Mol. Cells 2018; 41(2): 119-126
Published online January 29, 2018
https://doi.org/10.14348/molcells.2018.2235
© The Korean Society for Molecular and Cellular Biology
Correspondence to : *Correspondence: liulinbo@zzu.edu.cn
microRNA (miR)-612 shows anticancer activity in several types of cancers, yet its function in melanoma is still unclear. This study was undertaken to investigate the expression of miR-612 and its biological relevance in melanoma cell growth, invasion, and tumorigenesis. The expression and prognostic significance of miR-612 in melanoma were examined. The effects of miR-612 overexpression on cell proliferation, colony formation, tumorigenesis, and invasion were determined. Rescue experiments were conducted to identify the functional target gene(s) of miR-612. miR-612 was significantly downregulated in melanoma tissues compared to adjacent normal tissues. Low miR-612 expression was significantly associated with melanoma thickness, lymph node metastasis, and shorter overall, and disease-free survival of patients. Overexpression of miR-612 significantly decreased cell proliferation, colony formation, and invasion of SK-MEL-28 and A375 melanoma cells.
Keywords aggressiveness, downregulation, metastasis, microRNA, target
Melanoma is one of the most lethal malignancies worldwide, accounting for 80% of skin cancer-related deaths (Kircher et al., 2016; van Akkooi et al., 2016). It has a high metastatic potential and poor prognosis. The median survival for advanced melanoma is less than 1 year (Pal et al., 2016; Tas, 2012). Deciphering the mechanism governing the aggressiveness of melanoma is critical for the development of effective therapeutic strategies against this malignancy.
Espin encodes an actin-binding protein and is implicated in actin cytoskeleton remodeling. Mutations in
microRNAs (miRs) are a large family of small non-coding RNA molecules that negatively regulate gene expression on the post-transcriptional level, typically through binding to the 3′-untranslated region (UTR) of target mRNAs (Cheerla and Gevaert, 2017). Although thousands of miRs have been detected in cancers (Shu et al., 2017; Zhang et al., 2016), the functions of most of them in tumor progression are not elucidated. Several miRs have been shown to contribute to the aggressive phenotype of melanoma cells (Komina et al., 2016; Xu et al., 2016). For instance, inhibition of miR-4286 exerts antiproliferative and pro-apoptotic effects on melanoma cells (Komina et al., 2016). It was found that miR-9 can suppress the growth and invasion of malignant melanoma cells (Xu et al., 2016).
miR-612 is one less characterized miR. Recent studies have reported that miR-612 acts as a tumor suppressor in colorectal cancer (Sheng et al., 2015) and liver cancer (Tang et al., 2014). Overexpression of miR-612 can inhibit the epithelial-mesenchymal transition and metastasis in hepatocellular carcinoma (Tao et al., 2013). However, its expression and function in melanoma is still elusive. The aim of this study is to determine the expression and clinical relevance of miR-612 in melanoma and uncover its biological role in melanoma cell growth, invasion, and tumorigenesis.
In this study, 89 cases of melanoma tissues (37 primary melanomas and 52 metastatic melanomas) and matched adjacent normal tissues were collected from melanoma patients who underwent surgical resection at Qilu Hospital (China). This cohort included 56 males and 33 females, with a median age of 51 years (range 39–78 years). The patients receiving any anticancer treatment before operation were excluded. Tissue specimens were snap-frozen in liquid nitrogen immediately after surgery and stored at −80°C until use. This study was approved by the Ethics Committee of Shandong University (China), and written informed consent for research purposes was obtained from all patients.
Human melanoma cell lines (SK-MEL-28, SK-MEL-3, A375, HT-144, and Hs294T) were purchased from the American Type Culture Collection (ATCC, USA) and maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% heat-inactive fetal bovine serum (FBS; Gibco/Thermo Fisher Scientific, USA) in a CO2 incubator at 37°C. Human epidermal melanocytes were purchased from Scien-Cell Research Laboratories (USA) and cultured in Melanocyte Medium (ScienCell Research Laboratories) containing 10% FBS. HEK293T cells were purchased from the Institute of Biochemistry and Cell Biology of Chinese Academy of Sciences (China) and cultured in DMEM with 10% FBS.
Melanoma cell lines were plated in 96-well plates (1 × 104 cells/well) and exposed to different concentration of doxorubicin (0, 1, 2, 5, and 10 μM; Sigma-Aldrich, USA) for 24 h. The cell culture was added with 0.5 mg/ml 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Sigma-Aldrich). After incubation for 4 h at 37°C, dimethyl sulfoxide was added. Absorbance was measured at 570 nm. The half maximal inhibitory concentration (IC50) for doxorubicin was calculated.
Total RNA was extracted from tissue samples and cell lines using TRIzol reagent (Invitrogen, USA). The expression level of miR-612 was measured by qRT-PCR using the TaqMan MicroRNA Assay Kit (Applied Biosystems, USA). RNU6B was used as an endogenous control.
To generate a miR-612-expressing plasmid, a DNA fragment containing human miR-612 precursor and its flanking sequence was amplified by PCR and inserted into pcDNA3.1(+) vector. Human Espin-encoding cDNA (Origene Technologies, USA) lacking the 3′-UTR was amplified by PCR and cloned into pBABE-puro vector. For luciferase reporter assays, the entire
Transfection of the plasmids into cells was performed using Lipofectamine 2000 (Invitrogen) as per the manufacturer’s instructions. To generate miR-612 stable clones, melanoma cells were transfected with the miR-612-expressing plasmid and after 24 h, selected in the presence of 800 μg/ml G418 (Sigma-Aldrich) for 2 weeks. For co-expression of miR-612 and Espin, miR-612 stable cell lines were re-transfected with the pBABE-Espin plasmid and then selected in the presence of 800 μg/ml G418 and 2 μg/ml puromycin (Sigma-Aldrich) for 1 week. For knockdown of Espin, specific small interfering RNAs (siRNAs) targeting Espin (Santa Cruz Biotechnology, USA) were transfected at a concentration of 40 nM.
Cell proliferation was measured using the Cell Proliferation ELISA, BrdU (colorimetric) Kit (Roche Applied Science, USA), as described previously [12]. Absorbance was measured at 370 nm.
Colony formation assay was done as described previously [13]. Transfected cells (500 cells per well) were seeded onto 6-well plates and cultured for 10 days. Colonies were stained with 0.5% crystal violet (Sigma-Aldrich) solution and counted.
Cells were seeded in 6-wells plates (3 × 106 cells/well) and allowed to grow to confluence and incubated with 10 μg/mL mitomycin C (Sigma-Aldrich) for 2 h. A scratch wound was made with a 200-μl pipette tip. Cells were further cultured in DMEM medium containing 10% FBS. Pictures were taken at 0 and 48 h after scratching. The percentage of wound closure was determined from three independent experiments.
Cancer cell invasive ability was assessed using Transwell invasion assay (Li et al., 2016). In brief, 2 × 104 cells suspended in serum-free medium were seeded in the upper chamber of 24-well Transwell plates, which were precoated with Matrigel (BD Biosciences, USA). The lower chamber was filled with the cell culture medium containing 10% FBS. After incubation for 48 h, cells that had invaded through Matrigel membrane were fixed, stained with 0.1% crystal violet, and counted.
HEK293T cells were co-transfected with the
Tissue or cell lystates were prepared using radioimmunoprecipitation assay (RIPA) buffer (Beyotime, China) supplemented with protease and phosphatase inhibitors (Sigma-Aldrich). Protein samples were separated by SDS-polyacrylamide gel electrophoresis and transferred to nitrocellulose membranes. After blocking non-specific binding sites, membranes were incubated with anti-Espin (sc-393469, Santa Cruz Biotechnology; 1:500 dilution), anti-E-cadherin (ab15148, Abcam, Abcam, USA; 1:800 dilution), anti-vimentin (ab8978, Abcam; 1:500 dilution), and anti-β-actin (Sigma-Aldrich; 1:2000 dilution) antibodies, followed by incubation with horseradish peroxidase-conjugated secondary antibody (Santa Cruz Biotechnology). Protein bands were visualized using an enhanced chemiluminescence detection system (Thermo Fisher Scientific, USA). Densitometric analysis was carried out using the Quantity One software (Bio-Rad Laboratories, USA).
Data are expressed as mean ± standard deviation and analyzed by the Student’s
We first examined the levels of miR-612 in a set of melanoma tissues and their matched noncancerous normal tissues. As illustrated in Fig. 1A, the miR-612 transcript level was significantly lower in melanoma tissues than in paired adjacent normal tissues (
Next, we explored the function of miR-612 in melanoma progression. Compared to non-malignant melanocytes, miR-612 was commonly downregulated in melanoma cell lines (Fig. 2A). Overexpression of miR-612 (Fig. 2B) significantly reduced proliferation in both SK-MEL-28 and A375 melanoma cells, as determined by BrdU incorporation assays (
Next, we investigated whether miR-612 participated in the regulation of migration and invasion of melanoma cells.
To gain more insight into the role of miR-612 in melanoma, we attempted to identify functionally relevant targets. Using the TargetScan algorithm (
Next, we conducted rescue experiments to validate the involvement of Espin in the activity of miR-612 in melanoma. As determined by Western blot analysis, co-transfection with the Espin-expressing plasmid resulted in abundant expression of Espin in miR-612-overexpressing A375 cells (Fig. 5A).
Finally, we checked the role of miR-612/Espin axis in the regulation of drug sensitivity in melanoma cells. As illustrated in Fig. 6A, the IC50 for doxorubicin was reduced by 2- and 4-fold in miR-612-overexpressing SK-MEL-28 and A375 cells, respectively. Similarly, Espin knockdown led to a significant decline in the IC50 for doxorubicin in both SK-MEL-28 and A375 cells (Fig. 6B). However, the sensitivity of melanoma cells to paclitaxel was not affected by either miR-612 overexpression or Espin knockdown (data not shown).
It has been documented that miR-612 is downregulated in lung metastatic lesions of hepatocellular carcinoma (HCC) relative to primary HCC tissues (Tao et al., 2013). Another study demonstrated that miR-612 expression is significantly reduced in colorectal cancer (CRC) tissues, in particular metastatic CRC lesions (Sheng et al., 2015). miR-612 is dysregulated in pancreatic cancer (Mao et al., 2017). Consistently, our data showed that melanoma tissues had significantly decreased levels of miR-612, compared to paired adjacent normal tissues. Furthermore, low miR-612 expression correlated with aggressive parameters and predicted poor prognosis in melanoma patients. The clinical data suggest a link between miR-612 dysregulation and melanoma progression. To confirm the downregulation of miR-612 in tumor cells, we also examined the expression of miR-612 in a panel of melanoma cell lines. In line with clinical findings, miR-612 was generally downregulated in melanoma cells relative to normal human melanocytes.
Functionally, it was observed that ectopic expression of miR-612 restrained melanoma cell proliferation, colony formation, an invasion
Mechanistically, it was found that miR-612 negatively regulated the expression of Espin in melanoma cells. Luciferase reporter assays confirmed that miR-612 targeted the 3′-UTR of
In conclusion, miR-612 is downregulated and acts as a tumor suppression in melanoma. Targeting Espin represents an important mechanism for miR-612-mediated inhibition of aggressive phenotype of melanoma cells. Restoration of miR-612 could be a potential therapeutic strategy against melanoma.
Mol. Cells 2018; 41(2): 119-126
Published online February 28, 2018 https://doi.org/10.14348/molcells.2018.2235
Copyright © The Korean Society for Molecular and Cellular Biology.
Ying Zhu, Hao-liang Zhang, Qi-ying Wang, Min-jing Chen, and Lin-bo Liu*
Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
Correspondence to:*Correspondence: liulinbo@zzu.edu.cn
microRNA (miR)-612 shows anticancer activity in several types of cancers, yet its function in melanoma is still unclear. This study was undertaken to investigate the expression of miR-612 and its biological relevance in melanoma cell growth, invasion, and tumorigenesis. The expression and prognostic significance of miR-612 in melanoma were examined. The effects of miR-612 overexpression on cell proliferation, colony formation, tumorigenesis, and invasion were determined. Rescue experiments were conducted to identify the functional target gene(s) of miR-612. miR-612 was significantly downregulated in melanoma tissues compared to adjacent normal tissues. Low miR-612 expression was significantly associated with melanoma thickness, lymph node metastasis, and shorter overall, and disease-free survival of patients. Overexpression of miR-612 significantly decreased cell proliferation, colony formation, and invasion of SK-MEL-28 and A375 melanoma cells.
Keywords: aggressiveness, downregulation, metastasis, microRNA, target
Melanoma is one of the most lethal malignancies worldwide, accounting for 80% of skin cancer-related deaths (Kircher et al., 2016; van Akkooi et al., 2016). It has a high metastatic potential and poor prognosis. The median survival for advanced melanoma is less than 1 year (Pal et al., 2016; Tas, 2012). Deciphering the mechanism governing the aggressiveness of melanoma is critical for the development of effective therapeutic strategies against this malignancy.
Espin encodes an actin-binding protein and is implicated in actin cytoskeleton remodeling. Mutations in
microRNAs (miRs) are a large family of small non-coding RNA molecules that negatively regulate gene expression on the post-transcriptional level, typically through binding to the 3′-untranslated region (UTR) of target mRNAs (Cheerla and Gevaert, 2017). Although thousands of miRs have been detected in cancers (Shu et al., 2017; Zhang et al., 2016), the functions of most of them in tumor progression are not elucidated. Several miRs have been shown to contribute to the aggressive phenotype of melanoma cells (Komina et al., 2016; Xu et al., 2016). For instance, inhibition of miR-4286 exerts antiproliferative and pro-apoptotic effects on melanoma cells (Komina et al., 2016). It was found that miR-9 can suppress the growth and invasion of malignant melanoma cells (Xu et al., 2016).
miR-612 is one less characterized miR. Recent studies have reported that miR-612 acts as a tumor suppressor in colorectal cancer (Sheng et al., 2015) and liver cancer (Tang et al., 2014). Overexpression of miR-612 can inhibit the epithelial-mesenchymal transition and metastasis in hepatocellular carcinoma (Tao et al., 2013). However, its expression and function in melanoma is still elusive. The aim of this study is to determine the expression and clinical relevance of miR-612 in melanoma and uncover its biological role in melanoma cell growth, invasion, and tumorigenesis.
In this study, 89 cases of melanoma tissues (37 primary melanomas and 52 metastatic melanomas) and matched adjacent normal tissues were collected from melanoma patients who underwent surgical resection at Qilu Hospital (China). This cohort included 56 males and 33 females, with a median age of 51 years (range 39–78 years). The patients receiving any anticancer treatment before operation were excluded. Tissue specimens were snap-frozen in liquid nitrogen immediately after surgery and stored at −80°C until use. This study was approved by the Ethics Committee of Shandong University (China), and written informed consent for research purposes was obtained from all patients.
Human melanoma cell lines (SK-MEL-28, SK-MEL-3, A375, HT-144, and Hs294T) were purchased from the American Type Culture Collection (ATCC, USA) and maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% heat-inactive fetal bovine serum (FBS; Gibco/Thermo Fisher Scientific, USA) in a CO2 incubator at 37°C. Human epidermal melanocytes were purchased from Scien-Cell Research Laboratories (USA) and cultured in Melanocyte Medium (ScienCell Research Laboratories) containing 10% FBS. HEK293T cells were purchased from the Institute of Biochemistry and Cell Biology of Chinese Academy of Sciences (China) and cultured in DMEM with 10% FBS.
Melanoma cell lines were plated in 96-well plates (1 × 104 cells/well) and exposed to different concentration of doxorubicin (0, 1, 2, 5, and 10 μM; Sigma-Aldrich, USA) for 24 h. The cell culture was added with 0.5 mg/ml 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Sigma-Aldrich). After incubation for 4 h at 37°C, dimethyl sulfoxide was added. Absorbance was measured at 570 nm. The half maximal inhibitory concentration (IC50) for doxorubicin was calculated.
Total RNA was extracted from tissue samples and cell lines using TRIzol reagent (Invitrogen, USA). The expression level of miR-612 was measured by qRT-PCR using the TaqMan MicroRNA Assay Kit (Applied Biosystems, USA). RNU6B was used as an endogenous control.
To generate a miR-612-expressing plasmid, a DNA fragment containing human miR-612 precursor and its flanking sequence was amplified by PCR and inserted into pcDNA3.1(+) vector. Human Espin-encoding cDNA (Origene Technologies, USA) lacking the 3′-UTR was amplified by PCR and cloned into pBABE-puro vector. For luciferase reporter assays, the entire
Transfection of the plasmids into cells was performed using Lipofectamine 2000 (Invitrogen) as per the manufacturer’s instructions. To generate miR-612 stable clones, melanoma cells were transfected with the miR-612-expressing plasmid and after 24 h, selected in the presence of 800 μg/ml G418 (Sigma-Aldrich) for 2 weeks. For co-expression of miR-612 and Espin, miR-612 stable cell lines were re-transfected with the pBABE-Espin plasmid and then selected in the presence of 800 μg/ml G418 and 2 μg/ml puromycin (Sigma-Aldrich) for 1 week. For knockdown of Espin, specific small interfering RNAs (siRNAs) targeting Espin (Santa Cruz Biotechnology, USA) were transfected at a concentration of 40 nM.
Cell proliferation was measured using the Cell Proliferation ELISA, BrdU (colorimetric) Kit (Roche Applied Science, USA), as described previously [12]. Absorbance was measured at 370 nm.
Colony formation assay was done as described previously [13]. Transfected cells (500 cells per well) were seeded onto 6-well plates and cultured for 10 days. Colonies were stained with 0.5% crystal violet (Sigma-Aldrich) solution and counted.
Cells were seeded in 6-wells plates (3 × 106 cells/well) and allowed to grow to confluence and incubated with 10 μg/mL mitomycin C (Sigma-Aldrich) for 2 h. A scratch wound was made with a 200-μl pipette tip. Cells were further cultured in DMEM medium containing 10% FBS. Pictures were taken at 0 and 48 h after scratching. The percentage of wound closure was determined from three independent experiments.
Cancer cell invasive ability was assessed using Transwell invasion assay (Li et al., 2016). In brief, 2 × 104 cells suspended in serum-free medium were seeded in the upper chamber of 24-well Transwell plates, which were precoated with Matrigel (BD Biosciences, USA). The lower chamber was filled with the cell culture medium containing 10% FBS. After incubation for 48 h, cells that had invaded through Matrigel membrane were fixed, stained with 0.1% crystal violet, and counted.
HEK293T cells were co-transfected with the
Tissue or cell lystates were prepared using radioimmunoprecipitation assay (RIPA) buffer (Beyotime, China) supplemented with protease and phosphatase inhibitors (Sigma-Aldrich). Protein samples were separated by SDS-polyacrylamide gel electrophoresis and transferred to nitrocellulose membranes. After blocking non-specific binding sites, membranes were incubated with anti-Espin (sc-393469, Santa Cruz Biotechnology; 1:500 dilution), anti-E-cadherin (ab15148, Abcam, Abcam, USA; 1:800 dilution), anti-vimentin (ab8978, Abcam; 1:500 dilution), and anti-β-actin (Sigma-Aldrich; 1:2000 dilution) antibodies, followed by incubation with horseradish peroxidase-conjugated secondary antibody (Santa Cruz Biotechnology). Protein bands were visualized using an enhanced chemiluminescence detection system (Thermo Fisher Scientific, USA). Densitometric analysis was carried out using the Quantity One software (Bio-Rad Laboratories, USA).
Data are expressed as mean ± standard deviation and analyzed by the Student’s
We first examined the levels of miR-612 in a set of melanoma tissues and their matched noncancerous normal tissues. As illustrated in Fig. 1A, the miR-612 transcript level was significantly lower in melanoma tissues than in paired adjacent normal tissues (
Next, we explored the function of miR-612 in melanoma progression. Compared to non-malignant melanocytes, miR-612 was commonly downregulated in melanoma cell lines (Fig. 2A). Overexpression of miR-612 (Fig. 2B) significantly reduced proliferation in both SK-MEL-28 and A375 melanoma cells, as determined by BrdU incorporation assays (
Next, we investigated whether miR-612 participated in the regulation of migration and invasion of melanoma cells.
To gain more insight into the role of miR-612 in melanoma, we attempted to identify functionally relevant targets. Using the TargetScan algorithm (
Next, we conducted rescue experiments to validate the involvement of Espin in the activity of miR-612 in melanoma. As determined by Western blot analysis, co-transfection with the Espin-expressing plasmid resulted in abundant expression of Espin in miR-612-overexpressing A375 cells (Fig. 5A).
Finally, we checked the role of miR-612/Espin axis in the regulation of drug sensitivity in melanoma cells. As illustrated in Fig. 6A, the IC50 for doxorubicin was reduced by 2- and 4-fold in miR-612-overexpressing SK-MEL-28 and A375 cells, respectively. Similarly, Espin knockdown led to a significant decline in the IC50 for doxorubicin in both SK-MEL-28 and A375 cells (Fig. 6B). However, the sensitivity of melanoma cells to paclitaxel was not affected by either miR-612 overexpression or Espin knockdown (data not shown).
It has been documented that miR-612 is downregulated in lung metastatic lesions of hepatocellular carcinoma (HCC) relative to primary HCC tissues (Tao et al., 2013). Another study demonstrated that miR-612 expression is significantly reduced in colorectal cancer (CRC) tissues, in particular metastatic CRC lesions (Sheng et al., 2015). miR-612 is dysregulated in pancreatic cancer (Mao et al., 2017). Consistently, our data showed that melanoma tissues had significantly decreased levels of miR-612, compared to paired adjacent normal tissues. Furthermore, low miR-612 expression correlated with aggressive parameters and predicted poor prognosis in melanoma patients. The clinical data suggest a link between miR-612 dysregulation and melanoma progression. To confirm the downregulation of miR-612 in tumor cells, we also examined the expression of miR-612 in a panel of melanoma cell lines. In line with clinical findings, miR-612 was generally downregulated in melanoma cells relative to normal human melanocytes.
Functionally, it was observed that ectopic expression of miR-612 restrained melanoma cell proliferation, colony formation, an invasion
Mechanistically, it was found that miR-612 negatively regulated the expression of Espin in melanoma cells. Luciferase reporter assays confirmed that miR-612 targeted the 3′-UTR of
In conclusion, miR-612 is downregulated and acts as a tumor suppression in melanoma. Targeting Espin represents an important mechanism for miR-612-mediated inhibition of aggressive phenotype of melanoma cells. Restoration of miR-612 could be a potential therapeutic strategy against melanoma.
Zhihong Gao*, Xiaoyan Luo, Ting Shi, Bin Cai, Zhen Zhang, Zongming Cheng, and Weibing Zhuang
Mol. Cells 2012; 34(3): 239-249 https://doi.org/10.1007/s10059-012-0004-7Christopher J. Occhiuto, Jessica A. Moerland, Ana S. Leal, Kathleen A. Gallo, and Karen T. Liby
Mol. Cells 2023; 46(3): 176-186 https://doi.org/10.14348/molcells.2023.2191Min Ji Park, Eunji Jeong, Eun Ji Lee, Hyeon Ji Choi, Bo Hyun Moon, Keunsoo Kang, and Suhwan Chang
Mol. Cells 2023; 46(6): 351-359 https://doi.org/10.14348/molcells.2023.2174