1
|
Zhou W, Ruan H, Zhu L, Chen S, Yang M. Unveiling a Novel Glioblastoma Deep Molecular Profiling: Insight into the Cancer Cell Differentiation-Related Mechanisms. ACS OMEGA 2025; 10:10230-10250. [PMID: 40124014 PMCID: PMC11923693 DOI: 10.1021/acsomega.4c09586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/27/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND the sophisticated cellular heterogeneity of cell populations in glioblastoma (GBM) has been a key factor influencing tumor progression and response to therapy. The lack of more precise stratification based on cellular differentiation status poses a great challenge to therapeutic strategies. MATERIALS AND METHODS harnessing the bulk multiomics and single-nucleus RNA sequencing data available from the National Center for Biotechnology Information (NCBI) and The Cancer Genome Atlas (TCGA) Program repositories, we developed a novel and accurate GBM risk classification using an ensemble consensus clustering approach based on the junction of prognosis and trajectory analysis. Comprehensive cluster labeling and multiomics data characterization were also performed. RESULTS a novel GBM stratification model was constructed using 45 malignant cell fate genes: (a) energy metabolism-enhanced-type GBM; (b) invasion-enhanced-type GBM; (c) invasion-attenuated-type GBM; and (d) glycolysis-dominant energy metabolism-enhanced-type GBM. The biological plausibility of the model was verified through a range of comprehensive analyses of multiomics data, showing that cases with invasion-attenuated-type were the best prognosis and energy metabolism-enhanced-type the poorest. CONCLUSIONS the study has uncovered GBM complex cellular heterogeneity and a differentiated hierarchy of cell populations underlying tumorigenesis. This precise stratification system provided implications for further studies of individual therapies.
Collapse
Affiliation(s)
- Weili Zhou
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Hongtao Ruan
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Lihua Zhu
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Shunqiang Chen
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Muyi Yang
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| |
Collapse
|
2
|
Opila J, Krzysiek-Maczka G. Direct tool for quantitative analysis of cell/object dynamic behavior - metastasis and far beyond. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 229:107245. [PMID: 36455469 DOI: 10.1016/j.cmpb.2022.107245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/17/2022] [Accepted: 11/13/2022] [Indexed: 06/17/2023]
Abstract
INTRODUCTION The dynamics and depth of invasion as well as the ability of cancer cells to penetrate the walls of lymphatic or blood vessels represent critical survival-influencing factors in cancer patients. Depending on the cell type and tissue environment, cancer cell invasion differ in terms of motility mechanism and migration modes. Thus, there is the need of effective models allowing not only for single cell invasion potential assessment but also for collective migration and expansive growth evaluation in 3D microenvironment e.g. basement membranes. To meet this task, the specimens should be compared and analyzed in terms of the dynamics of movement and the evolution of the shape. OBJECTIVES Our main objective was development of the mathematical method that enables fast and credible calculation of parameters of shape and position, namely standard deviations (σX, σY), centroid position (μX, μY) and correlation coefficient ρ, based only on the contour of the aggregate. METHODS In order to accomplish this goal we measured geometrical properties of aggregates of RGM1 cells seeded in 3D Geltrex basement membrane. Referential microscopic images were taken 24 and 48 h after seeding and cell group dynamics was registered over 8 h periods using time lapse microscopy. RESULTS Based on gathered data, we managed to develop and fully test universal numerical tool allowing for estimation of statistical parameters of cell groups and aggregates which then allows for the precise evaluation of their behavior within microenvironment with time. CONCLUSION We conclude, that our tool is suitable for any research on the metastatic potential and motility of cancer cells in a given microenvironment, regardless of the migration mechanism, which together with the advanced analysis like cell single-cell transcriptomic, proteomic, and chromatin accessibility data may allow to identify precise targets for anti-cancer therapies, to predict the degree of malignancy of neoplastic lesions as well as it can be useful during architecting therapeutic strategies. Moreover, the developed tool seems to be broadly applicable for assessment of behavioural dynamics of any population.
Collapse
Affiliation(s)
- Janusz Opila
- Department of Applied Computer Sciences, The Faculty of Management, AGH University of Science and Technology, Cracow 30-059, Poland.
| | - Gracjana Krzysiek-Maczka
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, Cracow 31-531, Poland.
| |
Collapse
|
3
|
Abstract
Extracellular vesicles (EVs) refer to vesicles that are released by cells into the extracellular space. EVs mediate cell-to-cell communication via delivery of functional biomolecules between host and recipient cells. EVs can be categorised based on their mode of biogenesis and secretion and include apoptotic bodies, ectosomes or shedding microvesicles and exosomes among others. EVs have gained immense interest in recent years owing to their implications in pathophysiological conditions. Indeed, EVs have been proven useful in clinical applications as potential drug delivery vehicles and as source of diagnostic biomarkers. Despite the growing body of evidence supporting the clinical benefits, the processes involved in the biogenesis of EVs are poorly understood. Hence, it is critical to gain a deeper understanding of the underlying molecular machineries that ultimately govern the biogenesis and secretion of EVs. This chapter discusses the current knowledge on molecular mechanisms involved in the biogenesis of various subtypes of EVs.
Collapse
Affiliation(s)
- Taeyoung Kang
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Ishara Atukorala
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Suresh Mathivanan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
Ahn SB, Kamath KS, Mohamedali A, Noor Z, Wu JX, Pascovici D, Adhikari S, Cheruku HR, Guillemin GJ, McKay MJ, Nice EC, Baker MS. Use of a Recombinant Biomarker Protein DDA Library Increases DIA Coverage of Low Abundance Plasma Proteins. J Proteome Res 2021; 20:2374-2389. [PMID: 33752330 DOI: 10.1021/acs.jproteome.0c00898] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Credible detection and quantification of low abundance proteins from human blood plasma is a major challenge in precision medicine biomarker discovery when using mass spectrometry (MS). In this proof-of-concept study, we employed a mixture of selected recombinant proteins in DDA libraries to subsequently identify (not quantify) cancer-associated low abundance plasma proteins using SWATH/DIA. The exemplar DDA recombinant protein spectral library (rPSL) was derived from tryptic digestion of 36 recombinant human proteins that had been previously implicated as possible cancer biomarkers from both our own and other studies. The rPSL was then used to identify proteins from nondepleted colorectal cancer (CRC) EDTA plasmas by SWATH-MS. Most (32/36) of the proteins used in the rPSL were reliably identified from CRC plasma samples, including 8 proteins (i.e., BTC, CXCL10, IL1B, IL6, ITGB6, TGFα, TNF, TP53) not previously detected using high-stringency protein inference MS according to PeptideAtlas. The rPSL SWATH-MS protocol was compared to DDA-MS using MARS-depleted and postdigestion peptide fractionated plasmas (here referred to as a human plasma DDA library). Of the 32 proteins identified using rPSL SWATH, only 12 could be identified using DDA-MS. The 20 additional proteins exclusively identified using the rPSL SWATH approach were almost exclusively lower abundance (i.e., <10 ng/mL) proteins. To mitigate justified FDR concerns, and to replicate a more typical library creation approach, the DDA rPSL library was merged with a human plasma DDA library and SWATH identification repeated using such a merged library. The majority (33/36) of the low abundance plasma proteins added from the rPSL were still able to be identified using such a merged library when high-stringency HPP Guidelines v3.0 protein inference criteria were applied to our data set. The MS data set has been deposited to ProteomeXchange Consortium via the PRIDE partner repository (PXD022361).
Collapse
Affiliation(s)
- Seong Beom Ahn
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Karthik S Kamath
- Australian Proteome Analysis Facility (APAF), Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Abidali Mohamedali
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Zainab Noor
- ProCan, Children's Medical Research Institute, The University of Sydney, Westmead, Newtown, NSW 2042, Australia
| | - Jemma X Wu
- Australian Proteome Analysis Facility (APAF), Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Dana Pascovici
- Australian Proteome Analysis Facility (APAF), Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Subash Adhikari
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Harish R Cheruku
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Gilles J Guillemin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Matthew J McKay
- Australian Proteome Analysis Facility (APAF), Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Mark S Baker
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| |
Collapse
|
5
|
Yu K, Tang J, Wu JL, Li B, Wu SN, Zhang MY, Li QY, Zhang LJ, Pan YC, Ge QM, Shu HY, Shao Y. Risk factors for intraocular metastasis of primary liver cancer in diabetic patients: Alpha-fetoprotein and cancer antigen 125. World J Diabetes 2021; 12:158-169. [PMID: 33594335 PMCID: PMC7839172 DOI: 10.4239/wjd.v12.i2.158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/26/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND In rare instances, primary liver cancer can be associated with intraocular metastasis (IOM).
AIM To investigate the correlation between a diverse range of clinical characteristics and IOM in diabetic patients with primary liver cancer, and to determine potential risk factors in predicting IOM.
METHODS We recruited a total of 722 diabetic patients with primary liver cancer. The differences between the IOM and non-intraocular metastasis (NIOM) groups in these patients were assessed using the chi-squared test and Student’s t-test. Binary logistic regression analysis was subsequently used to determine risk factors. Finally, the diagnostic value of IOM in this cohort with primary liver cancer was analyzed by receiver operating characteristic (ROC) curve analysis.
RESULTS In all, 13 patients had IOM. There were no remarkable intergroup differences with respect to age, sex, histopathological sub-types, or blood biochemical parameters. However, the IOM group had significantly higher alpha-fetoprotein (AFP) and cancer antigen 125 (CA125) values than the NIOM group. Binary logistic regression identified AFP and CA125 to be significant risk factors for IOM in diabetic patients with primary liver cancer. ROC curve analysis showed that the area under the curve values for AFP and CA125 were 0.727 and 0.796, with the cut-off values of 994.20 ng/mL and 120.23 U/mL, respectively. The sensitivity and specificity for AFP were 92.3% and 59.9%, while those for CA125 were 84.6% and 70.1%, respectively.
CONCLUSION Elevated AFP and CA125 represent significant risk factors for IOM in diabetic patients with primary liver cancer.
Collapse
Affiliation(s)
- Kang Yu
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Jing Tang
- Department of Oncology, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou 412000, Hunan Province, China
| | - Jie-Li Wu
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Xiamen 361102, Fujian Province, China
| | - Biao Li
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Shi-Nan Wu
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Meng-Yao Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Qiu-Yu Li
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Li-Juan Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yi-Cong Pan
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Qian-Min Ge
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Hui-Ye Shu
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yi Shao
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
6
|
Lou X, Meng Y, Hou Y. A literature review on function and regulation mechanism of DKK4. J Cell Mol Med 2021; 25:2786-2794. [PMID: 33586359 PMCID: PMC7957263 DOI: 10.1111/jcmm.16372] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 12/16/2022] Open
Abstract
Dickkopf-related protein 4 (DKK4) is a member of the dickkopf family and an inhibitor of the Wnt/β-catenin signalling pathway. This review surveyed the single nucleotide polymorphisms (SNPs), copy number variations (CNVs), hypermethylation, regulation mechanism, correlation with clinicopathological parameters and chemotherapeutic resistance of DKK4. The signal pathways involved in DKK4 mainly include Wnt/β-catenin pathway and Wnt-JNK pathway independent β-catenin. DKK4 expression was upregulated in Renal Cell Carcinoma (RCC), Colorectal Cancer, Gastric Cancer (GC), Non-small Cell Lung Cancer (NSCLC) and Epithelial Ovarian Cancer (EOC), while downregulated in Hepatocellular Carcinoma (HCC). DKK4 is not only involved in tumour growth, invasion, migration and chemotherapy resistance, but also in osteoblastogenesis and secondary hair or meibomian gland formation. DKK4 has also been linked to schizophrenia.
Collapse
Affiliation(s)
- Xiaoli Lou
- Department of Central Laboratory, Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuchen Meng
- Department of Central Laboratory, Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanqiang Hou
- Department of Central Laboratory, Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Yun JA, Kim J, Baek YY, Park W, Park M, Kim S, Kim T, Choi S, Jeoung D, Lee H, Won MH, Kim JY, Ha KS, Kwon YG, Kim YM. N-Terminal Modification of the Tetrapeptide Arg-Leu-Tyr-Glu, a Vascular Endothelial Growth Factor Receptor-2 (VEGFR-2) Antagonist, Improves Antitumor Activity by Increasing its Stability against Serum Peptidases. Mol Pharmacol 2019; 96:692-701. [PMID: 31594790 DOI: 10.1124/mol.119.117234] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/03/2019] [Indexed: 02/14/2025] Open
Abstract
The tetrapeptide Arg-Leu-Tyr-Glu (RLYE), a vascular endothelial growth factor (VEGF) receptor-2 antagonist, has been used previously either alone or in combination with chemotherapeutic drugs for treating colorectal cancer in a mouse model. We analyzed the half-life of the peptide and found that because of degradation by aminopeptidases B and N, it had a short half-life of 1.2 hours in the serum. Therefore, to increase the stability and potency of the peptide, we designed the modified peptide, N-terminally acetylated RLYE (Ac-RLYE), which had a strongly stabilized half-life of 8.8 hours in serum compared with the original parent peptide. The IC50 value of Ac-RLYE for VEGF-A-induced endothelial cell migration decreased to approximately 37.1 pM from 89.1 pM for the parent peptide. Using a mouse xenograft tumor model, we demonstrated that Ac-RLYE was more potent than RLYE in inhibiting tumor angiogenesis and growth, improving vascular integrity and normalization through enhanced endothelial cell junctions and pericyte coverage of the tumor vasculature, and impeding the infiltration of macrophages into tumor and their polarization to the M2 phenotype. Furthermore, combined treatment of Ac-RLYE and irinotecan exhibited synergistic effects on M1-like macrophage activation and apoptosis and growth inhibition of tumor cells. These findings provide evidence that the N-terminal acetylation augments the therapeutic effect of RLYE in solid tumors via inhibition of tumor angiogenesis, improvement of tumor vessel integrity and normalization, and enhancement of the livery and efficacy of the coadministered chemotherapeutic drugs. SIGNIFICANCE STATEMENT: The results of this study demonstrate that the N-terminal acetylation of the tetrapeptide RLYE (Ac-RLYE), a novel vascular endothelial growth factor receptor-2 (VEGFR-2) inhibitor, significantly improves its serum stability, antiangiogenic activity, and vascular normalizing potency, resulting in enhanced therapeutic effect on solid tumors. Furthermore, the combined treatment of Ac-RLYE with the chemotherapeutic drug, irinotecan, synergistically enhanced its antitumor efficacy by improving the perfusion and delivery of the drug into the tumors and stimulating the conversion of the tumor-associated macrophages to an immunostimulatory M1-like antitumor phenotype.
Collapse
Affiliation(s)
- Jung-A Yun
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Joohwan Kim
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Yi-Yong Baek
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Wonjin Park
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Minsik Park
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Suji Kim
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Taesam Kim
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Seunghwan Choi
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Dooil Jeoung
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Hansoo Lee
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Moo-Ho Won
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Ji-Yoon Kim
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Kwon-Soo Ha
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Young-Guen Kwon
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Young-Myeong Kim
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| |
Collapse
|
8
|
Wei L, Zhang X, Wang J, Ye Q, Zheng X, Peng Q, Zheng Y, Liu P, Zhang X, Li Z, Liu C, Yan Q, Li G, Ma J. Lactoferrin deficiency induces a pro-metastatic tumor microenvironment through recruiting myeloid-derived suppressor cells in mice. Oncogene 2019; 39:122-135. [PMID: 31462711 DOI: 10.1038/s41388-019-0970-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 03/03/2019] [Accepted: 05/12/2019] [Indexed: 12/17/2022]
Abstract
Lactoferrin, an innate immunity molecule, is involved in anti-inflammatory, anti-microbial, and anti-tumor activities. We previously reported that lactoferrin is downregulated in specimens of nasopharyngeal carcinoma and negatively associated with tumor progression and metastasis of patients with nasopharyngeal carcinoma. However, the relationship between lactoferrin and the pro-metastatic microenvironment has not been reported yet. Here, by using the lactoferrin knockout mouse, we found that lactoferrin deficiency facilitated melanoma cells metastasizing to lungs, through recruiting myeloid-derived suppressor cells (MDSCs) in the lungs. Mechanistic studies showed that in the lung microenvironment of the lactoferrin knockout mice, the TLR9 signaling was the most repressed signaling. Lactoferrin can induce MDSCs differentiation and apoptosis, as well as upregulate TLR9 expression. TLR9 agonist or lactoferrin treatment can rescue this phenotype in the tumor metastasis mouse model. Our results suggest a protective role of lactoferrin in cancer metastasis, along with a deficiency in certain components of the innate immune system, may lead to a pro-metastatic tumor microenvironment.
Collapse
Affiliation(s)
- Lingyu Wei
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Third Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis of Ministry of Health, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, China.,Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xuemei Zhang
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Third Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jia Wang
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Qiurong Ye
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Xiang Zheng
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Qiu Peng
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Ying Zheng
- Center for Medical Research, Second Xiangya Hospital, Central South University, Changsha, China
| | - Peishan Liu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Xiaoyue Zhang
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Zhengshuo Li
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Can Liu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Qun Yan
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Guiyuan Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Third Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis of Ministry of Health, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, China
| | - Jian Ma
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Third Xiangya Hospital, Central South University, Changsha, China. .,Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China. .,Key Laboratory of Carcinogenesis of Ministry of Health, Changsha, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, China. .,Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
| |
Collapse
|
9
|
Wang P, Gao XY, Yang SQ, Sun ZX, Dian LL, Qasim M, Phyo AT, Liang ZS, Sun YF. Jatrorrhizine inhibits colorectal carcinoma proliferation and metastasis through Wnt/β-catenin signaling pathway and epithelial-mesenchymal transition. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:2235-2247. [PMID: 31371920 PMCID: PMC6627180 DOI: 10.2147/dddt.s207315] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/21/2019] [Indexed: 01/29/2023]
Abstract
Purpose Jatrorrhizine (JAT) is a natural protoberberine alkaloid, possesses detoxification, bactericidal and hypoglycemic activities. However, its anti-cancer mechanism is not clear. This study aimed to investigate the mechanism of JAT through which inhibits colorectal cancer in HCT-116 and HT-29 cells. Methods MTT assay and colony formation assay were used to check the cell proliferation ability. Cell apoptosis and cell cycle were measured by Hoechst 33342 staining and flow cytometry, respectively. Cell migration and invasion were detected by scratch wound healing assay and trans-well assay, respectively. Further, expression of related proteins was examined via Western blotting and the in vivo anti-cancer effect of JAT was confirmed by nude mice xenograft model. Results The research showed that JAT inhibited the proliferation of HCT-116 and HT-29 cells with IC50 values of 6.75±0.29 μM and 5.29±0.13 μM, respectively, for 72 hrs. It has also showed a time dependently, cell cycle arrested in S phase, promoted cell apoptosis and suppressed cell migration and invasion. In addition, JAT inhibited Wnt signaling pathway by reducing β-catenin and increasing GSK-3β expressions. Increased expression of E-cadherin, while decreased N-cadherin, indicating that JAT treatment suppressed the process of cell epithelial–mesenchymal transition (EMT). In HCT-116 nude mice xenograft model, JAT inhibited tumor growth and metastasis, and induced apoptosis of tumor cells. Conclusion This study demonstrated that JAT efficiently inhibited colorectal cancer cells growth and metastasis, which provides a new point for clinical treatment of colorectal cancer.
Collapse
Affiliation(s)
- Pan Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Xiao-Yan Gao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Si-Qian Yang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Zhi-Xin Sun
- College of Life Sciences, Wenzhou Medical University, Wenzhou 325035, People's Republic of China.,Department of Life Sciences, Zaozhuang No.1 Middle School, Zaozhuang, 277100, People's Republic of China
| | - Lu-Lu Dian
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Muhammad Qasim
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China.,Institute of Sustainable Halophyte Utilization, University of Karachi, Karachi 75270, Pakistan
| | - Aung Thu Phyo
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China.,Department of Biotechnology, Mandalay Technological University, Mandalay 05072, Myanmar
| | - Zong-Suo Liang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Yan-Fang Sun
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| |
Collapse
|
10
|
Kim SK, Kim K, Ryu JW, Ryu TY, Lim JH, Oh JH, Min JK, Jung CR, Hamamoto R, Son MY, Kim DS, Cho HS. The novel prognostic marker, EHMT2, is involved in cell proliferation via HSPD1 regulation in breast cancer. Int J Oncol 2018; 54:65-76. [PMID: 30365075 PMCID: PMC6254934 DOI: 10.3892/ijo.2018.4608] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/16/2018] [Indexed: 12/12/2022] Open
Abstract
Molecular classifications of breast cancer (BRC), such as human epidermal growth factor receptor 2 (HER2), luminal A and luminal B, have been developed to reduce unnecessary treatment by dividing patients with BRC into low- and high-risk progression groups. However, these methods do not cover all of the pathological characteristics of BRC, and investigations into novel prognostic/therapeutic markers are thus continually required. In this study, we identified the overexpression of the histone methyltransferase, euchromatic histone-lysine N-methyltransferase 2 (EHMT2) in BRC samples (n=1,222) and normal samples (n=113) derived from the TCGA portal by performing a BRC tissue microarray. EHMT2 overexpression was clearly associated with a poor prognosis in multiple cohorts of patients with BRC (total, n=1,644). Furthermore, the knockdown of EHMT2 expression affected cell apoptosis via the downregulation and re-localization of heat shock protein family D (Hsp60) member 1 (HSPD1). In addition, a statistically significant positive correlation between EHMT2 and HSPD1 expression was revealed in the clinical cohorts. On the whole, the findings of this study may assist the development of novel therapeutic strategies and provide a prognostic marker (EHMT2) for patients with BRC.
Collapse
Affiliation(s)
- Seon-Kyu Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305‑333, Republic of Korea
| | - Kwangho Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305‑333, Republic of Korea
| | - Jea-Woon Ryu
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305‑333, Republic of Korea
| | - Tae-Young Ryu
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305‑333, Republic of Korea
| | - Jung Hwa Lim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305‑333, Republic of Korea
| | - Jung-Hwa Oh
- Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea
| | - Jeong-Ki Min
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305‑333, Republic of Korea
| | - Cho-Rok Jung
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305‑333, Republic of Korea
| | - Ryuji Hamamoto
- Division of Molecular Modification and Cancer Biology, National Cancer Center, Tokyo 104‑0045, Japan
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305‑333, Republic of Korea
| | - Dae-Soo Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305‑333, Republic of Korea
| | - Hyun-Soo Cho
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305‑333, Republic of Korea
| |
Collapse
|
11
|
Kim K, Son MY, Jung CR, Kim DS, Cho HS. EHMT2 is a metastasis regulator in breast cancer. Biochem Biophys Res Commun 2018; 496:758-762. [PMID: 29337058 DOI: 10.1016/j.bbrc.2018.01.074] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/12/2022]
Abstract
Various modes of epigenetic regulation of breast cancer proliferation and metastasis have been investigated, but epigenetic mechanisms involved in breast cancer metastasis remain elusive. Thus, in this study, EHMT2 (a histone methyltransferase) was determined to be significantly overexpressed in breast cancer tissues and in Oncomine data. In addition, knockdown of EHMT2 reduced cell migration/invasion and regulated the expression of EMT-related markers (E-cadherin, Claudin 1, and Vimentin). Furthermore, treatment with BIX-01294, a specific inhibitor of EHMT2, affected migration/invasion in MDA-MB-231 cells. Therefore, our findings demonstrate functions of EHMT2 in breast cancer metastasis and suggest that targeting EHMT2 may be an effective therapeutic strategy for preventing breast cancer metastasis.
Collapse
Affiliation(s)
- Kwangho Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea; Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 305-333, Republic of Korea
| | - Cho-Rok Jung
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea; Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 305-333, Republic of Korea
| | - Dae-Soo Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea; Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 305-333, Republic of Korea
| | - Hyun-Soo Cho
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-333, Republic of Korea; Department of Functional Genomics, Korea University of Science and Technology, Daejeon, 305-333, Republic of Korea.
| |
Collapse
|
12
|
Novel prognostic marker PRMT1 regulates cell growth via downregulation of CDKN1A in HCC. Oncotarget 2017; 8:115444-115455. [PMID: 29383172 PMCID: PMC5777784 DOI: 10.18632/oncotarget.23296] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 12/05/2017] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a major type of liver cancer caused by the hepatitis B and C viruses, alcohol and exposure to aflatoxin. For HCC treatment, anticancer drugs have been widely used, but drug resistance in advanced HCC is an important problem, resulting in a continuous need for novel therapeutic targets. Therefore, in this study, we established a screening pipeline based on RNA-seq to screen novel therapeutic/prognostic targets in HCC and identified PRMT1 (Protein Arginine Methyltransferase 1). In the prognostic analysis, the overexpression of PRMT1 was clearly associated with poor prognosis in a number of HCC patient cohorts. Moreover, after PRMT1 knockdown, HCC cell lines exhibited cell growth and spheroid formation suppression, an increase in Sub-G1 cells by FACS analysis, and enrichment of the cell cycle pathway via functional enrichment analysis. With these results, we demonstrated that PRMT1 could be a novel prognostic marker and therapeutic target for HCC therapy.
Collapse
|
13
|
Yao Q, An Y, Hou W, Cao YN, Yao MF, Ma NN, Hou L, Zhang H, Liu HJ, Zhang B. LRP6 promotes invasion and metastasis of colorectal cancer through cytoskeleton dynamics. Oncotarget 2017; 8:109632-109645. [PMID: 29312635 PMCID: PMC5752548 DOI: 10.18632/oncotarget.22759] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/28/2017] [Indexed: 12/14/2022] Open
Abstract
Low density lipoprotein (LDL) receptor-related protein-6 (LRP6) is an important co-receptor of Wnt pathway, which plays a predominant role in development and progression of colorectal cancer. Recently, dysregulation of LRP6 has proved to be involved in the progression of cancers, but its biological role and clinical significance in colorectal cancer remain unclear. In present study, we revealed that phosphorylation of LRP6 was aberrantly upregulated in colorectal carcinoma correlating with TNM or Dukes staging and worse prognosis. In addition, phosphorylated LRP6 was positively correlated with nuclear accumulation of β-catenin. Overexpression or activation of LRP6 could activate Wnt signaling and promote tumor cell migration in vitro. The activation of LRP6 could induce microtubule dynamics and actin remodeling, probably through regulation of microtubule-associated protein 1B (MAP1B), microtubule actin cross-linking factor 1 (MACF1) and Rho GTPase--RhoA and Rac1. The investigation suggests that LRP6 may be a potential prognostic marker and therapeutic target in the progression of colorectal cancers.
Collapse
Affiliation(s)
- Qian Yao
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yu An
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wei Hou
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ya-Nan Cao
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Meng-Fei Yao
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ning-Ning Ma
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Lin Hou
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Hong Zhang
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Hai-Jing Liu
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Bo Zhang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
14
|
Gao C, Xing X, He Z, Chen S, Wang S, Li Q, Guo P, Zhang H, Li H, Chen L, Wang Q, Zhao J, Xiao Y, Chen W, Li D. Hypermethylation of PGCP gene is associated with human bronchial epithelial cells immortalization. Gene 2017; 642:505-512. [PMID: 29196255 DOI: 10.1016/j.gene.2017.11.063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/30/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023]
Abstract
Cell immortalization is the initial step for cancer development. To identify the differentially expressed genes regulated by DNA methylation over the course of human primary bronchial epithelial cell (HPBECs) immortalization, an immortalized HBE cell line (HBETT) was generated via introduction of an SV40 LT and a catalytic subunit of human telomerase reverse transcriptase (hTERT) into the HPBECs. Microarrays of mRNA and DNA methylation were performed to compare the transcriptomes and DNA methylomes between these two types of cells. The results from the mRNA microarray revealed many genes whose expression changed upon cell immortalization. We identified signatures including global hypomethylation, perturbation of ECM-receptor interaction, focal adhesion, and PI3K-Akt pathways associated with cell immortalization. Moreover, we revealed 155 differentiated methylation regions (DMRs) within the CpG islands (CGIs) of 42 genes and the perturbation of several key pathways that might be involved in HBE cell immortalization. Among these genes, the hypermethylation of the plasma glutamate carboxypeptidase (PGCP) gene appeared specifically in lung cancer tissues. The inhibition of PGCP expression by promoter hypermethylation was observed in both immortal HBETT cells and benzo[a]pyrene (Bap)-transformed HBE cells. In conclusion, these findings provide new insight into the epigenetic modifications that are critical in the transition and maintenance of cell immortalization.
Collapse
Affiliation(s)
- Chen Gao
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Xiumei Xing
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zhini He
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Shen Chen
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Shan Wang
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Qingye Li
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ping Guo
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Haiyan Zhang
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Huiyao Li
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Liping Chen
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Qing Wang
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jian Zhao
- Department of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Yongmei Xiao
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Wen Chen
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Daochuan Li
- Department of Toxicology, Guangzhou Key Laboratory of Environmental Pollution and Health Risk Assessment, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|