CUX1 is an animal gene. The name stands for Cut like homeobox 1. The term "cut" derives from the "cut wing" phenotype observed in a mutant of Drosophila melanogaster.[1] In mammals, a CCAAT-displacement activity was originally described in DNA binding assays.[2] The human gene was identified following purification of the CCAAT-displacement protein (CDP) and has been successively been called CDP, Cut-like 1 (CUTL1), CDP/Cut and finally, CUX1.[3] (Consequently, a literature search for CUX1 fails to bring up many of the earlier studies). Cut homeobox genes are present in all metazoans. In mammals, CUX1 is expressed ubiquitously in all tissues. A second gene, called CUX2, is expressed primarily in neuronal cells.[4]

Reviews on CUX1 edit

Multiple reviews have detailed the tissue-specific functions and genetic interactions in Drosophila and the early characterization of the CDP in mammalian cells,[5] expression and activity of CUX1 in the myeloid cell lineage,[6] multiple CUX1 isoforms, modes of DNA binding, early mouse models and cell-based assays,[7][8] roles of CUX1 in kidney development and homeostasis,[9] mechanisms by which CUX1 stimulates cell migration and invasion,[10] roles of CUX1 and CUX2 in neurons of the cortex upper layer[11][12] and the paradoxical implications of CUX1 in cancer.[13] In addition, a number of commentaries present more specific views and speculations.[14][15][16][17]

Genomic structure edit

The human CUX1 gene is large, encompassing more than 440,000 base pairs with two alternative first exons and an additional 23 exons.[18][19] The last exon has a weak polyadenylation site allowing RNA polymerase II often to continue transcribing until it reaches an additional 10 exons. Splicing of this longer transcript from exon 14 to exon 25 generates a mature mRNA that codes for a protein that was called CASP (Cut alternatively spliced product).[20] CASP localizes to the Golgi and does not seem to impact at all on CUX1 function.[21] However, because of the complex structure of the gene, most oligos in microarrays were derived from the most 3' exons that are unique to CASP. Thus, until the advent of RNA sequencing CUX1 expression data has been essentially limited to immunohistochemical analyses. Similarly, many guide RNAs in CRISPR-Cas screening studies target the CASP-specific exons and do not affect CUX1.

CUX1 protein isoforms, evolutionarily conserved domains and functional regions edit

The use of two transcription start sites, alternative splicing of some exons and proteolytic processing combine to generate several CUX1 protein isoforms[3][22][23][24][25][26][27] (reviewed[7]) The full-length protein, often referred to as p200 CUX1, contains five evolutionarily conserved domains: a coiled-coil (CC), three Cut domains (C1, C2 and C3), originally called Cut repeats (CRs), and a Cut homeodomain (HD).[3][28] The coiled-coil's function remains to be defined. The three Cut domains and the Cut homeodomain were originally characterized as DNA binding domains,[29][30][31][32] and were later found to be involved also in protein-protein interactions.[33][34][35][36] In addition to these conserved domains, the n-terminal 100 amino acids contains an autoinhibitory domain, while the carboxy-terminal region downstream of the homeodomain was shown to function as an active repression domain.[37][38] In mid to late G1, the full-length protein is proteolytically processed to generate a shorter isoform, p110 CUX1 that lacks approximately the n-terminal 747 amino acids and thus contains only 3 DNA binding domains: CUT domains 2 and 3 and the Cut homeodomain (C2C3HD).

DNA binding assays with histidine-tagged fusion proteins showed that one Cut domain is not sufficient for DNA binding, while several combination of domains were found to bind to DNA with distinct affinities and kinetics: CR1CR2, CR3HD and CR2CR3HD.[39] The C1C2 protein displayed very rapid "on" and "off" DNA binding rates, whereas any combination of a Cut domain with the Cut homeodomain exhibited slower binding kinetics.[39] Interestingly, the full-length CUX1 protein purified from insect cells exhibited DNA binding kinetics similar to that of CR1CR2, suggesting that the Cut homeodomain may not be active in the context of the full-length protein.[39]

Biochemical and cellular activities of p200 CUX1 edit

p200 CUX1 is a very abundant protein that binds to DNA with extremely fast kinetics (rapid "on" and "off" rates).[39] This is not consistent with a role as a classical transcription factor that binds stably to DNA and recruits a co-activator or a co-repressor, however, the p200 CUX1 protein may still be able to repress transcription by competition for binding site occupancy.[37] Indeed, CUX1 was originally purified as the CCAAT-displacement protein (CDP).[3][40] In addition to this potential role in transcriptional repression, recent studies revealed that p200 CUX1 functions as an auxiliary factor in base excision repair. In vitro, CUT domains of CUX1 stimulate the enzymatic activities of the 8-oxoguanine DNA glycosylase (OGG1) and the apurinic/apyrimidinic endonuclease 1 (APE1).[35][36][41][42] In agreement with results from in vitro DNA repair assays, a p200 CUX1-GFP fusion protein is rapidly recruited to DNA damage generated by laser microirradiation,[42] while CUX1 knockdown in human cells causes a delay in the repair of oxidative DNA damage and mono-alkylated bases in genomic DNA, and sensitizes cancer cells to treatments with H2O2,[35][41] ionizing radiation[42] and the mono-alkylating agent temozolomide.[36] Conversely, p200 CUX1 overexpression increases the resistance of cancer cells to the same treatments.[35][36][41][42] Structure/function analysis established that a fusion protein containing Cut domains 1 and 2 linked to a nuclear localization signal (C1C2-NLS) is rapidly recruited to DNA damage and is sufficient to accelerate the repair oxidative DNA damage and mono-alkylated bases in genomic DNA.[36][42] Since the C1C2-NLS protein is devoid of transcription activation potential,[35][42] these results suggest that Cut domains are directly involved in DNA repair.

Transcriptional and cellular activities of p110 CUX1 edit

The shorter p110 CUX1 isoform stably interacts with DNA and can function as transcriptional repressor or activator depending on promoter context.[43][44] Transcription and cell-based assays demonstrated a role for p110 CUX1 in stimulating cell cycle progression and cell proliferation,[24][45] strengthening of the spindle assembly checkpoint,[46] ensuring an efficient DNA damage response,[47] promoting cell migration and invasion,[44][48] and increasing resistance to apoptotic signals.[49] Of note, the p200 CUX1 protein was inactive in all these transcriptional and cell-based assays.

p110 CUX1 has been demonstrated to bind the protective allele of FTO single nucleotide polymorphisms (SNPs) rs8050136 or rs1421085 highly associated with human obesity, and promote the expression of RPGRIP1L.[50][51][52] RPGRIP1L hypomorphism in mice results in obesity,[53][54] suggesting an important role of CUX1 in body weight regulation.

CUX1 overexpression in human cancers edit

CUX1 is overexpressed in many cancers.[55] The comprehensive molecular characterization of human colon and rectal cancer performed by The Cancer Genome Atlas (TCGA) ranked CUX1 as the fifth gene on a scale showing a correlation between tumour aggressiveness and gene expression/somatic copy number alterations.[56] TCGA and REMBRANDT data also show shorter survival of glioblastoma patients with high CUX1 mRNA expression[57] (reviewed[36]). In smaller scale studies, immunohistochemical analyses on breast, pancreas and glioblastoma cancers reveal that CUX1 expression inversely correlates with relapse-free and overall survival.[36][48][49][58] An alternative CUX1 transcript that is initiated within intron 20 and codes for a p75 isoform is expressed specifically in the testis and thymus.[22][25] This transcript was found to be aberrantly expressed in many breast tumour cells lines and breast tumours.[22] Transgenic mice expressing this transcript in mammary epithelial cells were shown to develop mammary tumours with metastasis to the lung.[59]

Tumors in CUX1 transgenic mice edit

CUX1 transgenic mice develop tumors in multiple organs and tissues after a long latency period.[41][46][59][60][61][62] Transgenic mice expressing either p75, p110 or p200 CUX1 in mammary epithelial cells develop mammary tumours with a low penetrance (~20%) and with a long latency period.[41][59] Metastasis to the lung was observed in three cases of mammary tumors expressing p75 CUX1.[59]

The mechanisms of action differ widely between p110 CUX1 and p200 CUX1. Transgenic mice expressing p110 CUX1 in mammary epithelial cells develop mammary tumors that exhibit a sub-tetraploid chromosome number, suggesting passage through tetraploidy resulting from cytokinesis failure.[46][59] Cells do not usually survive as tetraploid with multiple centrosomes. Overexpression of p110 CUX1 was shown to activate a transcriptional program that reinforces the spindle assembly checkpoint and delays mitosis until extranumerary centrosomes have clustered to two poles, thereby enabling bipolar mitosis and survival of tetraploid cells. Yet, passage through a multipolar intermediate enriches for merotelic chromosome attachments, leading to chromosome mis-segregation and the rapid generation of aneuploid populations from which tumorigenic cells emerge.[46]

Tumors that develop in p200 CUX1 transgenic mice reveal a different mode of action, as 44% of these tumors harboured a spontaneous mutation activating the Kras gene.[41] The cooperation between RAS and p200 CUX1 was confirmed by lentiviral infections in the lung.[41] RAS oncogenes do not transform primary cells, but instead cause cellular senescence. This results from the elevated production of reactive oxygen species (ROS) that lead to oxidative DNA damage. Biochemical and cell-based assays demonstrated that the CUT domains within CUX1 and other Cut domain proteins stimulate the enzymatic activities of some enzymes of the base excision repair pathway and accelerate the repair of oxidative DNA damage.[35][36][41][42][63][64] Hence, human and primary rodent fibroblasts that are transfected with KRAS and CUX1 exhibit similar ROS levels as cells that receive only KRAS but show much reduced DNA damage.[41] In agreement with these findings, CUX1 knockdown is synthetic lethal in all cancer cells exhibiting high levels of ROS as a consequence of activating mutations in either KRAS, HRAS, BRAF or EGFR.[41][42]

Genetic studies defining CUX1 as a haploinsufficient tumor suppressor edit

Loss-of-heterozygosity (LOH) of the 7q22.1 chromosomal region, where CUX1 resides, was reported in 8–22% of various cancer,[65][66][67][68] and in close to 50% in therapy-associated leukemias.[69][70] As no inactivating mutations were found in the remaining allele,[71][72][73][74] the notion that CUX1 could be the tumor suppressor gene on 7q22.1 was dismissed for some time. However, refined mapping in leiomyomas and myeloid leukemias eventually pointed to CUX1 as the sole tumor suppressor gene in this region.[74][75][76][77][78][79] These genetic data suggest that CUX1 may be a haploinsufficient tumor suppressor gene. In addition to LOH events, large scale DNA sequencing identified inactivating point mutations in 1–5% of cancers where both alleles are present.[80] There is no tumor case where both CUX1 allele are inactivated.

What is the tumor suppressor function of CUX1? edit

While the biochemical activities of CUX1 that explain its role in tumor maintenance and progression have been well defined, the biochemical functions of CUX1 involved in tumor suppression have yet to be firmly established. Two mechanisms have been proposed. One study reported that CUX1 functions as a transcriptional activator of PIK3IP1, a gene that codes for the phosphoinositide-3-kinase interacting protein 1 (PIK3IP1), a direct inhibitor of the PI3K p110 catalytic subunit.[80][81] CUX1 knockdown caused a decrease in PIK3IP1 expression that was associated with an increase in PI3K signaling and AKT signaling.[80] A separate study, however, argued that CUX1 is itself a target of AKT signaling.[49] Activation of the PI3K–AKT signaling pathway by insulin-like growth factor 1 (IGF1) or by AKT2 overexpression led to the upregulation of CUX1 and was associated with resistance to apoptosis, whereas treatment of cells with the PI3K inhibitor LY294002 decreased CUX1 expression and increased apoptosis.[49] Whether the discrepancies in the results and conclusions from these two studies can be explained by differences in cell-types or other reasons remains to be verified. The auxiliary role of CUX1 in base excision repair has raised the possibility that its tumor suppressor function may be linked to its function in DNA repair. CUX1 knockdown delays DNA repair in multiple cell lines. Mouse embryo fibroblasts (MEFs) derived from a Cux1-/- knockout mouse exhibit increased genomic instability23, moreover Cux1+/−- heterozygous MEFs are haploinsufficient for DNA repair18. Yet, whether CUX1 hemizygosity augments the risk of cancer by increasing the frequency of mutations and/or genomic rearrangements remains to be formally tested.

References edit

  1. ^ Blanc R. "The production of wing scalloping in Drosophila melanogaster". University of California Publications in Zoology 1942, 49:1–31.
  2. ^ Barberis A, Superti-Furga G, and Busslinger M. "Mutually exclusive interaction of the CCAAT-binding factor and of a displacement protein with overlapping sequences of a histone gene promoter". Cell 1987, 50(3):347–59.
  3. ^ a b c d Neufeld EJ, Skalnik DG, Lievens PM, and Orkin SH. "Human CCAAT displacement protein is homologous to the Drosophila homeoprotein, cut". Nature Genetics 1992, 1(1):50–5.
  4. ^ Quaggin SE, Vandenheuvel GB, Golden K, Bodmer R, and Igarashi P. "Primary Structure, Neural-Specific Expression, and Chromosomal Localization Of Cux-2, a Second Murine Homeobox Gene Related to Drosophila Cut". Journal of Biological Chemistry 1996, 271(37):22624–34.
  5. ^ Nepveu A. "Role of the multifunctional CDP/Cut/Cux homeodomain transcription factor in regulating differentiation, cell growth and development". Gene 2001, 270(1–2):1–15.
  6. ^ Skalnik DG. "Transcriptional mechanisms regulating myeloid-specific genes". Gene 2002, 284(1–2):1–21.
  7. ^ a b Sansregret L, and Nepveu A. "The multiple roles of CUX1: Insights from mouse models and cell-based assays". Gene 2008, 412(1–2):84–94.
  8. ^ Hulea L, and Nepveu A. "CUX1 transcription factors: from biochemical activities and cell-based assays to mouse models and human diseases". Gene 2012, 497(1):18–26.
  9. ^ Alcalay NI, and Vanden Heuvel GB. "Regulation of cell proliferation and differentiation in the kidney". Frontiers in Bioscience 2009, 14:4978–91.
  10. ^ Kedinger V, and Nepveu A. "The roles of CUX1 homeodomain proteins in the establishment of a transcriptional program required for cell migration and invasion". Cell Adhesion & Migration 2010, 4(3):348–52.
  11. ^ Cubelos B, and Nieto M. "Intrinsic programs regulating dendrites and synapses in the upper layer neurons of the cortex". Communicative & Integrative Biology 2010, 3(6):483–6.
  12. ^ Weiss LA, and Nieto M. "The crux of Cux genes in neuronal function and plasticity". Brain Research 2019, 1705:32–42.
  13. ^ Ramdzan ZM, and Nepveu A. "CUX1, a haploinsufficient tumour suppressor gene overexpressed in advanced cancers". Nature Reviews Cancer 2014, 14(10):673–82.
  14. ^ Chapman HA. "Cathepsins as transcriptional activators? Developmental Cell 2004, 6(5):610–1.
  15. ^ Goulet B, and Nepveu A. "Complete and Limited Proteolysis in Cell Cycle Progression". Cell Cycle 2004, 3(8):986–9.
  16. ^ Michl P, and Downward J. "CUTL1: a key mediator of TGFbeta-induced tumor invasion". Cell Cycle 2006, 5(2):132–4.
  17. ^ Sansregret L, and Nepveu A. "Gene signatures of genomic instability as prognostic tools for breast cancer". Future Oncology 2011, 7(5):591–4.
  18. ^ "UCSC Genome Browser on Human Dec. 2013 (GRCh38/hg38) Assembly". University of California, Santa Cruz.
  19. ^ Zeng RW, Soucie E, Sung Moon N, Martin-Soudant N, Bérubé G, Leduy L, and Nepveu A. "Exon/intron structure and alternative transcripts of the CUTL1 gene". Gene 2000, 241(1):75–85.
  20. ^ Lievens PM, Tufarelli C, Donady JJ, Stagg A, and Neufeld EJ. "CASP, a novel, highly conserved alternative-splicing product of the CDP/cut/cux gene, lacks cut-repeat and homeo DNA-binding domains, and interacts with full-length CDP in vitro". Gene 1997, 197(1–2):73–81.
  21. ^ Gillingham AK, Pfeifer AC, and Munro S. "CASP, the alternatively spliced product of the gene encoding the CCAAT-displacement protein transcription factor, is a Golgi membrane protein related to giantin". Molecular Biology of the Cell 2002, 13(11):3761–74.
  22. ^ a b c Goulet B, Watson P, Poirier M, Leduy L, Berube G, Meterissian S, ... and Nepveu A. "Characterization of a tissue-specific CDP/Cux isoform, p75, activated in breast tumor cells". Cancer Research 2002, 62(22):6625–33.
  23. ^ Moon NS, Premdas P, Truscott M, Leduy L, Berube G, and Nepveu A. "S Phase-Specific Proteolytic Cleavage Is Required to Activate Stable DNA Binding by the CDP/Cut Homeodomain Protein". Molecular and Cellular Biology 2001, 21:6332–45.
  24. ^ a b Truscott M, Denault JB, Goulet B, Leduy L, Salvesen GS, and Nepveu A. "Carboxyl-terminal proteolytic processing of CUX1 by a caspase enables transcriptional activation in proliferating cells". Journal of Biological Chemistry 2007, 282(41):30216–26.
  25. ^ a b Vandenheuvel GB, Quaggin SE, and Igarashi P. "A Unique Variant Of a Homeobox Gene Related to Drosophila Cut Is Expressed In Mouse Testis". Biology of Reproduction 1996, 55(4):731–9.
  26. ^ Maitra U, Seo J, Lozano MM, and Dudley JP. "Differentiation-induced cleavage of Cutl1/CDP generates a novel dominant-negative isoform that regulates mammary gene expression". Molecular and Cellular Biology 2006, 26(20):7466–78.
  27. ^ "CUX1 cut like homeobox 1 [ Homo sapiens (human) ]". NCBI.
  28. ^ Blochlinger K, Bodmer R, Jack J, Jan LY, and Jan YN. "Primary structure and expression of a product from cut, a locus involved in specifying sensory organ identity in Drosophila". Nature 1988, 333(6174):629–35.
  29. ^ Harada R, Dufort D, Denis-Larose C, and Nepveu A. "Conserved cut repeats in the human cut homeodomain protein function as DNA binding domains". Journal of Biological Chemistry 1994, 269(3):2062–7.
  30. ^ Harada R, Berube G, Tamplin OJ, Denis-Larose C, and Nepveu A. "DNA-binding specificity of the cut repeats from the human cut-like protein". Molecular and Cellular Biology 1995, 15(1):129–40.
  31. ^ Aufiero B, Neufeld EJ, and Orkin SH. "Sequence-specific DNA binding of individual Cut repeats of the human CCAAT displacement/Cut homeodomain protein". Proceedings of the National Academy of Sciences of the United States of America 1994, 91:7757–61.
  32. ^ Andres V, Chiara MD, and Mahdavi V. "A new bipartite DNA-binding domain: cooperative interaction between the cut repeat and homeo domain of the cut homeo proteins". Genes & Development 1994, 8(2):245-57.
  33. ^ Santaguida M, Ding Q, Berube G, Truscott M, Whyte P, and Nepveu A. "Phosphorylation of the CCAAT displacement protein (CDP)/Cux transcription factor by cyclin A-Cdk1 modulates its DNA binding activity in G(2)". Journal of Biological Chemistry 2001, 276(49):45780–90.
  34. ^ Santaguida M, and Nepveu A. "Differential regulation of CDP/Cux p110 by cyclin A/Cdk2 and cyclin A/Cdk1". Journal of Biological Chemistry 2005, 280(38):32712–21.
  35. ^ a b c d e f Ramdzan ZM, Pal R, Kaur S, Leduy L, Berube G, Davoudi S, ... and Nepveu A. "The function of CUX1 in oxidative DNA damage repair is needed to prevent premature senescence of mouse embryo fibroblasts. Oncotarget 2015, 6(6):3613–26.
  36. ^ a b c d e f g h Kaur S, Ramdzan ZM, Guiot MC, Li L, Leduy L, Ramotar D, ... and Nepveu A. "CUX1 Stimulates APE1 Enzymatic Activity and Increases the Resistance of Glioblastoma Cells to the Mono-Alkylating Agent, Temozolomide". Neuro-oncology 2018, 20(4):484–93.
  37. ^ a b Mailly F, Berube G, Harada R, Mao PL, Phillips S, and Nepveu A. "The human cut homeodomain protein can repress gene expression by two distinct mechanisms: active repression and competition for binding site occupancy". Molecular and Cellular Biology 1996, 16(10):5346–57.
  38. ^ Truscott M, Raynal L, Wang Y, Berube G, Leduy L, and Nepveu A. "The N-terminal region of the CCAAT displacement protein (CDP)/Cux transcription factor functions as an autoinhibitory domain that modulates DNA binding". Journal of Biological Chemistry 2004, 279(48):49787–94.
  39. ^ a b c d Moon NS, Berube G, and Nepveu A. "CCAAT displacement activity involves Cut repeats 1 and 2, not the Cut homeodomain". Journal of Biological Chemistry 2000, 275(40):31325–34.
  40. ^ Skalnik DG, Strauss EC, and Orkin SH. "CCAAT displacement protein as a repressor of the myelomonocytic-specific gp91-phox gene promoter". Journal of Biological Chemistry 1991, 266:16736–44.
  41. ^ a b c d e f g h i j Ramdzan ZM, Vadnais C, Pal R, Vandal G, Cadieux C, Leduy L, ... and Nepveu A. "RAS Transformation Requires CUX1-Dependent Repair of Oxidative DNA Damage". PLOS Biology 2014, 12(3):e1001807.
  42. ^ a b c d e f g h Ramdzan ZM, Ginjala V, Pinder JB, Chung D, Donovan CM, Kaur S, ... and Nepveu A. "The DNA repair function of CUX1 contributes to radioresistance". Oncotarget 2017, 8(12):19021–38.
  43. ^ Harada R, Vadnais C, Sansregret L, Leduy L, Berube G, Robert F, and Nepveu A. "Genome-wide location analysis and expression studies reveal a role for p110 CUX1 in the activation of DNA replication genes". Nucleic Acids Research 2008, 36(1):189–202.
  44. ^ a b Kedinger V, Sansregret L, Harada R, Vadnais C, Cadieux C, Fathers K, ... and Nepveu A. "p110 CUX1 homeodomain protein stimulates cell migration and invasion in part through a regulatory cascade culminating in the repression of E-cadherin and occludin". Journal of Biological Chemistry 2009, 284(40):27701–11.
  45. ^ Sansregret L, Goulet B, Harada R, Wilson B, Leduy L, Bertoglio J, and Nepveu A. "The p110 isoform of the CDP/Cux transcription factor accelerates entry into S phase". Molecular and Cellular Biology 2006, 26(6):2441–55.
  46. ^ a b c d Sansregret L, Vadnais C, Livingstone J, Kwiatkowski N, Awan A, Cadieux C, ... and Nepveu A. "Cut homeobox 1 causes chromosomal instability by promoting bipolar division after cytokinesis failure". Proceedings of the National Academy of Sciences of the United States of America 2011, 108(5):1949–54.
  47. ^ Vadnais C, Davoudi S, Afshin M, Harada R, Dudley R, Clermont PL, ... and Nepveu A. "CUX1 transcription factor is required for optimal ATM/ATR-mediated responses to DNA damage". Nucleic Acids Research 2012, 40(10):4483–95.
  48. ^ a b Michl P, Ramjaun AR, Pardo OE, Warne PH, Wagner M, Poulsom R, ... and Downward J. "CUTL1 is a target of TGF(beta) signaling that enhances cancer cell motility and invasiveness". Cancer Cell 2005, 7(6):521–32.
  49. ^ a b c d Ripka S, Neesse A, Riedel J, Bug E, Aigner A, Poulsom R, ... and Michl P. "CUX1: target of Akt signalling and mediator of resistance to apoptosis in pancreatic cancer". Gut 2010, 59(8):1101–10.
  50. ^ Stratigopoulos, George; Padilla, Stephanie L.; LeDuc, Charles A.; Watson, Elizabeth; Hattersley, Andrew T.; McCarthy, Mark I.; Zeltser, Lori M.; Chung, Wendy K.; Leibel, Rudolph L. (2008-04-01). "Regulation of Fto/Ftm gene expression in mice and humans". American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 294 (4): R1185–R1196. doi:10.1152/ajpregu.00839.2007. ISSN 0363-6119. PMC 2808712. PMID 18256137.
  51. ^ Stratigopoulos, George; LeDuc, Charles A.; Cremona, Maria L.; Chung, Wendy K.; Leibel, Rudolph L. (January 2011). "Cut-like Homeobox 1 (CUX1) Regulates Expression of the Fat Mass and Obesity-associated and Retinitis Pigmentosa GTPase Regulator-interacting Protein-1-like (RPGRIP1L) Genes and Coordinates Leptin Receptor Signaling". Journal of Biological Chemistry. 286 (3): 2155–2170. doi:10.1074/jbc.m110.188482. ISSN 0021-9258. PMC 3023512. PMID 21037323.
  52. ^ Stratigopoulos, George; Leibel, Rudolph L. (December 2010). "FTO gains function". Nature Genetics. 42 (12): 1038–1039. doi:10.1038/ng1210-1038. ISSN 1546-1718. PMC 3818900. PMID 21102620.
  53. ^ Stratigopoulos, George; Martin Carli, Jayne F.; O’Day, Diana R.; Wang, Liheng; LeDuc, Charles A.; Lanzano, Patricia; Chung, Wendy K.; Rosenbaum, Michael; Egli, Dieter; Doherty, Daniel A.; Leibel, Rudolph L. (May 2014). "Hypomorphism for RPGRIP1L, a Ciliary Gene Vicinal to the FTO Locus, Causes Increased Adiposity in Mice". Cell Metabolism. 19 (5): 767–779. doi:10.1016/j.cmet.2014.04.009. ISSN 1550-4131. PMC 4131684. PMID 24807221.
  54. ^ Stratigopoulos, George; Burnett, Lisa Cole; Rausch, Richard; Gill, Richard; Penn, David Barth; Skowronski, Alicja A.; LeDuc, Charles A.; Lanzano, Anthony J.; Zhang, Pumin; Storm, Daniel R.; Egli, Dieter (2016-05-02). "Hypomorphism of Fto and Rpgrip1l causes obesity in mice". The Journal of Clinical Investigation. 126 (5): 1897–1910. doi:10.1172/JCI85526. ISSN 0021-9738. PMC 4855930. PMID 27064284.
  55. ^ "GRCh38 · COSMIC v90". Catalogue Of Somatic Mutations In Cancer.
  56. ^ Network TCGA. "Comprehensive molecular characterization of human colon and rectal cancer". Nature 2012, 487(7407):330–7.
  57. ^ The Cancer Genome Atlas Research N. "Comprehensive genomic characterization defines human glioblastoma genes and core pathways". Nature 2008, 455(7216):1061–8.
  58. ^ Ripka S, Konig A, Buchholz M, Wagner M, Sipos B, Kloppel G, ... and Michl P. WNT5A--target of CUTL1 and potent modulator of tumor cell migration and invasion in pancreatic cancer". Carcinogenesis 2007, 28(6):1178–87.
  59. ^ a b c d e Cadieux C, Kedinger V, Yao L, Vadnais C, Drossos M, Paquet M, and Nepveu A. "Mouse mammary tumor virus p75 and p110 CUX1 transgenic mice develop mammary tumors of various histologic types". Cancer Research 2009, 69(18):7188–97.
  60. ^ Cadieux C, Fournier S, Peterson AC, Bedard C, Bedell BJ, and Nepveu A. "Transgenic mice expressing the p75 CCAAT-displacement protein/Cut homeobox isoform develop a myeloproliferative disease-like myeloid leukemia". Cancer Research 2006, 66(19):9492–501.
  61. ^ Cadieux C, Harada R, Paquet M, Cote O, Trudel M, Nepveu A, and Bouchard M. "Polycystic kidneys caused by sustained expression of Cux1 isoform p75". Journal of Biological Chemistry 2008, 283(20):13817–24.
  62. ^ Siam R, Harada R, Cadieux C, Battat R, Vadnais C, and Nepveu A. "Transcriptional activation of the Lats1 tumor suppressor gene in tumors of CUX1 transgenic mice". Molecular Cancer 2009, 8:60–70.
  63. ^ Pal R, Ramdzan ZM, Kaur S, Duquette PM, Marcotte R, Leduy L, ... and Nepveu A. "CUX2 Functions As an Accessory Factor in the Repair of Oxidative DNA Damage". Journal of Biological Chemistry 2015, 290(37):22520–31.
  64. ^ Kaur S, Coulombe Y, Ramdzan ZM, Leduy L, Masson JY, and Nepveu A. "Special AT-rich Sequence-binding Protein 1 (SATB1) Functions as an Accessory Factor in Base Excision Repair". Journal of Biological Chemistry 2016, 291(43):22769–80.
  65. ^ Ozisik YY, Meloni AM, Surti U, and Sandberg AA. "Deletion 7q22 in uterine leiomyoma. A cytogenetic review". Cancer Genetics & Cytogenetics 1993, 71(1):1–6.
  66. ^ Zeng WR, Scherer SW, Koutsilieris M, Huizenga JJ, Filteau F, Tsui LC, and Nepveu A. "Loss Of Heterozygosity and Reduced Expression Of the Cutl1 Gene In Uterine Leiomyomas". Oncogene 1997, 14(19):2355–65.
  67. ^ Zeng WR, Watson P, Lin J, Jothy S, Lidereau R, Park M, and Nepveu A. "Refined mapping of the region of loss of heterozygosity on the long arm of chromosome 7 in human breast cancer defines the location of a second tumor suppressor gene at 7q22 in the region of the CUTL1 gene". Oncogene 1999, 18(11):2015–21.
  68. ^ Pedersen-Bjergaard J, Andersen MT, and Andersen MK. "Genetic pathways in the pathogenesis of therapy-related myelodysplasia and acute myeloid leukemia". Hematology / the Education Program of the American Society of Hematology American Society of Hematology Education Program 2007:392–7.
  69. ^ Zhang Y, and Rowley JD. "Chromatin structural elements and chromosomal translocations in leukemia". DNA Repair 2006, 5:1282–97.
  70. ^ Smith SM, Le Beau MM, Huo D, Karrison T, Sobecks RM, Anastasi J, ... and Larson RA. "Clinical-cytogenetic associations in 306 patients with therapy-related myelodysplasia and myeloid leukemia: the University of Chicago series". Blood 2003, 102(1):43–52.
  71. ^ Hindersin S, Niemeyer CM, Germing U, Göbel U, and Kratz CP. "Mutation analysis of CUTL1 in childhood myeloid neoplasias with monosomy 7". Leukemia Research 2007, 31:1323–4.
  72. ^ Patrikis M, Bryan E, Thomas N, Rice G, Quinn M, Baker M, and Campbell I. "Mutation analysis of CDP, TP53, and KRAS in uterine leiomyomas". Molecular Carcinogenesis 2003, 37:61–4.
  73. ^ Moon NS, Rong Zeng W, Premdas P, Santaguida M, Berube G, and Nepveu A. "Expression of N-terminally truncated isoforms of CDP/CUX is increased in human uterine leiomyomas". International Journal of Cancer 2002, 100(4):429–32.
  74. ^ a b Thoennissen NH, Krug UO, Lee DH, Kawamata N, Iwanski GB, Lasho T, ... and "Koeffler HP. Prevalence and prognostic impact of allelic imbalances associated with leukemic transformation of Philadelphia chromosome-negative myeloproliferative neoplasms". Blood 2010, 115(14):2882–90.
  75. ^ Schoenmakers EF, Bunt J, Hermers L, Schepens M, Merkx G, Janssen B, ... and van Kessel AG. "Identification of CUX1 as the recurrent chromosomal band 7q22 target gene in human uterine leiomyoma". Genes, Chromosomes & Cancer 2013, 52(1):11–23.
  76. ^ Klampfl T, Harutyunyan A, Berg T, Gisslinger B, Schalling M, Bagienski K, ... and Kralovics R. "Genome integrity of myeloproliferative neoplasms in chronic phase and during disease progression". Blood 2011, 118:167–76.
  77. ^ Thoennissen NH, Lasho T, Thoennissen GB, Ogawa S, Tefferi A, and Koeffler HP. "Novel CUX1 missense mutation in association with 7q- at leukemic transformation of MPN". American Journal of Hematology 2011, 86(8):703–5.
  78. ^ Jerez A, Sugimoto Y, Makishima H, Verma A, Jankowska AM, Przychodzen B, ... and Maciejewski JP. "Loss of heterozygosity in 7q myeloid disorders: clinical associations and genomic pathogenesis". Blood 2012, 119(25):6109–17.
  79. ^ McNerney ME, Brown CD, Wang X, Bartom ET, Karmakar S, Bandlamudi C, ... and White KP. "CUX1 is a haploinsufficient tumor suppressor gene on chromosome 7 frequently inactivated in acute myeloid leukemia". Blood 2013, 121(6):975–83.
  80. ^ a b c Wong CC, Martincorena I, Rust AG, Rashid M, Alifrangis C, Alexandrov LB, ... and Adams DJ. "Inactivating CUX1 mutations promote tumorigenesis". Nature Genetics 2014, 46(1):33–8.
  81. ^ He X, Zhu Z, Johnson C, Stoops J, Eaker AE, Bowen W, and DeFrances MC. "PIK3IP1, a negative regulator of PI3K, suppresses the development of hepatocellular carcinoma". Cancer Research 2008, 68(14):5591–8.