
Downregulation of kinetochore‑associated 1 gene increases lagging chromosomes and contributes to chromosomal instability in gastric cancer cells
- Authors:
- Published online on: August 4, 2025 https://doi.org/10.3892/mi.2025.258
- Article Number: 59
-
Copyright : © Ohsaki et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].
Abstract
Introduction
In 2020, ~770,000 patients succumbed due to gastric cancer (GC), rendering GC the fourth most common cause of cancer-related mortality worldwide, surpassed only by lung, colorectal and liver cancers (1). As GC exhibits histological and genetic heterogeneity, the development of biomarker-based molecularly targeted therapeutics has lagged behind that of other cancer types (2). However, The Cancer Genome Atlas research program has succeeded in characterizing GC at the genomic level, yielding a system for classifying GC into four molecular subtypes according to the Epstein-Barr virus-positive status, microsatellite instability, genomic stability and chromosomal instability (CIN), with the aim of simplifying the treatment and diagnosis of GC (2,3).
The CIN subtype accounts for ~50% of GC cases and is characterized by the amplification of receptor tyrosine kinase (RTK) genes, a high frequency of TP53 mutations (in 70% of cases) and CIN, which is indicated by a high frequency of aneuploidy (4,5). In addition, the CIN subtype is known to exhibit intratumoral heterogeneity, which is involved in tumor relapse owing to the acquisition of cellular insensitivity to targeted drugs (6-8). Therefore, elucidating the molecular mechanisms of CIN in GC is of utmost therapeutic importance. However, these mechanisms are not yet fully understood.
The kinetochore-associated 1 (KNTC1) gene encodes kinetochore-associated protein 1 (KNTC1), a protein component of the outer kinetochore that is essential for the association of chromosomes and spindle microtubules. KNTC1 forms a complex with ZW10 and ZWILCH during mitosis. This complex, which is known as the RZZ complex, is involved in the activation of the spindle assembly checkpoint (SAC), the kinetochore-dependent recruitment of the Mad1/Mad3 and dynein/dynactin complex and the formation of the kinetochore corona in the outermost layer of the kinetochore (9-14). In particular, the activation of the SAC delays anaphase when there is a lack of proper connection between kinetochores and spindle microtubules, allowing for homogenous chromosome segregation (15,16). The depletion or loss of function of various kinetochore proteins, including KNTC1, has been reported to cause lagging chromosomes, resulting in abnormal chromosome segregation and subsequent aneuploidy and CIN in Drosophila and Caenorhabditis elegans (16-18). However, the role of the KNTC1 gene in GC is poorly understood.
It was hypothesized that the abnormal function of KNTC1 may be associated with the mechanism of CIN in GC. Therefore, the present study investigated the role of KNTC1 in GC CIN.
Materials and methods
Cell lines and culture conditions
The human GC cell lines, NCI-N87 (exhibiting human epidermal growth factor receptor type 2 gene amplification; cat. no. CRL-5822, American Type Culture Collection), KATOIII [exhibiting fibroblast growth factor receptor type 2 gene amplification; cat. no. JCRB0611, Japan Collection of Research Bioresources (JCRB) Cell Bank] and MKN74 (without amplification of RTK genes; cat. no. JCRB0255, JCRB), and the human normal fibroblast cell line, TIG-1-20 (cat. no. JCRB0501, JCRB), were used in the present study. The cells were cultured in RPMI-1640 medium (cat. no. 30264-56; Nacalai Tesque, Inc.) containing a 10% fetal bovine serum and 0.5% penicillin and streptomycin mixture (cat. no. 09367-34; Nacalai Tesque, Inc.) at 37˚C in an atmosphere containing 5% CO2. The cell cultures were grown in a CO2 incubator (cat. no. MHE-S1301A2-PJ; PHC Holdings Corporation).
Measurement of the frequency of lagging chromosomes
The NCI-N87, KATOIII, MKN74 and TIG-1-20 cells were seeded into 4-well culture slides (cat. no. 192-004; Watson Bio Lab) and cultured for 24 h at 37˚C. After 24 h, the cells were transfected with small interfering RNA (siRNA) targeting KNTC1, as described below. Following transfection, the cells are incubated for an additional 3 days and fixed for 20 min at room temperature with 4% paraformaldehyde (cat. no. 006775-1L; Bioenno Tech, LLC). The slides were washed twice for 5 min each with phosphate-buffered saline (cat. no. 73111; Kanto Chemical Co.) and sealed using coverslips and VECTASHIELD Vibrance Antifade Mounting Medium with DAPI (cat. no. H-1800; Vector Laboratories, Inc.). A total of 50 cells in anaphase, defined according to visible sister chromatid separation, were then observed using a fluorescence microscope (BX53F; Olympus Corporation). Among these 50 cells, the number of lagging chromosomes was counted (Figs. 1 and S1), and the percentage was calculated.
Reverse transcriptionquantitative polymerase chain reaction (RTqPCR)
Total RNA was extracted from the NCI-N87, KATOIII, MKN74 and TIG-1-20 cells using an RNeasy mini kit (cat. no. 74104; Qiagen, Inc.). cDNA synthesis was performed using reverse transcription with Superscript Ⅳ VILO Master Mix with ezDNase (cat. no. 11766050; Invitrogen; Thermo Fisher Scientific, Inc.). cDNA synthesis reaction was performed at 25˚C for 10 min, 50˚C for 10 min, and 85˚C for 5 min. KNTC1 (Hs00938554_m1) and GAPDH (No. 1902206) primers were obtained from Thermo Fisher Scientific, Inc. (primer sequence information not available). qPCR was performed using TaqMan Fast Advanced Master Mix (cat. no. 4444556; Applied Biosystems; Thermo Fisher Scientific, Inc.) using the following reaction conditions: An initial denaturation at 95˚C for 20 sec, then 40 cycles at 95˚C for 3 sec and 60˚C for 30 sec. mRNA expression was analyzed using the 2-ΔΔCq calculation (19).
Western blot analysis
The NCI-N87, KATOIII, MKN74 and TIG-1-20 cells were lysed in RIPA buffer (cat. no. 08714-04; Nacalai Tesque, Inc.). The cell lysates were then treated using ultrasound and centrifuged at 20,630 x g for 10 min at room temperature. The lysates were mixed with sample buffer (cat. no. 30566-22; Nacalai Tesque, Inc.) with 2-mercapto ethanol and incubated at 100˚C for 3 min. Lysates containing 5-7 µg protein were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis using Mini Trans-Blot® Cell (cat. no. 153BR78145; Bio-Rad Laboratories, Inc.). The separated proteins were then transferred from the gel to polyvinylidene fluoride membranes (cat. no. 1704272; Bio-Rad Laboratories, Inc.) using the Trans-Blot®TurboTM System (cat. no. 690BR009070; Bio-Rad Laboratories, Inc.). The membranes were blocked for 5 min at room temperature with Every Blot blocking buffer (cat. no. 12010020; Bio-Rad Laboratories, Inc.) and incubated with anti-ZW10 (1:1,000; cat. no. 24561-1-AP, Proteintech, Inc.) and anti-α-tubulin (1:5,000; cat. no. 3873, clone DM1A, Cell Signaling Technology, Inc.) antibodies overnight at 4˚C. Following incubation, the membranes were washed in Tris-buffered saline containing 0.1% Tween-20 (TBS-T). The membranes were then incubated with appropriate secondary antibodies (1:5,000; anti-mouse IgG, cat. no. 7076, Cell Signaling Technology, Inc. and 1:5,000; anti-rabbit IgG, cat. no. 7074, Cell Signaling Technology, Inc.) for 1 h at room temperature and washed with TBS-T. Protein bands were detected using a ChemiDoc MP Imaging System (Bio-Rad Laboratories, Inc.). Quantitative analysis was performed using ImageLab software version 4.1 (Bio-Rad Laboratories, Inc.).
siRNA-mediated knockdown of KNTC1
The sequence (GGAAUGAUAUUGAGCUGCUAACAAA) of human KNTC1 siRNA was designed from Thermo Fisher Scientific, Inc. (cat. no. HSS114610). KNTC1 siRNA or negative control (cat. no. 12935-300; Invitrogen; Thermo Fisher Scientific, Inc.) was combined with Lipofectamine RNAiMAX (cat. no. 13778-030; Invitrogen; Thermo Fisher Scientific, Inc.) and incubated for 15 min at room temperature. The NCI-N87, KATOIII, MKN74 and TIG-1-20 cells were transfected with the KNTC1 siRNA or negative control-Lipofectamine RNAiMAX complexes and incubated at 37˚C for 3 days. At 3 days following the addition of the KNTC1 siRNA or negative control-Lipofectamine RNAiMAX complexes, the cells were harvested and processed for RT-qPCR and western blot analysis, and for the analysis of the frequency of lagging chromosomes.
Statistical analysis
All the statistical analyses were performed using Statcel 4 (https://oms-publ.main.jp/main/4steps4-hyo1/). The one-way ANOVA and Tukey-Kramer test were performed to assess differences in frequency of lagging chromosomes and KNTC1 mRNA expression. The Student's t test was used to assess the efficiency of KNTC1 knockdown and the frequency of lagging chromosomes and ZW10 protein expression following KNTC1 knockdown. Data are presented as the mean ± standard deviation. P-values #x003C;0.05 were considered to indicate statistically significant differences.
Results
Frequency of lagging chromosomes and expression levels of KNTC1 mRNA
To investigate the CIN status in GC cells, the frequency of lagging chromosomes was analyzed. The frequency of lagging chromosomes was significantly higher in the NCI-N87 and KATOIII cells, which exhibited the amplification of RTK genes, than in the MKN74 cells, which did not exhibit the amplification of RTK genes (Fig. 2). Only a small number of lagging chromosomes were observed in the TIG-1-20 cells.
The expression levels of KNTC1 mRNA were higher in the MKN74 cells than in the NCI-N87 and KATOIII cells, and were inversely associated with the frequency of lagging chromosomes (Fig. 3). In addition, the mRNA expression level of KNTC1 in the NCI-N87 cells was significantly lower than that in TIG-1-20 cells.
Frequency of lagging chromosomes and expression of ZW10 following KNTC1 knockdown
The present study then investigated whether the knockdown of KNTC1 increased the frequency of lagging chromosomes. The confirmation of the knockdown efficiency KNTC1 siRNA was performed using RT-qPCR (Fig. 4). The NCI-N87, KATOIII and MKN74 cells exhibited increased frequencies of lagging chromosomes following the knockdown of KNTC1 (Fig. 5A). These differences were statistically significant in the NCI-N87 and KATOIII cells. By contrast, no increases in lagging chromosomes were observed in the TIG-1-20 cells following the knockdown of KNTC1.
In addition, to determine whether ZW10 compensated for KNTC1 function, ZW10 protein expression was investigated following KNTC1 knockdown. However, no changes in ZW10 protein expression were observed in any of the cell lines tested (Fig. 6).
Discussion
Deletion or loss of function of various kinetochore proteins, including KNTC1, has been reported to cause chromosome segregation abnormalities and induce aneuploidy and CIN in Drosophila and C. elegans (16-18). However, the role of KNTC1 in GC CIN is poorly understood. In the present study, it was found that GC cells with the amplification of RTK genes, including NCI-N87 and KATOIII cells, exhibited lower mRNA expression levels of KNTC1 and KNTC1 expression exhibited an inverse association with the frequency of lagging chromosomes (Figs. 2 and 3). Moreover, the frequency of lagging chromosomes in the NCI-N87, KATOIII and MKN74 cells increased following the knockdown of KNTC1 (Fig. 5A). These findings suggest that the suppression of the expression of the KNTC1 gene may contribute to CIN in GC. In particular, suppression of the expression of KNTC1 in GC cells exhibiting the amplification of RTK genes may enhance CIN and could lead to intratumoral genetic heterogeneity (Fig. 5B). Therefore, the restoration of normal KNTC1 expression levels may improve patient prognosis by alleviating intratumoral genetic heterogeneity through appropriate kinetochore-microtubule attachments.
ZW10 is located in the cytoplasm and endoplasmic reticulum (ER) during the interphase and is involved in transport between the ER and Golgi apparatus (20). During mitosis, ZW10 and KNTC1 are recruited to the kinetochore, where they form the RZZ complex with ZWILCH (21). When KNTC1 is present, the RZZ complex activates the SAC and recruits dynein/dynactin during mitosis. Therefore, it was hypothesized that ZW10 may compensate for the loss of function of KNTC1. However, ZW10 protein expression levels were not altered when KNTC1 was knocked down in all cell lines (Fig. 6). This finding is consistent with the findings of a previous study demonstrating that ZW10 protein levels were not altered by the expression of a mutant KNTC1 gene, which severely affected the localization of ZW10 in Drosophila (22); this suggests that ZW10 does not compensate for the function of KNTC1. This finding also suggests that there may be no association between ZW10 and CIN.
The overexpression of the KNTC1 gene has been observed in several types of cancer, suggesting that KNTC1 promotes cell proliferation and viability (23-25). However, in the present study, no inhibition of cell proliferation or increase in apoptotic bodies were observed following KNTC1 knockdown in KATOIII or MKN74 cells (Data S1 and Fig. S2), although increases in lagging chromosomes were observed. In the NCI-N87 cells, differences in cell proliferation were observed at 72 h following KNTC1 knockdown, but no apoptosis was observed. This difference suggests that the role of the KNTC1 gene may vary by cancer type. In fact, mRNA expression data from The Human Protein Atlas revealed poor survival rates of patients with GC exhibiting a low expression of KNTC1 (Data S1 and Fig. S3). Previous findings that the suppression of the expression of the KNTC1 gene contributes to CIN may support these data (26-28). In the future, additional large-scale studies using clinical samples are warranted to clarify the association between the KNTC1 gene and CIN and its causal association with patient prognosis.
In conclusion, the present study demonstrated that the knockdown of KNTC1 increased the frequency of lagging chromosomes in GC cells. These finding suggest that the suppression of the expression of KNTC1 may contribute to CIN in GC.
Supplementary Material
Giemsa-stained image of the lagging chromosome in NCI-N87 cells. Chromosomes remained on the metaphase plate and formed bridges in anaphase (red arrows).
Cell proliferation following KNTC1 knockdown. (A) No decrease in cell proliferation was observed in the MKN74, KATOIII and TIG-1-20 cells following KNTC1 knockdown, whereas a decrease in cell proliferation was observed in the NCI-N87 cells. All data were analyzed using the Student's t-test. *P<0.05 vs. siControl. (B) No increase in apoptosis was observed following KNTC1 knockdown in all four cell lines. DAPI staining; 60X objective. KNTC1, kinetochore-associated 1 gene.
Kaplan-Meier plot showing KNTC1 mRNA expression levels and outcomes of prognoses in patients with gastric cancer. In gastric cancer, a low KNTC1 expression was found to be associated with a poor prognosis. P=0.047, low expression vs. high expression. Kaplan-Meier plots were obtained from The Human Protein Atlas version 23.0) (https://www.proteinatlas.org/ENSG00000184445-KNTC1/cancer/stomach+cancer#STAD_TCGA). KNTC1, kinetochore-associated 1 gene.
Supplementary materials and methods
Acknowledgements
Not applicable.
Funding
Funding: No funding was received.
Availability of data and materials
The data generated in the present study may be requested from the corresponding author.
Authors' contributions
KK conceptualized the study. DO analyzed the data and wrote the original draft of the manuscript. KK reviewed and edited the original draft of the manuscript. KK and DO confirm the authenticity of all the raw data. Both authors have read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Sung H, Ferlay J, Siegel RL, Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI View Article : Google Scholar | |
Sato Y, Okamoto K, Kawano Y, Kasai A, Kawaguchi T, Sagawa T, Sogabe M, Miyamoto H and Takayama T: Novel biomarkers of gastric cancer: Current research and future perspectives. J Clin Med. 12(4646)2023.PubMed/NCBI View Article : Google Scholar | |
Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 513:202–209. 2014.PubMed/NCBI View Article : Google Scholar | |
Ignatova EO, Kozlov E, Ivanov M, Mileyko V, Menshikova S, Sun H, Fedyanin M, Tryakin A and Stilidi I: Clinical significance of molecular subtypes of gastrointestinal tract adenocarcinoma. World J Gastrointest Oncol. 14:628–645. 2022.PubMed/NCBI View Article : Google Scholar | |
Sohn BH, Hwang JE, Jang HJ, Lee HS, Oh SC, Shim JJ, Lee KW, Kim EH, Yim SY, Lee SH, et al: Clinical significance of four molecular subtypes of gastric cancer identified by the cancer genome atlas project. Clin Cancer Res. 23:4441–4449. 2017.PubMed/NCBI View Article : Google Scholar | |
Nemtsova MV, Kuznetsova EB and Bure IV: Chromosomal instability in gastric cancer: Role in tumor development, progression, and therapy. Int J Mol Sci. 24(16961)2023.PubMed/NCBI View Article : Google Scholar | |
Grillo F, Fassan M, Sarocchi F, Fiocca R and Mastracci L: HER2 heterogeneity in gastric/gastroesophageal cancers: From benchside to practice. World J Gastroenterol. 22:5879–5887. 2016.PubMed/NCBI View Article : Google Scholar | |
Wakatsuki T, Yamamoto N, Sano T, Chin K, Kawachi H, Takahari D, Ogura M, Ichimura T, Nakayama I, Osumi H, et al: Clinical impact of intratumoral HER2 heterogeneity on trastuzumab efficacy in patients with HER2-positive gastric cancer. J Gastroenterol. 53:1186–1195. 2018.PubMed/NCBI View Article : Google Scholar | |
Gassmann R, Essex A, Hu JS, Maddox PS, Motegi F, Sugimoto A, O'Rourke SM, Bowerman B, McLeod I, Yates III JR, et al: A new mechanism controlling kinetochore–microtubule interactions revealed by comparison of two dynein-targeting components: SPDL-1 and the Rod/Zwilch/Zw10 complex. Genes Dev. 22:2385–2399. 2008.PubMed/NCBI View Article : Google Scholar | |
Lara-Gonzalez P, Westhorpe FG and Taylor SS: The spindle assembly checkpoint. Curr Biol. 22:R966–R980. 2012.PubMed/NCBI View Article : Google Scholar | |
Vallee RB, Varma D and Dujardin DL: ZW10 function in mitotic checkpoint control, dynein targeting, and membrane trafficking: Is dynein the unifying theme? Cell Cycle. 5:2447–2451. 2006.PubMed/NCBI View Article : Google Scholar | |
Cmentowski V, Ciossani G, d'Amico E, Wohlgemuth S, Owa M, Dynlacht B and Musacchio A: RZZ-Spindly and CENP-E form an integrated platform to recruit dynein to the kinetochore corona. EMBO J. 42(e114838)2023.PubMed/NCBI View Article : Google Scholar | |
Pereira C, Reis RM, Gama JB, Celestino R, Cheerambathur DK, Carvalho AX and Gassmann R: Self-assembly of the RZZ complex into filaments drives kinetochore expansion in the absence of microtubule attachment. Curr Biol. 28:3408–3421.e8. 2018.PubMed/NCBI View Article : Google Scholar | |
Barbosa J, Conde C and Sunkel C: RZZ-SPINDLY-DYNEIN: You got to keep 'em separated. Cell Cycle. 19:1716–1726. 2020.PubMed/NCBI View Article : Google Scholar | |
Zhang G, Lischetti T, Hayward DG and Nilsson J: Distinct domains in Bub1 localize RZZ and BubR1 to kinetochores to regulate the checkpoint. Nat Commun. 6(7162)2015.PubMed/NCBI View Article : Google Scholar | |
Lara-Gonzalez P, Pines J and Desai A: Spindle assembly checkpoint activation and silencing at kinetochores. Semin Cell Dev Biol. 117:86–98. 2021.PubMed/NCBI View Article : Google Scholar | |
Scaërou F, Aguilera I, Saunders R, Kane N, Blottière L and Karess R: The rough deal protein is a new kinetochore component required for accurate chromosome segregation in Drosophila. J Cell Sci. 112:3757–3768. 1999.PubMed/NCBI View Article : Google Scholar | |
Williams BC, Li Z, Liu S, Williams EV, Leung G, Yen TJ and Goldberg ML: Zwilch, a new component of the ZW10/ROD complex required for kinetochore functions. Mol Biol Cell. 14:1379–1391. 2003.PubMed/NCBI View Article : Google Scholar | |
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar | |
Hirose H, Arasaki K, Dohmae N, Takio K, Hatsuzawa K, Nagahama M, Tani K, Yamamoto A, Tohyama M and Tagaya M: Implication of ZW10 in membrane trafficking between the endoplasmic reticulum and Golgi. EMBO J. 23:1267–1278. 2004.PubMed/NCBI View Article : Google Scholar | |
Défachelles L, Raich N, Terracol R, Baudin X, Williams B, Goldberg M and Karess RE: RZZ and Mad1 dynamics in Drosophila mitosis. Chrom Res. 23:333–342. 2015.PubMed/NCBI View Article : Google Scholar | |
Williams BC and Goldberg ML: Determinants of drosophila zw10 protein localization and function. J Cell Sci. 107:785–798. 1994.PubMed/NCBI View Article : Google Scholar | |
Zhengxiang Z, Yunxiang T, Zhiping L and Zhimin Y: KNTC1 knockdown suppresses cell proliferation of colon cancer. 3 Biotech. 11(262)2021.PubMed/NCBI View Article : Google Scholar | |
Liu L, Chen H, Chen X, Yao C, Shen W and Jia C: KNTC1 as a putative tumor oncogene in pancreatic cancer. J Cancer Res Clin Oncol. 149:3023–3031. 2023.PubMed/NCBI View Article : Google Scholar | |
Liu CT, Min L, Wang YJ, Li P, Wu YD and Zhang ST: shRNA-mediated knockdown of KNTC1 suppresses cell viability and induces apoptosis in esophageal squamous cell carcinoma. Int J Oncol. 54:1053–1060. 2019.PubMed/NCBI View Article : Google Scholar | |
Bakhoum SF and Cantley LC: The multifaceted role of chromosomal instability in cancer and its microenvironment. Cell. 174:1347–1360. 2018.PubMed/NCBI View Article : Google Scholar | |
Bakhoum SF, Ngo B, Laughney AM, Cavallo JA, Murphy CJ, Ly P, Shah P, Sriram RK, Watkins TBK, Taunk NK, et al: Chromosomal instability drives metastasis through a cytosolic DNA response. Nature. 553:467–472. 2018.PubMed/NCBI View Article : Google Scholar | |
Sansregret L, Vanhaesebroeck B and Swanton C: Determinants and clinical implications of chromosomal instability in cancer. Nat Rev Clin Oncol. 15:139–150. 2018.PubMed/NCBI View Article : Google Scholar |