1
|
Deng Y, Huang X, Yang Y, Zhang Y, Zeng B, Bao Y, Cao L, Wang X, Ma L, Wang J. MFAP2 upregulation promotes ESCC metastasis via FAK-AKT signaling pathway. FASEB J 2024; 38:e70266. [PMID: 39698924 DOI: 10.1096/fj.202402411r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
Metastasis is the leading cause of mortality from esophageal squamous cell carcinoma (ESCC). By the time of diagnosis, most ESCC tumors have already invaded the lymph nodes or distant organs; however, it has been challenging to identify and confirm genes with a crucial role in ESCC metastasis. The microfibrillar-associated protein 2 (MFAP2) is upregulated in human ESCC, and its expression level was positively associated with poor overall and disease-free survival. Consistently, upregulation of MFAP2 promoted the metastasis and invasion of ESCC cells in vitro and in vivo. Conversely, these processes were reduced by MFAP2 knockdown. Mechanistically, MFAP2 was shown to bind to the FERM domain of focal adhesion kinase (FAK) and to alleviate FAK intramolecular inhibition, resulting in the enhanced binding affinity between FAK and integrin beta 4 (ITGB4) and activation of the FAK-AKT signaling pathway. Treatment of ESCC cells with the FAK inhibitor PND-1186 reduced MFAP2, induced the activation of the FAK-AKT pathway in vitro, and suppressed lung metastasis in a mouse model of ESCC. These findings support a major role for MFAP2 in promoting ESCC metastasis, in part via the activation of FAK-AKT signaling, and highlight the potential of MFAP2 as a promising therapeutic target for ESCC.
Collapse
Affiliation(s)
- Yiran Deng
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xu Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yiran Yang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yingcong Zhang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Bingjie Zeng
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yunxia Bao
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Leiqun Cao
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xianzhao Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Lifang Ma
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jiayi Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
2
|
Navasardyan I, Zaravinos A, Bonavida B. Therapeutic Implications of Targeting YY1 in Glioblastoma. Cancers (Basel) 2024; 16:2074. [PMID: 38893192 PMCID: PMC11171050 DOI: 10.3390/cancers16112074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
The transcription factor Yin Yang 1 (YY1) plays a pivotal role in the pathogenesis of glioblastoma multiforme (GBM), an aggressive form of brain tumor. This review systematically explores the diverse roles of YY1 overexpression and activities in GBM, including its impact on the tumor microenvironment (TME) and immune evasion mechanisms. Due to the poor response of GBM to current therapies, various findings of YY1-associated pathways in the literature provide valuable insights into novel potential targeted therapeutic strategies. Moreover, YY1 acts as a significant regulator of immune checkpoint molecules and, thus, is a candidate therapeutic target in combination with immune checkpoint inhibitors. Different therapeutic implications targeting YY1 in GBM and its inherent associated challenges encompass the use of nanoparticles, YY1 inhibitors, targeted gene therapy, and exosome-based delivery systems. Despite the inherent complexities of such methods, the successful targeting of YY1 emerges as a promising avenue for reshaping GBM treatment strategies, presenting opportunities for innovative therapeutic approaches and enhanced patient outcomes.
Collapse
Affiliation(s)
- Inesa Navasardyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Microbiology, Immunology & Molecular Genetics, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Apostolos Zaravinos
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), 1516 Nicosia, Cyprus;
- Department of Life Sciences, School of Sciences, European University Cyprus, 1516 Nicosia, Cyprus
| | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Hathi D, Chanswangphuwana C, Cho N, Fontana F, Maji D, Ritchey J, O'Neal J, Ghai A, Duncan K, Akers WJ, Fiala M, Vij R, DiPersio JF, Rettig M, Shokeen M. Ablation of VLA4 in multiple myeloma cells redirects tumor spread and prolongs survival. Sci Rep 2022; 12:30. [PMID: 34996933 PMCID: PMC8741970 DOI: 10.1038/s41598-021-03748-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a cancer of bone marrow (BM) plasma cells, which is increasingly treatable but still incurable. In 90% of MM patients, severe osteolysis results from pathological interactions between MM cells and the bone microenvironment. Delineating specific molecules and pathways for their role in cancer supportive interactions in the BM is vital for developing new therapies. Very Late Antigen 4 (VLA4, integrin α4β1) is a key player in cell–cell adhesion and signaling between MM and BM cells. We evaluated a VLA4 selective near infrared fluorescent probe, LLP2A-Cy5, for in vitro and in vivo optical imaging of VLA4. Furthermore, two VLA4-null murine 5TGM1 MM cell (KO) clones were generated by CRISPR/Cas9 knockout of the Itga4 (α4) subunit, which induced significant alterations in the transcriptome. In contrast to the VLA4+ 5TGM1 parental cells, C57Bl/KaLwRij immunocompetent syngeneic mice inoculated with the VLA4-null clones showed prolonged survival, reduced medullary disease, and increased extramedullary disease burden. The KO tumor foci showed significantly reduced uptake of LLP2A-Cy5, confirming in vivo specificity of this imaging agent. This work provides new insights into the pathogenic role of VLA4 in MM, and evaluates an optical tool to measure its expression in preclinical models.
Collapse
Affiliation(s)
- Deep Hathi
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Chantiya Chanswangphuwana
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Medicine, Division of Hematology, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Nicholas Cho
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Francesca Fontana
- Department of Medicine, Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dolonchampa Maji
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie Ritchey
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Julie O'Neal
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anchal Ghai
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kathleen Duncan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Walter J Akers
- Center for In Vivo Imaging and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mark Fiala
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ravi Vij
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - John F DiPersio
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael Rettig
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Monica Shokeen
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA. .,Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
4
|
Gómez de Segura I, Ahechu P, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Becerril S, Unamuno X, Mentxaka A, Baixauli J, Valentí V, Moncada R, Silva C, Frühbeck G, Catalán V. Decreased Levels of Microfibril-Associated Glycoprotein (MAGP)-1 in Patients with Colon Cancer and Obesity Are Associated with Changes in Extracellular Matrix Remodelling. Int J Mol Sci 2021; 22:ijms22168485. [PMID: 34445187 PMCID: PMC8395192 DOI: 10.3390/ijms22168485] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE The protein microfibril-associated glycoprotein (MAGP)-1 constitutes a crucial extracellular matrix protein. We aimed to determine its impact on visceral adipose tissue (VAT) remodelling during obesity-associated colon cancer (CC). METHODS Samples obtained from 79 subjects (29 normoponderal (NP) (17 with CC) and 50 patients with obesity (OB) (19 with CC)) were used in the study. Circulating concentrations of MAGP-1 and its gene expression levels (MFAP2) in VAT were analysed. The impact of inflammation-related factors and adipocyte-conditioned media (ACM) on MFAP2 mRNA levels in colon adenocarcinoma HT-29 cells were further analysed. The effects of MAGP-1 in the expression of genes involved in the extracellular matrix (ECM) remodelling and tumorigenesis in HT-29 cells was also explored. RESULTS Obesity (p < 0.01) and CC (p < 0.001) significantly decreased MFAP2 gene expression levels in VAT whereas an opposite trend in TGFB1 mRNA levels was observed. Increased mRNA levels of MFAP2 after the stimulation of HT-29 cells with lipopolysaccharide (LPS) (p < 0.01) and interleukin (IL)-4 (p < 0.01) together with a downregulation (p < 0.05) after hypoxia mimicked by CoCl2 treatment was observed. MAGP-1 treatment significantly enhanced the mRNA levels of the ECM-remodelling genes collagen type 6 α3 chain (COL6A3) (p < 0.05), decorin (DCN) (p < 0.01), osteopontin (SPP1) (p < 0.05) and TGFB1 (p < 0.05). Furthermore, MAGP-1 significantly reduced (p < 0.05) the gene expression levels of prostaglandin-endoperoxide synthase 2 (COX2/PTGS2), a key gene controlling cell proliferation, growth and adhesion in CC. Interestingly, a significant decrease (p < 0.01) in the mRNA levels of MFAP2 in HT-29 cells preincubated with ACM from volunteers with obesity compared with control media was observed. Conclusion: The decreased levels of MAGP-1 in patients with obesity and CC together with its capacity to modulate key genes involved in ECM remodelling and tumorigenesis suggest MAGP-1 as a link between AT excess and obesity-associated CC development.
Collapse
Affiliation(s)
- Iranzu Gómez de Segura
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
| | - Patricia Ahechu
- Department of Surgery, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (P.A.); (J.B.); (V.V.)
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
| | - Xabier Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
| | - Amaia Mentxaka
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
| | - Jorge Baixauli
- Department of Surgery, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (P.A.); (J.B.); (V.V.)
| | - Víctor Valentí
- Department of Surgery, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (P.A.); (J.B.); (V.V.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
| | - Rafael Moncada
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Department of Anesthesia, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Camilo Silva
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Correspondence: (G.F.); (V.C.); Tel.: +34-9-4825-5400 (ext. 4484) (G.F.); +34-9-4825-5400 (ext. 5133) (V.C.)
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.G.d.S.); (J.G.-A.); (A.R.); (B.R.); (S.B.); (X.U.); (A.M.)
- CIBEROBN, Instituto de Salud Carlos III, 31008 Pamplona, Spain; (R.M.); (C.S.)
- Obesity and Adipobiology Group, IdiSNA, 31008 Pamplona, Spain
- Correspondence: (G.F.); (V.C.); Tel.: +34-9-4825-5400 (ext. 4484) (G.F.); +34-9-4825-5400 (ext. 5133) (V.C.)
| |
Collapse
|
5
|
Shan Z, Wang W, Tong Y, Zhang J. Genome-Scale Analysis Identified NID2, SPARC, and MFAP2 as Prognosis Markers of Overall Survival in Gastric Cancer. Med Sci Monit 2021; 27:e929558. [PMID: 33758160 PMCID: PMC8006563 DOI: 10.12659/msm.929558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gastric cancer is the most common gastrointestinal tumor, and the rates of recurrence and metastasis are high. Research results on molecular biomarkers used for prognosis of gastric cancer remain inconclusive. This study aimed to explore the gene expression module of gastric cancer and to determine potential prognostic biomarkers. MATERIAL AND METHODS Three microarray datasets (GSE13911, GSE79973, and GSE29272) from Gene Expression Omnibus (GEO), including 206 pairs of gastric tumors and adjacent normal samples, were used for analysis of differentially expressed genes (DEGs). The 3 microarray datasets yielded 144 genes associated with the progression and prognosis of gastric cancer. After this, a risk score model was developed for result validation using an independent dataset from The Cancer Genome Atlas. RESULTS The validation of the independent dataset showed significantly increased NID2, SPARC, and MFAP2 expression in gastric tumor tissues, which were associated with poor outcomes in gastric cancer patients. Moreover, the high risk score obtained was associated with poor overall survival (HR: 1.787; 1.069-2.986; P=0.027). Subgroup analyses revealed that these significant prognostic values were detected in patients aged <65.0 years, tumors in the antrum/distal colon, grade 3 tumors, or TNM-M0 stages of cancer. CONCLUSIONS The findings of this study show that NID2, SPARC, and MFAP2 are upregulated in gastric tumor tissues and are significantly associated with poor overall survival. Therefore, the predictive values of the risk score model employed for the prognosis of gastric cancer could be improved by using these 3 upregulated DEGs.
Collapse
Affiliation(s)
- Zexing Shan
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Wentao Wang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Yilin Tong
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Jianjun Zhang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
6
|
Zhu X, Cheng Y, Wu F, Sun H, Zheng W, Jiang W, Shi J, Ma S, Cao H. MFAP2 Promotes the Proliferation of Cancer Cells and Is Associated With a Poor Prognosis in Hepatocellular Carcinoma. Technol Cancer Res Treat 2020; 19:1533033820977524. [PMID: 33280519 PMCID: PMC7724263 DOI: 10.1177/1533033820977524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Backgrounds: Microfibril-associated protein 2 (MFAP2) is an extracellular matrix protein
that regulates the function of microfibrils by interacting with fibrillin.
MFAP2 has been reported to play an important role in metabolic diseases and
has been shown to be significantly overexpressed in head and neck squamous
cell carcinoma and Hepatocellular carcinoma (HCC). However, the molecular
function and prognostic value of MFAP2 have never been reported in HCC or
other tumors. Methods: In the present study, expression characteristics of MFAP2 in HCC, its
influence on the development of HCC, as well as its function and potential
mechanism in HCC were verified by Quantitative reverse
transcription-polymerase chain reaction, bioinformatics data mining and in
vitro cell experiments. Results: MFAP2 was prominently high-expressed in HCC and associated with cancer
stages. HCC patients with higher MFAP2 expression displayed lower overall
survival (OS) and disease-specific survival(DSS), while there was no
significant difference in recurrence-free survival (RFS). In vitro
experiments showed that downregulation of MFAP2 inhibited proliferation,
migration level of HCC cells. Transcription factors, DNA methyltransferases,
immune factors may interact with MFAP2 mRNA to promote tumor progression in
HCC. Conclusion: These findings suggest that MFAP2 may play a key role in the development of
HCC. Therefore, MFAP2 may be a valuable prognostic marker and an effective
anticancer target in HCC.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of General Surgery, The East District of Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China.,Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Ye Cheng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Fan Wu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Haoyao Sun
- Department of Radio-Oncology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China
| | - Wubin Zheng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Wei Jiang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Junfeng Shi
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Shijie Ma
- Department of Gastroenterology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Nanjing, People's Republic of China
| | - Hongyong Cao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
7
|
Wu M, Ding Y, Jiang X, Chen Y, Wu N, Li L, Wang H, Huang Y, Xu N, Teng L. Overexpressed MAGP1 Is Associated With a Poor Prognosis and Promotes Cell Migration and Invasion in Gastric Cancer. Front Oncol 2020; 9:1544. [PMID: 32010630 PMCID: PMC6978879 DOI: 10.3389/fonc.2019.01544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/20/2019] [Indexed: 01/14/2023] Open
Abstract
Gastric cancer (GC) is a frequently occurring malignancy with high mortality rates. However, the underlying mechanism of GC progression is not very clear. The aim of this study is to reveal the inherent molecular mechanism and develop potential therapeutic targets for advanced GC. The microfibril-associated glycoprotein 1 (MAGP1), identified as a potential oncogene, was found upregulated in GC tissues and high MAGP1 expression was associated with aggressive clinicopathological features. Furthermore, the multivariate Cox regression analysis showed that high MAGP1 expression was an independent predictor of poor prognosis (HR = 2.37, 1.07-5.24; P = 0.033). Mechanistically, MAGP1 promoted the migration and invasiveness of GC cells. In addition, the genes co-expressed with MAGP1 were primarily enriched in focal adhesion and PI3K-Akt pathways. MAGP1 overexpression enhanced the phosphorylation of FAK, AKT, and mTOR, whereas its knockdown also inactivated these factors. Furthermore, the AKT inhibitor suppressed the phosphorylation of AKT, FAK, and mTOR in recMAGP1-treated AGS cells, as well as their migration and invasion capacities. Finally, correlation analysis indicated that MAGP1 is involved in AKT signaling in GC, and is clinically relevant. Taken together, MAGP1 is a promising prognostic marker and potential therapeutic target for advanced GC.
Collapse
Affiliation(s)
- Mengjie Wu
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yongfeng Ding
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoxia Jiang
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanyan Chen
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nan Wu
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Linrong Li
- Department of Otorhinolaryngology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haiyong Wang
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yingying Huang
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nong Xu
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lisong Teng
- Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Sun T, Wang D, Ping Y, Sang Y, Dai Y, Wang Y, Liu Z, Duan X, Tao Z, Liu W. Integrated profiling identifies SLC5A6 and MFAP2 as novel diagnostic and prognostic biomarkers in gastric cancer patients. Int J Oncol 2019; 56:460-469. [PMID: 31894266 PMCID: PMC6959404 DOI: 10.3892/ijo.2019.4944] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is one of the leading causes of malignancy‑associated mortality worldwide. However, the underlying molecular mechanisms of GC are unclear and the prognosis of GC is poor. Therefore, it is important and urgent to explore the underlying mechanisms and screen for novel diagnostic and prognostic biomarkers, as well as therapeutic targets. In the current study, scale‑free gene co‑expression networks were constructed using weighted gene co‑expression network analysis, the potential associations between gene sets and clinical features were investigated, and the hub genes were identified. The gene expression profiles of GSE38749 were downloaded from the Gene Expression Omnibus database. RNA‑seq and clinical data for GC from The Cancer Genome Atlas were utilized for verification. Furthermore, the expression of candidate biomarkers in gastric tissues was investigated. Survival analysis was performed using Kaplan‑Meier and log‑rank test. The predictive role of candidate biomarkers in GC was evaluated using a receiver operator characteristic (ROC) curve. Gene Ontology, gene set enrichment analysis and gene set variation analysis methods were used to interpret the function of candidate biomarkers in GC. A total of 29 modules were identified via the average linkage hierarchical clustering. A significant module consisting of 48 genes associated with clinical traits was found; three genes with high connectivity in the clinical significant module were identified as hub genes. Among them, SLC5A6 and microfibril‑associated protein 2 (MFAP2) were negatively associated with the overall survival, and their expression was elevated in GC compared with non‑tumor tissues. Additionally, ROC curves indicated that SLC5A6 and MFAP2 showed a good diagnostic power in discriminating cancerous from normal tissues. SLC5A6 and MFAP2 were identified as novel diagnostic and prognostic biomarkers in GC patients; both of these genes were first reported here in connection with GC and deserved further research.
Collapse
Affiliation(s)
- Tao Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Danhua Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Ying Ping
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yiwen Sang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yibei Dai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yiyun Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhenping Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiuzhi Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
9
|
Wei J, Sheng Y, Li J, Gao X, Ren N, Dong Q, Qin L. Genome-Wide Association Study Identifies a Genetic Prediction Model for Postoperative Survival in Patients with Hepatocellular Carcinoma. Med Sci Monit 2019; 25:2452-2478. [PMID: 30945699 PMCID: PMC6461006 DOI: 10.12659/msm.915511] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/19/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND As an important aspect of tumor heterogeneity, genetic variation may influence susceptibility and prognosis in different types of cancer. By exploring the prognostic value of genetic variation, this study aimed to establish a model for predicting postoperative survival and assessing the impact of variation on clinical outcomes in patients with hepatocellular carcinoma (HCC). MATERIAL AND METHODS A genome-wide association study of 367 patients with HCC was conducted to identify single nucleotide polymorphisms (SNPs) associated with prognosis. Identified predictors were further evaluated in 758 patients. Two prognostic models were established using Cox proportional hazards regression and Nomogram strategy, and validated in another 316 patients. The effect of the SNP rs2431 was analyzed in detail. RESULTS A prognostic model including 5 SNPs (rs10893585, rs2431, rs34675408, rs6078460, and rs6766361) was established and exhibited high predictive accuracy for HCC prognosis. The panel combined with tumor node metastasis (TNM) stage resulted in a significantly higher c-index (0.723) than the individual c-index values. Stratified by the Nomogram prediction model, the median overall survival for the low-risk and high-risk groups were 100.1 versus 30.8 months (P<0.001) in the training set and 82.2 versus 22.5 months (P<0.001) in the validation set. A closer examination of rs2431 revealed that it may regulate the expression of FNDC3B by disrupting a microRNA-binding site. CONCLUSIONS This study established prediction models based on genetic factors alone or in combination with TNM stage for postoperative survival in patients with HCC, and identified FNDC3B as a potential therapeutic target for combating HCC metastasis.
Collapse
Affiliation(s)
- Jinwang Wei
- Department of General Surgery, Huashan Hospital; Cancer Metastasis Institute; Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Yuanyuan Sheng
- Department of General Surgery, Huashan Hospital; Cancer Metastasis Institute; Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Jianhua Li
- Department of General Surgery, Huashan Hospital; Cancer Metastasis Institute; Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Xiaomei Gao
- Department of General Surgery, Huashan Hospital; Cancer Metastasis Institute; Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Ning Ren
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Qiongzhu Dong
- Department of General Surgery, Huashan Hospital; Cancer Metastasis Institute; Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| | - Lunxiu Qin
- Department of General Surgery, Huashan Hospital; Cancer Metastasis Institute; Institutes of Biomedical Sciences, Fudan University, Shanghai, P.R. China
| |
Collapse
|
10
|
Zaravinos A, Bonavida B, Chatzaki E, Baritaki S. RKIP: A Key Regulator in Tumor Metastasis Initiation and Resistance to Apoptosis: Therapeutic Targeting and Impact. Cancers (Basel) 2018; 10:287. [PMID: 30149591 PMCID: PMC6162400 DOI: 10.3390/cancers10090287] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/12/2018] [Accepted: 08/18/2018] [Indexed: 02/07/2023] Open
Abstract
RAF-kinase inhibitor protein (RKIP) is a well-established tumor suppressor that is frequently downregulated in a plethora of solid and hematological malignancies. RKIP exerts antimetastatic and pro-apoptotic properties in cancer cells, via modulation of signaling pathways and gene products involved in tumor survival and spread. Here we review the contribution of RKIP in the regulation of early metastatic steps such as epithelial⁻mesenchymal transition (EMT), migration, and invasion, as well as in tumor sensitivity to conventional therapeutics and immuno-mediated cytotoxicity. We further provide updated justification for targeting RKIP as a strategy to overcome tumor chemo/immuno-resistance and suppress metastasis, through the use of agents able to modulate RKIP expression in cancer cells.
Collapse
Affiliation(s)
- Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 2404, Cyprus.
- Centre for Risk and Decision Sciences (CERIDES), Nicosia 2404, Cyprus.
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Ekaterini Chatzaki
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece.
| | - Stavroula Baritaki
- Division of Surgical Oncology, School of Medicine, University of Crete, Heraklion, Crete 71500, Greece.
| |
Collapse
|
11
|
Wang JK, Wang WJ, Cai HY, Du BB, Mai P, Zhang LJ, Ma W, Hu YG, Feng SF, Miao GY. MFAP2 promotes epithelial-mesenchymal transition in gastric cancer cells by activating TGF-β/SMAD2/3 signaling pathway. Onco Targets Ther 2018; 11:4001-4017. [PMID: 30034240 PMCID: PMC6047603 DOI: 10.2147/ott.s160831] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Introduction Microfibril-associated protein 2 (MFAP2) is an extracellular matrix protein that interacts with fibrillin to modulate the function of microfibrils. MFAP2 has been reported to play a significant role in obesity, diabetes, and osteopenia, and has been shown to be upregulated in head and neck squamous cell carcinoma. However, the molecular function and prognostic value of MFAP2 have never been reported in gastric cancer (GC) or any other tumors. Methods The current study investigated the expression patterns, prognostic significance, functional role, and possible mechanisms of MFAP2 in GC. Results We demonstrated that MFAP2 was overexpressed in GC tissues, and its overexpression was significantly correlated with poor overall and disease-free survival in patients with GC. Moreover, we found that MFAP2 promoted the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) phenotype in GC cells. MFAP2 might modulate EMT of GC cells by activating the TGF-β/SMAD2/3 signaling pathway. Conclusion These findings provide novel evidence that MFAP2 plays a crucial role in the progression of GC. Therefore, MFAP2 may be a promising prognostic marker and a potent anticancer agent.
Collapse
Affiliation(s)
- Jian-Kai Wang
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Wen-Juan Wang
- Physical Examination Center, The Third People's Hospital of Gansu, Lanzhou, Gansu 730000, China
| | - Hong-Yi Cai
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Bin-Bin Du
- Department of Anorectal Surgery, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Ping Mai
- Department of Gastroenterology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Li-Juan Zhang
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Wen Ma
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Yong-Guo Hu
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Shi-Fang Feng
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| | - Guo-Ying Miao
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China,
| |
Collapse
|
12
|
Arhoma A, Chantry AD, Haywood-Small SL, Cross NA. SAHA-induced TRAIL-sensitisation of Multiple Myeloma cells is enhanced in 3D cell culture. Exp Cell Res 2017; 360:226-235. [PMID: 28890292 DOI: 10.1016/j.yexcr.2017.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Multiple Myeloma (MM) is currently incurable despite many novel therapies. Tumour Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) is a potential anti-tumour agent although effects as a single agent are limited. In this study, we investigated whether the Histone Deacetylase (HDAC) inhibitor SAHA can enhance TRAIL-induced apoptosis and target TRAIL resistance in both suspension culture, and 3D cell culture as a model of disseminated MM lesions that form in bone. METHODS The effects of SAHA and/or TRAIL in 6 Multiple Myeloma cell lines were assessed in both suspension cultures and in an Alginate-based 3D cell culture model. The effect of SAHA and/or TRAIL was assessed on apoptosis by assessment of nuclear morphology using Hoechst 33342/Propidium Iodide staining. Viable cell number was assessed by CellTiter-Glo luminescence assay, Caspase-8 and -9 activities were measured by Caspase-Glo™ assay kit. TRAIL-resistant cells were generated by culture of RPMI 8226 and NCI-H929 by acute exposure to TRAIL followed by selection of TRAIL-resistant cells. RESULTS TRAIL significantly induced apoptosis in a dose-dependent manner in OPM-2, RPMI 8226, NCI-H929, U266, JJN-3 MM cell lines and ADC-1 plasma cell leukaemia cells. SAHA amplified TRAIL responses in all lines except OPM-2, and enhanced TRAIL responses were both via Caspase-8 and -9. SAHA treatment induced growth inhibition that further increased in the combination treatment with TRAIL in MM cells. The co-treatment of TRAIL and SAHA reduced viable cell numbers all cell lines. TRAIL responses were further potentiated by SAHA in 3D cell culture in NCI-H929, RPMI 8226 and U266 at lower TRAIL + SAHA doses than in suspension culture. However TRAIL responses in cells that had been selected for TRAIL resistance were not further enhanced by SAHA treatment. CONCLUSIONS SAHA is a potent sensitizer of TRAIL responses in both TRAIL sensitive and resistant cell lines, in both suspension and 3D culture, however SAHA did not sensitise TRAIL-sensitive cell populations that had been selected for TRAIL-resistance from initially TRAIL-sensitive populations. SAHA may increase TRAIL sensitivity in insensitive cells, but not in cells that have specifically been selected for acquired TRAIL-resistance.
Collapse
Affiliation(s)
- A Arhoma
- Biomolecular Sciences Research Centre, Sheffield Hallam University, United Kingdom
| | - A D Chantry
- Biomolecular Sciences Research Centre, Sheffield Hallam University, United Kingdom; Mellanby Centre for Bone Research, University of Sheffield, United Kingdom
| | - S L Haywood-Small
- Biomolecular Sciences Research Centre, Sheffield Hallam University, United Kingdom
| | - N A Cross
- Biomolecular Sciences Research Centre, Sheffield Hallam University, United Kingdom
| |
Collapse
|
13
|
Shvartsur A, Givechian KB, Garban H, Bonavida B. Overexpression of RKIP and its cross-talk with several regulatory gene products in multiple myeloma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:62. [PMID: 28476134 PMCID: PMC5420138 DOI: 10.1186/s13046-017-0535-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/26/2017] [Indexed: 12/27/2022]
Abstract
Multiple myeloma (MM) is a clonal plasma-cell neoplastic disorder arising from an indolent premalignant disease known as monoclonal gammopathy of undetermined significance (MGUS). MM is a biologically complex heterogeneous disease reflected by its variable clinical responses of patients receiving the same treatment. Therefore, a molecular identification of stage-specific biomarkers will support a more individualized precise diagnostic/prognostic approach, an effective therapeutic regime, and will assist in the identification of novel therapeutic molecular targets. The metastatic suppressor/anti-resistance factor Raf-1 kinase inhibitor protein (RKIP) is poorly expressed in the majority of cancers and is often almost absent in metastatic tumors. RKIP inhibits the Raf/MEK/ERK1/2 and the NF-κB pathways. Whereby all tumors examined exhibited low levels of RKIP, in contrast, our recent findings demonstrated that RKIP is overexpressed primarily in its inactive phosphorylated form in MM cell lines and patient-derived tumor tissues. The underlying mechanism of RKIP overexpression in MM, in contrast to other tumors, is not known. We examined transcriptomic datasets on Oncomine platform (Life Technologies) for the co-expression of RKIP and other gene products in both pre-MM and MM. The transcription of several gene products was found to be either commonly overexpressed (i.e., RKIP, Bcl-2, and DR5) or underexpressed (i.e., Bcl-6 and TNFR2) in both pre-MM and MM. Noteworthy, a significant inverse correlation of differentially expressed pro-apoptotic genes was observed in pre-MM: overexpression of Fas and TNF-α and underexpression of YY1 versus expression of anti-apoptotic genes in MM: overexpression of YY1 and underexpression of Fas and TNF-α. Based on the analysis on mRNA levels and reported studies on protein levels of the above various genes, we have constructed various schemes that illustrate the possible cross-talks between RKIP (active/inactive) and the identified gene products that underlie the mechanism of RKIP overexpression in MM. Clearly, such cross-talks would need to be experimentally validated in both MM cell lines and patient-derived tumor tissues. If validated, the differential molecular signatures between pre-MM and MM might lead to a more precise diagnosis/prognosis of the disease and disease stages and will also identify novel molecular therapeutic targets for pre-MM and MM.
Collapse
Affiliation(s)
- Anna Shvartsur
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kevin B Givechian
- Department of Biological Sciences, USC Dana and David Dornsife College of Letters, Arts and Sciences at the University of Southern California, Los Angeles, CA, 90089, USA
| | - Hermes Garban
- California NanoSystems Institute (CnSI), University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
14
|
Inverse correlation between the metastasis suppressor RKIP and the metastasis inducer YY1: Contrasting roles in the regulation of chemo/immuno-resistance in cancer. Drug Resist Updat 2017; 30:28-38. [PMID: 28363333 DOI: 10.1016/j.drup.2017.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/28/2016] [Accepted: 01/04/2017] [Indexed: 02/06/2023]
Abstract
Several gene products have been postulated to mediate inherent and/or acquired anticancer drug resistance and tumor metastasis. Among these, the metastasis suppressor and chemo-immuno-sensitizing gene product, Raf Kinase Inhibitor Protein (RKIP), is poorly expressed in many cancers. In contrast, the metastasis inducer and chemo-immuno-resistant factor Yin Yang 1 (YY1) is overexpressed in many cancers. This inverse relationship between RKIP and YY1 expression suggests that these two gene products may be regulated via cross-talks of molecular signaling pathways, culminating in the expression of different phenotypes based on their targets. Analyses of the molecular regulation of the expression patterns of RKIP and YY1 as well as epigenetic, post-transcriptional, and post-translational regulation revealed the existence of several effector mechanisms and crosstalk pathways, of which five pathways of relevance have been identified and analyzed. The five examined cross-talk pathways include the following loops: RKIP/NF-κB/Snail/YY1, p38/MAPK/RKIP/GSK3β/Snail/YY1, RKIP/Smurf2/YY1/Snail, RKIP/MAPK/Myc/Let-7/HMGA2/Snail/YY1, as well as RKIP/GPCR/STAT3/miR-34/YY1. Each loop is comprised of multiple interactions and cascades that provide evidence for YY1's negative regulation of RKIP expression and vice versa. These loops elucidate potential prognostic motifs and targets for therapeutic intervention. Chiefly, these findings suggest that targeted inhibition of YY1 by specific small molecule inhibitors and/or the specific induction of RKIP expression and activity are potential therapeutic strategies to block tumor growth and metastasis in many cancers, as well as to overcome anticancer drug resistance. These strategies present potential alternatives for their synergistic uses in combination with low doses of conventional chemo-immunotherapeutics and hence, increasing survival, reducing toxicity, and improving quality of life.
Collapse
|
15
|
Expression of RKIP in chronic myelogenous leukemia K562 cell and inhibits cell proliferation by regulating the ERK/MAPK pathway. Tumour Biol 2015; 35:10057-66. [PMID: 25015191 DOI: 10.1007/s13277-014-2312-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/03/2014] [Indexed: 01/03/2023] Open
Abstract
RAF kinase inhibitor protein (RKIP) is a negative regulator of the RAS-mitogen-activated protein kinase/extracellular signal-regulated kinase signaling cascade. We investigated the expression of RKIP in chronic myelogenous leukemia (CML) K562 cells and the effects of RKIP on the characteristics of K562 cells. The recombinant plasmid pcDNA3.1-RKIP was established and transfected into K562 cells with the help of Lipofectamine 2000. At the same time, the RKIP-siRNA was transfected into K562 cells in another group. The expressions of RKIP in all groups were assayed by Western blot after 48 h. MTT (3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay was used to analyze the cell viability. Flow cytometry (FCM) was used to examine the cell cycle and cell apoptosis. Colony forming unit (CFU) assay was used to analyze the effect of RKIP on the clonogenic growth of CML cells. Western blot or luciferase reporter assay was used to detect the effect of RKIP on the level of phospho-ERK1/2 or the transcriptional activity of NF-κB. Western blot analysis showed that the plasmid pcDNA3.1-RKIP or RKIP-siRNA significantly enhanced or decreased RKIP expression (p < 0.01), respectively. In addition, MTT, FCM, and CFU assay indicated that the overexpression of RKIP significantly lowered the cell viability, cell proliferation and the clonogenic growth (p < 0.05), but improved cell apoptosis (p < 0.01). Western blot analysis or luciferase reporter assay showed that the level of phospho-ERK1/2 or the transcriptional activity of NF-κB was strongly inhibited by overexpression of RKIP. All these results could bring us a new perspective for biological therapy in myelogenous leukemia in the future.
Collapse
|