Skip to main content

Fez1/Lzts1 a new mitotic regulator implicated in cancer development

Abstract

Considerable evidence has accumulated suggesting that cancer has genetic origin, based on the development of genomic alterations, such as deletions, mutations, and/or methylations in critical genes for homeostasis of cellular functions, including cell survival, DNA replication and cell cycle control. Mechanism controlling the precise timing and sequence of cell cycle events as well as checkpoints insuring fidelity of those events are key targets that when disrupted could result in tumorigenesis. Mitosis is the process by which a cell duplicates its genetic information (DNA), in order to generate two, identical, daughter cells. In addition each daughter cell must receive one centrosome and the appropriate complements of cytoplasm and organelles. This process is conventionally divided in to five distinct stages: prophase, prometaphase, metaphase, anaphase and telophase that correspond to a different morphology of the cell. The entry into mitosis (M) is under the control of the cyclin dependent kinase Cdk1. During G2, the kinases Wee1 and Myt1 phosphorylate Cdk1 at T14/Y15 residues, rendering it inactive. The transition from G2 to M is promoted by the activation of Cdk1 via dephosphorylation by the Cdk1 phosphatase Cdc25C. Activated Cdk1 complexes translocate into the nucleus during prophase where phosphorylate numerous substrates in order to enhance their activation as the cells progresses trough prophase, prometaphase, and metaphase.

Recently we identified a new player: FEZ1/LZTS1 that contributes to the fine-tuning of the molecular events that determine progression through mitosis, and here will review its role in cancer development and in M phase regulation.

Background

Frequent loss of heterozygosity (LOH) at specific chromosomal regions in tumors implies the presence of tumor suppressor genes. One of these regions, located at the short arm of chromosome 8 (8p21.3–22) has been pointed out as frequently lost in different human malignancies as well as implicated in tumor progression. Investigation of this region, in primary esophageal cancer, has allowed the identification of the human FEZ1/LZTS1 (Leucine-Zipper Tumor Suppressor 1) gene at chromosome 8p22 [1]. LZTS1 gene show in non-cancerous cells a 6.8 kb mRNA including a 1.7 kb open reading frame (ORF), spanning exons 1–3, which encodes a 596-aa protein of 67 kDa [1]. Homology search of databases showed that the protein contains two Leucine-zipper motifs (Fig. 1) and it has 32% identity to the DNA binding domain of a cAmp-responsive activating transcription factor, Atf-5, although Lzts1 lacks the DNA recognition sub-domain usually found upstream of a Leucine-zipper motif in transcription factor. Moreover, searches for motifs and compositional analyses inside the Lzts1 protein sequence revealed the presence of multiple potential phosphorylation sites for different kinases (Fig. 1), such as PKA, CDC2 and PKC. The product of the LZTS1 gene is the founding member of a three member family proteins, all of which harbor a Leucine zipper Fez domain [2, 3]. Interestingly protein alignment of the three members (Lzts1, Lzts2 and Lzts3) showed significant sequences similarity, particularly in the C-terminal potential Leucine zipper regions. Although recently has been reported [4] that the nuclear export sequence identified in Lzts2 is not conserved in both the Lzts1 and the Lzts3 proteins, suggesting a potential role for Lzts2 in the regulation of nuclear and cytoplasmic trafficking, which may reveal a different function for this family member.

Figure 1
figure 1

Structural Functional Domains of Lzts1 protein. Putative phosphorylation motifs of various kinases cAMP-dependent protein kinase A (PKA, in blue), protein kinase C (PKC, in green), cGMP-dependent kinase (PKG, in prugna), casein kinases I and II (CK1 and CK2, in green and in red, respectively), CDC2 kinase (CDC2, in orange), tyrosine kinase (TyrKin, in magenta). The Leucine residues of Leucine zipper motifs are indicated by the gray color.

Expression pattern of LZTS1 in Primary Tumors and in Cancer Cell lines

Immunohistochemical studies of primary tumors indicated that Lzts1 protein expression in cancer cells is absent or markedly reduced in many different cancers compared with non-cancerous tissue [5–8]. Our previous studies showed that the expression of Lzts1 protein was undetectable or markedly reduced in 44% (39/88 cases) of primary gastric cancer with significant correlation to diffuse histotype (P < 0.001) [5], in 62% (37/60 cases) of primary transitional cell carcinoma of bladder with significant correlation to histopathological tumor grade (P < 0.005) [6], and in 64% (7/11 cases) of Bellini duct carcinoma [7]. Other studies showed that Lzts1 protein was lost or reduced in 68% (70/103 cases) of lung cancer with different histotypes, with a positive association with the tumor grade [8]. Overall, ~60% of different primary tumors showed loss or reduction of Lzts1 protein expression in cancer cells. Western blot and RT-PCR analyses in different cancer derived cell lines showed similar result [5–10], arguing that loss or reduction of LZTS1 is a frequent event in cancer development and progression. In addition LZTS1 is also mutated and/or deleted in several cancer types. We analyzed LZTS1 ORF (exon 1–3) in a total of 220 cancers, from 72 primary esophageal cancers, 18 esophageal cancer cell lines, 24 primary prostate cancers, 3 prostate cancer cell lines, 39 primary breast cancers, 25 breast cancer cell lines, 8 primary ovarian cancers, 4 leukemic cell lines, 1 cervical cancer cell line [1] and 26 primary gastric cancers [5], regardless of the presence or absence of LZTS1 expression. We found four point mutations in two primary esophageal cancers, in a prostate cancer cell line, and in a primary gastric carcinoma. In a primary esophageal tumor, alteration of TCC to CCC at codon 29 resulted in the substitution of Ser-29 with Pro-29, which is a predicted cAMP-dependent kinase phosphorylation site. In another primary esophageal cancer, alteration of AAG/Lys to GAG/Glu at codon 119 was found. Our LOH study indicated that these two patients had allelic losses at the D8S261 marker. Thus, these tumor cells retained the mutated LZTS1 allele and lost the normal LZTS1 allele by point mutation. The third point mutation was the change of CAG/Gln to TAG/Stop at codon 501 in a prostate cancer cell line, PC3, which resulted in coding of a putative 166-aa protein lacking the C terminus. The fourth mutation was a somatic missense mutation (CAC/His to CGC/Arg at LZTS1 codon 17) of one allele in a diffuse-type gastric carcinoma.

We identified, by RT-PCR, several internally truncated transcripts in esophageal cancer, prostate cancer, melanoma, and hematological malignancies (Table 1). The transcript from two independent esophageal cancers showed a frameshift within the ORF, which resulted in coding a short 76-aa protein [1]. We analyzed the nucleotide sequences around "breakpoints" of the deleted cDNAs and compared them to sequences from the full-length cDNA. The results showed that all of the acceptor sites contain the intronic AG flanking the exons, suggesting that the deleted transcripts in several tumors may be produced by alternative splicing.

Table 1

More recently a detailed DNA sequence analysis of LZTS1 was performed in a screening panel consisting of sporadic and hereditary prostate cancer (HPC) cases and unaffected controls. Twenty-four single nucleotide polymorphism (SNP), 15 of which were novel, were identified in germline DNA. Four coding SNP were identified. Eleven informative SNP were genotyped in 159 HPC probands, 245 sporadic prostate cancer cases, and 222 unaffected controls. Four of these SNP were statistically significant for association with prostate cancer (P < 0.04), supporting a role of LZTS1 in prostate cancer risk [11].

Effect of LZTS1 Gene Introduction Into Cancer Cells

Experimental studies using different cancer derived cell lines from human bladder, breast and prostate tumor, showed that reintroduction of LZTS1 results in inhibition of tumor cell growth in vitro and/or tumorigenicity in vivo.

In one study, introduction of full-length LZTS1 cDNA resulted in suppression of tumorigenicity in nude mice, in the inhibition of stable colony-forming efficiencies of ~50% of highly metastatic rat prostate cells and human prostate and embryonic kidney cells, and in the reduction of cell growth with accumulation of cells at late S to G2/M stage of cell cycle [12]. In our studies, using breast cancer (MCF7) and urothelial cancer derived cell lines (SW780) we observed inhibition of cancer cell growth in vitro and reduced tumorigenicity in vivo. In these studies we pointed out that LZTS1 inhibits cancer cell growth through the regulation of mitosis, interacting with CDK1, an important factor for G2/M transition of cell cycle [13, 6].

Lzts1 is a tumor suppressor in mammals

To investigate the function of LZTS1 we generated Lzts1 Knockout mice (Lzts1-/-) [14]. In animals at the age of 8–24 months (average 16 months), pathological analysis demonstrated in animals either heterozygous or knockout the onset of spontaneous neoplasms. The neoplasms in Lzts1-/- mice were breast tumors, hepatocellular carcinomas, lymphomas, soft tissue sarcomas, and lung adenomas, a spectrum suggesting that Lzts1 absence affects multiple cell types in vivo. In addition we used NMBA treatment to determine whether Lzts1 modulates carcinogen-induced malignancy development. Six weeks after NMBA administration all 20 Lzts1-/- mice (100%) and all 33 Lzts1+/- mice (100%) developed multiple tumors of the forestomach, whereas only 5 of 30 Lzts1+/+ mice (17%) developed tumors (Lzts1+/+ vs. Lzts1-/-, P < 0.001; Lzts1+/+ vs. Lzts1+/-, P < 0.001).

Histological sections showed an array of lesions typically seen in NMBA-treated animals, including hyperplasia, focal hyperplastic lesions, papillomas and carcinomas. Lzts1-/- and Lzts1+/- mice developed at high frequency invasive carcinoma of the forestomach, while Lzts1+/+ mostly showed thin and regular epithelia. All Lzts1+/- mice retained the wild type allele that was negative for mutations, a finding suggesting haploinsufficiency of Lzts1.

Lzts1: a novel regulator of the cell cycle

Biochemical investigation of the mouse Lzts1 gene using fibroblasts derived from Lzts1 wt and null embryos (MEF), revealed that Lzts1 wt and null MEFs displayed a normal progression along the different phases of the cell cycle but showed a faster M phase progression associated to a lower Cyclin B1/Cdk1 activity [14]. Using video time-lapse microscopy, we were able to demonstrate that the faster M progression of Lzts1-/- cells was due to the time they spent in prophase. The faster mitotic exit together with the lower Cdk1 activity resulted in an increase in the number of mitosis containing lagging chromosome with an increased hypertetraploid cell population [14]. The time to complete mitosis represents the key for proper DNA separation. Lzts1 absence decreases Cdk1 activity resulting in prompt mitotic exit accompanied by defects in DNA segregation. The mechanism whereby Lzts1 absence decreases Cdk1 activity was sought investigating the molecule responsible for Cdk1 activation: namely Cdc25C. Cdc25s family of phosphatase serves as a key activator of the Cdks/Cyclins [15]. The three mammals Cdc25 phosphatase (Cdc25A, B and C) are responsible for dephoshorylating Cdks/cyclins on pThr14 and/or pTyr15 residues. This dephosphorylation triggers the final activation of Cdks/cyclins activity during normal cell cycle progression.

Recently we discovered that mitotic Lzts1-/- MEFs showed a higher Cdk1-pY15/Cdk1 ratio accompanied by lower levels of endogenous Cdc25C that results in a lower association between endogenous Cdc25C and endogenous Cdk1.

It has been proposed that in G2/M arrested cells, Cdc25C may be efficiently ubiquitinated [16]. Therefore we tested whether Lzts1 might be involved in Cdc25C ubiquitination and we found that Lzts1 expression protects Cdc25C from proteosome degradation during M phase, thereby regulating Cdk1 activity [14] (Fig. 2).

Figure 2
figure 2

Progression through the different stages of mitosis in Lzts1+/+ and Lzts1-/- cells. During prophase the interaction between Lzts1 and Cdc25C allows high levels of Cdc25C expression and activity resulting in normal progression from prophase to metaphase. In Lzts1 deficient cell during prophase, Cdc25C is rapidly ubiquitinated and degraded thus determining a low activity of the cyclin B1/Cdk1 complex. As a consequence the cell progress faster through prophase and prometaphase and frequently undergo to chromosomes missegregation. Yellow arrows indicate lagging chromosomes in Lzts1-/- cells.

This findings are rather surprising and change the classical view of M phase entrance and progression that implicates a central role for the high Cdk1 activity necessary for the proper execution of M phase [17–19].

It is possible that decreased Cdk1 activity by changes in its inhibitory phosphorylation during G2 phase may result in G2 block of the cell cycle, whereas decreasing Cdk1 activity in M phase before the establishment of the metaphase plate causes a faster prometaphase progression and mitotic exit, as in Lzts1-/-cells. In turn this event would lead to higher percentage of improper chromosome segregation resulting in cell transformation.

One intriguing observation is that Lzts1 absence seems to affects Cdc25C levels in mitotic MEFs but not in cells returned in G1 after the completion of cell division [14]. Accordingly, endogenous Lzts1 and Cdc25C proteins associated better during M phase than in proliferating cells extracts [14]. These observations suggest that in mitotic cells one or both proteins undergo to post-translational modifications or bind yet unidentified factors specifically expressed during M phase that could favor their interaction.

Conclusion

Human tumor suppressors often function as negative regulator of the cell cycle. In fact their mutations can lead to failure of cell cycle checkpoint controls resulting in the accumulation of genetic changes contributing to a tumor phenotype. Several tumor suppressors are known to modulate the activities of Cdk/cyclin complexes. We focus here on a newly identified factor: Lzts1 which alterations lead to premature mitotic exit that results in chromosomal instability and aneuploidy. Being Lzts1 frequently down-regulated in many human tumors further investigations are needed in order to better understand its functions, eventually uncovering new opportunities for approaching cancer and other proliferation-related diseases.

References

  1. Ishii H, Baffa R, Numata S, Murakumo Y, Rattan S, Inoue H, Mori M, Fidanza V, Alder H, Croce CM: The FEZ1 gene at chromosome 8p22 encodes a leucine-zipper protein, and its expression is altered in multiple human tumors. Proc Natl Acad Sci USA 1999, 96: 3928–3933. 10.1073/pnas.96.7.3928

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  2. Cabeza-Arvelaiz Y, Thompson TC, Sepulveda JL, Chinaul AC: LAPSER1: a novel candidate tumor suppressor gene from 10q24.3. Oncogene 2001, 20: 6707–6717. 10.1038/sj.onc.1204866

    Article  CAS  PubMed  Google Scholar 

  3. Teufel A, Weinmann A, Galle PR, Lohse AW: In silico characterization of LZTS3, a potential tumor suppressor. Oncol Rep 2005, 14: 547–551.

    CAS  PubMed  Google Scholar 

  4. Thyssen G, Li TH, Lehmann L, Zhuo M, Sharma M, Sunz Z: LZTS2 is a novel beta-catenin-interacting protein and regulates the nuclear export of beta-catenin. Mol Cell Biol 2006, 26: 8857–8867. 10.1128/MCB.01031-06

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Vecchione A, Ishii H, Shiao YH, Trapasso F, Rugge M, Alder H, Croce CM, Baffa R: Altered expression of Fez1 protein in gastric carcinoma. Clin Cancer Res 2001, 7: 1546–1552.

    CAS  PubMed  Google Scholar 

  6. Vecchione A, Ishii H, Baldassarre G, Bassi P, Trapasso F, Alder H, Pagano F, Gomella LG, Croce CM, Baffa R: FEZ1/LZTS1 is down-regulated in high-grade bladder cancer, and its restoration suppresses tumorigenicity in transitional cell carcinoma cells. Am J Pathol 2002,160(4):1345–1352.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Vecchione A, Galetti TP, Gardiman M, Ishii H, Giarnieri E, Pagano F, Gomella LG, Croce CM, Baffa R: Collecting duct carcinoma of the kidney: an immunohistochemical study of 11 cases. BMC Urol 2004,9(4):11. 10.1186/1471-2490-4-11

    Article  Google Scholar 

  8. Nonaka D, Fabbri A, Roz L, Mariani L, Vecchione A, Moore GW, Tavecchio L, Croce CM, Sozzi G: Reduced FEZ1/LZTS1 expression and outcome prediction in lung cancer. Cancer Res 2005, 65: 1207–1212. 10.1158/0008-5472.CAN-04-3461

    Article  CAS  PubMed  Google Scholar 

  9. Ono K, Uzawa K, Nakatsuru M, Shiiba M, Mochida Y, Tada A, Bukawa H, Miyakawa A, Yokoe H, Tanzawa H: Down-regulation of FEZ1/LZTS1 gene with frequent loss of heterozygosity in oral squamous cell carcinomas. Int J Oncol 2003, 23: 297–302.

    CAS  PubMed  Google Scholar 

  10. Toyooka S, Fukuyama Y, Wistuba II, Tockman MS, Minna JD, Gazdar AF: Differential expression of FEZ1/LZTS1 gene in lung cancers and their cell cultures. Clin Cancer Res 2002, 8: 2292–2297.

    CAS  PubMed  Google Scholar 

  11. Hawkins GA, Mychaleckyi JC, Zheng SL, Faith DA, Kelly B, Isaacs SD, Wiley KE, Chang BL, Ewing CM, Bujnovszky P, Bleecker ER, Walsh PC, Meyers DA, Isaacs WB, Xu J: Germline sequence variants of the LZTS1 gene are associated with prostate cancer risk. Cancer Genet Cytogenet 2002, 137: 1–7. 10.1016/S0165-4608(02)00549-6

    Article  CAS  PubMed  Google Scholar 

  12. Cabeza-Arvelaiz Y, Sepulveda JL, Lebovitz RM, Thompson TC, Chinault AC: Functional identification of LZTS1 as a candidate prostate tumor suppressor gene on human chromosome 8p22. Oncogene 2001, 20: 4169–4179. 10.1038/sj.onc.1204539

    Article  CAS  PubMed  Google Scholar 

  13. Ishii H, Vecchione A, Murakumo Y, Baldassarre G, Numata S, Trapasso F, Alder H, Baffa R, Croce CM: FEZ1/LZTS1 gene at 8p22 suppresses cancer cell growth and regulates mitosis. Proc Natl Acad Sci USA 2001, 98: 10374–10379. 10.1073/pnas.181222898

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Vecchione A, Baldassarre G, Ishii H, Nicoloso MS, Belletti B, Petrocca F, Zanesi N, Fong LYY, Battista S, Guarnieri D, Baffa R, Alder H, Farber JL, Donovan PJ, Croce CM: Fez1/Lzts1 absence impairs Cdk1/Cdc25C interaction during mitosis and predisposes to cancer development. Cancer Cell 2007, 11: 275–289. 10.1016/j.ccr.2007.01.014

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  15. Rudolph J: Cdc25 Phosphatases: Structure, Specificity, and Mechanism. Biochemestry 2007, 46: 3595–3604. 10.1021/bi700026j

    Article  CAS  Google Scholar 

  16. Chen F, Zhang Z, Bower J, Lu Y, Leonard SS, Ding M, Castranova V, Piwinca-Worms H, Shi X: Arsenite-induced Cdc25C degradation is through the KEN-box and ubiquitin-proteasome pathway. Proc Natl Acad Sci USA 2002, 99: 1990–1995. 10.1073/pnas.032428899

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  17. Pines J: Mitosis: a matter of getting rid of the right protein at the right time. Trends Cell Biol 2006, 16: 55–63. 10.1016/j.tcb.2005.11.006

    Article  CAS  PubMed  Google Scholar 

  18. Nigg EA: Mitotic kinases as regulators of cell division and its checkpoints. Nat Rev Mol Cell Biol 2001, 2: 21–32. 10.1038/35048096

    Article  CAS  PubMed  Google Scholar 

  19. Smits VA, Medema RH: Checking out the G(2)/M transition. Biochim Biophys Acta 2001, 1519: 1–12.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thanks all the members of our labs for helpful discussion and suggestion. This work is supported by grants from AIRC and from Istituto Superiore di Sanità (Programma Oncotecnologico) to A.V. and G.B.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carlo M Croce.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Vecchione, A., Croce, C.M. & Baldassarre, G. Fez1/Lzts1 a new mitotic regulator implicated in cancer development. Cell Div 2, 24 (2007). https://doi.org/10.1186/1747-1028-2-24

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1747-1028-2-24

Keywords