Journal List > J Gynecol Oncol > v.31(2) > 1142868

Kunitomi, Kobayashi, Wu, Takeda, Tominaga, Banno, and Aoki: LAMC1 is a prognostic factor and a potential therapeutic target in endometrial cancer

Abstract

Objective

With the emerging significance of genetic profiles in the management of endometrial cancer, the identification of tumor-driving genes with prognostic value is a pressing need. The LAMC1 gene, encoding the laminin subunit gamma 1 (LAMC1) protein, has been reported to be involved in the progression of various malignant tumors. In this study, we aimed to investigate the role of LAMC1 in endometrial cancer and elucidate the underlying mechanism.

Methods

We evaluated the immunohistochemical expression of LAMC1 in atypical endometrial hyperplasia and endometrial cancer. Within the endometrial cancer cases, we analyzed the association of LAMC1 overexpression with clinicopathological factors and prognosis. Furthermore, to indentify genes influenced by LAMC1 overexpression, we transfected HEC50B and SPAC-S cells with siRNA targeting LAMC1 and conducted microarray gene expression assays.

Results

While none of the atypical endometrial hyperplasia specimens exhibited LAMC1 overexpression, endometrial cancer possessed a significantly higher LAMC1 overexpression rate. LAMC1 overexpression was strongly associated with histological type, lymphovascular space invasion, lymph node metastasis, advanced International Federation of Gynecology and Obstetrics stage, and poor overall survival in endometrial cancer. Gene expression microarray analysis identified 8 genes correlated with tumor progression (LZTFL1, TAPT1, SEL1L, PAQR6, NME7, TMEM109, CCDC58, and ANKRD40) that were commonly influenced in HEC50B and SPAC-S by LAMC1 silencing.

Conclusion

LAMC1 overexpression is a potent biomarker for identifying endometrial cancer patients needing aggressive adjuvant therapy. We elucidated 8 candidate genes that may mediate progression of LAMC1 overexpressing cancer. Further investigation of the underlying mechanism should lead to the discovery of new therapeutic targets.

INTRODUCTION

Among women, endometrial cancer is the sixth most common cancer worldwide [1], increasing its importance particularly in the developed countries reflecting their lifestyle changes. Traditionally, endometrial cancer has been classified into types 1 and 2 based on etiology and clinical behavior [23]. Type 1 tumors are estrogen-dependent well-differentiated adenocarcinomas, arising in the background of endometrial hyperplasia, comprising up to 80% of endometrial cancer cases. Contrarily, type 2 tumors are estrogen-independent, poorly differentiated adenocarcinomas, which occur de novo from atrophic endometria. Over many years, histological types, lymph node (LN) and distant metastasis have been regarded as the critical prognosis factors of endometrial cancer [45]. Since 2013, the classification of endometrial cancer has been drastically changed [6]; the importance of the tumor's genomic features in determining its aggressiveness and resistance to therapy has become apparent. Thus, to develop new therapeutic strategies, further investigation of tumor-driving genes is the pressing issue today.
Laminins are heterotrimeric extracellular matrix proteins that are composed of the alpha, beta, and gamma chains, which mediate cellular signaling through interaction with cell membrane receptors [7]. The effect of specific laminin isoform expressions on cancer progression has been vigorously studied; for instance, the LAMC2 gene, encoding the laminin subunit gamma 2 chain, is an established prognostic marker of various types of cancer [8-10]. In this study, we focused on the LAMC1 gene encoding the laminin subunit gamma 1 (LAMC1) chain, which has recently been reported to be involved in the carcinogenesis of fallopian tube cancer [11]. LAMC1 overexpression has been reported to be correlated with poor prognosis of hepatocellular carcinoma [1213] and meningioma [14], but its role in endometrial cancer remains to be elucidated.
By investigating the LAMC1 expression patterns in clinical specimens, we show that overexpression of LAMC1 in endometrial cancer is related to aggressive histological types, LN metastasis, advanced International Federation of Gynecology and Obstetrics (FIGO) stage, and poor prognosis. We also examine the downstream molecular changes caused by the overexpression of this new potential therapeutic target.

MATERIALS AND METHODS

1. Patients and tissue samples

Endometrial tissues were obtained from 97 patients with endometrial carcinoma and from 9 patients with atypical endometrial hyperplasia who underwent surgery at Keio University Hospital (Tokyo, Japan) from 1999 to 2013. All specimens were fixed in 10% phosphate-buffered formalin and embedded in paraffin. The 4 μm-thick sections were stained with hematoxylin and eosin to confirm the presence of tumor and to assess its histological characteristics. Written informed consent was obtained from each participant regarding the use of samples for research. The Ethics Committee of Keio University approved this study (approval No. 20130336).

2. Immunohistochemistry

The specimens were deparaffinized in xylene and rehydrated in a graded series of ethanol. Antigen retrieval was performed with citrate buffer (pH 6.0) for 20 minutes at 95°C. After blocking of endogenous peroxidase activity with 0.3% H2O2 in phosphate-buffered saline, the specimens were incubated overnight with anti-LAMC1 antibody (HPA001908, 1:500; Sigma-Aldrich, St. Louis, MO, USA) at 4°C. Indirect immunohistochemical staining was performed using N-Histofine Simple Stain MAX-PO (Nichirei Biosciences Inc., Tokyo, Japan). The specimens were counterstained with hematoxylin, dehydrated in a graded series of ethanol, dried and coverslipped.

3. Evaluation of immunohistochemical staining

Although laminin gamma 1 is a member of the extracellular matrix, the LAMC1 antibody stains its precursor, which localizes to the cytosol. We defined overexpression of LAMC1 as >10% of tumor cells or normal endometrial cells showing higher immunoreactivity compared to their stromal counterparts in 5 high-power fields in each section. Two investigators (H.K. and Y. K.) independently evaluated the slides in a blinded manner.

4. Cell line and culture

We used 2 human endometrial cancer cell lines: HEC50B cells were purchased from the American Type Culture Collection (Manassas, VA, USA); SPAC-S cells were kindly provided by Dr. Tokuichi Kawaguchi (Japanese Foundation for Cancer Research, Cancer Institute, Tokyo, Japan). HEC50B cells were maintained in Dulbecco's modified eagle medium supplemented with 10% fetal bovine serum (FBS). SPAC-S cells were cultured in 20% MCDB152 medium and 80% DM160 medium supplemented with 5% FBS, insulin-transferrin-selenium (5 μg/mL; Collaborative Research Co., Lexington, MA, USA), and epidermal growth factor (10 ng/mL; Takara Biomedicals, Shiga, Japan) [15].

5. siRNA LAMC1 inhibition

We purchased siRNA duplexes from Thermo Fisher Scientific; Silencer® Select siLAMC1 (siRNA ID s8077) and SilencerTM Select Negative Control No.1 siRNA (product No. 4390843) (Thermo Fisher Scientific, Waltham, MA, USA). Twenty-four hours after seeding, cells were transfected with siRNA duplexes for 48 hours using the Lipofectamine RNAiMAX reagent (Invitrogen, Waltham, MA, USA).

6. Microarray analysis

Total RNA from the HEC50B and SPAC-S cells was extracted using the RNeasy Mini Kit (Qiagen, Hilden, Germany) in combination with RNase-free DNase (Qiagen). The quality of RNA was assessed using the Agilent 2100 Bioanalyzer (Agilent, Santa Clara, CA, USA). The RNA samples were labeled using Low Input Quick Amp Labeling Kit (one-color; Agilent) and then hybridized to the Agilent SurePrint G3 Human Gene Expression version 3.0 Microarray (Agilent) according to the manufacturer's instruction. The arrays were scanned using the Agilent DNA microarray scanner G2505C (Agilent). The statistical software R (version 3.4.1; R Foundation, Vienna, Austria) and the Bioconductor package limma were used to process the raw data. Briefly, the raw data underwent background correction by the “normexp” method, followed by quantile normalization across samples. Probes that displayed low expression levels across all samples were excluded from the analysis. The empirical Bayes method implemented in limma [16] was used to identify genes that were differentially expressed (false discovery rate [FDR] <0.01) between the control (control siRNA) and the experimental (LAMC1 siRNA) groups. We performed gene ontology (GO) analysis using the Database for Annotation, Visualization and Integrated Discovery version 6.8 (DAVID, http://david.abcc.ncifcrf.gov). The p<0.05 was considered statistically significant.
Furthermore, we performed hierarchical clustering using R and identified differentially expressed genes (DEGs) possibly involved in cancer progression by referring to GeneCards (https://www.genecards.org) and The Human Protein Atlas (https://www.proteinatlas.org).

7. Quantitative real-time polymerase chain reaction (RT-PCR) analysis

Total RNA was obtained from cells as mentioned earlier, and 500 ng of each sample was reverse-transcribed using SuperScript VILO Master Mix (Thermo Fisher Scientific). RT-PCR analysis was performed in triplicate using SYBR Green Master Mix and Thermal Cycler Dice (Takara Biomedicals). The primer sets for the RT-PCR are listed in Supplementary Table 1.

8. Statistical analysis

Statistical analysis was performed using SPSS statistical software version 24.0 (IBM Japan, Tokyo, Japan). Categorical variables were reported as absolute numbers and percentages. Continuous variables were reported as median and range or as mean and standard deviation. Correlations between overexpression of LAMC1 and clinicopathological factors were evaluated by χ2 test. We calculated the progression-free and overall survival from the date of surgery to either the last follow-up or date of recurrence or death. The Kaplan-Meier method was used to estimate the probability of progression-free and overall survival, and log-rank test was used to compare between groups. The p<0.05 was considered statistically significant.

RESULTS

1. LAMC1 is overexpressed in endometrial carcinoma

The representative LAMC1 immunohistochemical staining results are shown in Fig. 1. In normal proliferative phase endometrium (obtained from the unaffected area of the resected specimen) and atypical endometrial hyperplasia, we observe only slight LAMC1 staining in the endometrial stroma and vascular endothelia, and LAMC1 expression in the endometrium did not exceed that in the stroma. On the contrary, the tumor cells in endometrial carcinoma (endometrioid carcinoma grades 1 and 3, and clear cell carcinoma) showed prominent LAMC1 expression, clearly exceeding that in the stroma. As described in Table 1, none of the atypical endometrial hyperplasia specimens showed LAMC1 overexpression (0/9), whereas overexpression of LAMC1 was observed in 76.3% (74/97) of the endometrial carcinoma specimens, representing a significant difference (p<0.001).
Fig. 1

LAMC1 expression pattern in endometrial tissues. (A) Normal endometrium (proliferative phase). (B) Atypical endometrial hyperplasia. (C) Endometrioid carcinoma grade 1. (D) Endometrioid carcinoma grade 3. (E) Clear cell carcinoma. (A,B) There were evaluated as negative. (C-E) There were evaluated as positive for LAMC1 overexpression. Scale bars, 100 μm.

LAMC1, laminin subunit gamma 1.
jgo-31-e11-g001
Table 1

LAMC1 overexpression in atypical endometrial hyperplasia and endometrial carcinoma

jgo-31-e11-i001
Overexpression of LAMC1 Negative Positive p-value*
Atypical endometrial hyperplasia 9 0 <0.001
Endometrial carcinoma 23 74
LAMC1, laminin subunit gamma 1.
*The χ2 test.

2. Relationship between LAMC1 overexpression and clinicopathological features in endometrial carcinoma

We analyzed the relationship between LAMC1 overexpression and clinicopathological features in patients with endometrial cancer. The age at diagnosis and body mass index did not differ among patients with or without LAMC1 overexpression (Supplementary Table 2). Focusing on the histological type, type 2 cases had a significantly higher LAMC1 overexpression rate (88.7% [47/53], p=0.002) than type 1 cases (61.4% [27/44]). Although grade 3 endometrioid carcinoma (81.5% [22/27]) had a higher LAMC1 overexpression rate than grades 1–2 (61.4% [27/44]), no statistical significance was noted among these 2 groups (p=0.113). Of note, in serous carcinoma and clear cell carcinoma, which are especially aggressive histological types, nearly all of the cases had LAMC1 overexpression (95.0% [17/18] and 100% [5/5], respectively). Furthermore, lymphovascular space invasion (LVSI), LN metastasis, and advanced FIGO stage were related to a significantly higher LAMC1 overexpression rate, while distant metastasis was not (Table 2).
Table 2

Relationship between overexpression of LAMC1 and clinicopathological factors

jgo-31-e11-i002
Overexpression of LAMC1 Negative (n=23) Positive (n=74) p-value*
Histology 0.002*
Type 1 (endometrioid grade 1–2) 17 27
Type 2 6 47
Endometrioid grade 3 5 22
Serous 1 17
Clear cell 0 5
Mixed 0 3
Lymphovascular space invasion 0.003*
Negative 15 25
Positive 7 49
NA 1 0
Lymph node metastasis 0.016*
Negative 22 48
Positive 1 24
NA 0 2
Distant metastasis 1.000
Negative 21 67
Positive 2 7
FIGO stage 0.007*
I–II 19 37
III–IV 4 37
FIGO, International Federation of Gynecology and Obstetrics; LAMC1, laminin subunit gamma 1; NA, not applicable.
*The χ2 test.

3. Association between LAMC1 overexpression and patient prognosis in endometrial carcinoma

The follow-up period of the 97 patients ranged from 3 to 122 months, with a median period of 81 months. A total of 22 patients experienced relapse of the disease, and 19 died. Patients with endometrial carcinoma overexpressing LAMC1 tended to have a shorter progression-free survival (p=0.201), and strikingly, had a significantly poor overall survival (Fig. 2, p=0.045).
Fig. 2

Kaplan-Meier curves representing the association between LAMC1 overexpression and (A) PFS and (B) OS rates. Log-rank tests indicated that patients with overexpression of LAMC1 had a poorer overall prognosis.

LAMC1, laminin subunit gamma 1; OS, overall survival; PFS, progression-free survival.
jgo-31-e11-g002

4. LAMC1 silencing in endometrial cancer cell lines

Several previous studies have indicated that LAMC1 silencing results in impaired proliferation, migration, and invasion in various cancer cell lines [131718]. However, the molecular mechanisms underlying these changes remain to be investigated. Thus, we aimed to identify genes influenced downstream of LAMC1 overexpression by manipulating its expression in endometrial cancer cells.
LAMC1 overexpression was prominent among type 2 endometrial carcinomas. Therefore, we chose HEC50B, which represents poorly differentiated endometrioid carcinoma [19], and SPAC-S, which represents serous carcinoma [15], to evaluate the effects of LAMC1 inhibition. The cells were transfected with either control siRNAs or siRNA targeting LAMC1, and total RNA was provided to microarray gene expression assays.
The results of the microarray analysis revealed that a total of 652 genes were differentially expressed (fold change >2, FDR <0.01) commonly in HEC50B and SPAC-S in response to LAMC1 knockdown, including 211 upregulated and 441 downregulated genes. Supplementary Fig. 1 shows the results of the GO term enrichment analysis. As to the biological process, the downregulated DEGs significantly enriched in cell cycle, posttranscriptional regulation of gene expression, and translation, suggesting that LAMC1 silencing may negatively impact endometrial cancer progression through these changes. Supplementary Fig. 2 presents the top 100 influenced genes showing common expression changes in HEC50B and SPAC-S. Of note, 8 independently expressed genes that are correlated with tumor progression according to GeneCards and the Human Protein Atlas [20] were influenced commonly in HEC50B and SPAC-S. Four favorable prognostic factors, LZTFL1, TAPT1, SEL1L, and PAQR6, were upregulated, whereas 4 unfavorable prognostic factors, NME7, TMEM109, CCDC58, and ANKRD40, were downregulated by LAMC1 knockdown (Table 3), suggesting their role in the tumor-promoting effect of LAMC1 overexpression. Quantitative PCR confirmed the expression changes of the aforementioned 8 genes by LAMC1 knockdown (Fig. 3).
Table 3

Genes correlated with tumor progression that are influenced commonly in HEC50B and SPAC-S by LAMC1 knockdown

jgo-31-e11-i003
Gene symbol Gene Log fold-change
HEC50B SPAC-S
LZTFL1 Leucine zipper transcription factor like 1 2.371 1.819
TAPT1 Transmembrane anterior posterior transformation 1 1.355 1.307
SEL1L SEL1L ERAD E3 ligase adaptor subunit 0.945 0.813
PAQR6 Progestin and adipoQ receptor family member 6 1.139 1.350
NME7 Non-metastatic cells 7 −3.253 −3.422
TMEM109 Transmembrane protein 109 −2.627 −2.830
CCDC58 Coiled-coil domain containing 58 −3.206 −2.706
ANKRD40 Ankyrin repeat domain 40 −2.410 −2.157
Fig. 3

Relative expression of genes in HEC50B and SPAC-S transfected with siRNA targeting LAMC1. Cells were transfected with either LAMC1 or control siRNAs for 48 hours. Silencing of LAMC1 and expression changes of genes identified by microarray analysis were confirmed. The p-values are as shown (Student's t-test). All quantitative data are expressed as means± standard deviation (n=3 experiments).

LAMC1, laminin subunit gamma 1.
jgo-31-e11-g003

DISCUSSION

In our present study, while none of the atypical endometrial hyperplasia specimens had LAMC1 overexpression, endometrial carcinoma had a significantly high LAMC1 overexpression rate, suggesting the involvement of LAMC1 in the carcinogenesis of endometrial cancer. Because LAMC1 was reported to be overexpressed in the serous tubal intraepithelial carcinoma [11], we first expected it to be an early diagnostic marker for endometrial cancer. However, given the fact that type 1 endometrial carcinoma has a relatively low LAMC1 overexpression rate and that type 2 endometrial carcinoma possesses a higher LAMC1 overexpression rate than type 1, LAMC1 may not be the initial driver gene of oncogenesis in endometrial carcinoma. Instead, it seems to be correlated with its aggressiveness. In line with this idea, our analysis indicated that endometrial carcinoma overexpressing LAMC1 is likely to have LVSI, LN metastasis, and advanced FIGO stage. Together with the fact that LAMC1 knockdown suppressed cell motility and invasion in endometrial cancer cells [17], our results suggest a relationship between LAMC1 overexpression and lymphatic invasion.
LAMC1 overexpression was correlated with a significantly poor overall survival in endometrial carcinoma. In addition, progression-free survival showed the same tendency but without statistical significance, and this discrepancy may be based on the low aggressiveness of LAMC1-negative endometrial carcinoma, allowing the cure or remission of the recurrent disease with subsequent therapy.
The downstream mechanism through which LAMC1 influences tumor aggressiveness is largely unknown. Here, we reported that knockdown of LAMC1 in endometrial cancer cells caused significant expression changes in LZTFL1, TAPT1, SEL1L, PAQR6, NME7, TMEM109, CCDC58, and ANKRD40 at the transcription levels. LZTFL1 is a tumor suppressor gene identified through the analysis of the hotspot of tumor suppressor genes at chromosome 3p21.3, and the lack of its expression is correlated with poorly differentiated histological types, metastasis, and poor prognosis in patients with gastric cancer [21]. In vitro studies have revealed that overexpression of LZTFL1 causes suppression of epithelial-mesenchymal transition, cell migration and invasion in cancer cells. Although the majority of its functions remain unclear, the interaction of LZTFL1 and beta-catenin nuclear translocation has been reported recently [22]. SEL1L is the human homolog of the Caenorhabditis elegans sel-1 gene, which acts as a tumor suppressor in breast cancer and pancreatic ductal adenocarcinoma. Overexpression of SEL1L inhibits tumorigenicity in vitro and in vivo through the modulation of the Notch receptor genes, Smad4, activin A and PTEN [23-26]. NME7 is a member of the NME/NDK family, which mainly acts as nucleoside-diphosphate kinases and protein histidine kinases. NME7 is widely known for its microtubule-organizing functions and role in ciliary cargo transport [27], and its participation in cancer biology has been overlooked. Its role in the chemosensitivity of myeloma was reported only recently [28]. The interaction between LAMC1 and these genes, and the specific molecules involved in the exhibition of tumor aggressiveness in LAMC1-overexpressing tumors, is to be further investigated.
In summary, our data indicate that overexpression of LAMC1 is strongly associated with histological type, LVSI, LN metastasis, advanced FIGO stage, and poor overall survival in endometrial cancer. These results suggest that LAMC1 is a potent biomarker for identifying patients needing aggressive adjuvant therapy. Gene expression microarray analysis elucidated 8 candidate genes (LZTFL1, TAPT1, SEL1L, PAQR6, NME7, TMEM109, CCDC58, and ANKRD40) that may be involved in cancer progression in the downstream of LAMC1 expression. Further elucidation of the underlying mechanism should lead to the discovery of new therapeutic targets.

ACKNOWLEDGEMENTS

The authors would like to acknowledge Ms. Kayoko Kobori, Mr. Toshihiro Arai and Ms. Eriko Arai for fundamental support.

Notes

Conflict of Interest No potential conflict of interest relevant to this article was reported.

Author Contributions

  • Conceptualization: K.H., K.Y., T.E.

  • Data curation: K.H., K.Y., W.R.C.

  • Formal analysis: K.H.

  • Funding acquisition: K.Y.

  • Investigation: K.H., K.Y., T.T.

  • Methodology: K.H., K.Y., T.T.

  • Project administration: K.H.

  • Resources: T.T.

  • Supervision: T.E., B.K., A.D.

  • Validation: K.Y., B.K.

  • Visualization: K.H., T.T.

  • Writing - original draft: K.H.

  • Writing - review & editing: K.Y., A.D.

References

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018; 68:394–424.
crossref pmid
2. Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecol Oncol. 1983; 15:10–17.
crossref pmid
3. Murali R, Soslow RA, Weigelt B. Classification of endometrial carcinoma: more than two types. Lancet Oncol. 2014; 15:e268–78.
crossref
4. Abu-Rustum NR, Zhou Q, Gomez JD, Alektiar KM, Hensley ML, Soslow RA, et al. A nomogram for predicting overall survival of women with endometrial cancer following primary therapy: toward improving individualized cancer care. Gynecol Oncol. 2010; 116:399–403.
crossref pmid
5. Salvesen HB, Haldorsen IS, Trovik J. Markers for individualised therapy in endometrial carcinoma. Lancet Oncol. 2012; 13:e353–61.
crossref
6. Cancer Genome Atlas Research Network. Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013; 497:67–73.
crossref
7. Domogatskaya A, Rodin S, Tryggvason K. Functional diversity of laminins. Annu Rev Cell Dev Biol. 2012; 28:523–553.
crossref pmid
8. Pyke C, Rømer J, Kallunki P, Lund LR, Ralfkiaer E, Danø K, et al. The gamma 2 chain of kalinin/laminin 5 is preferentially expressed in invading malignant cells in human cancers. Am J Pathol. 1994; 145:782–791.
pmid pmc
9. Pyke C, Salo S, Ralfkiaer E, Rømer J, Danø K, Tryggvason K. Laminin-5 is a marker of invading cancer cells in some human carcinomas and is coexpressed with the receptor for urokinase plasminogen activator in budding cancer cells in colon adenocarcinomas. Cancer Res. 1995; 55:4132–4139.
pmid
10. Marinkovich MP. Tumour microenvironment: laminin 332 in squamous-cell carcinoma. Nat Rev Cancer. 2007; 7:370–380.
pmid
11. Kuhn E, Kurman RJ, Soslow RA, Han G, Sehdev AS, Morin PJ, et al. The diagnostic and biological implications of laminin expression in serous tubal intraepithelial carcinoma. Am J Surg Pathol. 2012; 36:1826–1834.
crossref pmid pmc
12. Liétard J, Musso O, Théret N, L'Helgoualc'h A, Campion JP, Yamada Y, et al. Sp1-mediated transactivation of LamC1 promoter and coordinated expression of laminin-gamma1 and Sp1 in human hepatocellular carcinomas. Am J Pathol. 1997; 151:1663–1672.
pmid pmc
13. Zhang Y, Xi S, Chen J, Zhou D, Gao H, Zhou Z, et al. Overexpression of LAMC1 predicts poor prognosis and enhances tumor cell invasion and migration in hepatocellular carcinoma. J Cancer. 2017; 8:2992–3000.
crossref
14. Ke HL, Ke RH, Li B, Wang XH, Wang YN, Wang XQ. Association between laminin γ1 expression and meningioma grade, recurrence, and progression-free survival. Acta Neurochir (Wien). 2013; 155:165–171.
crossref pmid
15. Hirai Y, Kawaguchi T, Hasumi K, Kitagawa T, Noda T. Establishment and characterization of human cell lines from a serous papillary adenocarcinoma of the endometrium. Gynecol Oncol. 1994; 54:184–195.
crossref
16. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015; 43:e47.
crossref
17. Kashima H, Wu RC, Wang Y, Sinno AK, Miyamoto T, Shiozawa T, et al. Laminin C1 expression by uterine carcinoma cells is associated with tumor progression. Gynecol Oncol. 2015; 139:338–344.
crossref pmid pmc
18. Piovan C, Palmieri D, Di Leva G, Braccioli L, Casalini P, Nuovo G, et al. Oncosuppressive role of p53-induced miR-205 in triple negative breast cancer. Mol Oncol. 2012; 6:458–472.
crossref pmid pmc
19. Kuramoto H, Nishida M, Morisawa T, Hamano M, Hata H, Kato Y, et al. Establishment and characterization of human endometrial cancer cell lines. Ann N Y Acad Sci. 1991; 622:402–421.
crossref pmid
20. Uhlen M, Zhang C, Lee S, Sjöstedt E, Fagerberg L, Bidkhori G, et al. A pathology atlas of the human cancer transcriptome. Science. 2017; 357:eaan2507.
crossref
21. Wei Q, Zhou W, Wang W, Gao B, Wang L, Cao J, et al. Tumor-suppressive functions of leucine zipper transcription factor-like 1. Cancer Res. 2010; 70:2942–2950.
crossref pmid pmc
22. Wang L, Guo J, Wang Q, Zhou J, Xu C, Teng R, et al. LZTFL1 suppresses gastric cancer cell migration and invasion through regulating nuclear translocation of β-catenin. J Cancer Res Clin Oncol. 2014; 140:1997–2008.
crossref pmid
23. Orlandi R, Cattaneo M, Troglio F, Casalini P, Ronchini C, Ménard S, et al. SEL1L expression decreases breast tumor cell aggressiveness in vivo and in vitro. Cancer Res. 2002; 62:567–574.
pmid
24. Cattaneo M, Orlandini S, Beghelli S, Moore PS, Sorio C, Bonora A, et al. SEL1L expression in pancreatic adenocarcinoma parallels SMAD4 expression and delays tumor growth in vitro and in vivo. Oncogene. 2003; 22:6359–6368.
crossref pmid
25. Liu Q, Chen J, Mai B, Amos C, Killary AM, Sen S, et al. A single-nucleotide polymorphism in tumor suppressor gene SEL1L as a predictive and prognostic marker for pancreatic ductal adenocarcinoma in Caucasians. Mol Carcinog. 2012; 51:433–438.
crossref pmid
26. Liu Q, Chen J, Wang J, Amos C, Killary AM, Sen S, et al. Putative tumor suppressor gene SEL1L was downregulated by aberrantly upregulated hsa-miR-155 in human pancreatic ductal adenocarcinoma. Mol Carcinog. 2014; 53:711–721.
crossref pmid
27. Šedová L, Školníková E, Hodúlová M, Včelák J, Šeda O, Bendlová B. Expression profiling of Nme7 interactome in experimental models of metabolic syndrome. Physiol Res. 2018; 67:S543–50.
crossref
28. Zhu YX, Tiedemann R, Shi CX, Yin H, Schmidt JE, Bruins LA, et al. RNAi screen of the druggable genome identifies modulators of proteasome inhibitor sensitivity in myeloma including CDK5. Blood. 2011; 117:3847–3857.
crossref pmid pmc

SUPPLEMENTARY MATERIALS

Supplementary Table 1

Primers used for quantitative RT-PCR

Supplementary Table 2

Comparison of age and BMI in endometrial cancer patients with or without LAMC1 overexpression

Supplementary Fig. 1

Gene ontology analysis results of (A) upregulated and (B) downregulated differentially expressed genes with fold change >1.5.

Supplementary Fig. 2

Heatmap showing the top 100 commonly influenced genes in HEC50B and SPAC-S by siLAMC1.
TOOLS
ORCID iDs

Haruko Kunitomi
https://orcid.org/0000-0003-3045-337X

Yusuke Kobayashi
https://orcid.org/0000-0002-4503-2845

Ren-Chin Wu
https://orcid.org/0000-0003-1439-0874

Takashi Takeda
https://orcid.org/0000-0001-6686-5742

Eiichiro Tominaga
https://orcid.org/0000-0001-9243-0267

Kouji Banno
https://orcid.org/0000-0003-2610-2833

Daisuke Aoki
https://orcid.org/0000-0002-9596-8326

Similar articles