogene 2005;24:2684–94. different solid human tumours: association with survival. Br J Cancer 20. Ohira M, Kageyama H, Mihara M, Furuta S, Machida T, Shishikura T, 1999;79:1468–74. et al. Identification and characterization of a 500-kb homozygously 7. Utada Y, Emi M, Yoshimoto M, Kasumi F, Akiyama F, Sakamoto G, deleted region at 1p36.2-p36.3 in a neuroblastoma cell line. Onco- et al. Allelic loss at 1p34–36 predicts poor prognosis in node-negative gene 2000;19:4302–7. breast cancer. Clin Cancer Res 2000;6:3193–8. 21. Ejeskar K, Sjoberg RM, Abel F, Kogner P, Ambros PF, Martinsson T. 8. Ogunbiyi OA, Goodfellow PJ, Gagliardi G, Swanson PE, Birnbaum Fine mapping of a tumour suppressor candidate gene region in EH, Fleshman JW, et al. Prognostic value of chromosome 1p allelic 1p36.2–3, commonly deleted in neuroblastomas and germ cell loss in colon cancer. Gastroenterology 1997;113:761–6. tumours. Med Pediatr Oncol 2001;36:61–6. 9. Kambara T, Sharp GB, Nagasaka T, Takeda M, Sasamoto H, Naka- 22. Bagchi A, Papazoglu C, Wu Y, Capurso D, Brodt M, Francis D, et al. gawa H, et al. Allelic loss of a common microsatellite marker MYCL1: CHD5 is a tumor suppressor at human 1p36. Cell 2007;128:459–75. a useful prognostic factor of poor outcomes in colorectal cancer. Clin 23. Barbashina V, Salazar P, Holland EC, Rosenblum MK, Ladanyi M. Cancer Res 2004;10:1758–63. Allelic losses at 1p36 and 19q13 in gliomas: correlation with histologic 10. Di Vinci A, Infusini E, Peveri C, Risio M, Rossini FP, Giaretti W. classification, definition of a 150-kb minimal deleted region on 1p36, Deletions at chromosome 1p by fluorescence in situ hybridization and evaluation of CAMTA1 as a candidate tumor suppressor gene. are an early event in human colorectal tumorigenesis. Gastroenter- Clin Cancer Res 2005;11:1119–28. ology 1996;111:102–7. 24. Felsberg J, Erkwoh A, Sabel MC, Kirsch L, Fimmers R, Blaschke B, 11. Munn KE, Walker RA, Varley JM. Frequent alterations of chromo- et al. Oligodendroglial tumors: refinement of candidate regions on some 1 in ductal carcinoma in situ of the breast. Oncogene 1995;10: chromosome arm 1p and correlation of 1p/19q status with survival. 1653–7. Brain Pathol 2004;14:121–30. 12. Kuroki T, Fujiwara Y, Tsuchiya E, Nakamori S, Imaoka S, Kanematsu 25. Edstrom Elder E, Nord B, Carling T, Juhlin C, Backdahl M, Hoog A, T, et al. Accumulation of genetic changes during development and et al. Loss of heterozygosity on the short arm of chromosome 1 in progression of hepatocellular carcinoma: loss of heterozygosity of pheochromocytoma and abdominal paraganglioma. World J Surg chromosome arm 1p occurs at an early stage of hepatocarcinogen- 2002;26:965–71. esis. Genes Chromosomes Cancer 1995;13:163–7. 26. Poetsch M, Dittberner T, Woenckhaus C. Microsatellite analysis at 13. Williams AC, Harper SJ, Paraskeva C. Neoplastic transformation of a 1p36.3 in malignant melanoma of the skin: fine mapping in search of a human colonic epithelial cell line: in vitro evidence for the adenoma to possible tumour suppressor gene region. Melanoma Res 2003;13: carcinoma sequence. Cancer Res 1990;50:4724–30. 29–33. 14. Bader SA, Fasching C, Brodeur GM, Stanbridge EJ. Dissociation of 27. Girard L, Zochbauer-Muller S, Virmani AK, Gazdar AF, Minna JD. suppression of tumorigenicity and differentiation in vitro effected by Genome-wide allelotyping of lung cancer identifies new regions of transfer of single human chromosomes into human neuroblastoma allelic loss, differences between small cell lung cancer and non-small cells. Cell Growth Differ 1991;2:245–55. cell lung cancer, and loci clustering. Cancer Res 2000;60:4894–906.

www.aacrjournals.org Cancer Res; 72(23) December 1, 2012 OF7

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

Henrich et al.

28. Bieche I, Khodja A, Lidereau R. Deletion mapping of chromosomal 49. Henrich KO, Claas A, Praml C, Benner A, Mollenhauer J, Poustka A, region 1p32-pter in primary breast cancer. Genes Chromosomes et al. Allelic variants of CAMTA1 and FLJ10737 within a commonly Cancer 1999;24:255–63. deleted region at 1p36 in neuroblastoma. Eur J Cancer 2007;43: 29. Thorstensen L, Qvist H, Heim S, Liefers GJ, Nesland JM, Giercksky 607–16. KE, et al. Evaluation of 1p losses in primary carcinomas, local 50. Schraivogel D, Weinmann L, Beier D, Tabatabai G, Eichner A, Zhu JY, recurrences and peripheral metastases from colorectal cancer et al. CAMTA1 is a novel tumour suppressor regulated by miR-9/9() patients. Neoplasia 2000;2:514–22. in glioblastoma stem cells. EMBO J 2011;30:4309–22. 30. Berger AH, Knudson AG, Pandolfi PP. A continuum model for tumour 51. Asgharzadeh S, Pique-Regi R, Sposto R, Wang H, Yang Y, Shimada suppression. Nature 2011;476:163–9. H, et al. Prognostic significance of gene expression profiles of 31. Potts RC, Zhang P, Wurster AL, Precht P, Mughal MR, Wood WH 3rd, metastatic neuroblastomas lacking MYCN gene amplification. J Natl et al. CHD5, a brain-specific paralog of Mi2 chromatin remodeling Cancer Inst 2006;98:1193–203. enzymes, regulates expression of neuronal genes. PLoS One 2011;6: 52. Oberthuer A, Berthold F, Warnat P, Hero B, Kahlert Y, Spitz R, et al. e24515. Customized oligonucleotide microarray gene expression-based 32. Fujita T, Igarashi J, Okawa ER, Gotoh T, Manne J, Kolla V, et al. CHD5, classification of neuroblastoma patients outperforms current clinical a tumor suppressor gene deleted from 1p36.31 in neuroblastomas. J risk stratification. J Clin Oncol 2006;24:5070–8. Natl Cancer Inst 2008;100:940–9. 53. Vermeulen J, De Preter K, Naranjo A, Vercruysse L, Van Roy N, 33. VanMaerken T, Vandesompele J, Rihani A, De Paepe A, Speleman F. Hellemans J, et al. Predicting outcomes for children with neuroblas- Escape from p53-mediated tumor surveillance in neuroblastoma: toma using a multigene-expression signature: a retrospective SIO- switching off the p14(ARF)-MDM2-p53 axis. Cell Death Differ 2009; PEN/COG/GPOH study. Lancet Oncol 2009;10:663–71. 16:1563–72. 54. Warnat P, Oberthuer A, Fischer M, Westermann F, Eils R, Brors B. 34. Okawa ER, Gotoh T, Manne J, Igarashi J, Fujita T, Silverman KA, et al. Cross-study analysis of gene expression data for intermediate neu- Expression and sequence analysis of candidates for the 1p36.31 roblastoma identifies two biological subtypes. BMC Cancer 2007; tumor suppressor gene deleted in neuroblastomas. Oncogene 2008; 7:89. 27:803–10. 55. Henrich KO, Bauer T, Schulte J, Ehemann V, Deubzer H, Gogolin S, 35. Maser RS, Choudhury B, Campbell PJ, Feng B, Wong KK, Proto- et al. CAMTA1, a 1p36 tumor suppressor candidate, inhibits growth popov A, et al. Chromosomally unstable mouse tumours have geno- and activates differentiation programs in neuroblastoma cells. Can- mic alterations similar to diverse human cancers. Nature 2007;447: cer Res 2011;71:3142–51. 966–71. 56. Errani C, Zhang L, Sung YS, Hajdu M, Singer S, Maki RG, et al. A novel 36. Robbins CM, Tembe WA, Baker A, Sinari S, Moses TY, Beckstrom- WWTR1-CAMTA1 gene fusion is a consistent abnormality in epithe- Sternberg S, et al. Copy number and targeted mutational analysis lioid hemangioendothelioma of different anatomic sites. Genes Chro- reveals novel somatic events in metastatic prostate tumors. Genome mosomes Cancer 2011;50:644–53. Res 2011;21:47–55. 57. Nangaku M, Sato-Yoshitake R, Okada Y, Noda Y, Takemura R, 37. Gorringe KL, Choong DY, Williams LH, Ramakrishna M, Sridhar A, Qiu Yamazaki H, et al. KIF1B, a novel microtubule plus end-directed W, et al. Mutation and methylation analysis of the chromodomain- monomeric motor protein for transport of mitochondria. Cell 1994;79: helicase-DNA binding 5 gene in ovarian cancer. Neoplasia 2008;10: 1209–20. 1253–8. 58. Lyons DA, Naylor SG, Scholze A, Talbot WS. Kif1b is essential for 38. Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, et al. mRNA localization in oligodendrocytes and development of myelin- The consensus coding sequences of human breast and colorectal ated axons. Nat Genet 2009;41:854–8. cancers. Science 2006;314:268–74. 59. Zhao C, Takita J, Tanaka Y, Setou M, Nakagawa T, Takeda S, et al. 39. Mulero-Navarro S, Esteller M. Chromatin remodeling factor CHD5 is Charcot-Marie-Tooth disease type 2A caused by mutation in a silenced by promoter CpG island hypermethylation in human cancer. microtubule motor KIF1Bbeta. Cell 2001;105:587–97. Epigenetics 2008;3:210–5. 60. Yang HW, Chen YZ, Takita J, Soeda E, Piao HY, Hayashi Y. Genomic 40. Wang X, Lau KK, So LK, Lam YW. CHD5 is down-regulated through structure and mutational analysis of the human KIF1B gene which is promoter hypermethylation in gastric cancer. J Biomed Sci 2009; homozygously deleted in neuroblastoma at chromosome 1p36.2. 16:95. Oncogene 2001;20:5075–83. 41. Thompson PM, Gotoh T, Kok M, White PS, Brodeur GM. CHD5, a new 61. Alonso ME, Bello MJ, Arjona D, Gonzalez-Gomez P, Aminoso C, member of the chromodomain gene family, is preferentially Lopez-Marin I, et al. Mutational study of the 1p located genes expressed in the nervous system. Oncogene 2003;22:1002–11. p18ink4c, Patched-2, RIZ1 and KIF1B in oligodendrogliomas. Oncol 42. Garcia I, Mayol G, Rodriguez E, Sunol M, Gershon TR, Rios J, et al. Rep 2005;13:539–42. Expression of the neuron-specific protein CHD5 is an independent 62. Chen YY, Takita J, Chen YZ, Yang HW, Hanada R, Yamamoto K, et al. marker of outcome in neuroblastoma. Mol Cancer 2010;9:277. Genomic structure and mutational analysis of the human KIF1Balpha 43. Bouche N, Scharlat A, Snedden W, Bouchez D, Fromm H. A novel gene located at 1p36.2 in neuroblastoma. Int J Oncol 2003;23:737– family of calmodulin-binding transcription activators in multicellular 44. organisms. J Biol Chem 2002;277:21851–61. 63. Munirajan AK, Ando K, Mukai A, Takahashi M, Suenaga Y, Ohira M, 44. Henrich KO. CAMTA1 (calmodulin binding transcription activator 1). et al. KIF1Bbeta functions as a haploinsufficient tumor suppressor Atlas Genet Cytogenet Oncol Haematol 2011;15:441–2. gene mapped to chromosome 1p36.2 by inducing apoptotic cell 45. Tanas MR, Sboner A, Oliveira AM, Erickson-Johnson MR, Hespelt J, death. J Biol Chem 2008;283:24426–34. Hanwright PJ, et al. Identification of a disease-defining gene fusion in 64. Schlisio S, Kenchappa RS, Vredeveld LC, George RE, Stewart R, epithelioid hemangioendothelioma. Sci Transl Med 2011;3:98ra82. Greulich H, et al. The kinesin KIF1Bbeta acts downstream from EglN3 46. Henrich KO, Fischer M, Mertens D, Benner A, Wiedemeyer R, Brors B, to induce apoptosis and is a potential 1p36 tumor suppressor. Genes et al. Reduced expression of CAMTA1 correlates with adverse out- Dev 2008;22:884–93. come in neuroblastoma patients. Clin Cancer Res 2006;12:131–8. 65. Yeh IT, Lenci RE, Qin Y, Buddavarapu K, Ligon AH, Leteurtre E, et al. A 47. Ichimura K, Vogazianou AP, Liu L, Pearson DM, Backlund LM, Plant germline mutation of the KIF1B beta gene on 1p36 in a family with K, et al. 1p36 is a preferential target of chromosome 1 deletions in neural and nonneural tumors. Hum Genet 2008;124:279–85. astrocytic tumours and homozygously deleted in a subset of glio- 66. Zhang H, Zhai Y, Hu Z, Wu C, Qian J, Jia W, et al. Genome-wide blastomas. Oncogene 2008;27:2097–108. association study identifies 1p36.22 as a new susceptibility locus for 48. Kim MY, Yim SH, Kwon MS, Kim TM, Shin SH, Kang HM, et al. hepatocellular carcinoma in chronic hepatitis B virus carriers. Nat Recurrent genomic alterations with impact on survival in colorectal Genet 2010;42:755–8. cancer identified by genome-wide array comparative genomic 67. Caren H, Ejeskar K, Fransson S, Hesson L, Latif F, Sjoberg RM, et al. hybridization. Gastroenterology 2006;131:1913–24. A cluster of genes located in 1p36 are down-regulated in

OF8 Cancer Res; 72(23) December 1, 2012 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

1p36 Tumor Suppression—A Matter of Dosage?

neuroblastomas with poor prognosis, but not due to CpG island 86. Gallardo E, Navarro A, Vinolas N, Marrades RM, Diaz T, Gel B, et al. methylation. Mol Cancer 2005;4:10. miR-34a as a prognostic marker of relapse in surgically resected non- 68. Ohira M, Morohashi A, Inuzuka H, Shishikura T, Kawamoto T, small-cell lung cancer. Carcinogenesis 2009;30:1903–9. Kageyama H, et al. Expression profiling and characterization of 87. Welch C, Chen Y, Stallings RL. MicroRNA-34a functions as a poten- 4200 genes cloned from primary neuroblastomas: identification of tial tumor suppressor by inducing apoptosis in neuroblastoma cells. 305 genes differentially expressed between favorable and unfavor- Oncogene 2007;26:5017–22. able subsets. Oncogene 2003;22:5525–36. 88. Li Y, Guessous F, Zhang Y, Dipierro C, Kefas B, Johnson E, et al. 69. Astolfi A, Nannini M, Pantaleo MA, Di Battista M, Heinrich MC, Santini MicroRNA-34a inhibits glioblastoma growth by targeting multiple D, et al. A molecular portrait of gastrointestinal stromal tumors: an . Cancer Res 2009;69:7569–76. integrative analysis of gene expression profiling and high-resolution 89. Subramanian S, Thayanithy V, West RB, Lee CH, Beck AH, Zhu S, genomic copy number. Lab Invest 2010;90:1285–94. et al. Genome-wide transcriptome analyses reveal p53 inactivation 70. Ochiai H, Takenobu H, Nakagawa A, Yamaguchi Y, Kimura M, Ohira mediated loss of miR-34a expression in malignant peripheral nerve M, et al. Bmi1 is a MYCN target gene that regulates tumorigenesis sheath tumours. J Pathol 2010;220:58–70. through repression of KIF1Bbeta and TSLC1 in neuroblastoma. 90. Dijkstra MK, van Lom K, Tielemans D, Elstrodt F, Langerak AW, van 't Oncogene 2010;29:2681–90. Veer MB, et al. 17p13/TP53 deletion in B-CLL patients is associated 71. Huang R, Cheung NK, Vider J, Cheung IY, Gerald WL, Tickoo SK, with microRNA-34a downregulation. Leukemia 2009;23:625–7. et al. MYCN and MYC regulate tumor proliferation and tumorigenesis 91. Mraz M, Malinova K, Kotaskova J, Pavlova S, Tichy B, Malcikova J, directly through BMI1 in human neuroblastomas. FASEB J 2011;25: et al. miR-34a, miR-29c and miR-17–5p are downregulated in CLL 4138–49. patients with TP53 abnormalities. Leukemia 2009;23:1159–63. 72. Lee S, Nakamura E, Yang H, Wei W, Linggi MS, Sajan MP, et al. 92. Zenz T, Mohr J, Eldering E, Kater AP, Buhler A, Kienle D, et al. miR-34a Neuronal apoptosis linked to EglN3 prolyl hydroxylase and familial as part of the resistance network in chronic lymphocytic leukemia. pheochromocytoma genes: developmental culling and cancer. Blood 2009;113:3801–8. Cancer Cell 2005;8:155–67. 93. Pulikkan JA, Peramangalam PS, Dengler V, Ho PA, Preudhomme C, 73. Mellerick DM, Kassis JA, Zhang SD, Odenwald WF. Castor encodes a Meshinchi S, et al. C/EBPalpha regulated microRNA-34a targets novel zinc finger protein required for the development of a subset of E2F3 during granulopoiesis and is down-regulated in AML with CNS neurons in Drosophila. Neuron 1992;9:789–803. CEBPA mutations. Blood 2010;116:5638–49. 74. Caren H, Fransson S, Ejeskar K, Kogner P, Martinsson T. Genetic and 94. Hermeking H. The miR-34 family in cancer and apoptosis. Cell Death epigenetic changes in the common 1p36 deletion in neuroblastoma Differ 2010;17:193–9. tumours. Br J Cancer 2007;97:1416–24. 95. Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, 75. Liu Z, Yang X, Li Z, McMahon C, Sizer C, Barenboim-Stapleton L, Lee KH, et al. Transactivation of miR-34a by p53 broadly influences et al. CASZ1, a candidate tumor-suppressor gene, suppresses neu- gene expression and promotes apoptosis. Mol Cell 2007;26:745–52. roblastoma tumor growth through reprogramming gene expression. 96. He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, et al. A microRNA Cell Death Differ 2011;18:1174–83. component of the p53 tumour suppressor network. Nature 2007;447: 76. Liu Z, Yang X, Tan F, Cullion K, Thiele CJ. Molecular cloning and 1130–4. characterization of human Castor, a novel human gene upregulated 97. Raver-Shapira N, Marciano E, Meiri E, Spector Y, Rosenfeld N, during cell differentiation. Biochem Biophys Res Commun 2006;344: Moskovits N, et al. Transcriptional activation of miR-34a contributes 834–44. to p53-mediated apoptosis. Mol Cell 2007;26:731–43. 77. Vacalla CM, Theil T. Cst, a novel mouse gene related to Drosophila 98. Tarasov V, Jung P, Verdoodt B, Lodygin D, Epanchintsev A, Menssen Castor, exhibits dynamic expression patterns during neurogenesis A, et al. Differential regulation of microRNAs by p53 revealed by and heart development. Mech Dev 2002;118:265–8. massively parallel sequencing: miR-34a is a p53 target that induces 78. Fransson S, Martinsson T, Ejeskar K. Neuroblastoma tumors with apoptosis and G1-arrest. Cell Cycle 2007;6:1586–93. favorable and unfavorable outcomes: significant differences in mRNA 99. Bommer GT, Gerin I, Feng Y, Kaczorowski AJ, Kuick R, Love RE, et al. expression of genes mapped at 1p36.2. Genes Chromosomes p53-mediated activation of miRNA34 candidate tumor-suppressor Cancer 2007;46:45–52. genes. Curr Biol 2007;17:1298–307. 79. WangC,LiuZ,WooCW,LiZ,WangL,WeiJS,etal.EZH2 100. Yamakuchi M, Ferlito M, Lowenstein CJ. miR-34a repression of mediates epigenetic silencing of neuroblastoma suppressor SIRT1 regulates apoptosis. Proc Natl Acad Sci U S A 2008;105: genes CASZ1, CLU, RUNX3 and NGFR. Cancer Res 2012;72: 13421–6. 315–24. 101. Chen QR, Yu LR, Tsang P, Wei JS, Song YK, Cheuk A, et al. 80. Lodygin D, Tarasov V, Epanchintsev A, Berking C, Knyazeva T, Systematic proteome analysis identifies transcription factor YY1 as Korner H, et al. Inactivation of miR-34a by aberrant CpG methylation a direct target of miR-34a. J Proteome Res 2011;10:479–87. in multiple types of cancer. Cell Cycle 2008;7:2591–600. 102. Kaller M, Liffers ST, Oeljeklaus S, Kuhlmann K, Roh S, Hoffmann R, 81. Peurala H, Greco D, Heikkinen T, Kaur S, Bartkova J, Jamshidi M, et al. Genome-wide characterization of miR-34a induced changes in et al. MiR-34a expression has an effect for lower risk of protein and mRNA expression by a combined pulsed SILAC and and associates with expression patterns predicting clinical outcome microarray analysis. Mol Cell Proteomics 2011;10:M111 010462. in breast cancer. PLoS One 2011;6:e26122. 103. de Antonellis P, Medaglia C, Cusanelli E, Andolfo I, Liguori L, De Vita 82. Corney DC, Hwang CI, Matoso A, Vogt M, Flesken-Nikitin A, Godwin G, et al. MiR-34a targeting of Notch ligand delta-like 1 impairs AK, et al. Frequent downregulation of miR-34 family in human ovarian CD15þ/CD133þ tumor-propagating cells and supports neural dif- cancers. Clin Cancer Res 2010;16:1119–28. ferentiation in medulloblastoma. PLoS One 2011;6:e24584. 83. Jamieson NB, Morran DC, Morton JP, Ali A, Dickson EJ, Carter CR, 104. Guessous F, Zhang Y, Kofman A, Catania A, Li Y, Schiff D, et al. et al. MicroRNA molecular profi les associated with diagnosis, clin- microRNA-34a is tumor suppressive in brain tumors and glioma stem icopathologic criteria, and overall survival in patients with Resectable cells. Cell Cycle 2010;9:1031–6. Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2012;18: 105. Pang RT, Leung CO, Ye TM, Liu W, Chiu PC, Lam KK, et al. Micro- 534–45. RNA-34a suppresses invasion through downregulation of Notch1 84. Li N, Fu H, Tie Y, Hu Z, Kong W, Wu Y, et al. miR-34a inhibits migration and Jagged1 in cervical carcinoma and choriocarcinoma cells. and invasion by down-regulation of c-Met expression in human Carcinogenesis 2010;31:1037–44. hepatocellular carcinoma cells. Cancer Lett 2009;275:44–53. 106. Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, et al. The 85. Tazawa H, Tsuchiya N, Izumiya M, Nakagama H. Tumor-suppressive microRNA miR-34a inhibits prostate cancer stem cells and metas- miR-34a induces senescence-like growth arrest through modulation tasis by directly repressing CD44. Nat Med 2011;17:211–5. of the E2F pathway in human colon cancer cells. Proc Natl Acad Sci 107. Lal A, Thomas MP, Altschuler G, Navarro F, O'Day E, Li XL, et al. U S A 2007;104:15472–7. Capture of microRNA-bound mRNAs identifies the tumor suppressor

www.aacrjournals.org Cancer Res; 72(23) December 1, 2012 OF9

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

Henrich et al.

miR-34a as a regulator of growth factor signaling. PLoS Genet seminated neuroblastoma with favorable and unfavorable outcome. 2011;7:e1002363. Clin Cancer Res 2006;12:5118–28. 108. Tryndyak VP, Ross SA, Beland FA, Pogribny IP. Down-regulation of 122. Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, the microRNAs miR-34a, miR-127, and miR-200b in rat liver during et al. Molecular subclasses of high-grade glioma predict prognosis, hepatocarcinogenesis induced by a methyl-deficient diet. Mol Car- delineate a pattern of disease progression, and resemble stages in cinog 2009;48:479–87. neurogenesis. Cancer Cell 2006;9:157–73. 109. Zenz T, Habe S, Denzel T, Mohr J, Winkler D, Buhler A, et al. Detailed 123. Mukasa A, Ueki K, Matsumoto S, Tsutsumi S, Nishikawa R, Fujimaki analysis of p53 pathway defects in fludarabine-refractory chronic T, et al. Distinction in gene expression profiles of oligodendrogliomas lymphocytic leukemia (CLL): dissecting the contribution of 17p dele- with and without allelic loss of 1p. Oncogene 2002;21:3961–8. tion, TP53 mutation, p53-p21 dysfunction, and miR34a in a prospec- 124. Janoueix-Lerosey I, Novikov E, Monteiro M, Gruel N, Schleiermacher tive clinical trial. Blood 2009;114:2589–97. G, Loriod B, et al. Gene expression profiling of 1p35–36 genes in 110. Mraz M, Pospisilova S, Malinova K, Slapak I, Mayer J. MicroRNAs in neuroblastoma. Oncogene 2004;23:5912–22. chronic lymphocytic leukemia pathogenesis and disease subtypes. 125. Lastowska M, Viprey V, Santibanez-Koref M, Wappler I, Peters H, Leuk Lymphoma 2009;50:506–9. Cullinane C, et al. Identification of candidate genes involved in 111. Asslaber D, Pinon JD, Seyfried I, Desch P, Stocher M, Tinhofer I, et al. neuroblastoma progression by combining genomic and expression microRNA-34a expression correlates with MDM2 SNP309 polymor- microarrays with survival data. Oncogene 2007;26:7432–44. phism and treatment-free survival in chronic lymphocytic leukemia. 126. Wang Q, Diskin S, Rappaport E, Attiyeh E, Mosse Y, Shue D, et al. Blood 2010;115:4191–7. Integrative genomics identifies distinct molecular classes of neuro- 112. Wang X, Wang HK, McCoy JP, Banerjee NS, Rader JS, Broker TR, blastoma and shows that multiple genes are targeted by regional et al. Oncogenic HPV infection interrupts the expression of tumor- alterations in DNA copy number. Cancer Res 2006;66:6050–62. suppressive miR-34a through viral oncoprotein E6. RNA 2009;15: 127. Varambally S, Cao Q, Mani RS, Shankar S, Wang X, Ateeq B, et al. 637–47. Genomic loss of microRNA-101 leads to overexpression of histone 113. Li B, Hu Y, Ye F, Li Y, Lv W, Xie X. Reduced miR-34a expression in methyltransferase EZH2 in cancer. Science 2008;322:1695–9. normal cervical tissues and cervical lesions with high-risk human 128. Takeda O, Homma C, Maseki N, Sakurai M, Kanda N, Schwab M, papillomavirus infection. Int J Gynecol Cancer 2010;20:597–604. et al. There may be two tumor suppressor genes on chromosome arm 114. Chim CS, Wong KY, Qi Y, Loong F, Lam WL, Wong LG, et al. 1p closely associated with biologically distinct subtypes of neuro- Epigenetic inactivation of the miR-34a in hematological malignan- blastoma. Genes Chromosomes Cancer 1994;10:30–9. cies. Carcinogenesis 2010;31:745–50. 129. Tsafrir D, Bacolod M, Selvanayagam Z, Tsafrir I, Shia J, Zeng Z, et al. 115. Vogt M, Munding J, Gruner M, Liffers ST, Verdoodt B, Hauk J, et al. Relationship of gene expression and chromosomal abnormalities in Frequent concomitant inactivation of miR-34a and miR-34b/c by colorectal cancer. Cancer Res 2006;66:2129–37. CpG methylation in colorectal, pancreatic, mammary, ovarian, 130. Wolf M, Mousses S, Hautaniemi S, Karhu R, Huusko P, Allinen M, urothelial, and renal cell carcinomas and soft tissue sarcomas. et al. High-resolution analysis of gene copy number alterations in Virchows Arch 2011;458:313–22. human prostate cancer using CGH on cDNA microarrays: impact of 116. Cole KA, Attiyeh EF, Mosse YP, Laquaglia MJ, Diskin SJ, Brodeur copy number on gene expression. Neoplasia 2004;6:240–7. GM, et al. A functional screen identifies miR-34a as a candidate 131. Nigro JM, Misra A, Zhang L, Smirnov I, Colman H, Griffin C, et al. neuroblastoma tumor suppressor gene. Mol Cancer Res 2008;6: Integrated array-comparative genomic hybridization and expression 735–42. array profiles identify clinically relevant molecular subtypes of glio- 117. Feinberg-Gorenshtein G, Avigad S, Jeison M, Halevy-Berco G, Mar- blastoma. Cancer Res 2005;65:1678–86. doukh J, Luria D, et al. Reduced levels of miR-34a in neuroblastoma 132. Walker BA, Leone PE, Jenner MW, Li C, Gonzalez D, Johnson DC, are not caused by mutations in the TP53 binding site. Genes Chro- et al. Integration of global SNP-based mapping and expression arrays mosomes Cancer 2009;48:539–43. reveals key regions, mechanisms, and genes important in the path- 118. Rufini A, Agostini M, Grespi F, Tomasini R, Sayan BS, Niklison-Chirou ogenesis of multiple myeloma. Blood 2006;108:1733–43. MV, et al. p73 in Cancer. Genes Cancer 2011;2:491–502. 133. Knuutila S, Aalto Y, Autio K, Bjorkqvist AM, El-Rifai W, Hemmer S, 119. Oswald C, Stiewe T. In good times and bad: p73 in cancer. Cell Cycle et al. DNA copy number losses in human . Am J Pathol 2008;7:1726–31. 1999;155:683–94. 120. Agostini M, Tucci P, Killick R, Candi E, Sayan BS, Rivetti di Val Cervo 134. Baylin SB, Jones PA. A decade of exploring the cancer epigenome – P, et al. Neuronal differentiation by TAp73 is mediated by microRNA- biological and translational implications. Nat Rev Cancer 2011;11: 34a regulation of synaptic protein targets. Proc Natl Acad Sci U S A 726–34. 2011;108:21093–8. 135. Alimonti A, Carracedo A, Clohessy JG, Trotman LC, Nardella C, Egia 121. Fischer M, Oberthuer A, Brors B, Kahlert Y, Skowron M, Voth H, et al. A, et al. Subtle variations in Pten dose determine cancer suscepti- Differential expression of neuronal genes defines subtypes of dis- bility. Nat Genet 2010;42:454–8.

OF10 Cancer Res; 72(23) December 1, 2012 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 20, 2012; DOI: 10.1158/0008-5472.CAN-12-2230

1p36 Tumor Suppression−−A Matter of Dosage?

Kai-Oliver Henrich, Manfred Schwab and Frank Westermann

Cancer Res Published OnlineFirst November 20, 2012.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-12-2230

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2012/11/19/0008-5472.CAN-12-2230. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research.