1
|
Lee YJ, Choi YS, Kim S, Heo JY, Kim DS, Kim KD, Nam SM, Nam HS, Lee SH, Choi D, Cho MK. Overexpression of Dock180 and Elmo1 in Melanoma is Associated with Cell Survival and Migration. Ann Dermatol 2023; 35:439-450. [PMID: 38086358 PMCID: PMC10733078 DOI: 10.5021/ad.23.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 12/22/2023] Open
Abstract
BACKGROUND Melanoma is one of the most aggressive and metastatic skin cancers. Although overexpression of Dock180 and Elmo1 has been identified in various cancers, including glioma, ovarian cancer, and breast cancer, their expression and functions in melanoma remain unknown. OBJECTIVE This study aims to confirm the expression of Dock180 and Elmo1, their underlying mechanisms, and roles in melanoma. METHODS Both immunohistochemical staining and Western blotting were used to confirm expression of Dock180 and Elmo1 in human melanoma. To identify roles of Dock180 and Elmo1 in cell survival, apoptosis and migration, downregulation of Dock180 or Elmo1 in melanoma cells with small interfering RNA (siRNA) was performed. RESULTS We identified overexpression of Dock180 and Elmo1 in human melanoma compared to normal skin ex vivo. Inhibition of Dock180 or Elmo1 following siRNA in melanoma cells reduced cell viability and increased apoptosis as supported by increased proportion of cells with Annexin V-PE (+) staining and sub-G0/G1 peak in cell cycle analysis. Moreover, inhibition of Dock180 or Elmo1 regulated apoptosis-related proteins, showing downregulation of Bcl-2, caspase-3, and PARP and upregulation of Bax, PUMA, cleaved caspase-3, and cleaved PARP. Furthermore, knockdown of Dock180 and Elmo1 in melanoma cells reduced cell migration and changed cellular signaling pathways including ERK and AKT. Vemurafenib decreased cell viability in concentration-dependent manner, while transfection with Dock180- or Elmo1-specific siRNA in melanoma cells significantly reduced cell viability. CONCLUSION Our results suggest that both Dock180 and Elmo1 may be associated with cancer progression, and can be potential targets for treatment of melanoma.
Collapse
Affiliation(s)
- Yoon Jin Lee
- Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Yu Sung Choi
- Department of Dermatology, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Sooyoung Kim
- Department of Dermatology, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Jae Young Heo
- Department of Dermatology, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Dong Sung Kim
- Department of Dermatology, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Ki Dam Kim
- Department of Dermatology, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Seung Min Nam
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Hae Seon Nam
- Division of Molecular Cancer Research, Soonchunhyang Medical Research Institute, Soonchunhyang University, Cheonan, Korea
| | - Sang Han Lee
- Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Dongsic Choi
- Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Moon Kyun Cho
- Department of Dermatology, Soonchunhyang University Seoul Hospital, Seoul, Korea.
| |
Collapse
|
2
|
Yang G, Li H, Dong Z, Deng K, Lu Y. Nucleophosmin 1 associating with engulfment and cell motility protein 1 regulates hepatocellular carcinoma cell chemotaxis and metastasis. Open Med (Wars) 2023; 18:20230708. [PMID: 37251542 PMCID: PMC10224614 DOI: 10.1515/med-2023-0708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/24/2023] [Accepted: 04/17/2023] [Indexed: 05/31/2023] Open
Abstract
The chemokine, C-X-C motif chemokine ligand 12 (CXCL12) and its G-protein-coupled receptor (GPCR) and C-X-C chemokine receptor type 4 (CXCR4), are closely associated with promoting hepatocellular carcinoma (HCC) chemotaxis and metastasis. The binding of CXCL12 and CXCR4 depends on the heterotrimeric Gi proteins to regulate actin polymerisation and mobility in HCC. Although the role of GPCR/Gi signalling in carcinogenesis migration has been intensively studied, the detailed mechanism remains largely unknown. In this study, a small interfering RNA technique was used to knock down the Nucleophosmin 1 (NPM1) gene expression. Through the chemotaxis and invasion assays, wound healing, proliferation, filamentous-actin, immunofluorescence, immunohistochemical assays, and co-immunoprecipitation assays, we investigated the specific biological role and underlying mechanisms of the NPM1 in HCC. Additionally, dimethyl fumarate (DMF), a fumaric acid ester, was used to inhibit the HCC cell chemokines and metastasis by regulating ELMO1 and NPM1. Therefore, this study reported that NPM1 gene expression was upregulated in the HCC tissues and cell lines. The NPM1 knockdown significantly inhibited the proliferation, migration, and chemotaxis of the HepG2 cells in vitro. Further mechanistic studies suggested that the NPM1 interacts with ELMO1 and the CXCL12/CXCR4 pathway activates NPM1-dependent regulation of the ELMO1 localisation. Furthermore, the DMF significantly inhibited tumour metastasis induced by the NPM1/ELMO1 signalling pathway, as observed in in vitro cell functional experiments. These data suggested that as a potentially novel therapeutic approach, the simultaneous targeting of NPM1 and ELMO1 could effectively be used to treat HCC.
Collapse
Affiliation(s)
- Gangqi Yang
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, China
- General Surgery Department and Neurology Department, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Beijing100053, China
- Comprehensive Liver Cancer Center, 5th Medical Center of the PLA General Hospital, Beijing100039, China
- Guangdong Key Laboratory of Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Hongyan Li
- General Surgery Department and Neurology Department, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Beijing100053, China
| | - Zheng Dong
- Comprehensive Liver Cancer Center, 5th Medical Center of the PLA General Hospital, Beijing100039, China
| | - Kai Deng
- The First Affiliated Hospital of Chongqing Medical College, Chongqing400016, China
| | - Yinying Lu
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, China
- Comprehensive Liver Cancer Center, 5th Medical Center of the PLA General Hospital, Beijing100039, China
- Guangdong Key Laboratory of Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518055, China
| |
Collapse
|
3
|
Wen B, Li S, Ruan L, Yang Y, Chen Z, Zhang B, Yang X, Jie H, Li S, Zeng Z, Liu S. Engulfment and cell motility protein 1 fosters reprogramming of tumor-associated macrophages in colorectal cancer. Cancer Sci 2022; 114:410-422. [PMID: 36310143 PMCID: PMC9899619 DOI: 10.1111/cas.15628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/26/2022] [Accepted: 10/18/2022] [Indexed: 11/29/2022] Open
Abstract
Functional reprogramming of tumor-associated macrophages (TAMs) is crucial to their potent tumor-supportive capacity. However, the molecular mechanism behind the reprogramming process remains poorly understood. Here, we identify engulfment and cell motility protein 1 (ELMO1) as a crucial player for TAM reprogramming in colorectal cancer (CRC). The expression of ELMO1 in stromal but not epithelial tumor cells was positively associated with advanced clinical stage and poor disease-free survival in CRC. An increase in ELMO1 expression was specifically found in TAMs, but not in other multiple nonmalignant stromal cells. Gain- and loss-of-function assays indicated ELMO1 reprogrammed macrophages to a TAM-like phenotype through Rac1 activation. In turn, ELMO1-reprogrammed macrophages were shown to not only facilitate the malignant behaviors of CRC cells but exhibited potent phagocytosis of tumor cells. Taken together, our work underscores the importance of ELMO1 in determining functional reprogramming of TAMs and could provide new insights on potential therapeutic strategies against CRC.
Collapse
Affiliation(s)
- Bo Wen
- Department of Gastrointestinal SurgeryCentral Hospital of ShaoyangShaoyangChina
| | - Sheng Li
- Department of Gastrointestinal SurgeryCentral Hospital of ShaoyangShaoyangChina
| | - Lei Ruan
- Department of Colorectal Surgery and Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Yanping Yang
- Department of PathologyCentral Hospital of ShaoyangShaoyangChina
| | - Zilin Chen
- Department of Medical OncologyCentral Hospital of ShaoyangShaoyangChina
| | - Bin Zhang
- Department of Gastrointestinal SurgeryCentral Hospital of ShaoyangShaoyangChina
| | - Xin Yang
- Department of Colorectal Surgery and Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Haiqing Jie
- Department of Colorectal Surgery and Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Shujuan Li
- Department of PharmacyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhou, HenanChina
| | - Ziwei Zeng
- Department of Colorectal Surgery and Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina,University Clinic MannheimMedical Faculty Mannheim, University of HeidelbergMannheimGermany
| | - Sisi Liu
- Department of PathologyCentral Hospital of ShaoyangShaoyangChina
| |
Collapse
|
4
|
Tocci S, Ibeawuchi SR, Das S, Sayed IM. Role of ELMO1 in inflammation and cancer-clinical implications. Cell Oncol (Dordr) 2022; 45:505-525. [PMID: 35668246 DOI: 10.1007/s13402-022-00680-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Engulfment and cell motility protein 1 (ELMO1) is a key protein for innate immunity since it is required for the clearance of apoptotic cells and pathogenic bacteria as well as for the control of inflammatory responses. ELMO1, through binding with Dock180 and activation of the Rac1 signaling pathway, plays a significant role in cellular shaping and motility. Rac-mediated actin cytoskeletal rearrangement is essential for bacterial phagocytosis, but also plays a crucial role in processes such as cancer cell invasion and metastasis. While the role of ELMO1 in bacterial infection and inflammatory responses is well established, its implication in cancer is not widely explored yet. Molecular changes or epigenetic alterations such as DNA methylation, which ultimately leads to alterations in gene expression and deregulation of cellular signaling, has been reported for ELMO1 in different cancer types. CONCLUSIONS In this review, we provide an updated and comprehensive summary of the roles of ELMO1 in infection, inflammatory diseases and cancer. We highlight the possible mechanisms regulated by ELMO1 that are relevant for cancer development and progression and provide insight into the possible use of ELMO1 as a diagnostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Stefania Tocci
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | | | - Soumita Das
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
| | - Ibrahim M Sayed
- Department of Pathology, University of California San Diego, La Jolla, CA, USA. .,Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| |
Collapse
|
5
|
Xue R, Wang Y, Wang T, Lyu M, Mo G, Fan X, Li J, Yen K, Yu S, Liu Q, Xu J. Functional Verification of Novel ELMO1 Variants by Live Imaging in Zebrafish. Front Cell Dev Biol 2021; 9:723804. [PMID: 34993193 PMCID: PMC8724260 DOI: 10.3389/fcell.2021.723804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/17/2021] [Indexed: 02/02/2023] Open
Abstract
ELMO1 (Engulfment and Cell Motility1) is a gene involved in regulating cell motility through the ELMO1-DOCK2-RAC complex. Contrary to DOCK2 (Dedicator of Cytokinesis 2) deficiency, which has been reported to be associated with immunodeficiency diseases, variants of ELMO1 have been associated with autoimmune diseases, such as diabetes and rheumatoid arthritis (RA). To explore the function of ELMO1 in immune cells and to verify the functions of novel ELMO1 variants in vivo, we established a zebrafish elmo1 mutant model. Live imaging revealed that, similar to mammals, the motility of neutrophils and T-cells was largely attenuated in zebrafish mutants. Consequently, the response of neutrophils to injury or bacterial infection was significantly reduced in the mutants. Furthermore, the reduced mobility of neutrophils could be rescued by the expression of constitutively activated Rac proteins, suggesting that zebrafish elmo1 mutant functions via a conserved mechanism. With this mutant, three novel human ELMO1 variants were transiently and specifically expressed in zebrafish neutrophils. Two variants, p.E90K (c.268G>A) and p.D194G (c.581A>G), could efficiently recover the motility defect of neutrophils in the elmo1 mutant; however, the p.R354X (c.1060C>T) variant failed to rescue the mutant. Based on those results, we identified that zebrafish elmo1 plays conserved roles in cell motility, similar to higher vertebrates. Using the transient-expression assay, zebrafish elmo1 mutants could serve as an effective model for human variant verification in vivo.
Collapse
Affiliation(s)
- Rongtao Xue
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Wang
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | | | - Mei Lyu
- Laboratory of Immunology and Regeneration, School of Medicine, South China University of Technology, Guangzhou, China
| | - Guiling Mo
- GuangZhou KingMed Center For Clinical Laboratory Co., Ltd., International Biotech Island, Guangzhou, China
| | - Xijie Fan
- GuangZhou KingMed Center For Clinical Laboratory Co., Ltd., International Biotech Island, Guangzhou, China
| | - Jianchao Li
- Laboratory of Molecular and Structural Biology, School of Medicine, South China University of Technology, Guangzhou, China
| | - Kuangyu Yen
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- *Correspondence: Kuangyu Yen, ; Shihui Yu, ; Qifa Liu, ; Jin Xu,
| | - Shihui Yu
- GuangZhou KingMed Center For Clinical Laboratory Co., Ltd., International Biotech Island, Guangzhou, China
- *Correspondence: Kuangyu Yen, ; Shihui Yu, ; Qifa Liu, ; Jin Xu,
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Kuangyu Yen, ; Shihui Yu, ; Qifa Liu, ; Jin Xu,
| | - Jin Xu
- Laboratory of Immunology and Regeneration, School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Kuangyu Yen, ; Shihui Yu, ; Qifa Liu, ; Jin Xu,
| |
Collapse
|
6
|
Park YL, Choi JH, Park SY, Oh HH, Kim DH, Seo YJ, So JK, Song K, Cho MS, Chung MW, Hong JY, Kim KH, Myung E, Myung DS, Cho SB, Lee WS, Park D, Joo YE. Engulfment and cell motility 1 promotes tumor progression via the modulation of tumor cell survival in gastric cancer. Am J Transl Res 2020; 12:7797-7811. [PMID: 33437361 PMCID: PMC7791502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/25/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND/AIM Engulfment and cell motility 1 (ELMO1) protein has been implicated in phagocytosis of apoptotic cells, cell migration, neurite outgrowth, cancer cell invasion and metastasis, and poor prognosis in various cancers. We investigated the role of ELMO1 in mediating the oncogenic behavior of gastric cancer (GC) cells. We also investigated the correlation between expression of ELMO1 in GC tissues and various clinicopathological parameters. METHODS We studied the impact of ELMO1 on tumor cell behavior using the pcDNA-myc vector and small interfering RNA in AGS and SNU1750 GC cell lines. We performed western blotting and immunohistochemistry to investigate the expression of ELMO1 in GC cells and tissues. RESULTS ELMO1 overexpression inhibited apoptosis via the modulation of PARP, caspase-3 and caspase-7 in GC cells. ELMO1 overexpression led to significant increase in the number of migrating and invading GC cells. The expression of E-cadherin decreased and that of Snail increased in GC cells upon ELMO1 overexpression. Phosphorylation of PI3K/Akt and GSK-3β was increased and that of β-catenin was decreased upon ELMO1 overexpression in GC cells. These results were reversed after ELMO1 knockdown. ELMO1 expression was significantly associated with tumor size, cancer stage, lymph node metastasis and survival. ELMO1-positive tumors had significantly higher mean of Ki-67 labeling index than ELMO1-negative tumors. There was no significant relationship between ELMO1 expression and the mean value of the apoptotic index. CONCLUSIONS Our results indicate that ELMO1 promotes tumor progression by modulating tumor cell survival in human GC.
Collapse
Affiliation(s)
- Young-Lan Park
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Jung-Ho Choi
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Sun-Young Park
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Hyung-Hoon Oh
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Dong-Ho Kim
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Yoon-Jin Seo
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Jae-Kyoung So
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Kaeun Song
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Min-Seok Cho
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Min-Woo Chung
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Ji-Yun Hong
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Ki-Hyun Kim
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Eun Myung
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Dae-Seong Myung
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Sung-Bum Cho
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Wan-Sik Lee
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| | - Daeho Park
- School of Life Sciences and Bio Imaging Research Center, Gwangju Institute of Science and TechnologyGwangju, Republic of Korea
| | - Young-Eun Joo
- Department of Internal Medicine, Chonnam National University Medical SchoolGwangju, Republic of Korea
| |
Collapse
|
7
|
Wang Y, Li H, Li F. ELMO2 association with G αi2 regulates pancreatic cancer cell chemotaxis and metastasis. PeerJ 2020; 8:e8910. [PMID: 32292657 PMCID: PMC7144586 DOI: 10.7717/peerj.8910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/15/2020] [Indexed: 01/22/2023] Open
Abstract
Background Pancreatic cancer is a highly lethal disease. Nearly half of the patients have distant metastasis and remain asymptomatic. Emerging evidence suggests that the chemokine, CXCL12, has a role in cancer metastasis. The interaction between CXCL12 and CXCR4 activates heterotrimeric G proteins, which regulates actin polymerization and cancer cell migration. However, the molecular mechanisms underlying pancreatic cancer cell migration are still largely obscure. Here, we addressed the role of ELMO2 in chemotaxis and metastasis of pancreatic cancer cells. Methods Pancreatic cancer cell lines PANC-1 and AsPC-1 and siRNA-mediated knockdown of ELMO2 were used to determine the effects of ELMO2 on cancer cell chemotaxis, invasion, migration. Co-immunoprecipitation assays were carried out to identify interacting partners of ELMO2. Results ELMO2 knockdown inhibited pancreatic cancer cell chemotaxis, migration, invasion, and F-actin polymerization. Co-immunoprecipitation assays revealed that ELMO2 interacted with Gαi2 and that CXCL12 triggered Gα i2-dependent membrane translocation of ELMO2. Thus, ELMO2 is a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Yecheng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hongyan Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Intergrated analysis of ELMO1, serves as a link between tumour mutation burden and epithelial-mesenchymal transition in hepatocellular carcinoma. EBioMedicine 2019; 46:105-118. [PMID: 31324602 PMCID: PMC6710851 DOI: 10.1016/j.ebiom.2019.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/23/2019] [Accepted: 07/01/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is critical for cancer cell metastasis. Recently, EMT was reported to be associated with the inflammatory tumour microenvironment and, therefore, might be a predictive biomarker for immune checkpoint blockade agents. However, the underlying mechanism is still unclear. METHODS Patient survival data for our HCC cohort, TCGA and GEO datasets were determined by Kaplan-Meier analysis. The functional roles of ELMO1 in HCC were demonstrated by a series of in vitro and in vivo experiments. Gene microarray analysis was used to demonstrate potential mechanisms of ELMO1. Data retrieved from the TCGA datasets were used to determine the relationships of ELMO1, EMT and TMB. FINDINGS Here, we report an indispensable role for ELMO1 in linking EMT with tumour mutation burden (TMB), which is a promising biomarker for the immune checkpoint blockade agent response. Upregulated ELMO1 expression is associated with a poor prognosis in hepatocellular carcinoma (HCC), as well as increased cell growth, invasion, migration, angiogenesis and EMT in vitro and in vivo. Mechanistically, we provide evidence that ELMO1 regulates SOX10 expression and induces EMT through PI3K/Akt signalling. Moreover, ELMO1 is negatively associated with TMB, indicating a negative relationship between EMT and TMB. INTERPRETATION ELMO1 serves as a link between EMT and TMB, providing a mechanistic basis for the further development of ELMO1 as a therapeutic target against HCC and potentially a promising biomarker of the immune checkpoint blockade agent response. FUND: National Natural Science Foundation of China; Natural Science Foundation of Guangdong Province; Young Teacher Training Program of Sun Yat-sen University; Science and Technology Plan of Guangdong Province; Special Support Program of Guangdong Province, Science and Technology Innovation Youth Talent Support Program; the Pearl River Science and Technology New Talent of Guangzhou City; Medical Scientific Research Foundation of Guangdong Province.
Collapse
|
9
|
Yi J, Fan Y, Zhang L, Wang H, Mu T, Xie H, Gao H, Liu M, Li S, Tang H. MiR-HCC2 Up-regulates BAMBI and ELMO1 Expression to Facilitate the Proliferation and EMT of Hepatocellular Carcinoma Cells. J Cancer 2019; 10:3407-3419. [PMID: 31293644 PMCID: PMC6603416 DOI: 10.7150/jca.30858] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of gene expression regulators that participate in the occurrence and development of hepatocellular carcinoma (HCC), although the underlying mechanism by which they function in HCC has not been fully elucidated. Here, small RNA deep sequencing was used to identify aberrantly expressed miRNAs in HCC tissues, and a novel miRNA named miR-HCC2 was identified. RT-qPCR analysis demonstrated that miR-HCC2 displayed higher expression in HCC tissues than in adjacent non-tumor tissues. We documented that miR-HCC2 facilitated the growth, migration and invasion of HCC cells by accelerating cell cycle progression, incressing the expression of epithelial-to-mesenchymal transition (EMT)-associated marker vimentin but decreasing the expression of E-cadherin. MiR-HCC2 directly targeted the 3′ UTR of BAMBI and ELMO1 and up-regulated their expression. Both BAMBI and ELMO1 had the same patterns of expression with miR-HCC2 in HCC tissues. Additionally, blocking BAMBI or ELMO1 counteracted the phenotypic alterations elicited by miR-HCC2. Collectively, our investigation identified miR-HCC2 as a new positive modulator of HCC aggressiveness that may serve as a potential biomarker for the development of diagnostic and therapeutic approaches for HCC.
Collapse
Affiliation(s)
- Jianying Yi
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yajie Fan
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Le Zhang
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hong Wang
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Ting Mu
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hong Xie
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Huijie Gao
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Min Liu
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Shengping Li
- State Key Laboratory of Oncology in Southern China, Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, 651 Dong-Feng Road East, Guangzhou 510060, China
| | - Hua Tang
- Tianjin Life Science Research Center and Department of Pathogen Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
10
|
Pan C, Zhang Y, Meng Q, Dai G, Jiang Z, Bao H. Down Regulation of the Expression of ELMO3 by COX2 Inhibitor Suppresses Tumor Growth and Metastasis in Non-Small-Cell Lung Cancer. Front Oncol 2019; 9:363. [PMID: 31134158 PMCID: PMC6515945 DOI: 10.3389/fonc.2019.00363] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/18/2019] [Indexed: 01/04/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common malignancies. Studies have shown that engulfment and cell motility 3 (ELMO3) is highly expressed in NSCLC and can be used as a novel biomarker, but its underlying mechanism remains to be explored. The aim of this study was to investigate the mechanism by which ELMO3 may be down-regulated by COX-2 inhibitors to inhibit NSCLC. NSCLC tissue and adjacent normal lung tissue from 24 patients were used to detect the mRNA and protein expression of ELMO3, COX-2, and other related proteins by Western blot, RT-PCR, and Immunohistochemical analysis. Lewis Lung carcinoma (LLC) cells were used to investigate the effects and the mechanism of siELMO3 and COX-2 inhibitor. C57BL/6 mice inoculated with LLC cells by subcutaneous (s.c.) injection were used to detect the in vivo effects of cox-2 inhibitor. The expression of ELMO3 and cyclooxygenase-2 (COX-2) in human NSCLC tissues was significantly increased compared with that in the adjacent normal tissues. ELMO3 exhibited a positive correlation with COX-2 expression. Moreover, knockdown of ELMO3 suppressed the epithelial-mesenchymal transition (EMT), adhesion, and metastasis of Lewis lung carcinoma (LLC) cells. Importantly, Parecoxib, a selective inhibitor of COX-2, significantly reduced the expression of ELMO3 and EMT in LLC cells and LLC-bearing mice. Furthermore, it could inhibit the growth, adhesion and metastasis of LLC cells in vitro. Our results demonstrate that down regulation of ELMO3 suppressed growth and metastasis of lung cancer by inhibiting EMT. Parecoxib could reduce ELMO3 expression and suppress growth and metastasis of lung cancer, which might be a useful chemotherapeutic agent for inhibiting metastasis and recurrence of NSCLC.
Collapse
Affiliation(s)
- Cailong Pan
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yong Zhang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qinghai Meng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guoliang Dai
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhitao Jiang
- Department of Pharmacy Office, Zhangjiagang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Hongguang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Auerbach SS, Xu M, Merrick BA, Hoenerhoff MJ, Phadke D, Taxman DJ, Shah R, Hong HHL, Ton TV, Kovi RC, Sills RC, Pandiri AR. Exome Sequencing of Fresh-frozen or Formalin-fixed Paraffin-embedded B6C3F1/N Mouse Hepatocellular Carcinomas Arising Either Spontaneously or due to Chronic Chemical Exposure. Toxicol Pathol 2018; 46:706-718. [PMID: 30045675 DOI: 10.1177/0192623318789398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide; however, the mutational properties of HCC-associated carcinogens remain largely uncharacterized. We hypothesized that mechanisms underlying chemical-induced HCC can be characterized by evaluating the mutational spectra of these tumors. To test this hypothesis, we performed exome sequencing of B6C3F1/N HCCs that arose either spontaneously in vehicle controls ( n = 3) or due to chronic exposure to gingko biloba extract (GBE; n = 4) or methyleugenol (MEG; n = 3). Most archived tumor samples are available as formalin-fixed paraffin-embedded (FFPE) blocks, rather than fresh-frozen (FF) samples; hence, exome sequencing from paired FF and FFPE samples was compared. FF and FFPE samples showed 63% to 70% mutation concordance. Multiple known (e.g., Ctnnb1T41A, BrafV637E) and novel (e.g., Erbb4C559S, Card10A700V, and Klf11P358L) mutations in cancer-related genes were identified. The overall mutational burden was greater for MEG than for GBE or spontaneous HCC samples. To characterize the mutagenic mechanisms, we analyzed the mutational spectra in the HCCs according to their trinucleotide motifs. The MEG tumors clustered closest to Catalogue of Somatic Mutations in Cancer signatures 4 and 24, which are, respectively, associated with benzo(a)pyrene- and aflatoxin-induced HCCs in humans. These results establish a novel approach for classifying liver carcinogens and understanding the mechanisms of hepatocellular carcinogenesis.
Collapse
Affiliation(s)
- Scott S Auerbach
- 1 Division of the National Toxicology Program, NIEHS, Research Triangle Park, North Carolina, USA
| | - Miaofei Xu
- 1 Division of the National Toxicology Program, NIEHS, Research Triangle Park, North Carolina, USA
| | - B Alex Merrick
- 1 Division of the National Toxicology Program, NIEHS, Research Triangle Park, North Carolina, USA
| | - Mark J Hoenerhoff
- 1 Division of the National Toxicology Program, NIEHS, Research Triangle Park, North Carolina, USA.,2 Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Dhiral Phadke
- 3 Sciome LLC, Research Triangle Park, North Carolina, USA
| | - Debra J Taxman
- 3 Sciome LLC, Research Triangle Park, North Carolina, USA
| | - Ruchir Shah
- 3 Sciome LLC, Research Triangle Park, North Carolina, USA
| | - Hue-Hua L Hong
- 1 Division of the National Toxicology Program, NIEHS, Research Triangle Park, North Carolina, USA
| | - Thai-Vu Ton
- 1 Division of the National Toxicology Program, NIEHS, Research Triangle Park, North Carolina, USA
| | - Ramesh C Kovi
- 1 Division of the National Toxicology Program, NIEHS, Research Triangle Park, North Carolina, USA.,4 Experimental Pathology Laboratories Inc., Research Triangle Park, North Carolina, USA
| | - Robert C Sills
- 1 Division of the National Toxicology Program, NIEHS, Research Triangle Park, North Carolina, USA
| | - Arun R Pandiri
- 1 Division of the National Toxicology Program, NIEHS, Research Triangle Park, North Carolina, USA
| |
Collapse
|
12
|
McCauley HA, Chevrier V, Birnbaum D, Guasch G. De-repression of the RAC activator ELMO1 in cancer stem cells drives progression of TGFβ-deficient squamous cell carcinoma from transition zones. eLife 2017; 6:e22914. [PMID: 28219480 PMCID: PMC5319840 DOI: 10.7554/elife.22914] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/27/2017] [Indexed: 01/18/2023] Open
Abstract
Squamous cell carcinomas occurring at transition zones are highly malignant tumors with poor prognosis. The identity of the cell population and the signaling pathways involved in the progression of transition zone squamous cell carcinoma are poorly understood, hence representing limited options for targeted therapies. Here, we identify a highly tumorigenic cancer stem cell population in a mouse model of transitional epithelial carcinoma and uncover a novel mechanism by which loss of TGFβ receptor II (Tgfbr2) mediates invasion and metastasis through de-repression of ELMO1, a RAC-activating guanine exchange factor, specifically in cancer stem cells of transition zone tumors. We identify ELMO1 as a novel target of TGFβ signaling and show that restoration of Tgfbr2 results in a complete block of ELMO1 in vivo. Knocking down Elmo1 impairs metastasis of carcinoma cells to the lung, thereby providing insights into the mechanisms of progression of Tgfbr2-deficient invasive transition zone squamous cell carcinoma.
Collapse
Affiliation(s)
- Heather A McCauley
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, United States
| | - Véronique Chevrier
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, F-13009, CNRS, UMR7258, F-13009, Institut Paoli-Calmettes, F-13009, Aix-Marseille University, UM 105, F-13284, Marseille, France
| | - Daniel Birnbaum
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, F-13009, CNRS, UMR7258, F-13009, Institut Paoli-Calmettes, F-13009, Aix-Marseille University, UM 105, F-13284, Marseille, France
| | - Géraldine Guasch
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, United States
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, F-13009, CNRS, UMR7258, F-13009, Institut Paoli-Calmettes, F-13009, Aix-Marseille University, UM 105, F-13284, Marseille, France
| |
Collapse
|
13
|
Knockdown of ELMO3 Suppresses Growth, Invasion and Metastasis of Colorectal Cancer. Int J Mol Sci 2016; 17:ijms17122119. [PMID: 27999268 PMCID: PMC5187919 DOI: 10.3390/ijms17122119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 01/08/2023] Open
Abstract
The engulfment and cell motility (ELMOs) family of proteins plays a crucial role in tumor cell migration and invasion. However, the function of ELMO3 is poorly defined. To elucidate its role in the development and progression of colorectal cancer (CRC), we examined the expression of ELMO3 in 45 cases of paired CRC tumor tissues and adjacent normal tissues. Furthermore, we assessed the effect of the knockdown of ELMO3 on cell proliferation, cell cycle, migration, invasion and F-actin polymerization in HCT116 cells. The result shows that the expression of ELMO3 in CRC tissues was significantly increased in comparison to the adjacent normal colorectal tissues. Moreover, this overexpression was associated with tumor size (p = 0.007), tumor differentiation (p = 0.001), depth of invasion (p = 0.009), lymph node metastasis (p = 0.003), distant metastasis (p = 0.013) and tumor, node, metastasis (TNM)-based classification (p = 0.000). In in vitro experiments, the silencing of ELMO3 inhibited cell proliferation, invasion, metastasis, and F-actin polymerization, and induced Gap 1 (G1) phase cell cycle arrest. Our study demonstrates that ELMO3 is involved in the processes of growth, invasion and metastasis of CRC, and could be used a potential molecular diagnostic tool or therapy target of CRC.
Collapse
|