Introduction

In the multistep tumorigenic processes of gastrointestinal tumors, the accumulation of unhealthy mitochondria followed by reactive oxygen species (ROS)-mediated mitochondrial dysfunction stimulates malignant conversion1. Recently, we identified a novel p53-inducible protein, Mieap (Mitochondria-eating protein), as a crucial regulator of a novel mitochondrial quality control (MQC) system2,3, which consists of two mechanisms, including a repair process and an elimination process. The molecular mechanism underlying this repair process is intriguing; MALM, the Mieap-induced accumulation of lysosomal proteins within mitochondria, leads to a striking decrease in mitochondrial reactive oxygen species (mtROS) generation and an increase in mitochondrial ATP synthesis activities through the elimination of oxidized mitochondrial proteins and it differs completely from mitophagy (autophagosome-mediated autophagy)2,3,4. When MALM is inhibited, MIVs (Mieap-induced vacuoles) engulf and degrade the damaged, unhealthy mitochondria, behaving similarly to mitophagy3. Therefore, Mieap positively regulates mitochondrial quality by repairing or eliminating unhealthy mitochondria via MALM or MIV generation, respectively2,3,4. There is little doubt that the Mieap-mediated MQC function is critical for diverse physiological and pathophysiological conditions in vitro2,3,4,5.

Intestinal carcinoma is one of the leading causes of cancer-related deaths. The inactivation of adenomatous polyposis coli (APC) is an evoking event leading to the development of intestinal adenoma6,7; therefore, the ApcMin/+ mouse model is one of the best to investigate intestinal adenoma formation, which is directly implicated in human intestinal tumorigenic status8,9. To elucidate the involvement of the Mieap-regulated MQC function in tumorigenesis and malignant progression in vivo, in the present study, we utilized the ApcMin/+ murine intestinal tumor model7,8 and generated Mieap-deficient ApcMin/+ mice. We found that Mieap deficiency significantly accelerated the intestinal tumorigenic process, resulting in shorter lifespans and increased tumor multiplicity. Histopathologically, we confirmed the substantially increased number of intestinal high-grade adenomas and adenocarcinomas in the Mieap-deficient ApcMin/+ mice. Our results demonstrate that the loss of Mieap contributes remarkably to the malignant progression of intestinal tumors through the inactivation of the Mieap-mediated MQC function in ApcMin/+ mice.

Results

Mieap gene-deleted ApcMin/+ mice died earlier from severe anemia

To investigate the in vivo role of Mieap, we generated the Mieap-knockout mice as shown in Fig. 1. WT, Mieap+/− and Mieap−/− mice were born in expected Mendelian ratios. We did not observe any developmental defects in the Mieap+/− and Mieap−/− mice. The Mieap+/− and Mieap−/− mice were normally born and were able to grow and live after birth as well. These results prompted us to speculate that Mieap deficiency may play a facilitatory role in tumorigenic/carcinogenic processes. Since the p53/Mieap-regulated mitochondrial quality control pathway is frequently inactivated in primary cancer tissues of colorectal cancer patients (manuscript in preparation), we examined the role of Mieap in the ApcMin/+ murine intestinal tumor model.

Figure 1
figure 1

Generation of the Mieap-knockout mice.

(a) The schematic diagram of the construction of Mieap gene mutant. Mieap-knockout mice were generated by using the Cre/loxP recombination system. Briefly, the floxed and trapped alleles were generated using a single construct bearing a gene-trap cassette doubly flanked by LoxP and FRT located between exons 5 and 8 of the mouse Mieap gene. (b) Genotypic analyses of the Mieap gene. The Genotypes were determined by PCR using genomic DNA derived from wild-type (WT), Mieap heterozygous mutant (Mieap+/−) and Mieap homozygous mutant (Mieap−/−) mice. (c,d) Mieap expression. Mieap mRNA (c) and protein (d) expressions were examined by RT-PCR and western blot in the testes from wild-type (WT), Mieap heterozygous mutant (Mieap+/−) and Mieap homozygous mutant (Mieap−/−) mice.

To investigate the role of Mieap in intestinal tumorigenesis in vivo, we generated the ApcMin/+ mice with the Mieap+/− and Mieap−/− genetic background, ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice. To evaluate the effects of Mieap gene deletion (heterozygous and homozygous deletions) on the overall survival of ApcMin/+ mice, we monitored a cohort of ApcMin/+ (n = 37), ApcMin/+ Mieap+/− (n = 14) and ApcMin/+ Mieap−/− (n = 10) mice until death. Interestingly, Kaplan-Meier survival analysis demonstrated that the ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice had significantly much shorter survival times than the ApcMin/+ mice [Fig. 2; ApcMin/+ vs. ApcMin/+ Mieap+/−, P = 0.0007; ApcMin/+ vs. ApcMin/+ Mieap−/−, P < 0.0001, log-rank (Mantel-Cox) test]. Hematological and blood chemical analyses clearly revealed that the ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice had much severer anemia compared to the ApcMin/+ mice (Fig. 3a–g) because of intestinal hemorrhage.

Figure 2
figure 2

The ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice have shorter survival rates.

The overall survival of the ApcMin/+ (n = 37), ApcMin/+ Mieap+/− (n = 14) and ApcMin/+ Mieap−/− (n = 10) mice from birth to death was plotted using the Kaplan-Meier method. The log-rank (Mantel-Cox) P values are as follows: ApcMin/+ vs. ApcMin/+ Mieap+/−, P = 0.0007 (statistically significant); ApcMin/+ vs. ApcMin/+ Mieap−/−, P < 0.0001 (statistically significant); ApcMin/+ Mieap+/− vs. ApcMin/+ Mieap−/−, P = 0.4954.

Figure 3
figure 3

The ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice suffer much severer anemia compared to the ApcMin/+ mice.

Hematological and blood chemical analyses for the red blood cell count (RBC) (a), hemoglobin (HGB) (b), hematocrit (HCT) (c), mean corpuscular volume (MCV) (d), mean corpuscular hemoglobin (MCH) (e) and white blood cell count (WBC) (f), as well as spleen weight (g) data, strongly indicated that the ApcMin/+ Mieap+ /−, and ApcMin/+ Mieap−/− mice tended to have much severer anemia, compared to the ApcMin/+ mice. The data are presented as the mean ± SD from seventeen-week-old ApcMin/+, ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice (n = 15, each) (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001).

An increased number of intestinal polyps in Mieap gene-deleted ApcMin/+ mice

Since the ApcMin/+ mice die in anemia because of chronic intestinal tumor bleeding, we speculated that the Mieap-deficient ApcMin/+ mice (ApcMin/+ Mieap+/– mice and ApcMin/+ Mieap–/– mice) might burden much more tumors in the intestine. Therefore, we counted the number of intestinal polyps in the ApcMin/+ , ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice (n = 15, each) (Fig. 4). As shown in Fig. 4a–c, the Mieap gene-deleted ApcMin/+ mice (ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/−) had a greater number of intestinal polyps than the ApcMin/+ mice. Moreover, the intestinal polyps in the Mieap gene-deleted ApcMin/+ mice (ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/−) were significantly larger than those in the ApcMin/+ mice (Fig. 4d,e). These results suggest that Mieap plays a critical role in intestinal tumor suppression.

Figure 4
figure 4

The number of intestinal polyps was increased in the ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice.

(a) Representative examples of small intestines and colons from ApcMin/+, ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice. The substantially increased number of intestinal polyps in the small intestine (b,d) and colon (c,e) in the ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice compared to the ApcMin/+ mice. The data are presented as the mean ± SD from seventeen-week-old ApcMin/+, ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice (n = 15, each) (*P < 0.05; **P < 0.01; ***P <0.001; ****P < 0.0001).

Mieap gene deletion in ApcMin/+ mice promotes malignant tumor progression

To precisely evaluate the intestinal polyps in the ApcMin/+ , ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice (Fig. 4), all intestinal polyps were histopathologically diagnosed by two pathologists. In wild-type and Mieap−/− mice, intestinal tumors were not observed and there were few histological differences between these lines (Supplementary Fig. S1). Additionally, there were essentially no histological differences among the tumor-associated intestinal epithelia in the ApcMin/+, ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice. However, in the small intestines of the ApcMin/+ Mieap−/− mice, the tumor-free epithelia were slightly atrophic (Supplementary Fig. S2).

Neoplastic lesions among the intestinal polyps were categorized into three classes: Category 1, low-grade adenoma; Category 2, high-grade adenoma; and Category 3, adenocarcinoma (Fig. 5). The details of the definitions on each Category are described in the Methods section. We confirmed that the number of high-grade adenomas (Category 2) and adenocarcinomas (Category 3) in the small intestine (Fig. 5a), colon (Fig. 5b) and whole intestine (Fig. 5c) was substantially increased in accordance with the Mieap gene deletion in the ApcMin/+ mice (Fig. 5a–c). The majority of intestinal tumors in the ApcMin/+ mice were classified as Category 1, low-grade adenoma (Fig. 5a–c). Representative histopathology of a Category 1 tumor in the small intestine of ApcMin/+ mice is shown in Fig. 5d. There were distorted glandular architectures (Fig. 5d, upper panel) consisting of atypical cells with a modestly increased nuclear/cytoplasmic (N/C) ratio, but numerous goblet cells formed well-differentiated glandular structures (Fig. 5d, lower panel with inset). Focal penetration of the muscularis mucosae and crypt herniation were observed; however, the basement membrane remained intact (Fig. 5d).

Figure 5
figure 5

Substantial increase in the number of intestinal high-grade adenoma and adenocarcinoma in ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice.

The numbers of histopathologically diagnosed small intestinal (a), colonic (b) and intestinal (small intestine plus colon) (c) adenomas and adenocarcinomas categorized into three classes (Category 1, low-grade adenoma; Category 2, high-grade adenoma; Category 3, adenocarcinoma) in the ApcMin/+, ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice (n = 8, each). Representative histopathology (HE) of Category 1 (low-grade adenoma) in the small intestines of an ApcMin/+ mouse (d), Category 2 (high-grade adenoma) (e) and Category 3 (adenocarcinoma) (f) in the small intestine of an ApcMin/+ Mieap+/− mouse, Category 2 (high-grade adenoma) (g) and Category 3 (adenocarcinoma) (h) in the small intestine of an ApcMin/+ Mieap−/− mouse and Category 3 (adenocarcinoma) in the colons of the ApcMin/+ Mieap+/− (i). (d–i) Top panel, low-power field; bottom panel with insets, high-power field. Scale bars, 100μm.

In the Mieap gene-deleted ApcMin/+ mice, Category 1 intestinal tumors exhibited the same histology as that in Fig. 5d. In contrast to the Category 1 tumors, there were diversely shaped atypical/distorted glandular structures (Fig. 5e,g, upper panels) composed of dysplastic tumor cells with an increased N/C ratio (Fig. 5e,g, lower panels with insets) in the high-grade adenomas (Category 2) of the ApcMin/+ Mieap+/− (Fig. 5e) and ApcMin/+ Mieap−/− (Fig. 5g) mice. For Category 3, adenocarcinomas (Fig. 5f,h,i), there were apparent infiltrating irregularly shaped/sized tumor cell nests surrounded by fibrous and hyalinized stromata (Fig. 5f,h,i, lower panels with insets). Infiltrating tumor cells had large and dense nuclei and frequently exhibited less differentiated morphologies (Fig. 5f,h,i, lower panels with insets).

Interestingly, there were many irregularly shaped glandular tumor cell nests with abnormal mucous retention in the adenocarcinomas stemming from the colon (Fig. 5i). Colonic adenocarcinoma (Category 3) in the ApcMin/+ Mieap−/− mice showed the same histology as that in Fig. 5i. Moreover, to quantitatively confirm the increased tumor cell proliferative potentials in conjunction with the Mieap gene deletion in the ApcMin/+ mice, Ki67 labeling indices were determined in each category. It was obvious that the small intestinal high-grade adenoma (Category 2) and adenocarcinoma (Category 3) cell proliferative potentials of the Mieap gene-deleted ApcMin/+ mice were significantly higher than those of the ApcMin/+ mice (Fig. 6a). The colonic high-grade adenoma (Category 2) tumor cells in the ApcMin/+ Mieap−/− mice had an enhanced proliferative capacity compared to that in the ApcMin/+ Mieap+/− mice (Fig. 6b). Gross Ki67 labeling indices in each Category were shown in Supplementary Fig. S3.

Figure 6
figure 6

Higher tumor cell proliferative potentials in the intestinal adenomas and adenocarcinomas of the ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice.

Small intestinal (a) and colonic (b) tumor cell proliferative potentials in the ApcMin/+, ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice (n = 8, each) were histopathologically evaluated by Ki67 labeling indices (%), which indicated statistically significant increases in the high-grade adenomas (Category 2) and adenocarcinomas (Category 3) in the ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice compared to the ApcMin/+ mice. The data represent the mean Ki67-labeling indices ± SD (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001).

Unhealthy mitochondria and oxidative stress accumulate in the intestine and tumor of Mieap-deficient ApcMin/+ mice

Since Mieap is a critical regulator of mitochondrial quality control2,3,4 and Mieap deficiency leads to an accumulation of unhealthy mitochondria and an increase in mtROS in vitro2,3, we speculated that the accumulation of unhealthy mitocnondria and upregulation of oxidative stress in the intestine and tumor may be involved in the mechanism for the promoted tumor malignant progression in the Mieap-deficient ApcMin/+ mice. Therefore, we examined the status of the mitochondria in the intestine and tumor of ApcMin/+ mice and the ApcMin/+ Mieap−/− mice by performing electron microscopic analysis. As shown in Fig. 7a, compared with the small intestinal mucosal epithelial cells in the wild-type and ApcMin/+ mice, disruption of the cristae structure and integrity was remarkable in the ApcMin/+ Mieap−/− mice. We performed the quantitative analysis on 100 mitochondria for mitochondrial density that reflects the healthy status of the cristae structure. Mitochondrial density in the tumor-free mucosal epithelial cells and tumor cells was extremely low due to crista defects in the ApcMin/+ Mieap−/− mice (Fig. 7a,b). We confirmed that the same phenomenon occurred in the Mieap−/− mice (Supplementary Fig. S4).

Figure 7
figure 7

Unhealthy mitochondria and oxidative stress accumulate in the intestine and tumor of the ApcMin/+ Mieap−/− mice.

Electron microscopic analysis of the mitochondrial morphologies in wild-type (WT) normal mucosal epithelium (a, left panel), ApcMin/+ mice tumor-free mucosal epithelium (a, second from left panel) and small intestinal tumor cells (a, middle panel), ApcMin/+ Mieap−/− mice tumor-free mucosal epithelium (a, second from right panel) and small intestinal tumor cells (a, right panel). Densitometric image analysis of the internal mitochondria (internal cristae density) was performed in the WT normal epithelium, ApcMin/+ tumor-free epithelium, ApcMin/+ tumor cells, ApcMin/+ Mieap−/− tumor-free epithelium and ApcMin/+ Mieap−/− tumor cells in small intestine (b) (n = 100 mitochondria, each). In the tumor-free epithelial cells and small intestinal ApcMin/+ Mieap−/− tumor cells, internal cristae density was morphologically (a) and statistically (b) decreased compared to that in the WT and ApcMin/+ mice mucosal epithelial cells and tumor cells. The data represent the mean internal density/total area ratio ± SD (*P < 0.05; ****P < 0.0001). (c) Immuno-peroxidase (DAB) staining for nitrotyrosine (upper row) and immuno-alkaline phosphatase (ALP) staining for 8-OHdG (bottom row), hematoxylin counterstain. Representative small intestinal tumor histopathology of Category 1 (low-grade adenoma) in ApcMin/+ (left column) and ApcMin/+ Mieap−/− (second from left column) mice, Category 2 (high-grade adenoma, second from right column) and Category 3 (adenocarcinoma, right column) in the ApcMin/+ Mieap−/− mice. Scale bars: 500 nm (a), 100 μm (c).

We further examined the status of oxidative stress using anti-nitrotyrosine antibody and anti-8-OHdG antibody to show the cytoplasmic and nuclear oxidative stresses, respectively. As shown in Fig. 7c, compared with the ApcMin/+ mice, there was a substantial increase in both nitrotyrosine and 8-OHdG immunopositivity in the small intestinal tumors of the ApcMin/+ Mieap−/− mice (Fig. 7c) and the ApcMin/+ Mieap+/− mice (Supplementary Fig. S5). These results suggest that the Mieap deficiency leads to the accumulation of unhealthy mitochondria in the intestine of the ApcMin/+ mice and that unhealthy mitochondria are likely to increase oxidative stress, contributing to intestinal tumor progression.

Discussion

According to our recent studies, Mieap is a critical regulator of mitochondrial quality control under pathophysiological conditions2,3,4. Therefore, Mieap deficiency leads to an accumulation of unhealthy mitochondria and an increase in mtROS2,3. It has been reported that mtROS generated by unhealthy mitochondria participate in genomic DNA insults10, stabilizing the oxygen-sensitive transcription factor (HIF1α)11,12, matrix metalloproteinase (MMP) induction13,14 and oxidative stress-related signaling pathways, including NF-κB15,16. Accordingly, we specifically focused on Mieap as a key regulator in suppressing mtROS-mediated tumor progression. Therefore, we first hypothesized that Mieap deficiency (global knockout) increased the susceptibility of the mice to spontaneous tumorigenesis/carcinogenesis in multiple organs; however, strikingly, there was no difference in spontaneous carcinogenesis or in the long-term survival rate compared to the wild-type mice, at least until two years after birth (manuscript in preparation). These results suggest that Mieap deficiency plays a facilitatory role in tumorigenic/carcinogenic processes.

One of the most useful and established animal models used to investigate intestinal tumor progression is the ApcMin/+ mouse model8,17. ApcMin/+ mice recapitulate human intestinal adenoma formation with germline mutations in APC (adenomatous polyposis coli) and provide a strong animal model for studying intestinal tumorigenesis7,8,17. To investigate the role of Mieap in intestinal tumor progression in ApcMin/+ mice, we generated heterozygous (ApcMin/+ Mieap+/−) and homozygous (ApcMin/+ Mieap−/−) Mieap gene-deleted ApcMin/+ mice. A substantially increased number of intestinal polyps (in the small intestines and colon) in Mieap-deficient mice compared to general ApcMin/+ mice indicated that Mieap is a key suppressor of intestinal tumor formation. Through histopathological analysis of each intestinal polyp, we confirmed that Mieap deficiency remarkably promoted tumorigenesis (excluding adenomatous/polypoid hyperplasia18,19) as well as malignant progression (adenocarcinoma). Moreover, Mieap deficiency triggered an enhanced proliferative capacity in intestinal adenoma and adenocarcinoma cells. As a possible explanation for abnormal intestinal tumor progression in Mieap-deficient ApcMin/+ mice, we demonstrated the accumulation of morphologically abnormal mitochondria revealing decreased cristae density20,21.

Unhealthy mitochondria produce higher levels of ROS22,23. This could be due to abnormal electron transfer by dysfunctional respiratory chain proteins; impaired ATP production by dysfunctional ATP synthase proteins; and/or the decreased supply of NADH resulting from dysfunctional TCA cycle proteins. The generated ROS also oxidize mitochondrial proteins, including the core proteins of energy production themselves, leading to a vicious cycle and the acceleration of mitochondrial dysfunction22,23. Consistent with this mechanism, we also demonstrated the accumulation of cytoplasmic and nuclear oxidative insults in the Mieap-deficient intestinal tumors, as indicated by nitrotyrosine24,25 and 8-OHdG26,27 immunohistochemistry. Therefore, these results support our hypothesis that the accumulation of unhealthy mitochondria promotes intestinal tumor formation and progression via increased mtROS generation.

The localization of APC in the epithelia of intestinal villi and colorectal crypts, which shows a pronounced gradient in its expression levels from nearly negative at the bottom of the crypts to strongly positive at the luminal side28,29,30, provides insight into the actual molecular function(s). APC negatively regulates cell proliferation in the intestine by suppressing the canonical Wnt signaling pathway31, which stimulates the TCF-dependent transcription of Wnt-target genes, such as c-MYC32, EphB/ephrinB33 and cyclin D134, followed by β-catenin activation35. In addition to cell proliferation, APC inactivation has been reported to promote intestinal tumorigenesis through the downregulation of cell adhesion36,37. APC is associated with β-catenin, which links E-cadherin to α-catenin and the actin cytoskeleton and positively regulates cell adhesion by controlling the distribution of β-catenin and E-cadherin between the cell membrane and cytoplasm/nucleus38,39. We also observed a strong cytoplasmic/nuclear localization of β-catenin in the intestinal tumor cells of the Mieap-deficient ApcMin/+ mice compared to the Mieap wild-type ApcMin/+ mice (manuscript in preparation). Therefore, it is possible that Mieap-mediated mitochondrial quality control is involved in the Wnt/β-catenin signaling pathway.

We have also paid particular attention to the Hippo pathway, which is a downstream signaling event of cell-to-cell interaction and critically regulates appropriate cell survival40,41. Therefore, uncontrolled cell proliferation due to the dysregulation of the Hippo pathway (Hippo-OFF) is directly responsible for tumorigenesis42,43,44. Recently, it has been reported that cross-talk between the Wnt/β-catenin and Hippo pathways played a crucial role in balanced cell growth in the early development process and tissue homeostasis45,46,47. These studies have led to the same conceptual framework positing that cell-to-cell interaction mediated by adhesion molecules critically controls appropriate cell proliferation and cell death. To explain the malignant progression mediated by Mieap deficiency in intestinal tumors, we speculated that there was crosstalk between Mieap-mediated mitochondrial quality control and the Hippo pathway because it has been reported that mitochondrial dysfunction stimulated ROS production and inactivated the Hippo pathway through c-Jun amino (N)-terminal kinase (JNK) signaling48.

Our present results strongly indicate that Mieap-mediated mitochondrial quality control plays a key role in suppressing intestinal tumorigenesis and malignant progression in vivo. Indeed, the Mieap-related pathway was inactivated in more than 70% of colorectal cancer patients (manuscript in preparation). Regarding the actual mechanism(s) for p53-mediated tumor suppression, there is still missing information49,50. Our findings emphasize that Mieap-mediated tumor suppression may be a possible candidate for one of the critical functions of p53 in vivo.

Methods

Animal ethics statement

All animal experimental protocols were approved by the National Cancer Center Animal Ethics Committee (approved protocol No. T11-031) and the animal experiments were conducted in accordance with the institutional guidelines for animal experiments, which meet the ethical standards required by the law and the guidelines concerning experimental animals in Japan.

Mouse models

Wild-type (WT) C57BL6/J mice were obtained from CLEA Japan, Inc. (Tokyo, Japan). ApcMin/+ (C57BL/6J-ApcMin/J, Stock No. 002020) mice were obtained from The Jackson Laboratory (Bar Harbor, Maine, USA). The Mieap-knockout (Mieap−/−) mice were generated by using the Cre/loxP recombination system. Briefly, the floxed and trapped alleles were generated using a single construct bearing a gene-trap cassette doubly flanked by LoxP and FRT located between exons 5 and 8 of the mouse Mieap gene, which is located on chromosome 5. The Mieap homozygous (Mieap−/−) deficient mice were generated by mating between bleeding pairs of the Mieap heterozygous (Mieap+/−) mice. To determine the appropriate Mieap deficiencies, genomic DNA from the tails or fingers of the 3–4 week-old mice were genotyped by conventional genomic PCR using the Mieap knockout mice primers (forward, 5′-TCCCTTGAATCTTAACTTTGATGTC-3′; reverse, 5′-CTAAGACTGGCAGAAGACCAATAAG-3′). The Mieap expression was examined at mRNA and protein levels in the testes derived from the WT, Mieap+/− and Mieap−/− mice by RT-PCR (primers: Mieap forward, 5′-CGTGGAGACAATCAAGTGTC-3′; Mieap reverse, 5′-CAGCTATCTCTTCCTTCAGAT-3′; beta-2MG forward, 5′-TGGTGCTTGTCTCACTGACC-3′; beta-2MG reverse, 5′-CCGTTCTTCAGCATTTGGAT-3′) and western blot analysis (using rabbit polyclonal anti-mouse Mieap antibody and mouse monoclonal anti-beta actin antibody).

Mieap heterozygous (ApcMin/+ Mieap+/−) and homozygous (ApcMin/+ Mieap−/−) deficient ApcMin/+ mice were generated by mating between ApcMin/+ mice and the Mieap-deficient mice (Mieap+/− or Mieap−/− mice). To determine the appropriate Mieap deficiencies on ApcMin/+ mice, genomic DNA from the tails or fingers of the 3–4 week-old mice were genotyped by conventional genomic PCR using ApcMin/+ mice primers (The Jackson Laboratory protocol; wild-type forward, 5′-GCCATCCCTTCACGTTAG-3′; Min forward, 5′-TTCTGAGAAAGACAGAAGTTA-3′; common reverse, 5′-TTCCACTTTGGCATAAGGC-3′) and the Mieap knockout mice primers.

Survival studies

The overall survival of ApcMin/+ (n = 37), ApcMin/+ Mieap+ /− (n = 14) and ApcMin/+ Mieap−/− (n = 10) mice was calculated from birth to the ethical end point or death. Survival distribution was estimated using the Kaplan-Meier overall survival method.

Surgical procedures

Seventeen-week-old ApcMin/+, ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice (n = 15, each) were anesthetized using diethyl ether. Peripheral blood samples collected from the eye socket using K2EDTA capillary tubes (VITREX Medical, Herlev, Denmark) were hematologically analyzed for red blood cells (RBC), hemoglobin (HGB), hematocrit (HCT), mean cell volume (MCV), mean cell hemoglobin (MCH) and white blood cells (WBC) (CLEA Japan). The number and size of the intestinal polyps were calculated under a SMZ-10 stereoscopic microscope (Nikon, Tokyo, Japan).

Transmission electron microscopy

The small intestinal specimens were cut 1 mm3 from the WT, ApcMin/+ and ApcMin/+ Mieap−/− mice and were fixed in cold 2% paraformaldehyde/glutaraldehyde in 0.1 M PBS (pH 7.2) at 4 °C for 16 hours. After being rinsed with 0.1 M PBS at 4 °C for 90 min, the specimens were fixed in cold 1% osmium-tetroxide in 0.1 M PBS and then dehydrated with ethanol at 4 °C (from 50% to 100%: 10 min, each). Finally, the specimens were embedded in EPON 812 (TAAB Laboratories Equipment Ltd., Berks, England) and ultrathin sections were cut at 70 nm using a Leica UltraCut UCT microtome (Leica Microsystems, Wetzlar, Germany) and stained with uranium-lead. Digital electron microscopy images were captured using the H-7500 transmission electron microscope (HITACHI, Tokyo, Japan). The digital images were analyzed using ImageJ 1.49o software (National Institutes of Health, USA) on a Windows 7 computer.

Specimen handling

Intestinal (small intestinal and colonic) specimens were collected from 17-week-old WT, Mieap−/−, ApcMin/+, ApcMin/+ Mieap+/− and ApcMin/+ Mieap−/− mice (n = 8 each). The Swiss-roll intestinal surgical samples were routinely fixed in 10% formalin and embedded in paraffin. Serial 5 μm sections were cut from paraffin blocks and used for the histopathological studies.

Hematoxylin and eosin (HE) staining and histopathological evaluation

HE staining was performed using Dako Eosin (Code CS701, Dako, Glostrup, Denmark) and Dako REALTM Hematoxylin (Code S2020, Dako)51,52. Histopathologically, all intestinal specimens were evaluated by two pathologists (MT and TK) and the intestinal epithelial tumors (polyps) were categorized into the following three classes: Category 1, low-grade adenoma; Category 2, high-grade adenoma; and Category 3, adenocarcinoma. The defining histopathological features for each category are as follows. Category 1: mildly distorted glandular structures, branching villi and tubular crypt proliferation, mild nuclear and cellular atypism and intact basement membrane; Category 2: moderately or severely distorted glandular structures with branching villi, severe nuclear and cellular atypism, increased mitotic figures, increased atypical mucous retention and intact basement membrane; Category 3: evident infiltration of tumor cell nests with tumor stromal induction, increased mitotic figures, penetration of the basement membrane and invasion of the lamina propria or muscularis mucosae.

Antibodies

Rabbits were immunized with the recombinant amino (N)-terminal domain of mouse Mieap protein. Rabbit polyclonal anti-mouse Mieap antibody was subsequently purified on antigen affinity columns. Rabbit polyclonal antibodies against mouse Ki67 (catalog no. ab15580) were purchased from Abcam (Cambridge, MA, USA). A mouse monoclonal antibody against mouse 8-hydroxy-2′-deoxyguanosine (8-OHdG) (clone: N45.1, catalog no. MOG-100P)53 was purchased from the Japan Institute for the Control of Aging (JaICA), NIKKEN SEIL Co., Ltd (Shizuoka, Japan). A mouse monoclonal antibody against mouse nitrotyrosine (clone: 39B6, catalog no. NB110-96877) was purchased from Novus Biological USA (Littleton, CO, USA). A mouse monoclonal antibody against beta-actin (catalog no. A5316) was purchased from Sigma-Aldrich (St Louis, MO, USA).

Immunohistochemistry

Immunohistochemistry was performed using the EnVisionTM + Dual Link System-HRP and EnVisionTM G|2 System/AP Rabbit/Mouse (Permanent Red) system (Dako). For 8-OHdG immunohistochemistry, we selected alkaline phosphatase (ALP) staining to avoid non-specific antigen-antibody reaction by hydrogen peroxide. For antigen retrieval, sections were autoclaved in citric acid buffer (pH 6.0) at 121 °C for 10 min52,54. The sections were treated with 0.3% hydrogen peroxide in methanol for 30 min at room temperature to block endogenous peroxidase activity (for Ki67 and nitrotyrosine) and incubated with 5% bovine serum albumin (BSA) in 50 mM Tris-buffered saline (pH 7.4) containing 0.05% Triton X-100 (T-TBS) for 1 hour at room temperature to block non-specific protein binding sites54. The sections were then incubated at 4 °C with the primary antibodies diluted at 1:20 (anti-8-OHdG), 1:50 (anti-Nitrotyrosine) and 1:1000 (anti-Ki67) in T-TBS. After overnight incubation, the sections were incubated with EnVisionTM + Dual Link System-HRP reagents for 1 hour or EnVisionTM G|2 System/AP Rabbit/Mouse (Permanent Red) system reagents according to the manufacturer’s instruction at room temperature and treated with 0.02% 3,3′-diaminobenzidine, tetrahydrochloride (CAS no. 7411-49-6, catalog no. D006, DOJINDO LABORATORIES, Kumamoto, Japan) in 0.05 M Tris-HCl buffer (pH 7.6) containing 0.005% hydrogen peroxide or permanent red reagent according to the manufacturer’s instructions to visualize the reaction products54. Finally, sections were counterstained with Dako REALTM Hematoxylin (Dako)51. Digital HE and immunohistochemical images were captured on an Olympus BX43 microscope equipped with a DP27 digital camera and a D21-SAL stand-alone unit (Olympus Corporation, Tokyo, Japan) and compiled with Photoshop CS2 software (Adobe Systems Software Ireland Ltd., San Jose, CA, USA) on a Windows 7 computer.

Ki67 labeling index

The percentage of tumor cells with Ki67-positive nuclei among 100 cells was calculated in five fields of each intestinal tumor by pathologists and the mean ± standard deviation (SD) values were determined as Ki67-labeling indices55.

Statistical analysis and preparing graphs

All graphs and statistical analysis of all the experiments in this study were performed using GraphPad Prism version 6.03 for Windows (GraphPad Software Inc., La Jolla, CA, USA)41,43. Kaplan-Meier analysis and log-rank (Mantel-Cox) tests were used to compare the survival curves56. Other statistical analyses were performed using an unpaired Student’s t-test. P-values of less than 0.05 were considered to be significant41,43. Significant P-values are provided in the figure panels and result sections.

Additional Information

How to cite this article: Tsuneki, M. et al. Mieap suppresses murine intestinal tumor via its mitochondrial quality control. Sci. Rep. 5, 12472; doi: 10.1038/srep12472 (2015).