Lentivirus-mediated knockdown of rhomboid domain containing 1 inhibits colorectal cancer cell growth

Similar documents
Lentivirus-mediated Silencing of Rhomboid Domain Containing 1 Suppresses Tumor Growth and Induces Apoptosis in Hepatoma HepG2 Cells

RESEARCH COMMUNICATION. sirna Mediated Silencing of NIN1/RPN12 Binding Protein 1 Homolog Inhibits Proliferation and Growth of Breast Cancer Cells

HEK293FT cells were transiently transfected with reporters, N3-ICD construct and

SUPPLEMENTARY INFORMATION

HCC1937 is the HCC1937-pcDNA3 cell line, which was derived from a breast cancer with a mutation

Advances in Computer Science Research, volume 59 7th International Conference on Education, Management, Computer and Medicine (EMCM 2016)

RNA extraction, RT-PCR and real-time PCR. Total RNA were extracted using

Islet viability assay and Glucose Stimulated Insulin Secretion assay RT-PCR and Western Blot

Protocol for Gene Transfection & Western Blotting

A Hepatocyte Growth Factor Receptor (Met) Insulin Receptor hybrid governs hepatic glucose metabolism SUPPLEMENTARY FIGURES, LEGENDS AND METHODS

MTC-TT and TPC-1 cell lines were cultured in RPMI medium (Gibco, Breda, The Netherlands)

Annals of Oncology Advance Access published January 10, 2005

TFEB-mediated increase in peripheral lysosomes regulates. Store Operated Calcium Entry

Effect of Survivin-siRNA on Drug Sensitivity of Osteosarcoma Cell Line MG-63

Sestrin2 and BNIP3 (Bcl-2/adenovirus E1B 19kDa-interacting. protein3) regulate autophagy and mitophagy in renal tubular cells in. acute kidney injury

MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells

Plasmids Western blot analysis and immunostaining Flow Cytometry Cell surface biotinylation RNA isolation and cdna synthesis

RESEARCH COMMUNICATION

SUPPORTING MATREALS. Methods and Materials

microrna-200b and microrna-200c promote colorectal cancer cell proliferation via

Supplementary Information POLO-LIKE KINASE 1 FACILITATES LOSS OF PTEN-INDUCED PROSTATE CANCER FORMATION

RNAi-mediated downregulation of NOB1 suppresses the growth and colony-formation ability of human ovarian cancer cells

Construction of a hepatocellular carcinoma cell line that stably expresses stathmin with a Ser25 phosphorylation site mutation

KDR gene silencing inhibits proliferation of A549cells and enhancestheir sensitivity to docetaxel

SUPPLEMENTARY INFORMATION. Supplementary Figures S1-S9. Supplementary Methods

The Schedule and the Manual of Basic Techniques for Cell Culture

Reduction of angiocidin contributes to decreased HepG2 cell proliferation

Graveley Lab shrna knockdown followed by RNA-seq Biosample Preparation and Characterization Document

Supporting Information

Supplemental information

Supplementary data Supplementary Figure 1 Supplementary Figure 2

Argininosuccinate synthetase 1 suppression and arginine restriction inhibit cell

A549 and A549-fLuc cells were maintained in high glucose Dulbecco modified

Online Data Supplement. Anti-aging Gene Klotho Enhances Glucose-induced Insulin Secretion by Upregulating Plasma Membrane Retention of TRPV2

p47 negatively regulates IKK activation by inducing the lysosomal degradation of polyubiquitinated NEMO

(a) Significant biological processes (upper panel) and disease biomarkers (lower panel)

Supplementary Figure 1 Role of Raf-1 in TLR2-Dectin-1-mediated cytokine expression

PUMA gene transfection can enhance the sensitivity of epirubicin-induced apoptosis of MCF-7 breast cancer cells

Effects of AFP gene silencing on Survivin mrna expression inhibition in HepG2 cells

Impact of hyper-o-glcnacylation on apoptosis and NF-κB activity SUPPLEMENTARY METHODS

Graveley Lab shrna knockdown followed by RNA-seq Biosample Preparation and Characterization Document

Research on the inhibitory effect of metformin on human oral squamous cell carcinoma SCC-4 and CAL-27 cells and the relevant molecular mechanism.

Supplementary Materials and Methods

Supplementary Materials and Methods

Oncolytic Adenovirus Complexes Coated with Lipids and Calcium Phosphate for Cancer Gene Therapy

An epithelial-to-mesenchymal transition-inducing potential of. granulocyte macrophage colony-stimulating factor in colon. cancer

Positive nin one binding protein expression predicts poor outcome in prostate cancer

Erzsebet Kokovay, Susan Goderie, Yue Wang, Steve Lotz, Gang Lin, Yu Sun, Badrinath Roysam, Qin Shen,

General Laboratory methods Plasma analysis: Gene Expression Analysis: Immunoblot analysis: Immunohistochemistry:

mir 375 inhibits the proliferation of gastric cancer cells by repressing ERBB2 expression

Profiles of gene expression & diagnosis/prognosis of cancer. MCs in Advanced Genetics Ainoa Planas Riverola

ShRNA-mediated gene silencing of heat shock protein 70 inhibits human colon cancer growth

Cells and reagents. Synaptopodin knockdown (1) and dynamin knockdown (2)

Electron micrograph of phosphotungstanic acid-stained exosomes derived from murine

Supplementary Information

Functional characterisation of hepatitis B viral X protein/microrna-21 interaction in HBVassociated hepatocellular carcinoma

Analysis of small RNAs from Drosophila Schneider cells using the Small RNA assay on the Agilent 2100 bioanalyzer. Application Note

Soft Agar Assay. For each cell pool, 100,000 cells were resuspended in 0.35% (w/v)

Expression and clinical significance of ADAM17 protein in esophageal squamous cell carcinoma

Figure S1 Time-dependent down-modulation of HER3 by EZN No Treatment. EZN-3920, 2 μm. Time, h

Supplementary Fig. 1. Identification of acetylation of K68 of SOD2

Supplementary Figure 1: si-craf but not si-braf sensitizes tumor cells to radiation.

Overexpressing exogenous S100A13 gene and its effect on proliferation of human thyroid cancer cell line TT

Silencing Dicer expression enhances cellular proliferative and invasive capacities in human tongue squamous cell carcinoma

CD14 + S100A9 + Monocytic Myeloid-Derived Suppressor Cells and Their Clinical Relevance in Non-Small Cell Lung Cancer

Effect of ABCE1-silencing gene, transfected by electrotransfer, on the proliferation, invasion, and migration of human thyroid carcinoma SW579 cells

Berberine Sensitizes Human Ovarian Cancer Cells to Cisplatin Through mir-93/ PTEN/Akt Signaling Pathway

Supplemental Information

http / / cjbmb. bjmu. edu. cn Chinese Journal of Biochemistry and Molecular Biology COX-2 NTera-2 NTera-2 RT-PCR FasL caspase-8 caspase-3 PARP.

Supporting Information

Protection against doxorubicin-induced myocardial dysfunction in mice by cardiac-specific expression of carboxyl terminus of hsp70-interacting protein

Knockdown of ribosomal protein S15A induces human glioblastoma cell apoptosis

Epithelial interleukin-25 is a key mediator in Th2-high, corticosteroid-responsive

Essential Medium, containing 10% fetal bovine serum, 100 U/ml penicillin and 100 µg/ml streptomycin. Huvec were cultured in

Supplementary information

Downregulation of ubiquitin-associated protein 2-like with a short hairpin RNA inhibits human glioma cell growth in vitro

Supplementary Data Table of Contents:

Mapping the Ligand-binding Site on a GPCR Using Genetically-encoded Photocrosslinkers

Recombinant Protein Expression Retroviral system

B16-F10 (Mus musculus skin melanoma), NCI-H460 (human non-small cell lung cancer

http / /cjbmb. bjmu. edu. cn Chinese Journal of Biochemistry and Molecular Biology A431 . Western aza-dC FUT4-siRNA

Supplementary Figure S1. Venn diagram analysis of mrna microarray data and mirna target analysis. (a) Western blot analysis of T lymphoblasts (CLS)

Effect of sirna targeting EZH2 on cell viability and apoptosis of bladder cancer T24 cells

LncRNA LET function as a tumor suppressor in breast cancer development

Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells

Effect of starvation-induced autophagy on cell cycle of tumor cells

SUPPLEMENTAL MATERIAL. Supplementary Methods

RESEARCH ARTICLE. Ginsenoside-Rh2 Inhibits Proliferation and Induces Apoptosis of Human Gastric Cancer SGC-7901 Side Population Cells

Long noncoding RNA linc-ubc1 promotes tumor invasion and metastasis by regulating EZH2 and repressing E-cadherin in esophageal squamous cell carcinoma

MATERIALS AND METHODS. Neutralizing antibodies specific to mouse Dll1, Dll4, J1 and J2 were prepared as described. 1,2 All

DNA context and promoter activity affect gene expression in lentiviral vectors

Li et al. Journal of Experimental & Clinical Cancer Research (2018) 37:108

Prevalence and characterization of somatic mutations in Chinese aldosterone-producing adenoma. patients. Supplemental data. First author: Baojun Wang

Phosphate buffered saline (PBS) for washing the cells TE buffer (nuclease-free) ph 7.5 for use with the PrimePCR Reverse Transcription Control Assay

mirna Dr. S Hosseini-Asl

MyD88 expression is associated with paclitaxel resistance in lung cancer A549 cells

Effect of specific silencing of EMMPRIN on the growth and cell cycle distribution of MCF-7 breast cancer cells

Serum Amyloid A3 Gene Expression in Adipocytes is an Indicator. of the Interaction with Macrophages

Supplementary Materials for

SUPPLEMENT. Materials and methods

Transcription:

MOLECULAR MEDICINE REPORTS 12: 377-381, 2015 Lentivirus-mediated knockdown of rhomboid domain containing 1 inhibits colorectal cancer cell growth JUNYI HAN 1,2*, JUNCHAO BAI 3*, YAO YANG 2, HUA YIN 2, WEI GAO 2, AIGUO LU 2, FEI LIU 1, HAIYAN GE 2, ZHONGMIN LIU 4, JINYI WANG 4 and LAN ZHONG 1 Departments of 1 Gastroenterology and 2 General Surgery, Shanghai East Hospital, Tongji University, Shanghai 200120; 3 Department of General Surgery, People's Hospital of Dayao, Yunnan 675400; 4 Department of Thoracic Surgery, Shanghai East Hospital, Tongji University, Shanghai 200120, P.R. China Received January 16, 2014; Accepted August 12, 2014 DOI: 10.3892/mmr.2015.3365 Abstract. Rhomboid domain containing 1 (RHBDD1), is a member of the rhomboid protease family, which has a pivotal role in the progression of numerous severe malignancies. However, its role in colorectal carcinoma (CRC) remains to be elucidated. In the present study, RHBDD1 was shown to be widely expressed in CRC cell lines. Lentivirus mediated RNA interference was employed to knockdown RHBDD1 expression in RKO CRC cells. Functional analyses indicated that depletion of RHBDD1 expression resulted in significantly reduced CRC cell proliferation and colony formation, and induced a G0/G1 phase cell cycle arrest. The findings of the present study suggest that RHBDD1 may contribute to CRC tumorigenesis and serve as a potential therapeutic target in human CRC. Introduction Malignant colorectal cancer or carcinoma (CRC) originates from the epithelial cells of the colon or rectum. CRC is associated with a high risk of cancer morbidity and mortality and a previous genomic analysis demonstrated that colorectal and rectal cancers have considerably similar patterns (1). CRC is responsible for ~500,000 deaths annually (2), and in developed countries, one out of three cases of CRC are fatal (3). Genetic studies have resolved the regulation of cellular metabolism, proliferation, differentiation and survival in CRC (4). However, further research is required to fully understand the molecular changes associated with the pathophysiology of CRC. The effector molecules and signal transduction Correspondence to: Professor Lan Zhong or Professor Junyi Han, Department of Gastroenterology, Shanghai East Hospital, Tongji University, 150 Jimo Road, Shanghai 200120, P.R. China E-mail: lanzhongdr@163.com E mail: junyidr@163.com * Contributed equally Key words: RHBDD1, colorectal carcinoma, RNAi, proliferation pathways responsible for the development of CRC remain to be elucidated. Previous studies have indicated that conserved intramembrane proteolytic mechanisms are associated with the regulation of cellular processes, including transcriptional control, growth factor secretion and apoptosis (5,6). It has been well demonstrated that intramembrane proteases are involved in critical cellular processes, including apoptosis and signaling transduction (6 10). The rhomboid serine proteases are expressed in most species, and have been observed in both bacteria and humans (11), they are considered to be one of the most widely conserved membrane proteases (12). The first rhomboid serine protease was initially identified in a Drosophila genetics study, where it was shown to activate the upstream epidermal growth factor receptor (EGFR) signaling pathway (13). Previous research has also demonstrated that numerous yeast rhomboid proteases have a role in mitochondrial membrane remodeling (14). Rhomboid domain containing 1 (RHBDD1), a mammalian rhomboid protease highly expressed in testis, has previously been identified as a pro apoptotic member of the B cell lymphoma 2 family (15). RHBDD1 has been shown to be highly expressed in chronic myeloid leukemia patients, as compared with healthy controls (16). RHBDD1 exhibited a proteolytic activity in the tumor suppressor activated pathway 6, in both HCT116 and RKO colon cancer cells (15). Downregulation of RHBDD1 also demonstrated the ability to suppress proliferation and colony formation capability of HepG2 hepatocellular carcinoma cells (17). However, the precise function of RHBDD1 in CRC progression remains unclear. In the present study, RHBDD1 was confirmed to be highly expressed in numerous CRC cell lines. To determine the role of RHBDD1 in human CRC, a lentivirus mediated short hairpin RNA (shrna) was used to knockdown RHBDD1 expression in RKO CRC cells. The effects of RHBDD1 expression knockdown on CRC cell growth were then investigated. Materials and methods Cell Culture. SW480, SW620, RKO, DLD 1, HCT116 and HT 29 human CRC cell lines and HEK293T human embryonic kidney cells were obtained from the Cell Bank of the Chinese

378 HAN et al: RHBDD1 REGULATES CRC CELL GROWTH Academy of Sciences (Shanghai, China). SW480, SW620, RKO and DLD 1 cells were cultured in RPMI 1640, supplemented with 10% fetal bovine serum (FBS; HyClone Laboratories Inc., Logan, UT, USA). HCT116 and HT 29 cells were cultured in McCoy's 5A media, supplemented with 10% FBS (Sigma Aldrich, St. Louis, MO, USA). HEK293T cells were cultured in Dulbecco's modified Eagle's medium, supplemented with 10% FBS (HyClone Laboratories Inc.). Construction of recombinant lentivirus. The small interfering RNA (sirna) sequence for RHBDD1 (NM_032276) (5' GCTGGGATTCTTGTTGGACTA 3') was screened and validated, to confirm its use as the candidate sirna. A non silencing sirna sequence (5' CCAAGGAAGTGCAA TTGCATA 3') was used as a control. shrnas corresponding to both the RHBDD1 and control sirna sequences, were synthesized as 21 nt inverse repeats, separated by a 9 nt loop for each sequence and inserted downstream of the U6 promoter in the pfh L lentiviral vector (Shanghai Hollybio Co., Ltd., Shanghai, China). The lentiviruses were generated by triple transfection of 80% confluent HEK293T cells, with modified pfh L plasmid, and helper vectors pvsvg I and pcmvδr8.92 (Shanghai Hollybio Co., Ltd.), using Lipofectamine 2000 (Invitrogen Life Technologies, Carlsbad, CA, USA). The lentiviruses were harvested in serum free medium after three days, filtered and concentrated using primed Centricon Plus 20 filter devices (Millipore, Billerica, MA, USA). RNA extraction and quantitative polymerase chain reaction (qpcr). RKO cells were pre cultured and infected with the recombinant lentivirus for five days. Total RNA was extracted using TRIzol reagent (Gibco Life Technologies, Carlsbad, CA, USA), according to the manufacturer's instructions. Total RNA (5 mg) was reverse transcribed to produce the first strand of cdna, using 200 U/ml SuperScript II RT (Invitrogen, Carlsbad, CA, USA). RHBDD1 mrna expression levels were evaluated by qpcr using a Bio Rad Connect Real Time PCR platform (Bio Rad Laboratories, Hercules, CA, USA) with SYBR Green PCR core reagents (Bio-Rad Laboraties); β actin was used as an internal reference. The following primers were used: RHBDD1 forward, 5' GCAGGACTGAGTGAAGAAGAAC 3', and reverse, 5' GTGAGAGATGAAACCCGTAGG 3'; and β actin forward, 5' GTGGACATCCGCAAAGAC 3' and reverse, 5' AAAGGGTGTAACGCAACTA 3'. The RT-qPCR analysis was performed with the following amplification steps: initial denaturation at 95 C for 1 min, followed by 40 cycles of denaturation at 95 C for 5 sec and annealing extension at 60 C for 20 sec. The results are presented as cycle threshold (Ct) values, the threshold PCR cycle number at which the amplified product is first detected. The average Ct was calculated for both RHBDD1 and β actin, and the ΔCT was determined as the mean of the triplicate Ct values for RHBDD1, minus the mean of the triplicate Ct values for β actin. Western blot analysis. RKO cells, five days after lentiviral infection, were lysed in 2X SDS sample buffer [100 mm Tris Hcl (ph 6.8), 10 mm EDTA, 4% SDS, 10% glycine]. The proteins (3 µg) were loaded onto a polyacrylamide gel and were separated by SDS PAGE, followed by transfer to polyvinylidene difluoride membranes (Millipore). The blots were blocked with TBST containing 5% non-fat dry milk at room temperature for 1 h and then incubated with primary antibodies: rabbit anti-rhbdd1 (1:500 dilution; Sigma-Aldrich) and mouse anti-gapdh (1:3,000 dilution; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) at 4 C overnight. The blots were then probed with antibodies against RHBDD1 (1:500 dilution; Sigma Aldrich) and mouse anti GAPDH, (1:3,000 dilution; Santa Cruz Biotechnology, Inc.). Following washing three times (5 min each) with TBST, the blots were incubated with the corresponding horseradish peroxidase-conjugated secondary antibodies: goat anti-rabbit IgG (1:5,000 dilution; Santa Cruz Biotechnology, Inc.) and goat anti-mouse IgG (1:5,000 dilution; Santa Cruz Biotechnology, Inc.) at room temperature for 2 h. The blots were then visualized using a Super Enhanced Chemiluminescence Detection Reagent (Applygen Technologies Inc., Beijing, China). Colony formation assay. RKO cells were cultured in 24 well plates and treated with the recombinant lentiviruses at a multiplicity of infection (MOI) of 20. Following a 72 h incubation, the cells were washed, re cultured in the prepared 6 well plates (400 cells/well) and allowed to form natural colonies. Eight days later the cells in both groups were subjected to Giemsa staining. Briefly, the cells were washed and fixed using paraformaldehyde, the fixed cells were then washed twice with phosphate buffered saline (PBS), treated with Giemsa (Sigma Aldrich) for 10 min, washed three times with double distilled H 2 O, and then photographed using a digital camera. The number of colonies (>50 cells/colony) were counted. MTT Viability Assay. RKO cells were cultured in 24 well plates and inoculated with either RHBDD1 shrna or control shrna lentiviruses, at a MOI of 20. Following a 72 h incubation, the cells were washed, re cultured in the prepared 96 well plates (2,000 cells/well) and the cell viability was analyzed using MTT reagent (Sigma-Aldrich) and acidic isopropanol (10% SDS, 5% isopropanol and 0.01 mol/l HCl). Flow cytometric analysis. RKO cells were harvested by centrifugation at 404 x g for 5 min, 72 h following infection. The pellets were washed twice with cold PBS, fixed with cold 70% ethanol, centrifuged and resuspended with PBS. The pellets were washed twice with cold PBS, fixed with cold 70% ethanol at 4 C overnight and then resuspended in propidium iodide/rnase/pbs for incubation in the dark (room temperature for 30 min). The suspensions were filtered through a 400 mesh membrane and subjected to cell cycle analysis using a flow cytometer (BD Biosciences, San Jose, CA, USA). (BD Biosciences, San Jose, CA, USA). Statistical analysis. All statistical analyses were performed using SPSS version 13.0 (SPSS, Inc., Chicago, IL, USA)software. The differences between the groups were compared using a Student's t test, and data were expressed as the means ± standard deviation of triplicate experiments. A P<0.05 was considered to indicate a statistically significant difference. Results Knockdown of RHBDD1 expression by sirna. Preliminary studies were performed to determine the prime candidate

MOLECULAR MEDICINE REPORTS 12: 377-381, 2015 379 A B C D E Figure 1. Downregulation of rhomboid domain containing 1 (RHBDD1) by lentivirus-mediated small hairpin (sh)rna. (A) Quantitative polymerase chain reaction of RHBDD1 mrna in six colon cancer cell lines. (B) Western blot analysis of RHBDD1 protein in six colon cancer cell lines. (C) Representative graphs of RKO cells infected with lentivirus at multiplicity of infection of 20 (magnification, x400). (D Expression analysis of RHBDD1 mrna in RKO colorectal cancer cells with three treatments (control, Lv-shCon, Lv-shRHBDD1) by qpcr. (E) Expression analysis of RHBDD1 protein in RKO cells with three treatments (control, Lv-shCon, Lv-shRHBDD1) by western blotting. Con, uninfected; Lv-shCon, control lentivirus with non-silencing; Lv-shRHBDD1, RHBDD1-silencing lentivirus; GFP, green fluorescent protein. ** P<0.01, as compared to treatment with Lv-shCon. CRC cell line for further in vitro studies. The mrna and protein expression levels of RHBDD1 were analyzed in six CRC cell lines: SW480, SW620, RKO, DLD 1, HCT116 and HT 29. As shown in Fig. 1A and B, RHBDD1 was widely expressed in all of the cell lines tested. The RKO cell line, which had moderate RHBDD1 expression levels, is commonly used in colon cancer studies due to its high proliferation rate and low coefficient of MOI. Therefore, the following in depth in vitro investigations were conducted using RKO cells. To investigate the role of RHBDD1 in human CRC, control (Lv shcon) and RHBDD1 shrna (Lv shrhbdd1), lentiviruses were constructed. RKO cells were cultured and infected with either the Lv shcon or Lv shrhbdd1 lentivirus. The images were photographed following a 72 h incubation. An embedded green fluorescent protein tag was used to visualize the transfection efficiency of the lentiviruses. As shown in Fig. 1C, >80% of the cells were successfully infected with the recombinant lentiviruses. The specificity and efficiency of the RHBDD1 RNA interference (RNAi) treatment was further Figure 2. Knockdown of rhomboid domain containing 1 (RHBDD1) suppresses RKO colorectal cancer cell proliferation. The growth curve of RKO cells with three treatments was determined by MTT assay. OD, optical density; Con, uninfected; Lv-shCon, control lentivirus with non-silencing; Lv-shRHBDD1, RHBDD1-silencing lentivirus. ** P<0.01, as compared to treatment with Lv-shCon.

380 HAN et al: RHBDD1 REGULATES CRC CELL GROWTH A B Figure 3. Rhomboid domain containing 1 (RHBDD1) knockdown reduces colony formation capacity of RKO colorectal cancer cells. (A) Representative images of colonies captured by a light microscopy. (B) Statistical analysis of the number of colonies with Gimesa staining in RKO cells with three treatments. Con, uninfected; Lv-shCon, control lentivirus with non-silencing; Lv-shRHBDD1, RHBDD1-silencing lentivirus ** P<0.01, as compared to treatment with Lv-shCon. A B Figure 4. Knockdown of rhomboid domain containing 1 (RHBDD1) blocks cell cycle progression. (A) Representative images of cell cycle distribution by flow cytometric analysis. (B) Knockdown of RHBDD1 in RKO ovarian cancer cells induced cell cycle arrest in G0/G1 phase at 72 h after lentivirus infection. Con, uninfected; Lv-shCon, control lentivirus with non-silencing; Lv-shRHBDD1, RHBDD1-silencing lentivirus. ** P<0.01, as compared to treatment with Lv-shCon. verified as the control lentivirus, inserted with one irrelevant sequence, had no impact on RHBDD1 translation (Fig. 1D). The target lentivirus Lv shrhbdd1 markedly downregulated the endogenous RHBDD1 mrna expression levels, as compared with the control lentivirus (P<0.01). Knockdown efficiency was further confirmed using western blot analysis, there was little RHBDD1 protein expression detected following RHBDD1 specific knockdown by RNAi (Fig. 1E). Knockdown of RHBDD1 inhibits RKO cell proliferation. To explore the role of RHBDD1 in CRC tumorigenesis, the proliferation of RKO cells following RHBDD1 expression knockdown, was analyzed using an MTT assay. As shown in Fig. 2, the proliferation rate of Lv shrhbdd1 infected cells was markedly lower, as compared with the Lv shcon infected and uninfected cells (P<0.01). These results indicate that knockdown of RHBDD1 expression may inhibit RKO cell proliferation. Knockdown of RHBDD1 suppresses colony formation of RKO cells. The colony forming capacity of RKO cells was determined using a monolayer cell culture. As shown in Fig.3A, knockdown of RHBDD1 expression markedly suppressed colony formation in RKO cells, as determined by a colony formation assay. The number of colonies formed in RKO cells infected with Lv shrhbdd1, was significantly decreased, as compared with the Lv shcon infected and uninfected cells (Fig. 3B, p<0.01).

MOLECULAR MEDICINE REPORTS 12: 377-381, 2015 381 These results indicate that RHBDD1 may have an important role in the cell growth and tumorigenesis of CRC. Knockdown of RHBDD1 induces G0/G1 phase arrest in RKO cells. To elucidate whether RHBDD1 knockdown induced cell growth inhibition by affecting the progression of the cell cycle, flow cytometric analysis was performed in RKO cells. As shown in Figure 4, following infection with Lv shrhbdd1, an increased number of cells accumulated at the G0/G1 phase and the percentage of cells in the S and G2/M phases were reduced (P<0.01). These results indicate that RHBDD1 knockdown may inhibit the growth of RKO cells via cell cycle arrest. Discussion Since the initial identification of rhomboid proteases (18), there have been indications that they may have a fundamental function within numerous cell types. However little research has been performed to determine their role in CRC. Vertebrate rhomboid genes are grouped into three classes and HBDD1 is classed as an active cellular rhomboid. Previous research has demonstrated that in Drosophila that rhomboid proteases regulate EGFR signaling pathways (19,20), and that rhomboid families may have pivotal roles in the modulation of EGFR transactivation (21). The monoclonal antibodies Cetuximab and Panitumumab, which target EGFR, have been effective against CRC, in clinical practice (22). However, it remains unclear as to whether the rhomboid protease family is involved in CRC progression. A previous study in hepatoma cells suggested that RHBDD1 may be a positive regulator for HCC cell growth and apoptosis using recombinant lentivirus-mediated silencing of RHBDD1 in HepG2 cells (17). It is therefore conceivable that RHBDD1 has a similar, essential role in CRC tumorigenesis. In the present study, we noted that RHBDD1 was widely expressed in numerous human CRC cell lines. To illuminate the functional role of RHBDD1 in the CRC cells, the present study used lentivirus-mediated sirna to silence the expression of RHBDD1 in the RKO colon cancer cells. Knock down of RHBDD1 expression was found to markedly suppress cell proliferation and colony formation in the RKO cells. Furthermore, flow cytometric analysis revealed that depletion of RHBDD1 in the RKO cells led to cell cycle arrest in the G0/G1 phase. A previous study from a murine model indicated that RHBDD1 has anti apoptotic potential and that spermatogonia GC-1 cells, a mouse derived spermatogonia line, were more sensitive to apoptotic stimuli following knock down of RHBDD1 expression (23). Further investigation is required to elucidate the modulation of RHBDD1 on CRC cell apoptosis and its regulatory mechanism in CRC development and progression. In conclusion, the findings of the present study demonstrated that RHBDD1 could promote CRC cell growth via cell cycle control. RHBDD1 may serve as a potential therapeutic target in human CRC. Acknowledgements The authors of the present study are grateful for the financial support received from the Academic Leaders Training Program of Pudong Health Bureau of Shanghai (no. PEWd2010 05). References 1. Cancer Genome Atlas Network: Comprehensive molecular characterization of human colon and rectal cancer. Nature 487: 330 337, 2012. 2. Merika E, Saif MW, Katz A, Syrigos K and Morse M: Review. Colon cancer vaccines: an update. In Vivo 24: 607 628, 2010. 3. Cunningham D, Atkin W, Lenz HJ, et al: Colorectal cancer. Lancet 375: 1030 1047, 2010. 4. Fearon ER: Molecular genetics of colorectal cancer. Annu Rev Pathol 6: 479 507, 2011. 5. Lemberg MK: Intramembrane proteolysis in regulated protein trafficking. Traffic 12: 1109 1118, 2011. 6. Brown MS, Ye J, Rawson RB and Goldstein JL: Regulated intramembrane proteolysis: a control mechanism conserved from bacteria to humans. Cell 100: 391 398, 2000. 7. Kroos L and Yu YT: Regulation of sigma factor activity during Bacillus subtilis development. Curr Opin Microbiol 3: 553 560, 2000. 8. Weihofen A and Martoglio B: Intramembrane cleaving proteases: controlled liberation of proteins and bioactive peptides. Trends Cell Biol 13: 71 78, 2003. 9. Freeman M: Rhomboid proteases and their biological functions. Annu Rev Genet 42: 191 210, 2008. 10. Wolfe MS and Kopan R: Intramembrane proteolysis: theme and variations. Science 305: 1119 1123, 2004. 11. Urban S: Rhomboid proteins: conserved membrane proteases with divergent biological functions. Genes Dev 20: 3054 3068, 2006. 12. Koonin EV, Makarova KS, Rogozin IB, Davidovic L, Letellier MC and Pellegrini L: The rhomboids: a nearly ubiquitous family of intramembrane serine proteases that probably evolved by multiple ancient horizontal gene transfers. Genome Biol 4: R19, 2003. 13. Wasserman JD, Urban S and Freeman M: A family of rhomboid like genes: Drosophila rhomboid 1 and roughoid/rhomboid 3 cooperate to activate EGF receptor signaling. Genes Dev 14: 1651 1663, 2000. 14. McQuibban GA, Saurya S and Freeman M: Mitochondrial membrane remodelling regulated by a conserved rhomboid protease. Nature 423: 537 541, 2003. 15. Wang Y, Guan X, Fok KL, et al: A novel member of the Rhomboid family, RHBDD1, regulates BIK mediated apoptosis. Cell Mol Life Sci 65: 3822 3829, 2008. 16. Lin YN, Gui FM, Shen H, et al: Expression of RHBDD1 gene in patients with chronic myeloid leukemia and its clinical significance. Zhongguo Shi Yan Xue Ye Xue Za Zhi 21: 12 15, 2013 (Chinese). 17. Liu XN, Tang ZH, Zhang Y, et al: Lentivirus mediated silencing of rhomboid domain containing 1 suppresses tumor growth and induces apoptosis in hepatoma HepG2 cells. Asian Pac J Cancer Prev 14: 5 9, 2013. 18. Urban S and Dickey SW: The rhomboid protease family: a decade of progress on function and mechanism. Genome Biol 12: 231, 2011. 19. Lee JR, Urban S, Garvey CF and Freeman M: Regulated intracellular ligand transport and proteolysis control EGF signal activation in Drosophila. Cell 107: 161 171, 2001. 20. Urban S, Lee JR and Freeman M: Drosophila rhomboid 1 defines a family of putative intramembrane serine proteases. Cell 107: 173 182, 2001. 21. Zou H, Thomas SM, Yan ZW, Grandis JR, Vogt A and Li LY: Human rhomboid family 1 gene RHBDF1 participates in GPCR mediated transactivation of EGFR growth signals in head and neck squamous cancer cells. FASEB J 23: 425 432, 2009. 22. Normanno N, Tejpar S, Morgillo F, De Luca A, Van Cutsem E and Ciardiello F: Implications for KRAS status and EGFR targeted therapies in metastatic CRC. Nat Rev Clin Oncol 6: 519 527, 2009. 23. Wang Y, Song W, Li S, et al: GC 1 mrhbdd1 knockdown spermatogonia cells lose their spermatogenic capacity in mouse seminiferous tubules. BMC Cell Biol 10: 25, 2009.