1
|
Sittiju P, Wudtiwai B, Chongchai A, Hajitou A, Kongtawelert P, Pothacharoen P, Suwan K. Bacteriophage-based particles carrying the TNF-related apoptosis-inducing ligand (TRAIL) gene for targeted delivery in hepatocellular carcinoma. NANOSCALE 2024; 16:6603-6617. [PMID: 38470366 PMCID: PMC10977282 DOI: 10.1039/d3nr05660k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The TRAIL (Tumour Necrosis Factor-Related Apoptosis-Inducing Ligand) is a promising candidate for cancer treatment due to its unique ability to selectively induce programmed cell death, or apoptosis, in cancer cells while sparing healthy ones. This selectivity arises from the preferential binding of the TRAIL to death receptors on cancer cells, triggering a cascade of events that lead to their demise. However, significant limitations in using the TRAIL for cancer treatment are the administration of the TRAIL protein that can potentially lead to tissue toxicity (off-target) and the short half-life of the TRAIL in the body which may necessitate frequent and sustained administration; these can pose logistical challenges for long-term treatment regimens. We have devised a novel approach for surmounting these limitations by introducing the TRAIL gene directly into cancer cells, enabling them to produce the TRAIL locally and subsequently trigger apoptosis. A novel gene delivery system such as a bacteriophage-based particle TPA (transmorphic phage/AAV) was utilized to address these limitations. TPA is a hybrid M13 filamentous bacteriophage particle encapsulating a therapeutic gene cassette with inverted terminal repeats (ITRs) from adeno-associated viruses (AAVs). The particle also showed a tumour targeting ligand, CDCRGDCFC (RGD4C), on its capsid (RGD4C.TPA) to target the particle to cancer cells. RGD4C selectively binds to αvβ3 and αvβ5 integrins overexpressed on the surface of most of the cancer cells but is barely present on normal cells. Hepatocellular carcinoma (HCC) was chosen as a model because it has one of the lowest survival rates among cancers. We demonstrated that human HCC cell lines (Huh-7 and HepG2) express αvβ5 integrin receptors on their surface. These HCC cells also express death receptors and TRAIL-binding receptors. We showed that the targeted TPA particle carrying the transmembrane TRAIL gene (RGD4C.TPA-tmTRAIL) selectively and efficiently delivered the tmTRAIL gene to HCC cells resulting in the production of tmTRAIL from transduced cells and subsequently induced apoptotic death of HCC cells. This tumour-targeted particle can be an excellent candidate for the targeted gene therapy of HCC.
Collapse
Affiliation(s)
- Pattaralawan Sittiju
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Benjawan Wudtiwai
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Aitthiphon Chongchai
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Amin Hajitou
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Prachya Kongtawelert
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Peraphan Pothacharoen
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Keittisak Suwan
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
2
|
Huang XF, Fu LS, Cai QQ, Fan F. Prognostic and immunological role of sulfatide-related lncRNAs in hepatocellular carcinoma. Front Oncol 2023; 13:1091132. [PMID: 36816914 PMCID: PMC9929346 DOI: 10.3389/fonc.2023.1091132] [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: 11/06/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver. Long non-coding RNAs (lncRNAs) play important roles in the occurrence and development of HCC through multiple pathways. Our previous study reported the specific molecular mechanism for sulfatide regulation of integrin αV expression and cell adhesion in HCC cells through lncRNA AY927503. Next, it is necessary to identify more sulfatide-related lncRNAs, explore their clinical signifcance, and determine new targeted treatment strategies. Methods Microarrays were used to screen a complete set of lncRNAs with different expression profiles in sulfatide-treated cells. Sulfatide-related lncRNAs expression data and corresponding HCC patient survival information were obtained from the The Cancer Genome Atlas (TCGA) database, and the prognosis prediction model was constructed based on Cox regression analysis. Methylated RNA immunoprecipitation with next generation sequencing (MeRIP-seq) was used to detemine the effect of sulfatide on lncRNAs m6A modification. Tumor Immune Estimation Resource (TIMER) and Gene set nnrichment analysis (GSEA) were utilized to enrich the immune and functional pathways of sulfatide-related lncRNAs. Results A total of 85 differentially expressed lncRNAs (|Fold Change (FC)|>2, P<0.05) were screened in sulfatide-treated HCC cells. As a result, 24 sulfatide-related lncRNAs were highly expressed in HCC tissues, six of which were associated with poor prognosis in HCC patients. Based on thses data, a sulfatide-related lncRNAs prognosis assessment model for HCC was constructed. According to this risk score analysis, the overall survival (OS) curve showed that the OS of high-risk patients was significantly lower than that of low-risk patients (P<0.05). Notably, the expression difference in sulfatide-related lncRNA NRSN2-AS1 may be related to sulfatide-induced RNA m6A methylation. In addition, the expression level of NRSN2-AS1 was significantly positively correlated with immune cell infiltration in HCC and participated in the peroxisome and Peroxisome proliferator-activated receptor (PPAR) signaling pathways. Conclusions In conclusion, sulfatide-related lncRNAs might be promising prognostic and therapeutic targets for HCC.
Collapse
Affiliation(s)
- Xing Feng Huang
- Department of Biliary Tract Surgery, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Li Sheng Fu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Qian Qian Cai
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Fei Fan
- Department of The Second Ward of Special Treatment, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| |
Collapse
|
3
|
Byrne FL, Olzomer EM, Lolies N, Hoehn KL, Wegner MS. Update on Glycosphingolipids Abundance in Hepatocellular Carcinoma. Int J Mol Sci 2022; 23:ijms23094477. [PMID: 35562868 PMCID: PMC9102297 DOI: 10.3390/ijms23094477] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 11/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer. Low numbers of HCC patients being suitable for liver resection or transplantation and multidrug resistance development during pharmacotherapy leads to high death rates for HCC patients. Understanding the molecular mechanisms of HCC etiology may contribute to the development of novel therapeutic strategies for prevention and treatment of HCC. UDP-glucose ceramide glycosyltransferase (UGCG), a key enzyme in glycosphingolipid metabolism, generates glucosylceramide (GlcCer), which is the precursor for all glycosphingolipids (GSLs). Since UGCG gene expression is altered in 0.8% of HCC tumors, GSLs may play a role in cellular processes in liver cancer cells. Here, we discuss the current literature about GSLs and their abundance in normal liver cells, Gaucher disease and HCC. Furthermore, we review the involvement of UGCG/GlcCer in multidrug resistance development, globosides as a potential prognostic marker for HCC, gangliosides as a potential liver cancer stem cell marker, and the role of sulfatides in tumor metastasis. Only a limited number of molecular mechanisms executed by GSLs in HCC are known, which we summarize here briefly. Overall, the role GSLs play in HCC progression and their ability to serve as biomarkers or prognostic indicators for HCC, requires further investigation.
Collapse
Affiliation(s)
- Frances L. Byrne
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (F.L.B.); (E.M.O.); (K.L.H.)
| | - Ellen M. Olzomer
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (F.L.B.); (E.M.O.); (K.L.H.)
| | - Nina Lolies
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, Theodor Stern-Kai 7, 60590 Frankfurt, Germany;
| | - Kyle L. Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (F.L.B.); (E.M.O.); (K.L.H.)
| | - Marthe-Susanna Wegner
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia; (F.L.B.); (E.M.O.); (K.L.H.)
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, Theodor Stern-Kai 7, 60590 Frankfurt, Germany;
- Correspondence:
| |
Collapse
|
4
|
Chen MH, Qi B, Cai QQ, Sun JW, Fu LS, Kang CL, Fan F, Ma MZ, Wu XZ. LncRNA lncAY is upregulated by sulfatide via Myb/MEF2C acetylation to promote the tumorigenicity of hepatocellular carcinoma cells. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194777. [PMID: 34843988 DOI: 10.1016/j.bbagrm.2021.194777] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 01/17/2023]
Abstract
LncRNA (long noncoding RNA) is often dysregulated in tumors especially hepatocellular carcinoma (HCC). However, the dysregulation mechanism of lncRNAs is largely unknown. Here, we showed that lncRNA lncAY expression was stimulated in HCC by either endogenous or exogenous sulfatide. Elevated lncAY promoted HCC cell migration or angiogenesis, whereas lncAY silence suppressed HCC cell migration and proliferation. Interestingly, the activity of lncAY gene promoter was enhanced by sulfatide. Then Myb and MEF2C were identified as the transcription factors responsible for the stimulation of lncAY promoter activity and transcription by sulfatide. Both Myb and MEF2C enrichment on lncAY promoter was further confirmed, and their occupancy on lncAY promoter was strengthened by sulfatide for Myb or MEF2C was acetylated. Mutant Myb-K456A exhibited reduced acetylation and weak stimulation for lncAY transcription. However, Myb mutation K456/503A prevented Myb from acetylation induced by sulfatide. The mutant Myb K456/503A further was unable to occupy lncAY promoter and enhance lncAY transcription. In conclusion, this study demonstrated lncAY transcription was abnormally upregulated by sulfatide in HCC through Myb/MEF2C to promote HCC progression.
Collapse
Affiliation(s)
- Mei Hua Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR China
| | - Bing Qi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR China
| | - Qian Qian Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR China
| | - Jia Wen Sun
- Beng Bu Medical College, Bengbu, 233000, Anhui, PR China
| | - Li Sheng Fu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR China
| | - Chun Lan Kang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR China
| | - Fei Fan
- Department of the 3rd ward of Special Treatment, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, PR China
| | - Ming Zhe Ma
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, PR China
| | - Xing Zhong Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR China.
| |
Collapse
|
5
|
Cai Q, Liu Y, Zhu P, Kang C, Xu H, Qi B, Wang R, Dong Y, Wu XZ. SIN3B promotes integrin αV subunit gene transcription and cell migration of hepatocellular carcinoma. J Mol Cell Biol 2020; 11:421-432. [PMID: 30215728 PMCID: PMC7727265 DOI: 10.1093/jmcb/mjy050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/23/2018] [Accepted: 09/12/2018] [Indexed: 12/16/2022] Open
Abstract
Paired amphipathic helix protein (SIN3B) is a transcription corepressor for many genes. Here we show a different regulation mechanism of integrin αV gene expression by SIN3B in human hepatocellular carcinoma (HCC). We first observed a close relationship between Integrin αV and SIN3B expressions in HCC patients and tumor cell lines with different metastatic potentials. Overexpression of SIN3B significantly accelerated the cell migration rate of SMMC-7721, but failed when integrin αV expression was silenced. Interestingly, SIN3B stimulated integrin αV subunit promoter activity only in the presence of sulfatide. Importantly, SIN3B was identified in the complex with sulfatide by mass spectrometry. Fat blot assay indicated that SIN3B specifically interacted with sulfatide. Molecular modeling suggested that sulfatide induced the conformational change of SIN3B from compacted α-helices to a relaxed β-sheet in PAH2 domain. The data of immunoprecipitation and ChIP assay indicated that altered SIN3B lost the binding affinity with MAD1 and HDAC2, which reduced the recruitment of HDAC2 on integrin αV gene promoter and prevented the deacetylation of the histone 3. In conclusion, this study demonstrated that SIN3B promoted the transcriptional activation of the integrin αV subunit gene promoter by reducing interaction with HDAC2.
Collapse
Affiliation(s)
- Qianqian Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Yuanyuan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Ping Zhu
- Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Chunlang Kang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Heyang Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Bing Qi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Rong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Yiwei Dong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Xing Zhong Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| |
Collapse
|
6
|
Zhang N, Zhao SS, Zhang YX, Wang YC, Shao RG, Wang JX, He HW. A novel biphenyl compound IMB-S7 ameliorates hepatic fibrosis in BDL rats by suppressing Sp1-mediated integrin αv expression. Acta Pharmacol Sin 2020; 41:661-669. [PMID: 31932644 PMCID: PMC7470799 DOI: 10.1038/s41401-019-0325-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/31/2019] [Indexed: 01/15/2023]
Abstract
Chronic tissue injury with fibrosis results in the disruption of tissue architecture, organ dysfunction, and eventual organ failure. Therefore, the development of effective antifibrotic drugs is urgently required. IMB-S7 is novel biphenyl compound derived from bifendate (biphenyldicarboxylate) that is used for the treatment of chronic hepatitis in China. In the current study we investigated the potential of IMB-S7 as an antihepatic fibrosis agent. In bile duct ligation (BDL) rat model, oral administration of IMB-S7 (400 mg· kg-1· d-1, for 14 days) significantly ameliorated BDL-induced liver necrosis, bile duct proliferation, and collagen accumulation. We then showed that IMB-S7 treatment markedly suppressed the TGF-β/Smad pathway in human hepatic stellate cell line LX2 and mouse primary HSCs, as well as in liver samples of BDL rats, thus inhibiting the transcription of most fibrogenesis-associated genes, including TGF-β1, COL1A1, and ACTA2. Furthermore, IMB-S7 treatment significantly suppressed the expression of integrin αv at the mRNA and protein levels in TGF-β-treated LX2 cells and liver samples of BDL rats. Using integrin αv overexpression and silencing, we demonstrated that integrin αv activity correlated positively with the activation of TGF-β/Smad pathway. Based on dual luciferase assay and DNA affinity precipitation assay, we revealed that IMB-S7 inactivated integrin αv through competitively inhibiting the binding of Sp1, a transcription factor, to the integrin αv (ITGAV) promoter (-173/-163 bp). These results suggest that IMB-S7 inhibits HSCs activation and liver fibrosis through Sp1-integrin αv signaling, and IMB-S7 may be a promising candidate to combat hepatic fibrosis in the future.
Collapse
Affiliation(s)
- Na Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Shuang-Shuang Zhao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China
- The Joint Program in Infection and Immunity, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yi-Xuan Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Yu-Cheng Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Rong-Guang Shao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Ju-Xian Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China.
| | - Hong-Wei He
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China.
| |
Collapse
|
7
|
Zang D, Zhang C, Li C, Fan Y, Li Z, Hou K, Che X, Liu Y, Qu X. LPPR4 promotes peritoneal metastasis via Sp1/integrin α/FAK signaling in gastric cancer. Am J Cancer Res 2020; 10:1026-1044. [PMID: 32266108 PMCID: PMC7136906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 02/20/2020] [Indexed: 06/11/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies which has high incidence and mortality worldwide. Peritoneal dissemination is the main route of metastasis in advanced GC. However, few reliable diagnostic or prognostic biomarkers are available for peritoneal metastasis of GC. This study aimed to investigate the effect of lipid phosphate phosphatase-related protein type 4 (LPPR4) on the prognosis of peritoneal metastasis in GC, so as to explore the underlying molecular mechanisms and clinical significance of the process. Differentially expressed genes (DEGs) between tumor tissues and adjacent normal tissues were identified. The prognostic values of the DEGs were tested in two independent cohorts (TCGA-STAD cohort and GSE62254 cohort). Eight DEGs including LPPR4 with prognostic value in GC peritoneal metastasis were identified. The expression of LPPR4 increased in peritoneal metastasis of GC tissues, and high LPPR4 expression was associated with poor overall survival in GC. Loss- and gain-of functional experiments were performed to reveal that LPPR4 could promote the migration, invasion and adhesion abilities of GC cells in vitro. Tumor peritoneal dissemination was investigated in a mouse model to reveal that LPPR4 could promote peritoneal metastasis of GC cells in vivo. According to the Kyoto Encyclopedia of Genes and Genomics (KEGG) and gene set enrichment analysis (GSEA), LPPR4 was found to be related to focal adhesion, cell adhesion molecules (CAMs) and ECM-receptor interaction pathways. LPPR4 knockdown significantly inhibited the expression of integrin α1, integrin α2, integrin α5, integrin α6, integrin α7, p-FAK, p-Akt, p-Src and MMP2. Moreover, this process was regulated by the Specificity Protein 1 (Sp1) transcription factor. Taken together, LPPR4 plays an essential role in promoting peritoneal metastasis of GC through Sp1/integrin α/FAK signaling, and acts as a novel biomarker of prognosis of GC peritoneal metastasis. The results suggest that LPPR4 may serve as a new therapeutic target for patients with GC peritoneal metastasis.
Collapse
Affiliation(s)
- Dan Zang
- Department of Medical Oncology, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical UniversityShenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical UniversityShenyang 110001, China
| | - Chuang Zhang
- Department of Medical Oncology, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical UniversityShenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical UniversityShenyang 110001, China
| | - Ce Li
- Department of Medical Oncology, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical UniversityShenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical UniversityShenyang 110001, China
| | - Yibo Fan
- Department of Medical Oncology, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical UniversityShenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical UniversityShenyang 110001, China
| | - Zhi Li
- Department of Medical Oncology, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical UniversityShenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical UniversityShenyang 110001, China
| | - Kezuo Hou
- Department of Medical Oncology, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical UniversityShenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical UniversityShenyang 110001, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical UniversityShenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical UniversityShenyang 110001, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical UniversityShenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical UniversityShenyang 110001, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical UniversityShenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical UniversityShenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical UniversityShenyang 110001, China
| |
Collapse
|
8
|
Du J, Zhao Z, Zhao H, Liu D, Liu H, Chen J, Cheng B, Zhai X, Yin Z, Zhang Y, Ling C. Sec62 promotes early recurrence of hepatocellular carcinoma through activating integrinα/CAV1 signalling. Oncogenesis 2019; 8:74. [PMID: 31822656 PMCID: PMC6904485 DOI: 10.1038/s41389-019-0183-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Postsurgical recurrence within 2 years is the major cause of poor survival of hepatocellular carcinoma (HCC) patients. However, the molecular mechanism underlying HCC recurrence remains unclear. Here, we distinguish the function and mechanism of Sec62 in promoting HCC recurrence. The correlation between Sec62 and early recurrence was demonstrated in 60 HCC samples from a prospective study. HCC cells with Sec62 knockdown (Sec62KD) or overexpression (Sec62OE) were used to determine the potential of Sec62 in cell migration in vitro. Microarray analysis comparing Sec62KD or Sec62OE to their control counterparts was used to explore the mechanisms of Sec62-induced recurrence. A luciferase-labelled orthotopic nude mouse model of HCC with Sec62KD or Sec62OE was used to validate the potential of Sec62 in early HCC recurrence in vivo. We found that high expression of Sec62 was positively correlated with surgical recurrence in clinical HCC samples. Multivariate analysis revealed that Sec62 was an independent prognostic factor for early recurrence in postoperative HCC patients. Moreover, Sec62 promoted migration and invasion of HCC cells in vitro and postsurgical recurrence in vivo. Mechanically, integrinα/CAV1 signalling was identified as one of the targets of Sec62 in cell movement. Overexpression of integrin α partially rescued the Sec62 knockdown-induced inhibition of cell migration. Sec62 is a potentially prognostic factor for early recurrence in postoperative HCC patients and promotes HCC metastasis through integrinα/CAV1 signalling. Sec62 might be an attractive drug target for combating HCC postsurgical recurrence.
Collapse
Affiliation(s)
- Juan Du
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, 200433, China
| | - Zhihao Zhao
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, 200433, China
| | - Hetong Zhao
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, 200433, China
| | - Dong Liu
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, 200433, China
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai, 200433, China
| | - Jun Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Binbin Cheng
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, 200433, China
| | - Xiaofeng Zhai
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, 200433, China
| | - Zifei Yin
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, 200433, China
| | - Yani Zhang
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, 200433, China
| | - Changquan Ling
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
9
|
Kang CL, Qi B, Cai QQ, Fu LS, Yang Y, Tang C, Zhu P, Chen QW, Pan J, Chen MH, Wu XZ. LncRNA AY promotes hepatocellular carcinoma metastasis by stimulating ITGAV transcription. Am J Cancer Res 2019; 9:4421-4436. [PMID: 31285770 PMCID: PMC6599657 DOI: 10.7150/thno.32854] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 05/21/2019] [Indexed: 01/11/2023] Open
Abstract
Rationale: Tumor metastasis is the main cause for cancer-related death. However, the driving molecules of metastasis remain largely unknown. Here, we aim to identify long non-coding RNAs (lncRNAs) critical for human hepatocellular carcinoma (HCC) metastasis. Methods: Microarrays were used to screen a comprehensive set of lncRNAs with differential expression profiles in sulfatide-treated cells. Mass spectrometry, protein arrays, and RNA pull-down experiments were used to identify proteins that interacted with lncRNA. Epigenetic analysis was used to study lncRNA-mediated regulation mechanisms. Results: We identified lncRNA AY927503 (AY) as a metastasis-associated molecule that was highly expressed in human hepatocellular carcinoma (HCC) and correlated with metastatic events and poor prognosis in patients with HCC. AY promoted HCC cell migration, stemness, 5-fluorouracil resistance, and metastasis in mice. However, knockdown of integrin αV (ITGAV) abolished AY-stimulated migration, cell viability in HCC cells or tube formation. AY strongly promoted ITGAV transcription and αVβ3 expression by interacting with the ITGAV promoter specifically and stimulating its activity. AY was identified to interact with histone 1FX (H1FX), but deletion of the central domain of AY (AY∆371-522) abolished H1FX binding and ITGAV promoter stimulation. AY significantly enriched H3K4Me3 and acH3K9/14 but reduced H3K27Me3 and H1FX occupancy on the ITGAV promoter, which remodeled chromatin structures for RNA polymerase II recruitment. Knockdown of H1FX abrogated ITGAV transcription stimulated by AY. Conclusions: Our findings suggested that lncRNA AY promoted HCC metastasis via induction of chromatin modification for ITGAV transcription as a pioneer factor and was a potential molecular signature for metastasis or poor prognosis in patients with HCC.
Collapse
|
10
|
Cao Q, Chen X, Wu X, Liao R, Huang P, Tan Y, Wang L, Ren G, Huang J, Dong C. Inhibition of UGT8 suppresses basal-like breast cancer progression by attenuating sulfatide-αVβ5 axis. J Exp Med 2018; 215:1679-1692. [PMID: 29728441 PMCID: PMC5987921 DOI: 10.1084/jem.20172048] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/08/2018] [Accepted: 04/06/2018] [Indexed: 12/15/2022] Open
Abstract
Cao et al. show that UGT8 promotes BLBC progression through activating sulfatide–αVβ5 axis. ZA is identified as a direct inhibitor of UGT8 and suppresses BLBC progression, suggesting that inhibition of UGT8 offers a promising opportunity for treating BLBC. Basal-like breast cancer (BLBC) is associated with a poor clinical outcome as a result of the few treatment options and poor therapeutic response. Here, we report that elevated expression of urine diphosphate–galactose ceramide galactosyltransferase (UGT8) specifically occurs in BLBC and predicts poor prognosis in breast cancer patients. UGT8 expression is transcriptionally up-regulated by Sox10, triggering the sulfatide biosynthetic pathway; increased sulfatide activates integrin αVβ5-mediated signaling that contributes to BLBC progression. UGT8 expression promotes, whereas UGT8 knockdown suppresses tumorigenicity and metastasis. Importantly, we identify that zoledronic acid (ZA), a marketed drug for treating osteoporosis and bone metastasis, is a direct inhibitor of UGT8, which blocks the sulfatide biosynthetic pathway. Significantly, a clinically achievable dosage of ZA exhibits apparent inhibitory effect on migration, invasion, and lung metastasis of BLBC cells. Together, our study suggests that UGT8 is a potential prognostic indicator and druggable target of BLBC and that pharmacologic inhibition of UGT8 by ZA offers a promising opportunity for treating this challenging disease.
Collapse
Affiliation(s)
- Qianhua Cao
- Department of Pathology and Pathophysiology and Department of Surgical Oncology (Breast Center), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingyu Chen
- Department of Pathology and Pathophysiology and Department of Surgical Oncology (Breast Center), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuebiao Wu
- Department of Pathology and Pathophysiology and Department of Surgical Oncology (Breast Center), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruocen Liao
- Department of Pathology and Pathophysiology and Department of Surgical Oncology (Breast Center), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| | - Panpan Huang
- Department of Pathology and Pathophysiology and Department of Surgical Oncology (Breast Center), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanjia Tan
- Department of Pathology and Pathophysiology and Department of Surgical Oncology (Breast Center), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Wang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guoping Ren
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Huang
- Department of Pathology and Pathophysiology and Department of Surgical Oncology (Breast Center), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenfang Dong
- Department of Pathology and Pathophysiology and Department of Surgical Oncology (Breast Center), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China .,Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
11
|
Cai QQ, Dong YW, Qi B, Shao XT, Wang R, Chen ZY, He BM, Wu XZ. BRD1-Mediated Acetylation Promotes Integrin αV Gene Expression Via Interaction with Sulfatide. Mol Cancer Res 2018; 16:610-622. [PMID: 29453316 DOI: 10.1158/1541-7786.mcr-17-0527] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/16/2017] [Accepted: 01/26/2018] [Indexed: 11/16/2022]
Abstract
Integrin αV gene expression is often dysregulated in cancers especially in hepatocellular carcinoma (HCC); however, the mechanism of regulation is poorly understood. Here, it is demonstrated that sulfatide activated integrin αV gene transcription, through histone H3K9/14 acetylation at the promoter, and high integrin αV expression are closely associated with poor prognosis. To elucidate the mechanism of regulation of acetylation, sulfatide-bound proteins were screened by mass spectrometry (MS), and bromodomain containing protein 1 (BRD1) was identified as an interacting protein that also colocalized with sulfatide in HCC cells. BRD1 was also formed a complex with Sp1, which was recruited to the integrin αV gene promoter. Sulfatide was also found to induce BRD1, monocytic leukemia zinc finger (MOZ) and histone acetyltransferase binding to ORC1 (HBO1) acetyltransferase multiprotein complex recruitment to the integrin αV promoter, which is responsible for histone H3K9/14 acetylation. Finally, knockdown of BRD1 limited sulfatide-induced H3K9/14 acetylation and occupancy of MOZ or HBO1 on integrin αV gene promoter.Implications: This study demonstrates that sulfatide interaction with BRD1 mediates acetylation and is important for regulation of integrin αV gene expression. Mol Cancer Res; 16(4); 610-22. ©2018 AACR.
Collapse
Affiliation(s)
- Qian Qian Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Yi Wei Dong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Bing Qi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Xiao-Ting Shao
- Yu Ying Hospital, Wenzhou Medical University, Wenzhou, China
| | - Rong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Zhong Yi Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Bao Mei He
- Yu Ying Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xing Zhong Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China.
| |
Collapse
|
12
|
Wang R, Qi B, Dong YW, Cai QQ, Deng NH, Chen Q, Li C, Jin YT, Wu XZ. Sulfatide interacts with and activates integrin αVβ3 in human hepatocellular carcinoma cells. Oncotarget 2017; 7:36563-36576. [PMID: 27145276 PMCID: PMC5095021 DOI: 10.18632/oncotarget.9095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 04/16/2016] [Indexed: 12/12/2022] Open
Abstract
Integrin αVβ3 is a malignant driver of anchorage-independence and tumor angiogenesis, but its dysregulation in hepatocellular carcinoma (HCC) remains unclear. In this study, we observed that sulfatide significantly promoted integrin αV(ITGAV) expression and wound closure in HCC. We also noted that elevated sulfatide profoundly stimulated integrin αVβ3 clustering and signaling. In the cells with integrin αVβ3 clustering induced by sulfatide, integrin β3 subunit was phosphorylated. Simultaneously, focal adhesion kinase (FAK), Src and paxillin were also phosphorylated. Treatment with FAK inhibitor resulted in robust suppression of FAK-Y397 and Src-Y416 phosphorylation stimulated by sulfatide, but not suppression of integrin β3 phosphorylation. Src inhibitors repressed Src-Y416 and FAK Y861 and Y925 phosphorylation, but not FAK-Y397 and integrin β3 phosphorylation. After mutation of integrin β3 (Y773F and Y785F), FAK or Src phosphorylation failed to be stimulated by sulfatide. Moreover, β3 Y773 and Y785 phosphorylation was suppressed by insulin-like growth factor receptor knockdown even in cells stimulated by sulfatide. In assays of immunoprecipitation and immunostaining with integrin αV or β3 antibody, labeled sulfatide was found in the complex and co-localized with integrin αVβ3. Taken together, this study demonstrated that elevated sulfatide bound to integrin αVβ3 and induced clustering and phosphorylation of αVβ3 instead of matrix ligand binding, triggering outside-in signaling.
Collapse
Affiliation(s)
- Rong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, PR. China.,Key Laboratory of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR. China
| | - Bing Qi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, PR. China.,Key Laboratory of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR. China
| | - Yi Wei Dong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, PR. China.,Key Laboratory of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR. China
| | - Qian Qian Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, PR. China.,Key Laboratory of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR. China
| | - Nian Hui Deng
- Yu Ying Children's Hospital, Wenzhou Medical University, Wenzhou, PR. China
| | - Qi Chen
- Yu Ying Children's Hospital, Wenzhou Medical University, Wenzhou, PR. China
| | - Chao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, PR. China
| | - Yu Tong Jin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, PR. China
| | - Xing Zhong Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, PR. China.,Key Laboratory of Glycoconjugate Research, Ministry of Public Health, Shanghai, PR. China
| |
Collapse
|
13
|
Xiong S, Klausen C, Cheng JC, Leung PCK. TGFβ1 induces endometrial cancer cell adhesion and migration by up-regulating integrin αvβ3 via SMAD-independent MEK-ERK1/2 signaling. Cell Signal 2017; 34:92-101. [PMID: 28336232 DOI: 10.1016/j.cellsig.2017.03.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/14/2017] [Accepted: 03/19/2017] [Indexed: 11/26/2022]
Abstract
Endometrial cancer is the most common, and second most lethal, gynecological malignancy, and its rates of incidence and death are growing. This is likely attributable to increased numbers of high-risk type II endometrial cancers which account for ~30% of cases but ~75% of deaths due to their aggressive and metastatic behaviour. Histopathological and in vitro functional studies suggest that aberrant TGFβ1 signaling may contribute to endometrial cancer development and the acquisition of invasive/metastatic characteristics. However, little is known about the cellular and molecular mechanisms of TGFβ1 in high-risk endometrial cancers. In the present study, we examined the roles and mechanisms of TGFβ1 on cell adhesion and motility in type II endometrial cancer cell lines, KLE and HEC-1B. We show that treatment with TGFβ1 increases cell adhesion to vitronectin and transwell cell migration. We also demonstrate that TGFβ1 treatment increases integrin β3 and αv mRNA and protein levels via SMAD-independent MEK-ERK1/2 signaling. Importantly, siRNA depletion or antibody-mediated blocking of integrin αvβ3 reversed the effects of TGFβ1 on cell adhesion and migration. Our results suggest that TGFβ1-MEK-ERK1/2-integrin αvβ3 signaling could contribute to the invasive behaviour of high-risk endometrial cancer by promoting cell adhesion and migration.
Collapse
Affiliation(s)
- Siyuan Xiong
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Christian Klausen
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Jung-Chien Cheng
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada.
| |
Collapse
|
14
|
Chao KL, Kulakova L, Herzberg O. Gene polymorphism linked to increased asthma and IBD risk alters gasdermin-B structure, a sulfatide and phosphoinositide binding protein. Proc Natl Acad Sci U S A 2017; 114:E1128-E1137. [PMID: 28154144 PMCID: PMC5321033 DOI: 10.1073/pnas.1616783114] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The exact function of human gasdermin-B (GSDMB), which regulates differentiation and growth of epithelial cells, is yet to be elucidated. In human epidermal growth factor receptor 2 (HER2)-positive breast cancer, GSDMB gene amplification and protein overexpression indicate a poor response to HER2-targeted therapy. Genome-wide association studies revealed a correlation between GSDMB SNPs and an increased susceptibility to Crohn's disease, ulcerative colitis, and asthma. The N- and C-terminal domains of all gasdermins possess lipid-binding and regulatory activities, respectively. Inflammatory caspases cleave gasdermin-D in the interdomain linker but not GSDMB. The cleaved N-terminal domain binds phosphoinositides and cardiolipin, forms membrane-disrupting pores, and executes pyroptosis. We show that both full-length GSDMB and the N-terminal domain bind to nitrocellulose membranes immobilized with phosphoinositides or sulfatide, but not with cardiolipin. In addition, the GSDMB N-terminal domain binds liposomes containing sulfatide. The crystal structure of the GSDMB C-terminal domain reveals the structural impact of the amino acids encoded by SNPs that are linked to asthma and inflammatory bowel disease (IBD). A loop that carries the polymorphism amino acids corresponding to healthy individuals (Gly299:Pro306) exhibits high conformational flexibility, whereas the loop carrying amino acids found in individuals with increased disease risk (Arg299:Ser306) exhibits a well-defined conformation and higher positive surface charge. Apoptotic executioner caspase-3, -6, and -7, but not the inflammatory caspases, cleave GSDMB at 88DNVD91 within the N-terminal domain. Selective sulfatide binding may indicate possible function for GSDMB in the cellular sulfatide transport.
Collapse
Affiliation(s)
- Kinlin L Chao
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Liudmila Kulakova
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Osnat Herzberg
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850;
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742
| |
Collapse
|
15
|
MiR-124 inhibits the migration and invasion of human hepatocellular carcinoma cells by suppressing integrin αV expression. Sci Rep 2017; 7:40733. [PMID: 28094803 PMCID: PMC5240551 DOI: 10.1038/srep40733] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/09/2016] [Indexed: 12/13/2022] Open
Abstract
Tumor metastasis is the major cause of cancer-related death especially in human hepatocellular carcinoma (HCC). Although microRNAs have been implicated in tumor development, the roles of miR-124 in HCC metastasis are still not well understood. We conducted functional analysis in this study to investigate miR-124. We observed that miR-124 significantly retarded the wound healing and migration of HCC SMMC-7721 and BEL-7404 cells. Further analysis indicated miR-124 directly targeting the transcriptional factor Sp1 which is an important transcription factor for the integrin αV subunit gene transcription. Co-transfection of miR-124 with the luciferase reporter containing Sp1 3′ untranslated region (UTR) significantly suppressed the luciferase activities. While mutation of the binding site of miR-124 in Sp1 mRNA 3′UTR completely abrogated the suppression of miR-124. Overexpression of miR-124 resulted in robust downregulation of Sp1 and integrin αV expression at either mRNA or protein level. Ectopic expression of miR-124 in HCC dramatically repressed the wound healing and migration in vitro and tumor metastasis in mouse experiments. Our findings demonstrated that miR-124 played as an important role in regulation of integrin αV expression in HCC, and reintroduction of miR-124 might be an alternative therapeutic strategy for controlling integrin αV expression in HCC.
Collapse
|
16
|
Tian Y, Yang Y, Zhang X, Nakajima T, Tanaka N, Sugiyama E, Kamijo Y, Lu Y, Moriya K, Koike K, Gonzalez FJ, Aoyama T. Age-dependent PPARα activation induces hepatic sulfatide accumulation in transgenic mice carrying the hepatitis C virus core gene. Glycoconj J 2016; 33:927-936. [PMID: 27318478 PMCID: PMC6309323 DOI: 10.1007/s10719-016-9703-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 01/01/2023]
Abstract
Sulfatides, a type of glycosphingolipid, are associated with carcinogenesis. Peroxisome proliferator-activated receptor α (PPARα) is involved in the regulation of sulfatide metabolism as well as in cancer development. We previously reported that transgenic (Tg) mice expressing hepatitis C virus core protein (HCVcp) exhibited age-dependent PPARα activation and carcinogenesis in liver. However, the metabolism of sulfatides in hepatocellular carcinoma is unknown. To examine the relationship between sulfatide metabolism, carcinogenesis, HCVcp, and PPARα, age-dependent changes of these factors were examined in HCVcpTg, PPARα inhibitor-treated HCVcpTg, and non-Tg mice. The sulfatide content in liver, the hepatic expression of two key enzymes catalyzing the initial and last reactions in sulfatide synthesis, the hepatic expression of known sulfatide-transferring protein, oxidative stress, and hepatic PPARα expression and its activation were age-dependently increased in HCVcpTg mice. The increased synthesis and accumulation of sulfatides and PPARα activation were significantly enhanced in liver cancer lesions. These changes were attenuated by PPARα inhibitor treatment and not observed in non-Tg mice. These results suggest that HCVcp-induced age-dependent PPARα activation increases synthesis of sulfatides and the resulting sulfatide accumulation affects HCV-related liver cancer. The monitoring of hepatic sulfatide content and the modulation of sulfatide generation by intervention using a PPARα inhibitor might be useful for the prediction and prevention of HCV-related hepatocarcinogenesis, respectively.
Collapse
Affiliation(s)
- Yangyang Tian
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, 390-8621, Japan
| | - Yang Yang
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, 390-8621, Japan
| | - Xiaowei Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Takero Nakajima
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, 390-8621, Japan
| | - Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, 390-8621, Japan
| | - Eiko Sugiyama
- Department of Nutritional Science, Prefectural College, Nagano, Nagano, 380-8525, Japan
| | - Yuji Kamijo
- Department of Nephrology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
| | - Yu Lu
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, 390-8621, Japan
| | - Kyoji Moriya
- Department of Infection Control and Prevention, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Toshifumi Aoyama
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, 390-8621, Japan
| |
Collapse
|
17
|
Wu LH, Shao XT, Guo JX, Sun H, Chen Q, Pan J, Cai QQ, Dong YW, Chen ZY, Yan XM, Peng M, Zhong N, Wu XZ. Vimentin is important in the neural differentiation of PC12 cells promoted by sialylation. Glycoconj J 2016; 34:51-59. [DOI: 10.1007/s10719-016-9727-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/14/2016] [Accepted: 08/22/2016] [Indexed: 12/19/2022]
|
18
|
Shinderman-Maman E, Cohen K, Weingarten C, Nabriski D, Twito O, Baraf L, Hercbergs A, Davis PJ, Werner H, Ellis M, Ashur-Fabian O. The thyroid hormone-αvβ3 integrin axis in ovarian cancer: regulation of gene transcription and MAPK-dependent proliferation. Oncogene 2015; 35:1977-87. [PMID: 26165836 DOI: 10.1038/onc.2015.262] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 05/27/2015] [Accepted: 06/05/2015] [Indexed: 12/18/2022]
Abstract
Ovarian carcinoma is the fifth common cause of cancer death in women, despite advanced therapeutic approaches. αvβ3 integrin, a plasma membrane receptor, binds thyroid hormones (L-thyroxine, T4; 3,5,3'-triiodo-L-thyronine, T3) and is overexpressed in ovarian cancer. We have demonstrated selective binding of fluorescently labeled hormones to αvβ3-positive ovarian cancer cells but not to integrin-negative cells. Physiologically relevant T3 (1 nM) and T4 (100 nM) concentrations in OVCAR-3 (high αvβ3) and A2780 (low αvβ3) cells promoted αv and β3 transcription in association with basal integrin levels. This transcription was effectively blocked by RGD (Arg-Gly-Asp) peptide and neutralizing αvβ3 antibodies, excluding T3-induced β3 messenger RNA, suggesting subspecialization of T3 and T4 binding to the integrin receptor pocket. We have provided support for extracellular regulated kinase (ERK)-mediated transcriptional regulation of the αv monomer by T3 and of β3 monomer by both hormones and documented a rapid (30-120 min) and dose-dependent (0.1-1000 nM) ERK activation. OVCAR-3 cells and αvβ3-deficient HEK293 cells treated with αvβ3 blockers confirmed the requirement for an intact thyroid hormone-integrin interaction in ERK activation. In addition, novel data indicated that T4, but not T3, controls integrin's outside-in signaling by phosphorylating tyrosine 759 in the β3 subunit. Both hormones induced cell proliferation (cell counts), survival (Annexin-PI), viability (WST-1) and significantly reduced the expression of genes that inhibit cell cycle (p21, p16), promote mitochondrial apoptosis (Nix, PUMA) and tumor suppression (GDF-15, IGFBP-6), particularly in cells with high integrin expression. At last, we have confirmed that hypothyroid environment attenuated ovarian cancer growth using a novel experimental platform that exploited paired euthyroid and severe hypothyroid serum samples from human subjects. To conclude, our data define a critical role for thyroid hormones as potent αvβ3-ligands, driving ovarian cancer cell proliferation and suggest that disruption of this axis may present a novel treatment strategy in this aggressive disease.
Collapse
Affiliation(s)
- E Shinderman-Maman
- Translational Hemato-Oncology Laboratory, The Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - K Cohen
- Translational Hemato-Oncology Laboratory, The Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - C Weingarten
- Translational Hemato-Oncology Laboratory, The Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - D Nabriski
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Endocrinology, Meir Medical Center, Kfar-Saba, Israel
| | - O Twito
- Department of Endocrinology, Meir Medical Center, Kfar-Saba, Israel
| | - L Baraf
- Department of Endocrinology, Meir Medical Center, Kfar-Saba, Israel
| | - A Hercbergs
- Radiation Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - P J Davis
- Department of Medicine, Albany Medical College, Albany, NY, USA
| | - H Werner
- Department of Human Molecular Genetics and Biochemistry.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - M Ellis
- Translational Hemato-Oncology Laboratory, The Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - O Ashur-Fabian
- Translational Hemato-Oncology Laboratory, The Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
19
|
Berghauser Pont LM, Kleijn A, Kloezeman JJ, van den Bossche W, Kaufmann JK, de Vrij J, Leenstra S, Dirven CM, Lamfers ML. The HDAC Inhibitors Scriptaid and LBH589 Combined with the Oncolytic Virus Delta24-RGD Exert Enhanced Anti-Tumor Efficacy in Patient-Derived Glioblastoma Cells. PLoS One 2015; 10:e0127058. [PMID: 25993039 PMCID: PMC4436250 DOI: 10.1371/journal.pone.0127058] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 04/10/2015] [Indexed: 01/12/2023] Open
Abstract
Background A phase I/II trial for glioblastoma with the oncolytic adenovirus Delta24-RGD was recently completed. Delta24-RGD conditionally replicates in cells with a disrupted retinoblastoma-pathway and enters cells via αvβ3/5 integrins. Glioblastomas are differentially sensitive to Delta24-RGD. HDAC inhibitors (HDACi) affect integrins and share common cell death pathways with Delta24-RGD. We studied the combination treatment effects of HDACi and Delta24-RGD in patient-derived glioblastoma stem-like cells (GSC), and we determined the most effective HDACi. Methods SAHA, Valproic Acid, Scriptaid, MS275 and LBH589 were combined with Delta24-RGD in fourteen distinct GSCs. Synergy was determined by Chou Talalay method. Viral infection and replication were assessed using luciferase and GFP encoding vectors and hexon-titration assays. Coxsackie adenovirus receptor and αvβ3 integrin levels were determined by flow cytometry. Oncolysis and mechanisms of cell death were studied by viability, caspase-3/7, LDH and LC3B/p62, phospho-p70S6K. Toxicity was studied on normal human astrocytes. MGMT promotor methylation status, TCGA classification, Rb-pathway and integrin gene expression levels were assessed as markers of responsiveness. Results Scriptaid and LBH589 acted synergistically with Delta24-RGD in approximately 50% of the GSCs. Both drugs moderately increased αvβ3 integrin levels and viral infection in responding but not in non-responding GSCs. LBH589 moderately increased late viral gene expression, however, virus titration revealed diminished viral progeny production by both HDACi, Scriptaid augmented caspase-3/7 activity, LC3B conversion, p62 and phospho-p70S6K consumption, as well as LDH levels. LBH589 increased LDH and phospho-p70S6K consumption. Responsiveness correlated with expression of various Rb-pathway genes and integrins. Combination treatments induced limited toxicity to human astrocytes. Conclusion LBH589 and Scriptaid combined with Delta24-RGD revealed synergistic anti-tumor activity in a subset of GSCs. Both HDACi moderately augmented viral infection and late gene expression, but slightly reduced progeny production. The drugs differentially activated multiple cell death pathways. The limited toxicity on astrocytes supports further evaluation of the proposed combination therapies.
Collapse
Affiliation(s)
| | - Anne Kleijn
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Rotterdam, The Netherlands
| | - Jenneke J. Kloezeman
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Rotterdam, The Netherlands
| | | | - Johanna K. Kaufmann
- Department of Neurosurgery, Harvey Cushing Neuro-Oncology Laboratories, Brigham & Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jeroen de Vrij
- Department of Neurosurgery, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Sieger Leenstra
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Rotterdam, The Netherlands
- Department of Neurosurgery, Elisabeth Hospital, Tilburg, The Netherlands
| | - Clemens M.F. Dirven
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Rotterdam, The Netherlands
| | - Martine L.M. Lamfers
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
20
|
Xu XH, Huang XW, Qun L, Li YN, Wang Y, Liu C, Ma Y, Liu QM, Sun K, Qian F, Jin L, Wang J. Two functional loci in the promoter of EPAS1 gene involved in high-altitude adaptation of Tibetans. Sci Rep 2014; 4:7465. [PMID: 25501874 PMCID: PMC4264014 DOI: 10.1038/srep07465] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/24/2014] [Indexed: 12/21/2022] Open
Abstract
EPAS1 involves in the hypoxic response and is suggested to be responsible for the genetic adaptation of high-altitude hypoxia in Tibetans. However, the detailed molecular mechanism remains unknown. In this study, a single nucleotide polymorphism rs56721780:G>C and an insertion/deletion (indel) polymorphism -742 indel in the promoter region showed divergence between Tibetans and non-Tibetan lowlanders. rs56721780:G>C regulated the transcription of EPAS1 by IKAROS family zinc finger 1 (IKZF1), which was identified as a new transcriptional repressor for EPAS1 gene. It demonstrated that the C allele of rs56721780:G>C decreased the binding of IKZF1, leading to the attenuated transcriptional repression of EPAS1 gene. The insertion at -742 indel provided a new binding site for Sp1 and was related to the activation of EPAS1 promoter. Further functional analysis revealed that lysyl oxidase (LOX) gene, which was reported to be responsible for extracellular matrix protein cross-linking of amnion previously, was a direct target of EPAS1. The CC genotype at rs56721780:G>C and the insertion genotype at -742 indel were found associated with higher EPAS1 and LOX expression levels in amnion, as well as higher birth weight of Tibetan newborns, suggesting that EPAS1 gene might play important roles in the development of amnion, fetus growth and high-altitude adaptation of Tibetans.
Collapse
Affiliation(s)
- Xiang-Hong Xu
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai 200438, P.R. China
| | - Xue-Wen Huang
- 1] Clinical laboratory of Huadong Sanatorium, Dajishan, Meiyuan Garden, Wuxi, Jiangsu 214065, P.R. China [2] Public Health Bureau for Shigatse District, 5 Keji Road, Shigatse District, Tibet 857000, P.R. China
| | - Li Qun
- Department of Gynecology and Obstetrics, The People's Hospital of Shigatse District, 28 Shanghai Middle Road, Shigatse District, Tibet 857000, P.R. China
| | - Ya-Nan Li
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai 200438, P.R. China
| | - Yi Wang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai 200438, P.R. China
| | - Chao Liu
- School of Life Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai 200438, P.R. China
| | - Yanyun Ma
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai 200438, P.R. China
| | - Qing-Mei Liu
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai 200438, P.R. China
| | - Kang Sun
- 1] School of Life Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai 200438, P.R. China [2] Center for Reproductive Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200135, P.R. China
| | - Feng Qian
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai 200438, P.R. China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai 200438, P.R. China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai 200438, P.R. China
| |
Collapse
|
21
|
Wang R, Wu XZ. Roles of galactose 3′-O- sulfation in signaling. Glycoconj J 2014; 31:549-54. [DOI: 10.1007/s10719-014-9558-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/28/2014] [Accepted: 09/05/2014] [Indexed: 01/23/2023]
|
22
|
Dong YW, Wang R, Cai QQ, Qi B, Wu W, Zhang YH, Wu XZ. Sulfatide epigenetically regulates miR-223 and promotes the migration of human hepatocellular carcinoma cells. J Hepatol 2014; 60:792-801. [PMID: 24333181 DOI: 10.1016/j.jhep.2013.12.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 11/22/2013] [Accepted: 12/02/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS The biological relevance and regulation mechanism of aberrant miR-223 expression in human hepatocellular carcinoma (HCC) remain unknown. Our aim was to investigate miR-223 regulation in HCC. METHODS miR-223 and integrin αV dysregulation were verified in 57 HCC specimens. Immunohistochemical analysis of integrin αV and sulfatide levels was performed on another cohort of 103 HCC samples. Epigenetic analysis was used to explore the effect of sulfatide on miR-223 transcription. Orthotopic growth, and intrahepatic and pulmonary metastasis of tumors derived from SMMC-7721 cells expressing miR-223 or cerebroside sulfotransferase were monitored in mice. RESULTS miR-223 was reduced in HCC specimens and highly metastatic cell lines. Enhanced miR-223 expression had a negative effect on integrin αV-mediated cell migration. In vivo assays of metastasis in an orthotopically implanted model demonstrated that miR-223 effectively inhibited HCC metastasis. Further analysis demonstrated that integrin αV is negatively regulated by miR-223. Moreover, the integrin αV subunit was significantly positively correlated with highly expressed sulfatide in 103 HCC specimens. Intriguingly, miR-223 expression was suppressed by sulfatide in HCC in association with reduced recruitment of acetylated histone H3 and C/EBPα to the pre-miR-223 gene promoter, where monocytic leukemia zinc finger (MOZ) protein, a MYST-type histone acetyltransferase, lost its attachment. The expression of histone deacetylases, HDAC9 and HDAC10, were greatly stimulated by sulfatide and their recruitment to miR-223 gene promoter was enhanced. CONCLUSIONS Downregulation of miR-223 in HCC is associated with the epigenetic regulation by highly expressed sulfatide and involved in tumor metastasis.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Cell Movement
- Down-Regulation
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Heterografts
- Humans
- Integrin alphaV/genetics
- Integrin alphaV/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Mice
- Mice, Nude
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Neoplasm Metastasis
- Promoter Regions, Genetic
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Sulfoglycosphingolipids/metabolism
Collapse
Affiliation(s)
- Yi Wei Dong
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Rong Wang
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Qian Qian Cai
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Bing Qi
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Wei Wu
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China
| | - Yong Hu Zhang
- People's Hospital of Beilun District, Ningbo, Zhejiang, China
| | - Xing Zhong Wu
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Key Lab of Glycoconjugate Research, Ministry of Public Health, Shanghai, China.
| |
Collapse
|
23
|
Lin X, Shang X, Manorek G, Fofana M, Stephen B H. Integrin αV modulates the cellular pharmacology of copper and cisplatin by regulating expression of the influx transporter CTR1. Oncoscience 2014; 1:185-195. [PMID: 25594011 PMCID: PMC4278295 DOI: 10.18632/oncoscience.22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 03/23/2014] [Indexed: 01/07/2023] Open
Abstract
The αV integrin is expressed in most cancer cells where it regulates a diverse array of cellular functions essential to the initiation, progression and metastasis of solid tumors. However, little is known about how αV integrin modulates cellular sensitivity to chemotherapeutic agents, particularly the platinum drugs. In this study, we found that down-regulation of αV sensitized human M21 cells to cisplatin (cDDP) through up-regulation of the copper influx transporter CTR1. Cells selected for low αV integrin expression (M21L) were more sensitive to cDDP, accompanied by increase in CTR1 mRNA and CTR1 protein levels, more intracellular cDDP accumulation and cDDP DNA adduct formation. Basal copper (Cu) content, Cu uptake, and Cu cytotoxicity were also increased. Transfection of a luciferase reporter construct containing the hCTR1 promoter sequence revealed an increase of the hCTR1 transcription activity in M21L cells. The basis for the increased hCTR1 transcription was related to an increase in the steady-state level of Sp1, a transcription factor known to drive hCTR1 expression. These results indicate that the αV integrin modulates sensitivity of human cells to the cytotoxic effect of cDDP by regulating expression of the Cu transporter CTR1, and introduce the concept that αV expression is linked to Cu homeostasis.
Collapse
Affiliation(s)
- Xinjian Lin
- Department of Medicine and UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Xiying Shang
- Department of Medicine and UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Gerald Manorek
- Department of Medicine and UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Mariama Fofana
- Department of Medicine and UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Howell Stephen B
- Department of Medicine and UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA
| |
Collapse
|