Purpose:

Chromosomal instability (CIN) is a common phenomenon in colorectal cancer, but its role and underlying cause remain unknown. We have identified that mitotic regulator microtubule-associated protein 9 (MAP9) is a critical regulator of CIN in colorectal cancer. We thus studied the effect of MAP9 loss on colorectal cancer in Map9-knockout mice and in cell lines.

Experimental Design:

We generated colon epithelial–specific Map9-knockout mice and evaluated colorectal cancer development. Effect of Map9 knockout on colorectal cancer progression was determined in chemical or ApcMin/+-induced colorectal cancer. Molecular mechanism of MAP9 was determined using spectral karyotyping, microtubule assays, and whole-genome sequencing (WGS). Clinical significance of MAP9 was examined in 141 patients with CRC.

Results:

Spontaneous colonic tumors (9.1%) were developed in colon epithelium–specific Map9-knockout mice at 17 months, but none was observed in wild-type littermates. Map9 deletion accelerated colorectal cancer formation both in ApcMin/+ mice and azoxymethane-treated mice, and reduced survival in ApcMin/+ mice. Mechanistically, MAP9 stabilized microtubules and mediated mitotic spindle assembly. MAP9 also maintained the spindle pole integrity and protected K-fiber from depolymerization at spindle poles. MAP9 loss induced severe mitosis failure, chromosome segregation errors, and aneuploidy, leading to transformation of normal colon epithelial cells. WGS confirmed enhanced CIN in intestinal tumors from Map9 knockout ApcMin/+ mice. In patients with colorectal cancer, MAP9 was frequently silenced and its downregulation was associated with poor survival.

Conclusions:

MAP9 is a microtubule stabilizer that contributes to spindle stability and inhibits colorectal tumorigenesis, supporting the role of MAP9 as a tumor suppressor for preventing CIN in colorectal cancer.

Translational Relevance

Chromosomal instability (CIN) is a major type of genomic instability that occurs at a high frequency (65%–70%) in colorectal cancer. However, its molecular basis remains poorly understood. In this study, we identified the MAP9 as a spindle-associated protein that plays a gatekeeper role in colorectal cancer development. Intestinal-specific Map9 knockout in mice is sufficient to induce CIN and spontaneous colorectal tumorigenesis, as well as promoting tumor progression in chemical- or ApcMin/+-induced colorectal cancer. MAP9 loss initiates CIN and induces transformation of normal colon epithelial cells. In human colorectal cancer, MAP9 is frequently silenced, and its loss is associated with poorer survival. These data highlight the role of MAP9 inactivation as an important early event in malignant transformation of colorectal cancer.

Colorectal cancer is the third most common cancer worldwide and the second most common overall cause of cancer death (1, 2). Nevertheless, the mechanism leading to colorectal cancer initiation and development remains elusive. Chromosomal instability (CIN) is a major type of genomic instability, in which either the whole or parts of chromosomes are duplicated or deleted, leading to a range of karyotypic abnormalities (3). One main form of CIN, the recurrent missegregation of whole chromosomes during cell division, leads to aneuploidy, a hallmark of most solid tumors especially colorectal cancer (4). CIN can arise from a myriad of events, such as failed correction of chromosome error, an elevated formation of improper chromosome spindle attachment caused by abnormal spindle microtubule dynamics and/or spindle geometry defects (3). CIN contributes to disease progression at multiple stages of tumor evolution and a potential therapeutic target for intervention (5). Despite the importance of CIN to tumorigenesis, underlying mechanisms that drive CIN and its precise role in cancer initiation and development remain poorly understood.

Through a genome-wide screening, we identified that microtubule-associated protein 9 (MAP9), also known as aster-associated protein, was frequently inactivated in colorectal cancer. MAP9 localizes to microtubules in interphase and associates with the mitotic spindle during the whole process of mitosis, and the loss of MAP9 induced severe mitotic defects with delayed mitotic progression (6). However, the role of MAP9 in induction of CIN and oncogenic transformation is unknown. In this study, we demonstrated for the first time that MAP9 is an indispensable spindle microtubule stabilizer and chromosomal fidelity in the colonic epithelium. Knockout of Map9 triggers mitosis failure, aneuploidy, and CIN, leading to spontaneous colorectal cancer development in mice. MAP9 is commonly silenced in human colorectal cancer and is associated with poor survival of patients with colorectal cancer.

Primary colorectal cancer tumor and normal tissue samples

A total of 141 patients with histologically confirmed colorectal cancer who underwent surgery at the Beijing University Cancer Hospital (Beijing, China) were enrolled in the study. Biopsy samples from primary colorectal cancer tumor and adjacent normal were obtained from patients with colorectal cancer prior to any therapeutic intervention. In addition, 20 age-matched normal colon mucosae from healthy subjects were collected as normal control. All the patients provided informed consent for collecting specimens for study. The study protocols were approved by the Clinical Research Ethics Committee of Prince of Wales Hospital and the Chinese University of Hong Kong. This study was carried out in accordance with the Declaration of Helsinki of the World Medical Association. For survival analysis, the tumor/normal ratio of MAP9 mRNA was calculated as follows: |${{\rm{2}}^{( {\Delta \Delta {C_{\rm{t}}}( {{\rm{Tumor }}( {\Delta {C_{\rm{t}}}( {MAP9\, - \,\,\beta \hyphen ACTIN} )} ){\rm{ }} - {\rm{ Normal}}( {\Delta {C_{\rm{t}}}( {MAP9{\rm{ }} - {\rm{ }}\beta \hyphen ACTIN} )} )} )} )}}$|⁠. If tumor/normal ratio < 1, patients were defined as low MAP9 expression group. If tumor/normal ratio > 1, patients were defined as high MAP9 expression group.

Spectral karyotyping analysis

Spectral karyotyping (SKY) analysis was used to uncover chromosomal abnormalities (7). Briefly, metaphase spreads were prepared on glass slides, denatured, and hybridized with denatured SKY probes (SkyPaint mixture, Applied Spectral Imaging).

Microtubule regrowth assay

Microtubule regrowth assay was performed as described previously (8). To completely depolymerize microtubules, 3 μmol/L nocodazole (Sigma-Aldrich) was added to cells for 3 hours. Nocodazole was then washed out with prewarmed culture medium and regrowth of microtubules and spindle reassembly were observed by live-cell imaging.

Microtubule cold-stable assays

Microtubule cold-stable assay was performed as described previously (9). Briefly, cells were arrested in mitosis with 0.32 μmol/L nocodazole (Sigma-Aldrich) for 2.5 hours and released into 10 μmol/L MG132 (Sigma-Aldrich) for 40 minutes to enrich metaphase cells. Then cells were incubated for 10 minutes on ice before fixation and immunofluorescence staining.

Tubulin poleward flux experiment

Kinetochore microtubule turnover was determined as described previously (10). Briefly, cells stably expressing photoactivatable GFP (PA-GFP)-tubulin were incubated with 5 μmol/L DRAQ5 DNA dye (Abcam) for 5 minutes, which is a cell-permeable far-red fluorescent DNA dye for live cells. Spindles with metaphase plate were identified with the DRAQ5 signal. PA-GFP-tubulin was activated in thin stripes (2 pixels wide) on one side of the metaphase plate for 15 seconds from a 405 nm laser (100%). GFP fluorescence was captured every 5 seconds for 300 seconds. The poleward microtubule flux rate was manually calculated.

Generation of constitutive Map9Δ/+ and Map9Δ/Δ mice

Map9-deficient mice containing a “knockout-first” allele were generated by Beijing Biocytogen Co., Ltd (Supplementary Fig. S1A and S1B; ref. 11). The “knockout-first” conditional allele was generated by inserting a gene trap cassette through homologous recombination in mouse embryonic stem cells into intron 4 of mouse Map9 gene, leading to disruption of Map9 gene. Cassettes were removed by Flp-mediated excision by crossing with mice expressing Flp (B6.129S4-Gt(ROSA)26Sortm1(FLP1)Dym/RainJ; Beijing Biocytogen Co., Ltd), resulting in a floxed Map9 allele, in which the critical exon 5 of Map9 is flanked by loxP sites (Map9flox/+). To generate constitutive Map9-knockout mice (Map9Δ/+ and Map9Δ/Δ), germline Cre-mediated excision was performed by cross-mating Map9flox/flox mice with mice harboring the cytomegalovirus (CMV) promoter−Cre (CMV-Cre; B6.C-Tg(CMV-cre)1Cgn/J; Beijing Biocytogen Co. Ltd). Genotyping was performed by PCR of tail-snip DNA using genotyping primers (Supplementary Fig. S1C; Supplementary Table S1). Absence of Map9 mRNA in Map9Δ/Δ mice was confirmed by qPCR (Supplementary Fig. S1D). All mice were housed in a pathogen-free barrier environment for the duration of the study. All experimental procedures were approved by the Animal Ethics Committee of the Army Medical University (Chongqing, China).

Generation of Map9-deficient mouse embryonic fibroblasts

The isolation of mouse embryonic fibroblasts (MEF) was performed as described previously (12, 13). Map9Δ/+ and Map9Δ/Δ MEFs were isolated from embryonic day 13–14 fetuses after intercross of constitutive Map9Δ/+ mice. Wild-type (WT) MEFs were obtained by cross-mating WT mice.

Generation of intestinal-specific Map9-knockout mouse

CDX2-CreERT2 transgenic mice was purchased from The Jackson Laboratory, which express Cre-ERT2 in adult epithelium of the distal intestinal tract under the direction of caudal type homeobox 2 (CDX2) gene promoter/enhancer regions. Cre-ERT2 can only gain access to the nuclear compartment after exposure to tamoxifen. Map9flox/flox mice were bred to CDX2-CreERT2 to generate Map9flox/+CDX2-CreERT2 mice and Map9flox/floxCDX2-CreERT2 mice (intestinal-specific Map9−/+ and Map9−/− mice; Supplementary Fig. S1A–S1C). All experimental procedures were approved by the Animal Ethics Committee of the Army Medical University (Chongquing, China).

Azoxymethane-induced colorectal cancer model

To induce colon epithelium–specific deletion of Map9, 7-week-old intestinal-specific Map9−/+ and Map9−/− mice were intraperitoneally injected with tamoxifen (Sigma-Aldrich) dissolved in corn oil (100 mg/kg) for 4 consecutive days. Two weeks after first tamoxifen dose, mice were injected intraperitoneally with 10 mg/kg azoxymethane (AOM; Sigma-Aldrich) once a week for 6 consecutive weeks (14). Mice were sacrificed 25 weeks after the first dosing of AOM and subjected to a complete necropsy. Intestine examination, tumor measurement, and histology inspection were performed. All experimental procedures were approved by the Animal Ethics Committee of the Army Medical University (Chongquing, China).

Other additional experimental procedures are provided in the Supplementary Information.

MAP9 is commonly silenced in colorectal cancer

MAP9 mRNA expression was downregulated in 68% (96/141) of primary colorectal cancer tissues as compared with their adjacent normal tissues (P < 0.0001; Supplementary Fig. S2A). We analyzed MAP9 mRNA levels in the Broad Institute Cancer Cell Line Encyclopedia (CCLE) database. Among 58 colorectal cancer cell lines in CCLE, there are only 4 colorectal cancer cell lines with MAP9 mRNA reads per kilobase per million mapped reads > 1 (SNU503, SNUC4, SNU1033, CW2; 6.9%; Supplementary Fig. S2B). By qRT-PCR, MAP9 mRNA was silenced in all 7 commercial human colorectal cancer cell lines (CL-14, DLD-1, HCT116, HT-29, LS 180 SW480, and SW620) and in all 5 primary human-derived colorectal cancer cells (colorectal cancer spheroid POP92 and POP66, colorectal cancer organoid PDO 828, PDO 1482, PDO 816), but was readily expressed in normal human colon tissues and two immortalized normal human colonic epithelial cells HCEC 1CT (1CT), HCEC 2CT (2CT; Supplementary Fig. S2C). The downregulation of MAP9 expression was validated in The Cancer Genome Atlas (TCGA) cohort of patients with colorectal cancer (Supplementary Fig. S2D).

Map9 knockout drives spontaneous colorectal tumorigenesis in mice

Given it is downregulated in colorectal cancer, we hypothesized that MAP9 might function as a tumor suppressor in colorectal cancer. We generated intestinal epithelium–specific, inducible Map9-knockout (Map9−/−) mice by crossing Map9flox/flox mice to CDX2-CreERT2 mice (Fig. 1A; Supplementary Fig. S1A–S1C; ref. 15). Tamoxifen injection activates CreERT2, leading to excision of floxed exon 5 in the intestinal epithelium (Supplementary Fig. S1A). Cre-mediated excision of the floxed exon 5 results in a frameshift and a premature stop codon in exon 6, generating truncated MAP9 protein lacking C terminus, a domain essential for microtubule-binding ability of MAP9 (Supplementary Fig. S1B; ref. 6). At 15 months postinjection, intestinal-specific Map9−/− mice spontaneously developed colonic dysplasia (5/22, 22.7%) and colon cancer (2/22, 9.1%; Fig. 1B and C). Moreover, intestinal-specific heterozygous Map9+/− mice also developed colonic dysplasia (4/21, 19.0%), whereas their 23 WT littermates (Map9+/+) showed normal colon histology at the same age (Fig. 1C).

Figure 1.

Map9 deficiency drives colon tumorigenesis in mice. A, Scheme for the experimental design of three transgenic mice models. B, Scheme for the experimental course of Map9 spontaneous colorectal cancer model. C, Representative hematoxylin and eosin staining of colon harvested at 17 months of age from Map9 spontaneous colorectal cancer model. D, The average body weight, intestinal tumor number, and intestinal tumor burden from ApcMin/+, Map9Δ/+ApcMin/+, Map9Δ/ΔApcMin/+ mice at 3 months of age in ApcMin/+ mice model. Tumor burden was calculated as the sum of average diameter of all intestinal tumors in each mouse. E, Dissection micrographs of representative intestines and hematoxylin and eosin–stained sections of tumors from ApcMin/+, Map9Δ/+ApcMin/+, Map9Δ/ΔApcMin/+ mice at 3 months of age in ApcMin/+ mice model. F, Kaplan–Meier overall survival curves of ApcMin/+ and Map9Δ/ΔApcMin/+ mice. *, P = 0.016 compared with ApcMin/+ mice (log rank test). G, Scheme for the experimental course of AOM-induced colon cancer model. Two weeks after the first dose of tamoxifen, mice were injected with the carcinogen AOM once a week for 6 consecutive weeks. Twenty-five weeks after the first dose of AOM, mice were analyzed for colon tumor formation. H, The average body weight, colon tumor number, and colon tumor burden from WT, intestinal-specific Map9−/− and Map9−/+ mice at the endpoint in AOM model. I, Dissection micrographs of representative intestines and hematoxylin and eosin staining of colon tumors from WT, intestinal epithelium–specific Map9−/− and Map9−/+ mice at the endpoint in AOM-induced colon cancer model. All histogram data represent mean ± SD. *, P < 0.05; **, P < 0.01. NS, not significant.

Figure 1.

Map9 deficiency drives colon tumorigenesis in mice. A, Scheme for the experimental design of three transgenic mice models. B, Scheme for the experimental course of Map9 spontaneous colorectal cancer model. C, Representative hematoxylin and eosin staining of colon harvested at 17 months of age from Map9 spontaneous colorectal cancer model. D, The average body weight, intestinal tumor number, and intestinal tumor burden from ApcMin/+, Map9Δ/+ApcMin/+, Map9Δ/ΔApcMin/+ mice at 3 months of age in ApcMin/+ mice model. Tumor burden was calculated as the sum of average diameter of all intestinal tumors in each mouse. E, Dissection micrographs of representative intestines and hematoxylin and eosin–stained sections of tumors from ApcMin/+, Map9Δ/+ApcMin/+, Map9Δ/ΔApcMin/+ mice at 3 months of age in ApcMin/+ mice model. F, Kaplan–Meier overall survival curves of ApcMin/+ and Map9Δ/ΔApcMin/+ mice. *, P = 0.016 compared with ApcMin/+ mice (log rank test). G, Scheme for the experimental course of AOM-induced colon cancer model. Two weeks after the first dose of tamoxifen, mice were injected with the carcinogen AOM once a week for 6 consecutive weeks. Twenty-five weeks after the first dose of AOM, mice were analyzed for colon tumor formation. H, The average body weight, colon tumor number, and colon tumor burden from WT, intestinal-specific Map9−/− and Map9−/+ mice at the endpoint in AOM model. I, Dissection micrographs of representative intestines and hematoxylin and eosin staining of colon tumors from WT, intestinal epithelium–specific Map9−/− and Map9−/+ mice at the endpoint in AOM-induced colon cancer model. All histogram data represent mean ± SD. *, P < 0.05; **, P < 0.01. NS, not significant.

Close modal

Map9 knockout accelerates colorectal tumorigenesis in ApcMin/+ mice and in AOM-treated C57/BL6 mice

To confirm the role of Map9 deficiency in promoting colorectal tumorigenesis, constitutive Map9-knockout (Map9Δ/Δ) mice were cross-mated with ApcMin/+ mice, a mouse model of colorectal tumorigenesis (Fig. 1A). As ApcMin/+ mice typically develop intestinal adenomas by 3 months of age, we investigated whether Map9 deficiency can promote tumorigenesis in the ApcMin/+ mice model (Fig. 1A). Macroscopic examination of mice at 3 months of age revealed that WT (Apc+/+), Map9Δ/+ (Map9Δ/+Apc+/+), or Map9Δ/Δ (Map9Δ/ΔApc+/+) mice developed no visible tumors, whereas 50% ApcMin/+ mice (5/10) developed tumors (3/10 with multiple tumors; Fig. 1D and E; Supplementary Fig. S3; Supplementary Table S2). Importantly, nearly all age-matched Map9Δ/+ApcMin/+ and Map9Δ/ΔApcMin/+ mice developed intestinal tumors (Map9Δ/+ApcMin/+: 2/10 with multiple tumors; Map9Δ/ΔApcMin/+: 5/8 with multiple tumors; Fig. 1D and E; Supplementary Table S2). Map9Δ/ΔApcMin/+ mice had 3-fold more tumors than ApcMin/+ mice, accompanied by significant body weight loss (Fig. 1D). Average tumor burden (sum of all tumor size per mouse) in Map9Δ/ΔApcMin/+ mice was two times greater than in ApcMin/+ mice (Fig. 1D; ref. 14). Map9Δ/ΔApcMin/+ mice showed markedly decreased overall survival compared with ApcMin/+ mice (P = 0.016; Fig. 1F).

Intestinal-specific Map9−/−, Map9−/+, and WT mice were treated with colonic carcinogen AOM to induce colorectal carcinogenesis (Fig. 1A and G; ref. 14). Tumor incidence was significantly higher in Map9−/− mice (100%) and Map9−/+ mice (100%) compared with WT mice (67%; Supplementary Table S2). Both intestinal-specific Map9−/+ and Map9−/− mice had a significant increase in tumor number and burden compared with control mice (Fig. 1H and I). These findings further confirmed that Map9 knockout promoted colon tumorigenesis in mice.

MAP9 deficiency causes mitosis failure

In light of the observed tumor-promoting effect of Map9 deficiency in mice, we investigated the molecular mechanisms of MAP9 in colon cancer. The subcellular localization of MAP9 was examined in the immortalized normal human colonic epithelial cells 1CT, 2CT, and immortalized normal human gastric epithelial GES1 cells using Flag-tagged MAP9. MAP9 colocalized with α-tubulin in interphase as well as during the whole process of mitosis in all three cell lines (Fig. 2A; Supplementary Fig. S4A). The colocalization of MAP9 with microtubules was confirmed by super-resolution fluorescence imaging analysis under stochastic optical reconstitution microscopy (STORM; Fig. 2B). In addition, MAP9 closely surrounded the pericentriolar γ-tubulin ring complex (γ-TuRC; Fig. 2C), an essential component of pericentriolar material (PCM) targeted to centrosomes (16). From these findings, we inferred that MAP9 might function as a microtubule-associated protein involved in chromosomal microtubule assembly and dynamics. We thus tested the function of MAP9 in mitosis. Knockdown of MAP9 in 1CT, 2CT, and GES1 cells delayed mitotic progression to varying extents as evidenced by increased G2–M phase cells' accumulation (Fig. 2D; Supplementary Fig. S4B).

Figure 2.

MAP9 is a spindle-associated protein and regulates the process of mitosis in intestinal epithelial cells. A, Subcellular localization of Flag-tagged MAP9 in metaphase and anaphase in 1CT cells. Scale bars, 5 μm. B, 1CT cells in interphase imaged with STORM. Scale bars, 1,000 nm. C, MAP9 tightly surrounded γ-TuRC. D, MAP9 knockdown led to cell-cycle arrest at G2–M phase, as indicated by flow cytometry. Three replicates were performed, with 1 × 105 cells analyzed per replicate. All histogram data represent mean ± SD (*, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001. NS, not significant).

Figure 2.

MAP9 is a spindle-associated protein and regulates the process of mitosis in intestinal epithelial cells. A, Subcellular localization of Flag-tagged MAP9 in metaphase and anaphase in 1CT cells. Scale bars, 5 μm. B, 1CT cells in interphase imaged with STORM. Scale bars, 1,000 nm. C, MAP9 tightly surrounded γ-TuRC. D, MAP9 knockdown led to cell-cycle arrest at G2–M phase, as indicated by flow cytometry. Three replicates were performed, with 1 × 105 cells analyzed per replicate. All histogram data represent mean ± SD (*, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001. NS, not significant).

Close modal

MAP9 functions as microtubule stabilizer and is essential for mitotic spindle assembly and stability

We next investigated underlying role of MAP9 on cell mitosis in 1CT, 2CT, and GES1 cells. We observed that bipolar spindles in metaphase were significantly shorter in MAP9-knockdown cells (Fig. 3A); conversely, ectopic expression of MAP9 led to longer bipolar spindles (Fig. 3B), indicating that MAP9 has microtubule-stabilizing activity (17). Notably, monopolar spindle formation was induced after ectopic expression of MAP9 in metaphase cells (1CT: 62.6%; GES1: 53.8%). This phenomenon was also observed in a previous study (6), which is probably due to the toxic effect for the cells under high constitutive expression of MAP9. As MAP9 colocalized with chromosomal microtubules during mitosis, we carried out a microtubule regrowth assay to examine its role in spindle assembly by live-cell imaging using α-tubulin-monomeric red fluorescent protein (mRFP)–expressing cells. In control cells, more than two microtubule asters were formed within 5 minutes regrowth after nocodazole release, which then quickly coalesced and progressively organized into a bipolar spindle (Fig. 3C and D; Supplementary Fig. S4C; Supplementary Video S1). However, in MAP9-knockdown cells, two or less microtubule asters appeared within same time frame with less densely packed and short microtubules, thus forming a much smaller spindle (Fig. 3C and D; Supplementary Fig. S4C; Supplementary Video S2). In addition, in approximately 20% of MAP9-knockdown GES1 cells, the spindle collapsed along with PCM fragmentation (Supplementary Fig. S4C). Hence, MAP9 is required for proper spindle assembly and spindle stability.

Figure 3.

MAP9 acts as microtubule stabilizer and regulates spindle dynamics. A, Quantification of metaphase spindle length in MAP9-knockdown and control cells. The metaphase bipolar spindles assembled in MAP9-knockdown cells were significantly shorter than in controls. Metaphase cells containing bipolar spindles of three experiments (N = 100). B, Quantification of the metaphase spindle length in MAP9-overexpressing (OE) and control GES1 cells. Metaphase cells containing bipolar spindles of three experiments (N = 150). C, Representative live-cell images of control and MAP9-knockdown 1CT cells expressing α-tubulin-mRFP after nocodazole washout. Green arrows indicated the formed microtubule asters. D, Quantifications of the number of microtubule asters per cell monitored at the indicated times after nocodazole washout. E, Representative live-cell images of control and MAP9-knockdown 1CT cells coexpressing H2B-GFP and RFP-tubulin during mitosis. F, Spindle length and chromosome thickness in 1CT cells coexpressing H2B-eGFP and RFP-tubulin during mitosis at the indicated times. The bipolar spindle in MAP9-knockdown cells collapsed (51, 75, 126, 159, 174, 216, and 237 minutes) along with chromosome decondensation 7 times within 4-hour observation. All scale bars, 5 μm. All histogram data represent mean ± SD. Compared with control group (****, P < 0.0001).

Figure 3.

MAP9 acts as microtubule stabilizer and regulates spindle dynamics. A, Quantification of metaphase spindle length in MAP9-knockdown and control cells. The metaphase bipolar spindles assembled in MAP9-knockdown cells were significantly shorter than in controls. Metaphase cells containing bipolar spindles of three experiments (N = 100). B, Quantification of the metaphase spindle length in MAP9-overexpressing (OE) and control GES1 cells. Metaphase cells containing bipolar spindles of three experiments (N = 150). C, Representative live-cell images of control and MAP9-knockdown 1CT cells expressing α-tubulin-mRFP after nocodazole washout. Green arrows indicated the formed microtubule asters. D, Quantifications of the number of microtubule asters per cell monitored at the indicated times after nocodazole washout. E, Representative live-cell images of control and MAP9-knockdown 1CT cells coexpressing H2B-GFP and RFP-tubulin during mitosis. F, Spindle length and chromosome thickness in 1CT cells coexpressing H2B-eGFP and RFP-tubulin during mitosis at the indicated times. The bipolar spindle in MAP9-knockdown cells collapsed (51, 75, 126, 159, 174, 216, and 237 minutes) along with chromosome decondensation 7 times within 4-hour observation. All scale bars, 5 μm. All histogram data represent mean ± SD. Compared with control group (****, P < 0.0001).

Close modal

We next examined the consequence of MAP9 loss on mitosis following spindle assembly by live-cell imaging. Control cells quickly assembled a bipolar spindle and completed mitosis within 1.5 hours (Fig. 3E and F; Supplementary Videos S3 and S4). The spindle length of control cells was kept stable in metaphase. Mitotic chromosomes then underwent condensation and became fully condensed prior to the segregation of chromosomes. In contrast, MAP9-knockdown cells remained arrested in mitosis during the 4-hour observation period (Fig. 3E and F; Supplementary Videos S5 and S6). The spindle length of MAP9-knockdown cells in metaphase was approximately half of that in control cells. Although the formation of bipolar spindle and the alignment and condensation of chromosomes were also observed in MAP9-knockdown cells, these spindles were highly unstable and underwent repeated cycles of collapse and reassembly (Fig. 3E and F).

Consistent with the compromised spindle dynamics, more than half of the bipolar spindles in MAP9-knockdown cells were abnormal, presented as asymmetrically curved bending of kinetochore fibers (K-fibers; Supplementary Fig. S5A). We further assessed whether MAP9 affects correction of improper chromosome–spindle attachment using monastrol washout assay. The Eg5 inhibitor monastrol induces monopolar spindles along with many syntelic attachments, a major type of erroneous kinetochore attachments during spindle assembly (18, 19). The percentage of cells without complete chromosome alignment on the metaphase plate was elevated upon release from monastrol in MAP9-knockdown cells (Supplementary Fig. S5B).

MAP9 participates in spindle pole stability

K-fibers are microtubule bundles, of which the minus ends are associated with γ-TuRC in pericentriolar matrix and undergo controlled depolymerization at spindle poles to modulate poleward movement of chromosomes during mitosis (20, 21). Because γ-TuRC was closely surrounded by MAP9, we proposed that MAP9 might affect K-fiber formation and stability. Indeed, we observed that K-fiber length in monopolar spindles assembled in the presence of monastrol was substantially reduced in MAP9-knockdown cells (Fig. 4A and B; Supplementary Fig. S5C). Correspondingly, the percentage of cells with extremely short K-fibers was dramatically increased in MAP9-knockdown cells (Fig. 4B), indicating that MAP9 is required for K-fiber minus-end stability. Consistently, microtubule cold-stable assay showed that K-fibers depolymerized much faster upon cold exposure at their minus ends in MAP9-knockdown cells compared with control cells, with approximately 20% of MAP9-knockdown cells having less than 20 microtubules left, compared with <10% of control cells (Fig. 4C and D; Supplementary Fig. S5D). The minus ends of K-fibers attaching to spindle poles depolymerize continuously while the plus ends undergo polymerization, leading to a characteristic tubulin flux from the plus end toward the minus end (Fig. 4E, left; ref. 22). Using cells stably expressing PA-GFP-tubulin, microtubule poleward flux was measured by tracking marks photobleached into green fluorescent spindles in metaphase (Fig. 4E). As shown in Fig. 4F, poleward flux in metaphase spindles in most of MAP9-knockdown cells quickly moved from the metaphase plate to spindle pole within 90 seconds, whereas no obvious poleward flux in metaphase spindles was observed in control cells. Quantitative evaluation showed that poleward flux rates were significantly higher in the metaphase spindles of MAP9-knockdown cells (Fig. 4G). Thus, in the absence of MAP9, K-fiber minus ends are more prone to depolymerization, leading to unstable and shortened K-fibers undergoing a faster flux (Fig. 4E, right). MAP9 likely has no effect on plus ends, as its disruption would abolish microtubule poleward flux (8). Finally, monastrol washout assay demonstrated a significant increase in spindle pole defocusing and PCM fragmentation in MAP9-knockdown cells (Fig. 4H–J; Supplementary Fig. S5E), further confirming the critical role of MAP9 in spindle pole stability and integrity.

Figure 4.

MAP9 maintains the integrity and stability of spindle pole. A, Top: Schematic diagram of K-fiber length assay. Bottom: Representative immunofluorescence images of monopolar spindles in 1CT cells in K-fiber length assay. B, Top: The percentage of cells with all K-fibers extremely short was assessed in 1CT and GES1 cells. Each group contained three replicates. Each replicate was determined in approximately 100 cells with the induced monopolar spindle. Bottom: K-fiber length was assessed in 1CT and GES1 cells. N = 200 K-fibers from the monopolar spindles of three experiments. C, Top: Schematic diagram of cold-stable microtubule assay. Bottom: Representative immunofluorescence images of metaphase spindles in 1CT cells after cold exposure. D, The graph showed the percentage of 100 metaphase-like cells with intact K-fibers (N > 40), K-fibers remnants (20 < N < 40), or almost no K-fibers (N < 20). E, Schematic diagram of tubulin poleward flux assay. F, Live-cell images showed that the photoactivated marks on K-fibers moved toward the spindle pole in control and MAP9-knockdown 1CT cells. Metaphase spindles were identified with the red fluorescent DRAQ5 DNA dye signal. PA-GFP-tubulin was then activated in the thin stripes just above the metaphase plate. The progression of the photoactivated tubulin toward the spindle pole, which corresponded to the poleward microtubule flux rate, was manually measured. The white dashed lines indicated the locations where PA-GFP-tubulin was activated on the side of the metaphase plate. G, Quantification of the tubulin poleward flux rate in the metaphase spindles in 1CT and GES1 cells. Flux was measured in 25 metaphase cells in each condition. H, Schematic diagram of monastrol washout assay. I, Representative immunofluorescence images of 1CT cells with PCM fragmentation or spindle pole defocusing after monastrol washout. J, The percentage of cells with PCM fragmentation or spindle pole defocusing was assessed after monastrol washout in 1CT and GES1 cells. Each group contained 4 replicates. Each replicate was assessed in 100 metaphase cells. All scale bars, 5 μm. All histogram data represent mean ± SD. Compared with control group (**, P < 0.01; ***, P < 0.001; and ****, P < 0.0001).

Figure 4.

MAP9 maintains the integrity and stability of spindle pole. A, Top: Schematic diagram of K-fiber length assay. Bottom: Representative immunofluorescence images of monopolar spindles in 1CT cells in K-fiber length assay. B, Top: The percentage of cells with all K-fibers extremely short was assessed in 1CT and GES1 cells. Each group contained three replicates. Each replicate was determined in approximately 100 cells with the induced monopolar spindle. Bottom: K-fiber length was assessed in 1CT and GES1 cells. N = 200 K-fibers from the monopolar spindles of three experiments. C, Top: Schematic diagram of cold-stable microtubule assay. Bottom: Representative immunofluorescence images of metaphase spindles in 1CT cells after cold exposure. D, The graph showed the percentage of 100 metaphase-like cells with intact K-fibers (N > 40), K-fibers remnants (20 < N < 40), or almost no K-fibers (N < 20). E, Schematic diagram of tubulin poleward flux assay. F, Live-cell images showed that the photoactivated marks on K-fibers moved toward the spindle pole in control and MAP9-knockdown 1CT cells. Metaphase spindles were identified with the red fluorescent DRAQ5 DNA dye signal. PA-GFP-tubulin was then activated in the thin stripes just above the metaphase plate. The progression of the photoactivated tubulin toward the spindle pole, which corresponded to the poleward microtubule flux rate, was manually measured. The white dashed lines indicated the locations where PA-GFP-tubulin was activated on the side of the metaphase plate. G, Quantification of the tubulin poleward flux rate in the metaphase spindles in 1CT and GES1 cells. Flux was measured in 25 metaphase cells in each condition. H, Schematic diagram of monastrol washout assay. I, Representative immunofluorescence images of 1CT cells with PCM fragmentation or spindle pole defocusing after monastrol washout. J, The percentage of cells with PCM fragmentation or spindle pole defocusing was assessed after monastrol washout in 1CT and GES1 cells. Each group contained 4 replicates. Each replicate was assessed in 100 metaphase cells. All scale bars, 5 μm. All histogram data represent mean ± SD. Compared with control group (**, P < 0.01; ***, P < 0.001; and ****, P < 0.0001).

Close modal

MAP9 protects chromosomal stability

MAP9-knockdown 1CT, 2CT, and GES1 cells exhibited high rate of lagging chromosomes with increased chromosome segregation errors (Fig. 5A). Because aneuploidy is an obligate outcome of CIN, conventional karyotyping was performed to assess MAP9-knockdown–induced aneuploidy. All control cells had a normal modal chromosome copy number with a low percentage of mode deviation (Supplementary Fig. S6A and S6B). In contrast, MAP9-knockdown cells showed a significantly higher percentage of deviation from the mode compared with control cells (Supplementary Fig. S6A and S6B). To further identify numerical and structural chromosome abnormalities induced by MAP9 insufficiency, we performed SKY analysis on 1CT and 2CT cells after stable MAP9 knockdown. As shown in Fig. 5B–D, the aneuploidy frequency was nine times more in MAP9-knockdown 1CT cells (59.3% vs. 6.0%) and two times more in MAP9-knockdown 2CT cells (30.0% vs. 10.7%) compared with control cells. Thus, MAP9 insufficiency in cells clearly results in CIN.

Figure 5.

Knockdown of MAP9 induced chromosomal abnormalities. A, Anaphase cells of MAP9-knockdown and control cells. The percentage of anaphase cells with lagging chromosomes was shown in the right plot. Each group contained three replicates. Each replicate was assessed in approximately 50 anaphase cells. Scale bars, 5 μm. B, Representative SKY metaphases (arrayed chromosomes) from MAP9-knockdown and control 1CT and 2CT cells harvested 10 days after lentiviral transduction. Red arrows indicated chromosome gains, and green arrows indicated chromosome losses. C, Top, the aneuploidy frequency by SKY analysis. Bottom, the distribution of numerical and structural aberrations in MAP9-knockdown and control cells by SKY analysis. D, Tables recapitulating the karyotypes of 150 cells analyzed in each condition by SKY analysis. Each row represented an individual cell. Red represented gain of chromosomes, dark gray represented loss of chromosomes, and orange showed structural aberrations. All histogram data represent mean ± SD. Compared with control group (**, P < 0.01; ***, P < 0.001; and ****, P < 0.0001).

Figure 5.

Knockdown of MAP9 induced chromosomal abnormalities. A, Anaphase cells of MAP9-knockdown and control cells. The percentage of anaphase cells with lagging chromosomes was shown in the right plot. Each group contained three replicates. Each replicate was assessed in approximately 50 anaphase cells. Scale bars, 5 μm. B, Representative SKY metaphases (arrayed chromosomes) from MAP9-knockdown and control 1CT and 2CT cells harvested 10 days after lentiviral transduction. Red arrows indicated chromosome gains, and green arrows indicated chromosome losses. C, Top, the aneuploidy frequency by SKY analysis. Bottom, the distribution of numerical and structural aberrations in MAP9-knockdown and control cells by SKY analysis. D, Tables recapitulating the karyotypes of 150 cells analyzed in each condition by SKY analysis. Each row represented an individual cell. Red represented gain of chromosomes, dark gray represented loss of chromosomes, and orange showed structural aberrations. All histogram data represent mean ± SD. Compared with control group (**, P < 0.01; ***, P < 0.001; and ****, P < 0.0001).

Close modal

To test the effect on CIN under varying MAP9-knockdown efficiencies in vitro, we transfected the 1CT cells with different amounts of MAP9 siRNA. Karyotype analysis revealed no changes in chromosome abnormalities with 20% MAP9 knockdown (Supplementary Fig. S6C). Approximately 50% knockdown led to a moderate increase in aneuploidy, and to a greater extent in cells with >80% knockdown of MAP9 (Supplementary Fig. S6C). Thus, approximately 50% loss of MAP9 could initiate genome abnormalities in normal colon epithelial cells.

We further examined the effects of the loss of Map9 on CIN by using MEFs derived from Map9Δ/+, Map9Δ/Δ, and WT (Map9+/+) mice (Supplementary Fig. S7A–S7C). Conventional karyotyping showed that a large percentage of Map9Δ/+ and Map9Δ/Δ MEFs were aneuploid (70%–80%; Supplementary Fig. S7D; Supplementary Table S3). SKY data confirmed that the percentage of Map9Δ/Δ MEFs with numerical aberrations was almost three times higher than that of WT MEFs (Fig. 6A–C). Increased CIN induced by knockout of Map9 was also evidenced by elevated rates of lagging chromosomes in anaphase of Map9Δ/+ and Map9Δ/Δ MEFs (Fig. 6D and E). Moreover, Map9Δ/+ and Map9Δ/Δ MEFs showed a significant increase in the percentage of cells with detached centrosomes and PCM fragmentation compared with WT MEFs (Fig. 6D and E). Of note, MEF cells appear to be more sensitive to effects of Map9 loss compared with intestinal epithelial cells. The heterozygous knockout led to striking changes on CIN and spindle pole stability, which nearly matches homozygous knockout in MEFs. We reasoned that this may be attributed to differential response of MEF cells from intestinal epithelial cells to Map9 loss, for example, differences in DNA damage repair ability. Collectively, these findings indicate that loss of Map9 is sufficient to induce chromosomal abnormalities in MEF cells.

Figure 6.

Increased CIN and spindle instability in Map9 deficient MEFs. A, Representative images of SKY analysis obtained from Map9Δ/+, Map9Δ/Δ, and WT MEFs (green, chromosome loss; red, chromosome gain). B, Top, the aneuploidy frequency of Map9Δ/Δ and WT MEFs by SKY analysis. Bottom, the distribution of numerical and structural aberrations in Map9Δ/Δ and WT MEFs by SKY analysis. C, Tables recapitulating the karyotypes of 100 cells analyzed in Map9Δ/Δ and WT MEFs by SKY analysis. Each row represented an individual cell. Red represented gain of chromosomes, dark gray represented loss of chromosomes, and orange showed structural aberrations. D, The representative images of spindles in metaphase and anaphase in Map9Δ/+, Map9Δ/Δ, and WT MEFs. Green arrows indicated PCM fragmentation or spindle pole defocusing. Pink arrows indicated the misalignment of chromosomes in metaphase or lagging chromosomes in anaphase. E, Left, quantification of spindle pole defocusing and PCM fragmentation in metaphase in MEFs. Right, quantification of lagging chromosome in anaphase in MEFs. Each group contained three replicates. Each replicate was assessed in approximately 100 metaphase or approximately 50 anaphase MEF cells. F,Map9 insufficiency promoted aneuploidy in mice splenocytes. Left, qPCR analysis of Map9 and p53 mRNA expression in cultured splenocytes obtained from the mice at 3 to 4 months of age. Right, quantification of aneuploidy rate. p53Δ/+ group contained two replicates, and the other groups contained three replicates/group. Each replicate was assessed in 37 to 64 metaphase splenocyte cells. All scale bars, 5 μm. All histogram data represent mean ± SD (*, P < 0.05; **, P < 0.01; and ****, P < 0.0001; NS, not significant).

Figure 6.

Increased CIN and spindle instability in Map9 deficient MEFs. A, Representative images of SKY analysis obtained from Map9Δ/+, Map9Δ/Δ, and WT MEFs (green, chromosome loss; red, chromosome gain). B, Top, the aneuploidy frequency of Map9Δ/Δ and WT MEFs by SKY analysis. Bottom, the distribution of numerical and structural aberrations in Map9Δ/Δ and WT MEFs by SKY analysis. C, Tables recapitulating the karyotypes of 100 cells analyzed in Map9Δ/Δ and WT MEFs by SKY analysis. Each row represented an individual cell. Red represented gain of chromosomes, dark gray represented loss of chromosomes, and orange showed structural aberrations. D, The representative images of spindles in metaphase and anaphase in Map9Δ/+, Map9Δ/Δ, and WT MEFs. Green arrows indicated PCM fragmentation or spindle pole defocusing. Pink arrows indicated the misalignment of chromosomes in metaphase or lagging chromosomes in anaphase. E, Left, quantification of spindle pole defocusing and PCM fragmentation in metaphase in MEFs. Right, quantification of lagging chromosome in anaphase in MEFs. Each group contained three replicates. Each replicate was assessed in approximately 100 metaphase or approximately 50 anaphase MEF cells. F,Map9 insufficiency promoted aneuploidy in mice splenocytes. Left, qPCR analysis of Map9 and p53 mRNA expression in cultured splenocytes obtained from the mice at 3 to 4 months of age. Right, quantification of aneuploidy rate. p53Δ/+ group contained two replicates, and the other groups contained three replicates/group. Each replicate was assessed in 37 to 64 metaphase splenocyte cells. All scale bars, 5 μm. All histogram data represent mean ± SD (*, P < 0.05; **, P < 0.01; and ****, P < 0.0001; NS, not significant).

Close modal

To evaluate the role of Map9 in the context of loss of p53 function, we further investigated the effect of Map9 deficiency on CIN in splenocytes from Map9Δ/+, Map9Δ/Δ, p53Δ/+, Map9Δ/+p53Δ/+, Map9Δ/Δp53Δ/+, and WT mice at 3–4 months of age. Map9 expression was inversely correlated with aneuploidy rate in mouse splenocytes (Fig. 6F; Supplementary Table S3). The aneuploidy frequency of Map9Δ/Δp53Δ/+ compound mutant mice (58%) was significantly higher than either Map9Δ/Δ (39%) or p53Δ/+ (23%) single mutant mice (Fig. 6F). The same trend was observed in Map9Δ/+p53Δ/+ mice (Fig. 6F), suggesting the loss of p53 function exacerbates CIN induced by Map9 knockout. We observed an aneuploidy frequency of 17% in WT splenocytes, similar to that reported by others (23–29).

MAP9 silencing leads to the transformation of normal human colon epithelial cells

CIN has been shown to drive tumorigenic phenotypes such as anchorage-independent growth and metastasis by selectively eliminating or acquiring chromosome segments that harbor the key regulator genes (30, 31). Protumorigenic effect of CIN is a long-term effect, as it requires adaptation to aneuploidy and subsequent tumor evolution (32). We thus first examined the effect of long-term MAP9 silencing (>50 days) on 1CT and 2CT cells. Anchorage-independent growth is a hallmark of cellular transformation (33). Although parental 1CT and 2CT cells did not grow in soft agar (33), multiple colonies were formed in long-term MAP9-knockdown 1CT and 2CT cells (Fig. 7A). Matrigel cell invasion assays also showed that long-term MAP9 knockdown enhanced invasiveness of 1CT and 2CT cells (Fig. 7B). In contrast, short-term MAP9 silencing (3–4 days) failed to elicit neither of these effects (Supplementary Fig. S8A and S8B), suggesting that MAP9 loss led to transformation of normal human colon epithelial cells via CIN. Nevertheless, our data do not exclude potential contributions from CIN-independent effect of MAP9.

Figure 7.

MAP9 knockdown caused in vitro transformation of normal colon epithelial cells and induced CIN in vivo. A, 1CT and 2CT cells with long-term MAP9-knockdown acquired anchorage-independent growth ability as determined by a soft-agar colony formation assay. Each group contained five replicates. B, Effect of long-term MAP9 knockdown on cell invasion. Each group contained 4 replicates. C, The altered somatic CNV size in tumors from ApcMin/+ and Map9Δ/ΔApcMin/+ mice. D, Representative altered somatic CNV region in tumor (T1) from ApcMin/+ and tumor (T3) from Map9Δ/ΔApcMin/+ mice. Red dot represented copy-number gains and green dot represented normal genomic regions. E, Somatic SVs in tumors from ApcMin/+ and Map9Δ/ΔApcMin/+ mice. SVs included interchromosomal translocations (CTX), intrachromosomal translocations (ITX), and deletions (DEL). F, Circos plot of SVs in mice tumors. G, Somatic mutation numbers in tumors from ApcMin/+ and Map9Δ/ΔApcMin/+ mice. Left, mutations in CDS region. Right, all mutation types. H, Kaplan–Meier curves of patients with colorectal cancer, stratified by MAP9 mRNA level. Patients with colorectal cancer were divided into two groups: low MAP9 expression group (tumor/normal ratio < 1) and high MAP9 expression group (tumor/normal ratio > 1). The tumor/normal ratio of MAP9 mRNA was calculated as follows: |${{\rm{2}}^{( {\Delta \Delta {C_{\rm{t}}}( {{\rm{Tumor }}( {\Delta {C_{\rm{t}}}( {MAP9\, - \,\,\beta - ACTIN} )} ){\rm{ }} - {\rm{ Normal}}( {\Delta {C_{\rm{t}}}( {MAP9{\rm{ }} - {\rm{ }}\beta - ACTIN} )} )} )} )}}$|⁠. All histogram data represent mean ± SD. Compared with control group, *, P < 0.05; **, P < 0.01; and ***, P < 0.001. NS, not significant.

Figure 7.

MAP9 knockdown caused in vitro transformation of normal colon epithelial cells and induced CIN in vivo. A, 1CT and 2CT cells with long-term MAP9-knockdown acquired anchorage-independent growth ability as determined by a soft-agar colony formation assay. Each group contained five replicates. B, Effect of long-term MAP9 knockdown on cell invasion. Each group contained 4 replicates. C, The altered somatic CNV size in tumors from ApcMin/+ and Map9Δ/ΔApcMin/+ mice. D, Representative altered somatic CNV region in tumor (T1) from ApcMin/+ and tumor (T3) from Map9Δ/ΔApcMin/+ mice. Red dot represented copy-number gains and green dot represented normal genomic regions. E, Somatic SVs in tumors from ApcMin/+ and Map9Δ/ΔApcMin/+ mice. SVs included interchromosomal translocations (CTX), intrachromosomal translocations (ITX), and deletions (DEL). F, Circos plot of SVs in mice tumors. G, Somatic mutation numbers in tumors from ApcMin/+ and Map9Δ/ΔApcMin/+ mice. Left, mutations in CDS region. Right, all mutation types. H, Kaplan–Meier curves of patients with colorectal cancer, stratified by MAP9 mRNA level. Patients with colorectal cancer were divided into two groups: low MAP9 expression group (tumor/normal ratio < 1) and high MAP9 expression group (tumor/normal ratio > 1). The tumor/normal ratio of MAP9 mRNA was calculated as follows: |${{\rm{2}}^{( {\Delta \Delta {C_{\rm{t}}}( {{\rm{Tumor }}( {\Delta {C_{\rm{t}}}( {MAP9\, - \,\,\beta - ACTIN} )} ){\rm{ }} - {\rm{ Normal}}( {\Delta {C_{\rm{t}}}( {MAP9{\rm{ }} - {\rm{ }}\beta - ACTIN} )} )} )} )}}$|⁠. All histogram data represent mean ± SD. Compared with control group, *, P < 0.05; **, P < 0.01; and ***, P < 0.001. NS, not significant.

Close modal

We utilized the Human Cancer PathwayFinder RT² Profiler PCR Array to profile the expression of 84 genes involved in transformation and tumorigenesis in long-term MAP9-knockdown 1CT cells. The data demonstrated that long-term knockdown of MAP9 altered gene expression involved in DNA damage, apoptosis, cell cycle, cellular senescence, and angiogenesis pathways (Supplementary Fig. S8C; Supplementary Table S4). Gene set enrichment analysis of RNA sequencing data from TCGA colorectal cancer cohort revealed that genes promoting cell survival and growth such as telomere extension, mRNA capping, and activation of prereplicative complex and DNA repair pathways are upregulated in MAP9-low (<25th percentile) compared with MAP9-high tumors (>75th percentile; Supplementary Fig. S8D; Supplementary Table S5), consistent with the expression profiles in 1CT cells with long-term knockdown of MAP9. Taken together, MAP9 loss in normal colon epithelial induces CIN and oncogenic signaling, thereby causing cellular transformation.

Tumors from Map9-knockout ApcMin/+ mice harbor chromosomal abnormalities

To elucidate the effect of Map9 knockout on the colorectal cancer genome, we performed whole genome sequencing (WGS) on three colon tumors from Map9Δ/ΔApcMin/+ mice and two colon tumors from ApcMin/+ mice. We observed increased trend of somatic copy-number variation (CNV) sizes in colon tumors from Map9Δ/ΔApcMin/+ mice compared with ApcMin/+ mice (Fig. 7C and D; Supplementary Table S6); however, there was no significant difference due to the limited number of mice assessed and the big CNV size variations (Supplementary Fig. S8E). More importantly, colon tumors from Map9Δ/ΔApcMin/+ mice had significantly more somatic structural variations (SV; Fig. 7E and F; Supplementary Table S6). The number of interchromosomal translocations (CTX) in Map9Δ/ΔApcMin/+ tumors was twice as much as ApcMin/+ tumors, whereas intrachromosomal translocations (ITX) were only found in Map9Δ/ΔApcMin/+ tumors (Fig. 7E). However, no significant difference was observed for somatic mutations (Fig. 7G; Supplementary Table S6), suggesting the effect of MAP9 is specific to chromosomal abnormalities. Collectively, Map9 knockout in vivo generates CIN, which in turn drives colorectal tumorigenesis.

Patient with colorectal cancer with MAP9 downregulation has poor outcomes

We evaluated the clinical impact of MAP9 in 141 patients with colorectal cancer (Supplementary Table S7). Low MAP9 mRNA expression (tumor/normal ratio) was associated with poorer survival of patients with colorectal cancer after adjustment for age, gender, and tumor–node–metastasis (TNM) stage (HR, 2.36; 95% confidence interval, 1.05–5.34; P = 0.04). Kaplan–Meier survival curves showed that overall survival of patients with colorectal cancer with low MAP9 mRNA expression was significantly shorter than that of other patients (P = 0.03, log-rank test; Fig. 7H). After stratification by TNM stage, patients with colorectal cancer with low MAP9 mRNA expression had significantly shorter survival in early stage (I/II/III; P = 0.02), but not advanced stage (IV) colorectal cancer (Supplementary Fig. S8F).

A majority of sporadic colorectal cancer arise through the CIN pathway (65%–70%); however, its underlying molecular mechanism remains poorly understood. In this study, we identified a novel spindle-associated protein MAP9 that is frequently inactivated in colorectal cancer, inferring that MAP9 may have tumor-suppressive function. We showed that MAP9 expression is essential for maintaining chromosomal integrity, and its loss drives CIN in the colon epithelium and induces cellular transformation. Deletion of Map9 in the colonic epithelium triggers spontaneous colorectal cancer and accelerates carcinogen- or ApcMin/+-driven colorectal cancer, implying that the silencing of MAP9 is a driver event in CIN-associated colorectal cancer development.

Despite the high prevalence of CIN in colorectal cancer, mutations in mitosis-associated genes are rare. Here, we uncovered that inappropriate silencing of MAP9 may be an important mechanism by which CIN can be triggered in colorectal cancer. MAP9 is downregulated in a majority of primary colorectal cancer (∼70%). MAP9 knockdown in human normal colon epithelial cells (1CT/2CT) and MEFs induced severe chromosome segregation error, aneuploidy, and a CIN phenotype. Long-term MAP9 silencing in 1CT/2CT cells led to acquisition of anchorage-independent growth capacity and enhanced invasiveness, key hallmarks of cellular transformation. Our in vitro data were recapitulated in vivo using Map9-knockout mice. Intestinal-specific knockout of Map9 in mice resulted in the spontaneous formation of colonic dysplasia and colorectal cancer. Moreover, Map9 loss also significantly increased tumor incidence, tumor number, and tumor load in genetically (ApcMin/+) and carcinogen (AOM)-driven colorectal cancer mouse models. WGS revealed an increased CNVs and chromosomal translocations in colon tumors from Map9-knockout ApcMin/+ mice compared with those from control ApcMin/+ mice, consistent with in vitro data. These transgenic mice results demonstrated that Map9 inactivation promoted colon tumorigenesis in vivo. Given the gatekeeper role of MAP9 in chromosomal integrity, the transcriptional silence of MAP9 likely has a significant role-promoting colorectal cancer through induction of CIN. Consistent with tumor-suppressive function of MAP9 in colorectal cancer, patients with colorectal cancer with low MAP9 expression were associated with poorer outcome. Hence, our data provide solid evidence that Map9 inactivation plays a causal role in colorectal tumorigenesis through the induction of CIN.

Around 50% MAP9 loss could induce genome instability in vitro. On the other hand, tumor-promoting effect of heterozygous versus homozygous Map9 knockout varies according to the model utilized in vivo. In AOM or spontaneous model, heterozygous Map9 knockout could increase tumor burden or induce transformation; whereas only homozygous knockout of Map9 enhanced tumorigenesis in ApcMin/+ mice. This discrepancy might be attributed to the short duration of the study (∼3 months) compared with the other two (>6 months). In our cohort of patients with colorectal cancer, we analyzed the distribution of MAP9 expression levels by tumor/normal ratio, revealing that more than 40% of patients have at least approximately 50% loss of MAP9 expression (Supplementary Fig. S8G). This indicates that a large number of human colorectal cancers harbor MAP9 loss that may reach the threshold to cause CIN.

MAP9 belongs to MAP family that regulates microtubule dynamics (6). We observed that MAP9 is localized to spindle microtubules during mitosis, suggesting that it might play a role in the mitosis processes. Indeed, MAP9 knockdown caused shortened spindles in metaphase that were extremely unstable and repeatedly collapsed and reassembled. Conversely, the ectopic expression of MAP9 led to spindle lengthening. We further demonstrated that disrupted spindle dynamics caused by MAP9 deficiency initiates CIN via the following pathways: (i) spindle pole defocusing and PCM fragmentation; (ii) chromosomes lagging; and (iii) failure to correct K-fiber misattachment. Mechanistically, MAP9 tightly surrounds γ-TuRC to protect K-fiber from depolymerization at their minus end at spindle poles. This is evidenced by shortened and curved K-fibers in MAP9-knockdown cells, whereas MAP9 overexpression has a stabilizing effect on microtubules. Consistent with our data, other MAP proteins have a wide range of functions that revolve around microtubules, including stabilization of microtubules by inducing polymerization or inhibiting depolymerization, destabilization of microtubules by preventing their assembly, and mediating interactions of microtubules with other proteins (34). This in turn controls spindle length during mitosis, which is critical for proper segregation of chromosomes (35). Taken together, our results suggested that MAP9 acts as a microtubule stabilizer and contributed to spindle stability by interacting and stabilizing spindle microtubules.

In summary, we identified a novel tumor suppressor, MAP9, in colorectal cancer. Loss of MAP9 led to compromised spindle stability and proper chromosomal segregation, contributing to CIN, aneuploidy, and ultimately colorectal tumorigenesis (Supplementary Fig. S9). This study established the molecular mechanisms by which this key CIN regulator normally maintains chromosomal stability and prevents cancer.

No potential conflicts of interest were disclosed.

Conception and design: S. Wang, J. Huang, J. Yu

Development of methodology: S. Wang, J. Huang, J. Zhai, W. Deng, Y. Zhang, G. Wang, J.W. Shay, J. Yu

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Wang, J. Huang, L. Zhao, J. Zhai, Y. Zhou, W. Deng, S. Gao, X.Y. Guan, J. Yu

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Wang, J. Huang, C.C. Wong, W. Deng, Y. Zeng, H.H. He

Writing, review, and/or revision of the manuscript: S. Wang, J. Huang, C.C. Wong, H.H. He, J.W. Shay, J. Yu

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): W. Deng, H. Wei, S.H. Wong, J.W. Shay

Study supervision: J. Yu

Other (animal experiment): C. Li

This study was supported by HMRF Hong Kong (03140856), National Key R&D Program of China (2017YFE0190700), Science and Technology Program Grant Shenzhen (JCYJ20170413161534162), Science and Technology Program Grant Shenzhen (JCYJ20170413161534162), RGC-GRF Hong Kong (14111216, 14163817), Vice-Chancellor's Discretionary Fund CUHK, CUHK direct grant, and Shenzhen Virtual University Park Support Scheme to CUHK Shenzhen Research Institute.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Torre
LA
,
Bray
F
,
Siegel
RL
,
Ferlay
J
,
Lortet-Tieulent
J
,
Jemal
A
. 
Global cancer statistics, 2012
.
CA Cancer J Clin
2015
;
65
:
87
108
.
2.
Arnold
M
,
Sierra
MS
,
Laversanne
M
,
Soerjomataram
I
,
Jemal
A
,
Bray
F
. 
Global patterns and trends in colorectal cancer incidence and mortality
.
Gut
2017
;
66
:
683
91
.
3.
Burrell
RA
,
McGranahan
N
,
Bartek
J
,
Swanton
C
. 
The causes and consequences of genetic heterogeneity in cancer evolution
.
Nature
2013
;
501
:
338
45
.
4.
Grady
WM
,
Carethers
JM
. 
Genomic and epigenetic instability in colorectal cancer pathogenesis
.
Gastroenterology
2008
;
135
:
1079
99
.
5.
Geigl
JB
,
Obenauf
AC
,
Schwarzbraun
T
,
Speicher
MR
. 
Defining ‘chromosomal instability'
.
Trends Genet
2008
;
24
:
64
9
.
6.
Saffin
J-M
,
Venoux
M
,
Prigent
C
,
Espeut
J
,
Poulat
F
,
Giorgi
D
, et al
ASAP, a human microtubule-associated protein required for bipolar spindle assembly and cytokinesis
.
Proc Natl Acad Sci U S A
2005
;
102
:
11302
7
.
7.
Padilla-Nash
HM
,
Barenboim-Stapleton
L
,
Difilippantonio
MJ
,
Ried
T
. 
Spectral karyotyping analysis of human and mouse chromosomes
.
Nat Protoc
2007
;
1
:
3129
42
.
8.
Meunier
S
,
Vernos
I
. 
K-fibre minus ends are stabilized by a RanGTP-dependent mechanism essential for functional spindle assembly
.
Nat Cell Biol
2011
;
13
:
1406
14
.
9.
Foley
EA
,
Maldonado
M
,
Kapoor
TM
. 
Formation of stable attachments between kinetochores and microtubules depends on the B56-PP2A phosphatase
.
Nat Cell Biol
2011
;
13
:
1265
71
.
10.
Kabeche
L
,
Compton
DA
. 
Cyclin A regulates kinetochore microtubules to promote faithful chromosome segregation
.
Nature
2013
;
502
:
110
3
.
11.
Skarnes
WC
,
Rosen
B
,
West
AP
,
Koutsourakis
M
,
Bushell
W
,
Iyer
V
, et al
A conditional knockout resource for the genome-wide study of mouse gene function
.
Nature
2011
;
474
:
337
42
.
12.
Xu
J
.
Preparation, culture, and immortalization of mouse embryonic fibroblasts
.
Hoboken, NJ
:
John Wiley & Sons, Inc
; 
2001
.
p.
141
8
.
13.
Jozefczuk
J
,
Drews
K
,
Adjaye
J
. 
Preparation of mouse embryonic fibroblast cells suitable for culturing human embryonic and induced pluripotent stem cells
.
J Vis Exp
2012
;
pii
:
3854
.
14.
Neufert
C
,
Becker
C
,
Neurath
MF
. 
An inducible mouse model of colon carcinogenesis for the analysis of sporadic and inflammation-driven tumor progression
.
Nat Protoc
2007
;
2
:
1998
2004
.
15.
Feng
Y
,
Sentani
K
,
Wiese
A
,
Sands
E
,
Green
M
,
Bommer
GT
, et al
Sox9 induction, ectopic Paneth cells, and mitotic spindle axis defects in mouse colon adenomatous epithelium arising from conditional biallelic Apc inactivation
.
Am J Pathol
2013
;
183
:
493
503
.
16.
Schiebel
E
. 
Gamma-tubulin complexes: binding to the centrosome, regulation and microtubule nucleation
.
Curr Opin Cell Biol
2000
;
12
:
113
8
.
17.
Zou
J
,
Hallen
MA
,
Yankel
CD
,
Endow
SA
. 
A microtubule-destabilizing kinesin motor regulates spindle length and anchoring in oocytes
.
J Cell Biol
2008
;
180
:
459
66
.
18.
Gregan
J
,
Polakova
S
,
Zhang
L
,
Tolić-Nørrelykke
IM
,
Cimini
D
. 
Merotelic kinetochore attachment: causes and effects
.
Trends Cell Biol
2011
;
21
:
374
81
.
19.
Lampson
MA
,
Renduchitala
K
,
Khodjakov
A
,
Kapoor
TM
. 
Correcting improper chromosome-spindle attachments during cell division
.
Nat Cell Biol
2004
;
6
:
232
7
.
20.
Bickenson
AF
. 
CYTOSKELETON A stabilizing influence for K-fibres
.
Nat Rev Mol Cell Biol
2011
;
13
:
2
3
.
21.
Akhmanova
A
,
Hoogenraad
CC
. 
Microtubule minus-end-targeting proteins
.
Curr Biol
2015
;
25
:
R162
71
.
22.
Cimini
D
,
Wan
X
,
Hirel
CB
,
Salmon
ED
. 
Aurora kinase promotes turnover of kinetochore microtubules to reduce chromosome segregation errors
.
Curr Biol
2006
;
16
:
1711
8
.
23.
Wang
P
,
Lushnikova
T
,
Odvody
J
,
Greiner
TC
,
Jones
SN
,
Eischen
CM
. 
Elevated Mdm2 expression induces chromosomal instability and confers a survival and growth advantage to B cells
.
Oncogene
2008
;
27
:
1590
8
.
24.
Gonzalez-Loyola
A
,
Fernandez-Miranda
G
,
Trakala
M
,
Partida
D
,
Samejima
K
,
Ogawa
H
, et al
Aurora B overexpression causes aneuploidy and p21(Cip1) repression during tumor development
.
Mol Cell Biol
2015
;
35
:
3566
78
.
25.
Weaver
BAA
,
Silk
AD
,
Montagna
C
,
Verdier-Pinard
P
,
Cleveland
DW
. 
Aneuploidy acts both oncogenically and as a tumor suppressor
.
Cancer Cell
2007
;
11
:
25
36
.
26.
Faggioli
F
,
Wang
T
,
Vijg
J
,
Montagna
C
. 
Chromosome-specific accumulation of aneuploidy in the aging mouse brain
.
Hum Mol Genet
2012
;
21
:
5246
53
.
27.
Britigan
EMC
,
Wan
J
,
Zasadil
LM
,
Ryan
SD
,
Weaver
BA
. 
The ARF tumor suppressor prevents chromosomal instability and ensures mitotic checkpoint fidelity through regulation of Aurora B
.
Mol Biol Cell
2014
;
25
:
2761
73
.
28.
Lushnikova
T
,
Bouska
A
,
Odvody
J
,
Dupont
WD
,
Eischen
CM
. 
Aging mice have increased chromosome instability that is exacerbated by elevated Mdm2 expression
.
Oncogene
2011
;
30
:
4622
31
.
29.
Dawar
S
,
Lim
Y
,
Puccini
J
,
White
M
,
Thomas
P
,
Bouchier-Hayes
L
, et al
Caspase-2-mediated cell death is required for deleting aneuploid cells
.
Oncogene
2017
;
36
:
2704
14
.
30.
Gao
C
,
Su
Y
,
Koeman
J
,
Haak
E
,
Dykema
K
,
Essenberg
C
, et al
Chromosome instability drives phenotypic switching to metastasis
.
Proc Natl Acad Sci U S A
2016
;
113
:
14793
8
.
31.
Fujiwara
T
,
Bandi
M
,
Nitta
M
,
Ivanova
EV
,
Bronson
RT
,
Pellman
D
. 
Cytokinesis failure generating tetraploids promotes tumorigenesis in p53-null cells
.
Nature
2005
;
437
:
1043
7
.
32.
Sheltzer
JM
,
Ko
JH
,
Replogle
JM
,
Habibe Burgos
NC
,
Chung
ES
,
Meehl
CM
, et al
Single-chromosome gains commonly function as tumor suppressors
.
Cancer Cell
2017
;
31
:
240
55
.
33.
Roig
AI
,
Eskiocak
U
,
Hight
SK
,
Kim
SB
,
Delgado
O
,
Souza
RF
, et al
Immortalized epithelial cells derived from human colon biopsies express stem cell markers and differentiate in vitro
.
Gastroenterology
2010
;
138
:
1012
272
.
34.
Petry
S
. 
Mechanisms of mitotic spindle assembly
.
Annu Rev Biochem
2016
;
85
:
659
83
.
35.
Dumont
S
,
Mitchison
TJ
. 
Force and length in the mitotic spindle
.
Curr Biol
2009
;
19
:
R749
61
.