1
|
Song D, Pan S, Jin W, Wu R, Zhao T, Jiang J, Zhu M. Minoxidil delivered via a stem cell membrane delivery controlled release system promotes hair growth in C57BL/6J mice. Front Bioeng Biotechnol 2024; 11:1331754. [PMID: 38260729 PMCID: PMC10800965 DOI: 10.3389/fbioe.2023.1331754] [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: 11/01/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Objective: Umbilical cord-derived mesenchymal stem cell membrane-loaded minoxidil (MXD) nanoparticles (STCM-MXD-NPs) were prepared to investigate their effects on hair growth in C57BL/6J mice. Methods: STCM-MXD-NPs were obtained by freeze-thawing and differential centrifugation, and their effects on hair growth were evaluated using C57BL/6J mice. The mRNA and protein expression levels of vascular endothelial growth factor (VEGF) and insulin-like growth factor-1 (IGF-1) were detected by real-time polymerase chain reaction and enzyme-linked immunosorbent assays, respectively. Protein expression levels of marker of proliferation Ki-67 (MKI67) and β-catenin (CTNNB) in skin tissue were detected by immunohistochemistry. Results: STCM-MXD-NPs improved MXD solubility. They released the drug slowly, increasing its transdermal properties, accumulation in the skin, and content in the hair bulb tissues with a better efficacy than that of ordinary MXD. Moreover, STCM-MXD-NPs significantly upregulated the mRNA and protein levels of VEGF and IGF-1 and promoted the protein expression of MKI67 and CTNNB in mouse skin tissues, promoting mouse hair growth. Conclusion: Stem cell membrane-loaded MXD nanoparticles with slow-release properties increased MXD accumulation in the skin by improving its transdermal properties, increasing VEGF, IGF-1, MKI67, and CTNNB expression levels and promoting hair growth in C57BL/6J mice.
Collapse
Affiliation(s)
- Dandan Song
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shouxi Pan
- Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Wenxia Jin
- Lanzhou Institute of Biological Products Co., Ltd., Lanzhou, China
| | - Ronghui Wu
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Tianqi Zhao
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mingji Zhu
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
Bai P, Fan T, Wang X, Zhao L, Zhong R, Sun G. Modulating MGMT expression through interfering with cell signaling pathways. Biochem Pharmacol 2023; 215:115726. [PMID: 37524206 DOI: 10.1016/j.bcp.2023.115726] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Guanine O6-alkylating agents are widely used as first-line chemotherapeutic drugs due to their ability to induce cytotoxic DNA damage. However, a major hurdle in their effectiveness is the emergence of chemoresistance, largely attributed to the DNA repair pathway mediated by O6-methylguanine-DNA methyltransferase (MGMT). MGMT plays an important role in removing the alkyl groups from lethal O6-alkylguanine (O6-AlkylG) adducts formed by chemotherapeutic alkylating agents. By doing so, MGMT enables tumor cells to evade apoptosis and develop drug resistance toward DNA alkylating agents. Although covalent inhibitors of MGMT, such as O6-benzylguanine (O6-BG) and O6-(4-bromothenyl)guanine (O6-4-BTG or lomeguatrib), have been explored in clinical settings, their utility is limited due to severe delayed hematological toxicity observed in most patients when combined with alkylating agents. Therefore, there is an urgent need to identify new targets and unravel the underlying molecular mechanisms and to develop alternative therapeutic strategies that can overcome MGMT-mediated tumor resistance. In this context, the regulation of MGMT expression via interfering the specific cell signaling pathways (e.g., Wnt/β-catenin, NF-κB, Hedgehog, PI3K/AKT/mTOR, JAK/STAT) emerges as a promising strategy for overcoming tumor resistance, and ultimately enhancing the efficacy of DNA alkylating agents in chemotherapy.
Collapse
Affiliation(s)
- Peiying Bai
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Tengjiao Fan
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China; Department of Medical Technology, Beijing Pharmaceutical University of Staff and Workers, Beijing 100079, China
| | - Xin Wang
- Department of Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
3
|
Dincel AS, Jørgensen NR. New Emerging Biomarkers for Bone Disease: Sclerostin and Dickkopf-1 (DKK1). Calcif Tissue Int 2023; 112:243-257. [PMID: 36165920 DOI: 10.1007/s00223-022-01020-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/26/2022] [Indexed: 01/25/2023]
Abstract
A healthy skeleton depends on a continuous renewal and maintenance of the bone tissue. The process of bone remodeling is highly controlled and consists of a fine-tuned balance between bone formation and bone resorption. Biochemical markers of bone turnover are already in use for monitoring diseases and treatment involving the skeletal system, but novel biomarkers reflecting specific biological processes in bone and interacting tissues may prove useful for diagnostic, prognostic, and monitoring purposes. The Wnt-signaling pathway is one of the most important pathways controlling bone metabolism and consequently the action of inhibitors of the pathway such as sclerostin and Dickkopf-related protein 1 (DKK1) have crucial roles in controlling bone formation and resorption. Thus, they might be potential markers for clinical use as they reflect a number of physiological and pathophysiological events in bone and in the cross-talk with other tissues in the human body. This review focuses on the clinical utility of measurements of circulating sclerostin and DKK1 levels based on preanalytical and analytical considerations and on evidence obtained from published clinical studies. While accumulating evidence points to clear associations with a number of disease states for the two markers, and thus, the potential for especially sclerostin as a biochemical marker that may be used clinically, the lack of standardization or harmonization of the assays still hampers the clinical utility of the markers.
Collapse
Affiliation(s)
- Aylin Sepinci Dincel
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, Ankara, Turkey
- Department of Clinical Biochemistry, Rigshospitalet, Valdemar Hansens Vej 13 Glostrup, 2600, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niklas Rye Jørgensen
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, Ankara, Turkey.
- Department of Clinical Biochemistry, Rigshospitalet, Valdemar Hansens Vej 13 Glostrup, 2600, Copenhagen, Denmark.
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
4
|
Kauerová T, Pérez-Pérez MJ, Kollar P. Salicylanilides and Their Anticancer Properties. Int J Mol Sci 2023; 24:ijms24021728. [PMID: 36675241 PMCID: PMC9861143 DOI: 10.3390/ijms24021728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Salicylanilides are pharmacologically active compounds with a wide spectrum of biological effects. Halogenated salicylanilides, which have been used for decades in human and veterinary medicine as anthelmintics, have recently emerged as candidates for drug repurposing in oncology. The most prominent example of salicylanilide anthelmintic, that is intensively studied for its potential anticancer properties, is niclosamide. Nevertheless, recent studies have discovered extensive anticancer potential in a number of other salicylanilides. This potential of their anticancer action is mediated most likely by diverse mechanisms of action such as uncoupling of oxidative phosphorylation, inhibition of protein tyrosine kinase epidermal growth factor receptor, modulation of different signaling pathways as Wnt/β-catenin, mTORC1, STAT3, NF-κB and Notch signaling pathways or induction of B-Raf V600E inhibition. Here we provide a comprehensive overview of the current knowledge about the proposed mechanisms of action of anticancer activity of salicylanilides based on preclinical in vitro and in vivo studies, or structural requirements for such an activity.
Collapse
Affiliation(s)
- Tereza Kauerová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackého tř. 1946/1, 612 42 Brno, Czech Republic
| | | | - Peter Kollar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackého tř. 1946/1, 612 42 Brno, Czech Republic
- Correspondence: ; Tel.: +420-541-562-892
| |
Collapse
|
5
|
Guo Q, Lai Y, Chu J, Chen X, Gao M, Sang C, Dong J, Pu J, Ma C. LRP6 Polymorphisms Is Associated With Sudden Cardiac Death in Patients With Chronic Heart Failure in the Chinese Han Population. Front Cardiovasc Med 2022; 8:815595. [PMID: 35187114 PMCID: PMC8854291 DOI: 10.3389/fcvm.2021.815595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 6 (LRP6) plays a critical role in cardiovascular homeostasis. The deficiency of LRP6 is associated with a high risk of arrhythmias. However, the association between genetic variations of LRP6 and sudden cardiac death (SCD) remains unknown. This study aims to explore the association between common variants of LRP6 and the prognosis of chronic heart failure (CHF) patients. From July 2005 to December 2009, patients with CHF were enrolled from 10 hospitals in China. The single-nucleotide polymorphism (SNP) rs2302684 was selected for the evaluation of the effect of LRP6 polymorphisms on the survival in patients with CHF. A total of 1,437 patients with CHF were finally included for the analysis. During a median follow-up of 61 months (range 0.4–129 months), a total of 546 (38.0%) patients died, including 201 (36.8%) cases with SCD and 345 (63.2%) cases with non-SCD. Patients carrying A allele of rs2302684 had an increased risk of all-cause death (adjusted HR 1.452, 95% CI 1.189–1.706; P < 0.001) and SCD (adjusted HR 1.783, 95% CI 1.337–2.378; P < 0.001). Therefore, the SNP rs2302684 T>A in LRP6 indicated higher risks of all-cause death and SCD in patients with CHF. LRP6 could be added as a novel predictor of SCD and might be a potential therapeutic target in the prevention of SCD in the CHF population.
Collapse
Affiliation(s)
- Qi Guo
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yiwei Lai
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jianmin Chu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuhua Chen
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingyang Gao
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Caihua Sang
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jianzeng Dong
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jielin Pu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Shanghai East Hospital, Tongji University, Shanghai, China
- *Correspondence: Jielin Pu
| | - Changsheng Ma
- Department of Cardiology, National Clinical Research Center for Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Changsheng Ma
| |
Collapse
|
6
|
Yao X, Yan Z, Wang X, Jiang H, Qian Y, Fan C. The influence of reduced graphene oxide on stem cells: a perspective in peripheral nerve regeneration. Regen Biomater 2021; 8:rbab032. [PMID: 34188955 PMCID: PMC8226110 DOI: 10.1093/rb/rbab032] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 05/13/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022] Open
Abstract
Graphene and its derivatives are fascinating materials for their extraordinary electrochemical and mechanical properties. In recent decades, many researchers explored their applications in tissue engineering and regenerative medicine. Reduced graphene oxide (rGO) possesses remarkable structural and functional resemblance to graphene, although some residual oxygen-containing groups and defects exist in the structure. Such structure holds great potential since the remnant-oxygenated groups can further be functionalized or modified. Moreover, oxygen-containing groups can improve the dispersion of rGO in organic or aqueous media. Therefore, it is preferable to utilize rGO in the production of composite materials. The rGO composite scaffolds provide favorable extracellular microenvironment and affect the cellular behavior of cultured cells in the peripheral nerve regeneration. On the one hand, rGO impacts on Schwann cells and neurons which are major components of peripheral nerves. On the other hand, rGO-incorporated composite scaffolds promote the neurogenic differentiation of several stem cells, including embryonic stem cells, mesenchymal stem cells, adipose-derived stem cells and neural stem cells. This review will briefly introduce the production and major properties of rGO, and its potential in modulating the cellular behaviors of specific stem cells. Finally, we present its emerging roles in the production of composite scaffolds for nerve tissue engineering.
Collapse
Affiliation(s)
- Xiangyun Yao
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, 600 Yishan Road, Shanghai 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Zhiwen Yan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, 600 Yishan Road, Shanghai 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Xu Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, 600 Yishan Road, Shanghai 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Huiquan Jiang
- College of Fisheries and Life Science, Shanghai Ocean University, 999 Metro loop Road Shanghai, China
| | - Yun Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, 600 Yishan Road, Shanghai 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, 600 Yishan Road, Shanghai 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
7
|
Xu C, Shi H, Jiang X, Fan Y, Huang D, Qi X, Cheng Q. ZEB1 Mediates Bone Marrow Mesenchymal Stem Cell Osteogenic Differentiation Partly via Wnt/β-Catenin Signaling. Front Mol Biosci 2021; 8:682728. [PMID: 34109218 PMCID: PMC8183571 DOI: 10.3389/fmolb.2021.682728] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/14/2021] [Indexed: 01/28/2023] Open
Abstract
Zinc finger E-box-binding homebox 1 (ZEB1) is a zinc-finger transcription factor best known for its role in promoting the epithelial-mesenchymal transition, which is also related to osteogenesis. Here, ZEB1 was investigated for its role in the commitment of bone marrow mesenchymal stem cells (BMSCs) to osteoblasts. In vitro, ZEB1 expression decreased following osteogenic differentiation. Furthermore, silencing of ZEB1 in BMSCs promoted osteogenic activity and mineralization. The increase in osteogenic differentiation induced by si-ZEB1 could be partly rescued by the inhibition of Wnt/β-catenin (si-β-catenin). In vivo, knockdown of ZEB1 in BMSCs inhibited the rapid bone loss of ovariectomized (OVX) mice. ZEB1 expression has also been negatively associated with bone mass and bone formation in postmenopausal women. In conclusion, ZEB1 is an essential transcription factor in BMSC differentiation and may serve as a potential anabolic strategy for treating and preventing postmenopausal osteoporosis (PMOP).
Collapse
Affiliation(s)
- Cuidi Xu
- Department of Osteoporosis and Bone Disease, Huadong Hospital Affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute, Shanghai, China
| | - Hongli Shi
- Department of Osteoporosis and Bone Disease, Huadong Hospital Affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute, Shanghai, China
| | - Xin Jiang
- Department of Osteoporosis and Bone Disease, Huadong Hospital Affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute, Shanghai, China
| | - Yongqian Fan
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Donghui Huang
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Xinming Qi
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qun Cheng
- Department of Osteoporosis and Bone Disease, Huadong Hospital Affiliated to Fudan University, Research Section of Geriatric Metabolic Bone Disease, Shanghai Geriatric Institute, Shanghai, China
| |
Collapse
|
8
|
Silencing Celsr2 inhibits the proliferation and migration of Schwann cells through suppressing the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2020; 533:623-630. [PMID: 32988580 DOI: 10.1016/j.bbrc.2020.09.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 01/26/2023]
Abstract
After a peripheral nerve injury, the remaining Schwann cells undergo proliferation and adopt a migratory phenotype to prepare for the regeneration of nerves. Celsr2 has been reported to play an important role in the development and maintenance of the function of the nervous system. However, the role and mechanism of Celsr2 during peripheral nerve regeneration remain unknown. Here, we showed that after sciatic nerve injury, Celsr2 mRNA and protein were significantly increased in nerve tissues. In addition, silencing Celsr2 decreased the ki67-positve portion and the migration distance of Schwann cells in vivo. In vitro, the results of MTT and EdU staining, transwell and wound healing assays indicated that Celsr2 siRNA-transfected primary Schwann cells showed significant decrease in proliferation and migration compared to that seen in negative control (NC)-transfected cells. Furthermore, we found that Wnt/β-catenin luciferase activity was reduced, as were the expression of β-catenin in the nucleus and the mRNA levels of its downstream genes Cyclin D1 and MMP-7 in Celsr2 siRNA-transfected primary Schwann cells. Further investigations showed that silencing Celsr2 inhibited the phosphorylation of GSK3β. Moreover, specific activators of the Wnt/β-catenin pathway, LiCl or mutant β-catenin (S33Y), partially reversed the inhibitory effect of Celsr2 siRNA. Taken together, our data indicated that silencing Celsr2 inhibited Schwann cells migration and proliferation through the suppressing Wnt/β-catenin pathway, providing a potential target for peripheral nerve regeneration.
Collapse
|
9
|
Newcastle disease virus mediated apoptosis and migration inhibition of human oral cancer cells: A probable role of β-catenin and matrix metalloproteinase-7. Sci Rep 2019; 9:10882. [PMID: 31350432 PMCID: PMC6659693 DOI: 10.1038/s41598-019-47244-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/09/2019] [Indexed: 12/19/2022] Open
Abstract
Cancer cell metastasis and its dissemination are most enigmatic and challenging aspects in the development of its therapeutics. Newcastle disease virus (NDV) is a well-studied avian paramyxovirus frequently isolated from birds and rarely from mammals. Since the first report of its oncolytic property, many NDV strains were studied for its effect in various cancer cells. In the present study, NDV strain Bareilly was characterized for its apoptotic potential and migration inhibition in human oral cancer cells. The NDV mediated apoptosis was confirmed by flow cytometry, DNA laddering, and immunoblotting. Moreover, NDV decreased the mitochondrial membrane potential suggesting an intrinsic pathway of apoptosis in oral cancer cells. NDV infection in oral cancer cells results in migration inhibition by a reduction in levels of MMP-7. MMP-7 is one of the key target genes of β-catenin. While overexpression of MMP-7 reversed the inhibitory effect of NDV mediated migration suggested its possible involvement. Wnt/β-catenin is an essential pathway for cell growth, differentiation, and metastasis. The involvement of the Wnt/β-catenin pathway in NDV infection has never been reported. Our results showed that NDV dysregulates Wnt/β-catenin by down-regulation of p-Akt and p-GSK3β leading to degradation of β-catenin. Furthermore, NDV infection leads to a reduction in cytoplasmic and nuclear levels of β-catenin. The study will provide us with a better insight into the molecular mechanism of NDV mediated oncolysis and the key cellular partners involved in the process.
Collapse
|
10
|
Niu J, Wang X, Liang C, Zhang YD, Liu FY, Li HY, Xie SQ, Sun H, Fang D. Suppression of epidermal growth factor receptor-mediated β-catenin nuclear accumulation enhances the anti-tumor activity of phosphoinositide 3-kinase inhibitor in breast cancer. Cell Biol Int 2019; 43:931-939. [PMID: 31124219 DOI: 10.1002/cbin.11183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/04/2019] [Accepted: 05/21/2019] [Indexed: 12/20/2022]
Abstract
Phosphoinositide 3-kinase (PI3K) signaling is frequently deregulated in breast cancer and plays a critical role in tumor progression. However, resistance to PI3K inhibitors in breast cancer has emerged, which is due to the enhanced β-catenin nuclear accumulation. Until now, the mechanisms underlying PI3K inhibition-induced β-catenin nuclear accumulation remains largely unknown. In the present study, we found inhibition of PI3K with LY294002 promoted β-catenin nuclear accumulation in MCF-7 and MDA-MB-231 breast cancer cells. Combining PI3K inhibitor LY294002 with XAV-939, an inhibitor against β-catenin nuclear accumulation, produced an additive anti-proliferation effect against breast cancer cells. Subsequent experiments suggested β-catenin nuclear accumulation induced by PI3K inhibition depended on the feedback activation of epidermal growth factor receptor (EGFR) signaling pathway in breast cancer cells. Inhibition of EGFR phosphorylation with Gefitinib enhanced anti-proliferation effect of PI3K inhibitor LY294002 in MCF-7 and MDA-MB-231 cells. Taken together, our findings may elucidate a possible mechanism explaining the poor outcome of PI3K inhibitors in breast cancer treatment.
Collapse
Affiliation(s)
- Jie Niu
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Xiao Wang
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Chao Liang
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Yi-Dan Zhang
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Fan-Ye Liu
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Hai-Ying Li
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Song-Qiang Xie
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China.,School of Pharmacy, Institute of Chemical Biology, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Hua Sun
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Dong Fang
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| |
Collapse
|
11
|
Zhang Y, Wang S, Wang C, Xiao J, Zhang S, Zhou H. High expression of FAM13A was associated with increasing the liver cirrhosis risk. Mol Genet Genomic Med 2019; 7:e543. [PMID: 30604588 PMCID: PMC6418360 DOI: 10.1002/mgg3.543] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/20/2018] [Accepted: 12/04/2018] [Indexed: 12/17/2022] Open
Abstract
Aim Liver cirrhosis is a consequence of chronic liver disease, and it may be caused by multiple influences of both genetic and environmental factors. Family with sequence similarity 13 member A (FAM13A) has been previously associated with lung function in several lung diseases, including chronic obstructive pulmonary disease, asthma, lung cancer, and pulmonary fibrosis. The aim of this study was to explore whether FAM13A polymorphisms confer susceptibility to liver cirrhosis. Methods FAM13A expression was evaluated in liver cirrhosis tissues by immunohistochemistry staining. The relationship between FAM13A gene polymorphism and liver cirrhosis was determined by association analysis. The genotypes were assessed in the Agena MassARRAY platform. Statistical analysis was performed using chi‐squared test/Fisher's exact test, genetic model analysis, and haplotype analysis. Results The results showed that the expression of FAM13A is obvious higher in the liver cirrhosis tissue cells than in the normal liver tissue cells. Moreover, association analysis results indicated that the minor allele “A” of rs3017895 was positively associated with high risk of liver cirrhosis in the allele model by the chi‐squared test (OR = 1.32, 95%CI = 1.03–1.68, p = 0.028). Logistic regression analyses revealed that the risk of liver cirrhosis was significantly higher in subjects with the G/A‐G/G genotype of rs3017895 than those with A/A genotype under the dominant model and log additive model, and the T/A‐A/A genotype of rs1059122 was positively associated with higher liver cirrhosis than T/T genotype based on dominant model respectively. In addition, haplotype analysis showed that the G‐A haplotype of rs3017895‐rs1059122 of the FAM13A gene significantly increased the risk of liver cirrhosis. Conclusion Our findings demonstrated that the high expression of FAM13A may be associated with an increased risk of liver cirrhosis.
Collapse
Affiliation(s)
- Yingai Zhang
- Institute of Tropical Agriculture and Forestry, Hainan University, Haikou, China.,Central Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Shunlan Wang
- Central Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Chan Wang
- Central Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Jingchuan Xiao
- Central Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Shufang Zhang
- Central Laboratory, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Hailong Zhou
- Institute of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| |
Collapse
|
12
|
Wei D, Yun LIS, Dejun X, Cong L, He JH, Yan L. Curcumin combining with si-MALAT1 inhibits the invasion and migration of colon cancer SW480 cells. BRAZ J PHARM SCI 2019. [DOI: 10.1590/s2175-97902019000118276] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Dai Wei
- Gangzhou People’s Hospital, China
| | | | | | - Liu Cong
- Gangzhou People’s Hospital, China
| | - Jin-Hua He
- Central Hospital of Panyu District, China
| | - Lin Yan
- Gangzhou People’s Hospital, China
| |
Collapse
|
13
|
Zhou J, Chen S, Guo H, Xia L, Liu H, Qin Y, He C. Electroacupuncture Prevents Ovariectomy-Induced Osteoporosis in Rats: A Randomised Controlled Trial. Acupunct Med 2018; 30:37-43. [PMID: 22378584 DOI: 10.1136/acupmed-2011-010114] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background Electroacupuncture (EA) treatment has been shown to increase bone mineral density (BMD) in ovariectomised (OVX) rats; however, the underlying mechanisms remain unclear. Objective To systematically evaluate the effects of EA on OVX rats and the Wnt/β-catenin signalling pathway. Methods Three-month-old female Sprague–Dawley rats were randomly divided into three different groups (n=10 each): sham operated control (sham operated), ovariectomy (OVX) and ovariectomy with EA treatment (OVX+EA). Rats in the OVX+EA group received 12-week EA treatments. Results Serum bone-specific alkaline phosphatase level (p<0.01), BMD of the proximal femoral metaphysis and the fifth lumbar (L5) vertebral body (both, p<0.05) and maximum load and energy to failure of L5 vertebral body (both p<0.01) were significantly higher in the OVX+EA group than in the OVX group. Trabecular area, trabecular width and trabecular number were significantly higher in the OVX+EA group by 66.9%, 29.2% and 30.3%, respectively, than in the OVX group (all, p<0.01). Trabecular separation was 31.9% lower in the OVX+EA group than in the OVX group (p<0.01). Quantitative real-time reverse transcription polymerised chain reaction indicated that the expressions of mRNAs for low-density lipoprotein receptor-related protein 5 and β-catenin were significantly increased in the OVX+EA group, as compared with the OVX group (p<0.01 and p<0.05, respectively). Conclusion This study demonstrates that EA can prevent OVX-induced bone loss and deterioration of bone architecture and strength by stimulating the Wnt/β-catenin signalling pathway. These findings suggest that EA may bet a promising adjunct method for inhibiting OVX-induced osteoporosis in clinical settings.
Collapse
Affiliation(s)
- Jun Zhou
- Rehabilitation Key Laboratory of Sichuan Province, Department of Rehabilitation, West China Hospital, Sichuan University, Sichuan, People's Republic of China
| | - Shiju Chen
- Rehabilitation Key Laboratory of Sichuan Province, Department of Rehabilitation, West China Hospital, Sichuan University, Sichuan, People's Republic of China
| | - Hua Guo
- Rehabilitation Key Laboratory of Sichuan Province, Department of Rehabilitation, West China Hospital, Sichuan University, Sichuan, People's Republic of China
| | - Lu Xia
- Rehabilitation Key Laboratory of Sichuan Province, Department of Rehabilitation, West China Hospital, Sichuan University, Sichuan, People's Republic of China
| | - Huifang Liu
- Rehabilitation Key Laboratory of Sichuan Province, Department of Rehabilitation, West China Hospital, Sichuan University, Sichuan, People's Republic of China
| | - Yuxi Qin
- Rehabilitation Key Laboratory of Sichuan Province, Department of Rehabilitation, West China Hospital, Sichuan University, Sichuan, People's Republic of China
| | - Chengqi He
- Rehabilitation Key Laboratory of Sichuan Province, Department of Rehabilitation, West China Hospital, Sichuan University, Sichuan, People's Republic of China
| |
Collapse
|
14
|
Liu Y, Yang T, Chen T, Hao J, Gai Y, Zhang W. (R)‑dehydroxyabscisic alcohol β‑D‑apiofuranosyl‑(1ˮ→6')‑β‑D‑glucopyranoside enhances the osteoblastic differentiation of ST2 cells via the BMP/WNT pathways. Mol Med Rep 2018; 19:461-467. [PMID: 30483786 PMCID: PMC6297791 DOI: 10.3892/mmr.2018.9690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 10/19/2018] [Indexed: 01/28/2023] Open
Abstract
Lonicera japonica has been used in traditional Chinese medicine as an important medicinal plant, with the ability to inhibit osteoclast development and bone loss. However, it is not clear which active ingredient exerts these effects. (R)-dehydroxyabscisic alcohol β-D-apiofuranosy l-(1ˮ→6’)-β-D-glucopyranoside (DAG) is an active constituent isolated from Lonicera japonica. In the present study, the ST2 bone marrow stromal cell line was treated by DAG at different concentrations and the osteoblastic differentiation was explored by ELISA assay, Von Kossa staining, Alizarin Red S staining, reverse transcription-quantitative polymerase chain reaction and western blot analysis. The results revealed that DAG promoted osteoblastic differentiation, as evidenced by increasing mineralization and alkaline phosphatase (ALP) activity, as well as the expression of genes encoding bone differentiation markers, including Alp, osteopontin (Opn) and osteocalcin (Ocn). In addition, DAG upregulated the gene expression of bone morphogenetic protein (Bmp)-2, Bmp4, Wnt family member (Wnt)-1, Wnt3 and runt-related transcription factor 2 (Runx2), as well as the protein expression of phosphorylated-mothers against decapentaplegic homolog (Smad) 1, Smad5 Smad8, β-catenin and Runx2 in ST2 cells. The osteogenic effects induced by DAG were attenuated by the BMP antagonist Noggin and the WNT signaling pathway inhibitor Dickkopf related protein-1. The data indicated that DAG promoted the osteoblastic differentiation of ST2 cells, at least partially through regulating the BMP/WNT signaling pathways. This provides scientific rationale for the development of DAG as a treatment for bone loss-associated diseases, such as osteoporosis.
Collapse
Affiliation(s)
- Yadong Liu
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Tao Yang
- Department of Joint Surgery and Sports Medicine, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Ting Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jun Hao
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yu Gai
- Department of Pathology, Dalian Municipal Central Hospital Affiliated to Dalian Medical University, Dalian, Liaoning 116033, P.R. China
| | - Weiguo Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
15
|
Yuan H, Wei R, Xiao Y, Song Y, Wang J, Yu H, Fang T, Xu W, Mao S. RHBDF1 regulates APC-mediated stimulation of the epithelial-to-mesenchymal transition and proliferation of colorectal cancer cells in part via the Wnt/β-catenin signalling pathway. Exp Cell Res 2018; 368:24-36. [PMID: 29654741 DOI: 10.1016/j.yexcr.2018.04.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/31/2022]
Abstract
The human rhomboid family-1 gene (RHBDF1) is an oncogene in breast and head and neck squamous cancers. Here, we show that RHBDF1 plays a significant role in colorectal cancer (CRC) formation and that the RHBDF1 expression level is higher in CRC than in corresponding normal tissues. Moreover, RHBDF1 promotes cell proliferation, invasion and migration in vitro. Furthermore, through overexpression and silencing of RHBDF1 and the mediator complex, our study demonstrates that RHBDF1 may positively regulate adenomatous polyposis coli (APC) in the Wnt/β-catenin signalling pathway to increase the expression levels of MMP-14 and Twist, which act as important epithelial-to-mesenchymal transition (EMT) stimulating factors. Additionally, RHBDF1 may regulate c-myc and CyclinD1 expression to influence cell proliferation. Finally, RHBDF1 overexpression and silencing influence CRC growth in BALB/c nude mice. In summary, our findings demonstrate that the regulatory effects of RHBDF1 on EMT and on cell proliferation are partially attributable to the Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Huiping Yuan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
| | - Ran Wei
- The First Clinical Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuhong Xiao
- The Second Clinical Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yi Song
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
| | - Jia Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
| | - Huihuan Yu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
| | - Ting Fang
- The Second Clinical Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Wei Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China.
| | - Shengxun Mao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China.
| |
Collapse
|
16
|
Ma Y, Zheng W, Chen H, Shao X, Lin P, Liu X, Li X, Ye H. Glucosamine promotes chondrocyte proliferation via the Wnt/β‑catenin signaling pathway. Int J Mol Med 2018; 42:61-70. [PMID: 29568900 PMCID: PMC5979785 DOI: 10.3892/ijmm.2018.3587] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 01/22/2018] [Indexed: 02/06/2023] Open
Abstract
The present study investigated the mechanism underlying the effects of glucosamine (GlcN) on the proliferation of chondrocytes isolated from the knee cartilage of Sprague-Dawley rats. Chondrocytes were treated with various concentrations of GlcN or without GlcN. The effects of GlcN on chondrocyte proliferation were determined using reverse transcription-polymerase chain reaction, western blot analysis and immunohistochemistry. The results indicated that GlcN significantly improved chondrocyte viability, accelerated G1/S transition during progression of the cell cycle and promoted the expression of cell cycle regulatory proteins, including cyclin D1, cyclin-dependent kinase (CDK)4 and CDK6, thus indicating that GlcN may promote chondrocyte proliferation. Furthermore, GlcN upregulated the expression levels of Wnt-4, Frizzled-2 and β-catenin, and downregulated the expression of glycogen synthase kinase-3. GlcN also promoted β-catenin translocation; β-catenin is able to activate numerous downstream target genes, including cyclin D1. To determine the role of the Wnt/β-catenin signaling pathway in chondrocyte proliferation, the Wnt/β-catenin signaling pathway was inhibited using Dickkopf-1 (DKK-1), after which chondrocytes were treated with GlcN. The results demonstrated that the expression levels of β-catenin and cyclin D1 were decreased in chondrocytes treated with DKK-1 and GlcN. These results suggested that GlcN may promote chondrocyte proliferation via the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yuhuan Ma
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Wenwei Zheng
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Houhuang Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xiang Shao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Pingdong Lin
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xianxiang Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xihai Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Hongzhi Ye
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
17
|
Song Z, Han X, Shen L, Zou H, Zhang B, Liu J, Gong A. PTEN silencing enhances neuronal proliferation and differentiation by activating PI3K/Akt/GSK3β pathway in vitro. Exp Cell Res 2018; 363:179-187. [PMID: 29305963 DOI: 10.1016/j.yexcr.2018.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/03/2017] [Accepted: 01/02/2018] [Indexed: 01/09/2023]
Abstract
The failure of neuronal proliferation and differentiation is a major obstacle for neural repair and regeneration after traumatic central nervous system (CNS) injury. PTEN acts as an intrinsic brake on the neuronal cells, but its roles and mechanism still remain to be clarified. Herein, for the first time we confirmed that PTEN had a dual effect on the neuronal cells in vitro. Firstly, we found that PTEN knockdown significantly promoted cell proliferation and differentiation. Then, PTEN knockdown activated PI3K/Akt and Wnt/β-catenin pathways in vitro. Further evidence revealed that GSK3β as a key node involved in PTEN controlling cell proliferation and differentiation in PC12 cells. In addition, we identified that PTEN-GSK3β pathway modulated neuronal proliferation via β-catenin. Taken together, these results suggest that PTEN silencing enhances neuronal proliferation and differentiation by activating PI3K/Akt/GSK3β pathway that it may be a promising therapeutic approach for CNS injury.
Collapse
Affiliation(s)
- Zhiwen Song
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xiu Han
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Liming Shen
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Hongjun Zou
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining 272000, China
| | - Jinbo Liu
- Department of Orthopedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| | - Aihua Gong
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
18
|
Han B, Meng X, Chen H, Chen L, Liu X, Wang H, Liu D, Gao F, Lin L, Ming J, Sun B, Yin S, Wang R, Wu P, Cai J, Jiang C. Epigenetic silencing of miR-338 facilitates glioblastoma progression by de-repressing the pyruvate kinase M2-β-catenin axis. Aging (Albany NY) 2017; 9:1885-1897. [PMID: 28858851 PMCID: PMC5611983 DOI: 10.18632/aging.101271] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 07/27/2017] [Indexed: 12/29/2022]
Abstract
Glioblastoma is the most malignant type of brain tumor, and its high invasiveness and multiplication severely shortens patients' overall survival. The embryonic pyruvate kinase M2 (PKM2) isoform is highly expressed in human cancer. We used computational target gene prediction, in vitro cell culture, immunoblotting, quantitative real-time PCR, ATP measurements, luciferase reporter assays, wound-healing assays, Transwell assays, RNA immunoprecipitation PCR, co-immunoprecipitation, flow cytometry and tumor xenografts to study the regulation of the PKM2/β-catenin axis in glioma. PKM2 was predicted to be a potential target of miR-338. MiR-338 was downregulated in high-grade gliomas due to hypermethylation of CpG islands in its promoter, and ectopic expression of miR-338 inhibited cell proliferation, invasion and ATP generation. MiR-338 inhibited PKM2 expression by binding to the 3' untranslated region of PKM2, which ultimately prevented binding of PKM2 to β-catenin and reduced T-cell factor/lymphoid enhancer factor reporter gene transcriptional activity. MiR-338 also inhibited PKM2 expression, attenuated glioma growth and prolonged survival in an animal model. These results confirm that miR-338, a tumor suppressor, suppresses the PKM2/β-catenin axis and is downregulated in glioblastoma. This provides a theoretical basis for glioma-targeting therapy.
Collapse
Affiliation(s)
- Bo Han
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Hui Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Lingchao Chen
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xing Liu
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
- Beijing Neurosurgical Institute, Beijing 100050, China
| | - Hongjun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Daming Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Fei Gao
- Department of Laboratory Diagnosis, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Lin Lin
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Jianguang Ming
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Bo Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Shi Yin
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Ruijia Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Pengfei Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin 150086, China
- Chinese Glioma Cooperative Group (CGCG), Beijing 100050, China
| |
Collapse
|
19
|
Long Noncoding RNA BC032913 as a Novel Therapeutic Target for Colorectal Cancer that Suppresses Metastasis by Upregulating TIMP3. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:469-481. [PMID: 28918047 PMCID: PMC5545770 DOI: 10.1016/j.omtn.2017.07.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 07/09/2017] [Accepted: 07/09/2017] [Indexed: 01/08/2023]
Abstract
Long noncoding RNAs (lncRNAs) have been shown to play critical roles in the biology of various cancers. However, their expression patterns and biological functions in human colorectal cancer (CRC) remain largely unknown. The aim of this study was to explore lncRNA profiles in CRC and investigate key lncRNAs involved in CRC tumorigenesis and progression. The microarray data of six CRC and matched non-cancerous tissues revealed distinct lncRNA profiles, including 899 upregulated and 1,646 downregulated lncRNAs (p < 0.05, fold change > 2.0). Furthermore, we found that the lncRNA BC032913 was generally underexpressed in 115 CRC samples compared with normal tissues. Reduced BC032913 levels were significantly associated with an advanced tumor, lymph nodes, distant metastasis (TNM) stage and a higher risk of lymph node and distant metastases. BC032913 downregulation indicated poor overall survival in CRC patients. Moreover, BC032913 enhanced the mRNA and protein expression of TIMP3 and inhibited Wnt/β-catenin pathway activity, thus suppressing CRC metastasis in vitro and in vivo. Collectively, the obtained data show that BC032913 plays an inhibitory role in CRC aggression by upregulating TIMP3, followed by inactivation of the Wnt/β-catenin pathway. Our findings indicate that the novel lncRNA BC032913 could serve as a novel prognostic marker and effective therapeutic target for CRC.
Collapse
|
20
|
Khalili S, Rasaee MJ, Bamdad T. 3D structure of DKK1 indicates its involvement in both canonical and non-canonical Wnt pathways. Mol Biol 2017. [DOI: 10.1134/s0026893317010095] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
21
|
Mulligan WA, Wegner KA, Keil KP, Mehta V, Taketo MM, Vezina CM. Beta-catenin and estrogen signaling collaborate to drive cyclin D1 expression in developing mouse prostate. Differentiation 2016; 93:66-71. [PMID: 27918915 DOI: 10.1016/j.diff.2016.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/31/2016] [Accepted: 11/07/2016] [Indexed: 12/17/2022]
Abstract
Androgen, beta-catenin (CTNNB1), and estrogen pathways stimulate proliferative growth of developing mouse prostate but how these pathways interact is not fully understood. We previously found that androgens induce CTNNB1 signaling in mouse urogenital sinus (UGS) epithelium from which prostatic ductal epithelium derives. Others have shown that low estradiol concentrations induce UGS epithelial proliferative growth. Here, we found that CTNNB1 signaling overlaps cyclin D1 (CCND1) expression in prostatic buds and we used a genetic approach to test whether CTNNB1 signaling induces CCND1 expression. We observed an unexpected sexually dimorphic response to hyperactive CCNTB1 signaling: in male mouse UGS it increased Ccnd1 mRNA abundance without increasing its protein abundance but in female UGS it increased Ccnd1 mRNA and protein abundance, suggesting a potential role for estrogens in stabilizing CCND1 protein. Treating wild type male UGS explants with androgen and either 17β-estradiol or a proteasome inhibitor increased CCND1 protein and KI67 labeling in prostatic bud epithelium. Together, our results are consistent with an epithelial proliferative growth mechanism linking CTNNB1-driven Ccnd1 transcription and estrogen-mediated CCND1 protein stabilization.
Collapse
Affiliation(s)
- William A Mulligan
- George M. O'Brien Benign Urology Center, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA; School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - Kyle A Wegner
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - Kimberly P Keil
- School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - Vatsal Mehta
- School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - M Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University Yoshida-Konoé-cho, Sakyo, Kyoto 606-8501, Japan
| | - Chad M Vezina
- George M. O'Brien Benign Urology Center, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA; School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA; Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA.
| |
Collapse
|
22
|
Masoumi KC, Daams R, Sime W, Siino V, Ke H, Levander F, Massoumi R. NLK-mediated phosphorylation of HDAC1 negatively regulates Wnt signaling. Mol Biol Cell 2016; 28:346-355. [PMID: 27903773 PMCID: PMC5231902 DOI: 10.1091/mbc.e16-07-0547] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/25/2016] [Accepted: 11/21/2016] [Indexed: 01/15/2023] Open
Abstract
Primary embryonic fibroblast cells isolated from NLK-deficient mice proliferate faster and have a shorter cell cycle than wild-type cells. Nemo-like kinase and HDAC1 together negatively regulate Wnt signaling via Tcf/Lef transcription repression and prevent aberrant proliferation of fibroblast cells. The Wnt signaling pathway is essential in regulating various cellular processes. Different mechanisms of inhibition for Wnt signaling have been proposed. Besides β-catenin degradation through the proteasome, nemo-like kinase (NLK) is another molecule that is known to negatively regulate Wnt signaling. However, the mechanism by which NLK mediates the inhibition of Wnt signaling was not known. In the present study, we used primary embryonic fibroblast cells isolated from NLK-deficient mice and showed that these cells proliferate faster and have a shorter cell cycle than wild-type cells. In NLK-knockout cells, we observed sustained interaction between Lef1 and β-catenin, leading to elevated luciferase reporter of β-catenin/Lef1–mediated transcriptional activation. The mechanism for the reduced β-catenin/Lef1 promoter activation was explained by phosphorylation of HDAC1 at serine 421 via NLK. The phosphorylation of HDAC1 was achieved only in the presence of wild-type NLK because a catalytically inactive mutant of NLK was unable to phosphorylate HDAC1 and reduced the luciferase reporter of β-catenin/Lef1–mediated transcriptional activation. This result suggests that NLK and HDAC1 together negatively regulate Wnt signaling, which is vital in preventing aberrant proliferation of nontransformed primary fibroblast cells.
Collapse
Affiliation(s)
| | - Renée Daams
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund 22381, Sweden
| | - Wondossen Sime
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund 22381, Sweden
| | - Valentina Siino
- Department of Immunotechnology, Lund University, Lund 22381, Sweden
| | - Hengning Ke
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund 22381, Sweden.,Cancer Research Institute, General Hospital, Ningxia Medical University, Yinchuan 750004, China
| | - Fredrik Levander
- Department of Immunotechnology, Lund University, Lund 22381, Sweden.,National Bioinformatics Infrastructure Sweden, Department of Immunotechnology, Lund University, Lund 22381, Sweden
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund 22381, Sweden
| |
Collapse
|
23
|
Silencing NKD2 by Promoter Region Hypermethylation Promotes Esophageal Cancer Progression by Activating Wnt Signaling. J Thorac Oncol 2016; 11:1912-1926. [DOI: 10.1016/j.jtho.2016.06.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 06/20/2016] [Accepted: 06/23/2016] [Indexed: 12/12/2022]
|
24
|
Hsieh CH, Hsu HH, Shibu MA, Day CH, Bau DT, Ho CC, Lin YM, Chen MC, Wang SH, Huang CY. Down-regulation of β-catenin and the associated migration ability by Taiwanin C in arecoline and 4-NQO-induced oral cancer cells via GSK-3β activation. Mol Carcinog 2016; 56:1055-1067. [PMID: 27648737 DOI: 10.1002/mc.22570] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 08/24/2016] [Accepted: 09/16/2016] [Indexed: 12/20/2022]
Abstract
Cancer is one of the leading causes of death worldwide, and oral squamous cell carcinoma (OSCC) accounts for almost a sixth of all reported cancers. Arecoline, from areca nut is known to enhance carcinogenesis in oral squamous cells. The objective of this study is to determine the effect of Taiwanin C, from Taiwania cryptomerioides Hayata against Arecoline-associated carcinogenesis. An OSCC model was created in C57BL/6J Narl mice by administrating 0.5 mg mL-1 arecoline with 0.2 mg mL-1 4-NQO carcinogen for 8 and 28 wk to mimic the etiology of oral cancer patients in Asia. Mice were sacrificed and two cell lines, T28 from the tumor and N28 cancerous cell line from the surrounding non tumor area, were established. Taiwanin C showed effective anti-tumor activity in nude mice models. Taiwanin C significantly inhibited the cell viability of T28 cells in a dose dependent manner, but did not inflict any effect on N28 normal cells. Taiwanin C treatment inhibited the migration ability of T28 cells in a dose dependent manner as determined by wound healing and migration assays. Taiwanin C also reduced the levels of β-catenin and its downstream metastatic proteins, Tbx3 and c-Myc. Besides, Taiwanin C inhibited the nuclear accumulation of β-catenin and induced β-catenin degradation via proteasome-mediated pathway. Moreover, Taiwanin C enhanced GSK-3β and reduced the p-ser9 GSK-3β protein level to inactivate Wnt signaling. Taken together, Taiwanin C blocked the cell migration effects of T28 cells mediated through the activation of GSK-3β to enhance protein degradation and reduce nuclear accumulation of β-catenin. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cheng-Hong Hsieh
- Department of Health and Nutritional Biotechnology, Asia University, Taichung, Taiwan
| | - Hsi-Hsien Hsu
- Division of Colorectal Surgery, Mackay Memorial Hospital, Taipei, Taiwan.,Mackay Medicine, Nursing and Management College, Taipei, Taiwan
| | | | | | - Da-Tian Bau
- Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Chu Ho
- HK. Zen Heart Group Biopharmaceutical Co., Limited, Wanchai, Hong Kong.,Zen Transmission Foundation of Medical Culture and Education, Taichung, Taiwan, ROC
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan.,Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management College, Taipei, Taiwan
| | - Ming-Cheng Chen
- Division of Colorectal Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shu-Huai Wang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Department of Health and Nutritional Biotechnology, Asia University, Taichung, Taiwan.,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
25
|
Jin Y, Cui D, Ren J, Wang K, Zeng T, Gao L. CACNA2D3 is downregulated in gliomas and functions as a tumor suppressor. Mol Carcinog 2016; 56:945-959. [PMID: 27583705 DOI: 10.1002/mc.22548] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 08/22/2016] [Accepted: 08/29/2016] [Indexed: 01/08/2023]
Abstract
CACNA2D3, an auxiliary member of the alpha-2/delta subunit three family of the voltage-dependent calcium channel complex, plays a critical role in tumor suppression. However, its role in glioma carcinogenesis remains largely unknown. Here, we investigated the putative tumor suppressive role of CACNA2D3 in gliomas. Downregulation of CACNA2D3 was frequently detected in glioma tissues and cells compared with their non-tumorigenic counterparts, and correlated with poor survival. To investigate the underlying mechanism of CACNA2D3 in the development and progression of glioma, we performed CACNA2D3 ectopic expression in glioma cells (U87 and U251) and knockdown of CACNA2D3 in LN229 cells and conducted in vitro and in vivo functional assays. Our findings showed that increased intracellular calcium (Ca2+ ) mediated by overexpression of CACNA2D3 induced mitochondrial-mediated apoptosis, upregulation of NLK (through the Wnt/Ca2+ pathway) and inhibition of the epithelial-to-mesenchymal transition. Ectopic expression of CACNA2D3 inhibited cell proliferation, migration, invasion, and tumor growth in vitro and in vivo, whereas CACNA2D3 depletion inhibited cell viability and invasion. Furthermore, we confirmed that CACNA2D3 increased NLK expression in vitro by immunostaining and found that downregulation of CACNA2D3 in glioma cells and high-grade glioma tissue was accompanied by increased methylation. A reporter assay showed increased luciferase activity in NLK knockdown glioma cells and transcriptional activity of β-cantenin/TCF was remarkably enhanced, which further confirmed that NLK antagonizes Wnt signaling-mediated anchorage-dependent and independent cell proliferation and invasion. This mechanism may contribute to a better understanding of glioma cancer pathogenesis and facilitate the development of new therapeutic strategies for the treatment of this disease. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yi Jin
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Daming Cui
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jie Ren
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Ke Wang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Tao Zeng
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Liang Gao
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
26
|
Requirement of Smad4 from Ocular Surface Ectoderm for Retinal Development. PLoS One 2016; 11:e0159639. [PMID: 27494603 PMCID: PMC4975478 DOI: 10.1371/journal.pone.0159639] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 07/06/2016] [Indexed: 11/28/2022] Open
Abstract
Microphthalmia is characterized by abnormally small eyes and usually retinal dysplasia, accounting for up to 11% of the blindness in children. Right now there is no effective treatment for the disease, and the underlying mechanisms, especially how retinal dysplasia develops from microphthalmia and whether it depends on the signals from lens ectoderm are still unclear. Mutations in genes of the TGF-β superfamily have been noted in patients with microphthalmia. Using conditional knockout mice, here we address the question that whether ocular surface ectoderm-derived Smad4 modulates retinal development. We found that loss of Smad4 specifically on surface lens ectoderm leads to microphthalmia and dysplasia of retina. Retinal dysplasia in the knockout mice is caused by the delayed or failed differentiation and apoptosis of retinal cells. Microarray analyses revealed that members of Hedgehog and Wnt signaling pathways are affected in the knockout retinas, suggesting that ocular surface ectoderm-derived Smad4 can regulate Hedgehog and Wnt signaling in the retina. Our studies suggest that defective of ocular surface ectoderm may affect retinal development.
Collapse
|
27
|
Rastogi I, Rajanna S, Webb A, Chhabra G, Foster B, Webb B, Puri N. Mechanism of c-Met and EGFR tyrosine kinase inhibitor resistance through epithelial mesenchymal transition in non-small cell lung cancer. Biochem Biophys Res Commun 2016; 477:937-944. [PMID: 27396618 DOI: 10.1016/j.bbrc.2016.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 07/01/2016] [Indexed: 10/21/2022]
Abstract
According to currently available estimates from Cancer Research UK, 14.1 million new lung cancer cases were diagnosed and a staggering 8.2 million people worldwide died from lung cancer in 2012. EGFR and c-Met are two tyrosine kinase receptors most commonly overexpressed or mutated in Non-small Cell Lung Cancer (NSCLC) resulting in increased proliferation and survival of lung cancer cells. Tyrosine kinase inhibitors (TKIs), such as erlotinib, approved by the FDA as first/second line therapy for NSCLC patients have limited clinical efficacy due to acquired resistance. In this manuscript, we investigate and discuss the role of epithelial mesenchymal transition (EMT) in the development of resistance against EGFR and c-Met TKIs in NSCLC. Our findings show that Zeb-1, a transcriptional repressor of E-Cadherin, is upregulated in TKI-resistant cells causing EMT. We observed that TKI-resistant cells have increased gene and protein expression of EMT related proteins such as Vimentin, N-Cadherin, β-Catenin and Zeb-1, while expression of E-Cadherin, an important cell adhesion molecule, was suppressed. We also confirmed that TKI-resistant cells display mesenchymal cell type morphology, and have upregulation of β-Catenin which may regulate expression of Zeb-1, a transcriptional repressor of E-Cadherin in TKI-resistant NSCLC cells. Finally, we show that down-regulating Zeb-1 by inducing miR-200a or β-Catenin siRNA can increase drug sensitivity of TKI-resistant cells.
Collapse
Affiliation(s)
- Ichwaku Rastogi
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Illinois, United States
| | - Supriya Rajanna
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Illinois, United States
| | - Andrew Webb
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Illinois, United States
| | - Gagan Chhabra
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Illinois, United States
| | - Brad Foster
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Illinois, United States
| | - Brian Webb
- Thermo Fisher Scientific, Rockford, Illinois, United States
| | - Neelu Puri
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Illinois, United States.
| |
Collapse
|
28
|
Qian G, Hu Z, Xu H, Müller S, Wang D, Zhang H, Kim S, Chen Z, Saba NF, Shin DM, Wang AY, Chen ZG. A novel prediction model for human papillomavirus-associated oropharyngeal squamous cell carcinoma using p16 and subcellular β-catenin expression. J Oral Pathol Med 2016; 45:399-408. [PMID: 26493274 PMCID: PMC4841757 DOI: 10.1111/jop.12378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND p16 overexpression is a highly sensitive yet moderately specific biomarker for predicting human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC). Nuclear β-catenin translocation has been linked to HPV-positive OPSCC. However, whether the strategy of combining β-catenin with p16 can better predict HPV-associated OPSCC remains unknown. METHODS We evaluated the expression of p16 and β-catenin (nuclear and membrane) by immunohistochemistry staining in 101 OPSCC tissues and HPV status by HPV DNA in situ hybridization. Logistic regression models were used to evaluate single or multiple biomarkers for HPV prediction. The prediction power, sensitivity, and specificity were determined by receiver operating characteristic (ROC) analyses. RESULTS Our data showed that upon univariate analysis, p16 and nuclear β-catenin were positively correlated with HPV status, while membrane β-catenin was inversely correlated with HPV status (P < 0.01). p16 showed the highest HPV predictive power, with area under the curve (AUC) of 0.9074 compared to 0.6762 for nuclear β-catenin and 0.7635 for membrane β-catenin, respectively, indicating differential accuracies for HPV prediction. Multivariable analysis showed that p16 was significantly correlated with HPV, while nuclear and membrane β-catenin showed marginal significance. The three-biomarker model was similarly sensitive (98.9% vs. 100%) but more specific (88.9% vs. 81%) than p16 alone, which also showed a good predictive value for overall (P = 0.0002) survival and disease-free (P = 0.0158) survival. CONCLUSION Our study suggests a novel model of combining p16 and subcellular β-catenin for prediction of HPV-associatred OPSCC, and this finding deserves further validation.
Collapse
Affiliation(s)
- Guoqing Qian
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Zhongliang Hu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
- Department of Pathology, Xiangya Hospital; Department of Pathology, Xiangya Medical School; Central South University, Changsha, Hunan, 410078, China
| | - Hong Xu
- Ocean NanoTech, San Diego, California, 92126, USA
| | - Susan Müller
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Dongsheng Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Hongzheng Zhang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Sungjin Kim
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, 30322, USA
| | - Zhengjia Chen
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, 30322, USA
| | - Nabil F. Saba
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Dong M. Shin
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | | | - Zhuo Georgia Chen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| |
Collapse
|
29
|
Kho AT, Sharma S, Davis JS, Spina J, Howard D, McEnroy K, Moore K, Sylvia J, Qiu W, Weiss ST, Tantisira KG. Circulating MicroRNAs: Association with Lung Function in Asthma. PLoS One 2016; 11:e0157998. [PMID: 27362794 PMCID: PMC4928864 DOI: 10.1371/journal.pone.0157998] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 06/08/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND MicroRNAs are key transcriptional and network regulators previously associated with asthma susceptibility. However, their role in relation to asthma severity has not been delineated. OBJECTIVE We hypothesized that circulating microRNAs could serve as biomarkers of changes in lung function in asthma patients. METHODS We isolated microRNAs from serum samples obtained at randomization for 160 participants of the Childhood Asthma Management Program. Using a TaqMan microRNA array containing 754 microRNA primers, we tested for the presence of known asthma microRNAs, and assessed the association of the individual microRNAs with lung function as measured by FEV1/FVC, FEV1% and FVC%. We further tested the subset of FEV1/FVC microRNAs for sex-specific and lung developmental associations. RESULTS Of the 108 well-detected circulating microRNAs, 74 (68.5%) had previously been linked to asthma susceptibility. We found 22 (20.3%), 4 (3.7%) and 8 (7.4%) microRNAs to be associated with FEV1/FVC, FEV1% and FVC%, respectively. 8 (of 22) FEV1/FVC, 3 (of 4) FEV1% and 1 (of 8) FVC% microRNAs had functionally validated target genes that have been linked via genome wide association studies to asthma and FEV1 change. Among the 22 FEV1/FVC microRNAs, 9 (40.9%) remain associated with FEV1/FVC in boys alone in a sex-stratified analysis (compared with 3 FEV1/FVC microRNAs in girls alone), 7 (31.8%) were associated with fetal lung development, and 3 (13.6%) in both. Ontology analyses revealed enrichment for pathways integral to asthma, including PPAR signaling, G-protein coupled signaling, actin and myosin binding, and respiratory system development. CONCLUSIONS Circulating microRNAs reflect asthma biology and are associated with lung function differences in asthmatics. They may represent biomarkers of asthma severity.
Collapse
Affiliation(s)
- Alvin T. Kho
- Children’s Hospital Informatics Program, Boston Children’s Hospital and Harvard Medical School, Boston MA 02115, United States of America
| | - Sunita Sharma
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO 80045, United States of America
| | - Joshua S. Davis
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States of America
- Pulmonary and Critical Care Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, United States of America
| | - Joseph Spina
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States of America
| | - Dagnie Howard
- Oregon Health & Science University, Portland, OR 97239, United States of America
| | - Kevin McEnroy
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States of America
| | - Kip Moore
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States of America
| | - Jody Sylvia
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States of America
| | - Weiliang Qiu
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States of America
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States of America
- Partners Personalized Medicine, Partners HealthCare System, Boston, MA 02115, United States of America
| | - Kelan G. Tantisira
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States of America
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, United States of America
| |
Collapse
|
30
|
Ota M, Gonja H, Koike R, Fukuchi S. Multiple-Localization and Hub Proteins. PLoS One 2016; 11:e0156455. [PMID: 27285823 PMCID: PMC4902230 DOI: 10.1371/journal.pone.0156455] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/13/2016] [Indexed: 12/11/2022] Open
Abstract
Protein-protein interactions are fundamental for all biological phenomena, and protein-protein interaction networks provide a global view of the interactions. The hub proteins, with many interaction partners, play vital roles in the networks. We investigated the subcellular localizations of proteins in the human network, and found that the ones localized in multiple subcellular compartments, especially the nucleus/cytoplasm proteins (NCP), the cytoplasm/cell membrane proteins (CMP), and the nucleus/cytoplasm/cell membrane proteins (NCMP), tend to be hubs. Examinations of keywords suggested that among NCP, those related to post-translational modifications and transcription functions are the major contributors to the large number of interactions. These types of proteins are characterized by a multi-domain architecture and intrinsic disorder. A survey of the typical hub proteins with prominent numbers of interaction partners in the type revealed that most are either transcription factors or co-regulators involved in signaling pathways. They translocate from the cytoplasm to the nucleus, triggered by the phosphorylation and/or ubiquitination of intrinsically disordered regions. Among CMP and NCMP, the contributors to the numerous interactions are related to either kinase or ubiquitin ligase activity. Many of them reside on the cytoplasmic side of the cell membrane, and act as the upstream regulators of signaling pathways. Overall, these hub proteins function to transfer external signals to the nucleus, through the cell membrane and the cytoplasm. Our analysis suggests that multiple-localization is a crucial concept to characterize groups of hub proteins and their biological functions in cellular information processing.
Collapse
Affiliation(s)
- Motonori Ota
- Graduate School of Information Sciences, Nagoya University, Nagoya, Japan
- * E-mail:
| | - Hideki Gonja
- Graduate School of Information Sciences, Nagoya University, Nagoya, Japan
| | - Ryotaro Koike
- Graduate School of Information Sciences, Nagoya University, Nagoya, Japan
| | - Satoshi Fukuchi
- Faculty of Engineering, Maebashi Institute of Technology, Maebashi, Japan
| |
Collapse
|
31
|
Abstract
Wnt signaling encompasses multiple and complex signaling cascades and is involved in many developmental processes such as tissue patterning, cell fate specification, and control of cell division. Consequently, accurate regulation of signaling activities is essential for proper embryonic development. Wnt signaling is mostly silent in the healthy adult organs but a reactivation of Wnt signaling is generally observed under pathological conditions. This has generated increasing interest in this pathway from a therapeutic point of view. In this review article, the involvement of Wnt signaling in cardiovascular development will be outlined, followed by its implication in myocardial infarct healing, cardiac hypertrophy, heart failure, arrhythmias, and atherosclerosis. The initial experiments not always offer consensus on the effects of activation or inactivation of the pathway, which may be attributed to (i) the type of cardiac disease, (ii) timing of the intervention, and (iii) type of cells that are targeted. Therefore, more research is needed to determine the exact implication of Wnt signaling in the conditions mentioned above to exploit it as a powerful therapeutic target.
Collapse
|
32
|
Ke H, Masoumi KC, Ahlqvist K, Seckl MJ, Rydell-Törmänen K, Massoumi R. Nemo-like kinase regulates the expression of vascular endothelial growth factor (VEGF) in alveolar epithelial cells. Sci Rep 2016; 6:23987. [PMID: 27035511 PMCID: PMC4817507 DOI: 10.1038/srep23987] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/17/2016] [Indexed: 01/08/2023] Open
Abstract
The canonical Wnt signaling can be silenced either through β-catenin-mediated ubiquitination and degradation or through phosphorylation of Tcf and Lef by nemo-like kinase (NLK). In the present study, we generated NLK deficient animals and found that these mice become cyanotic shortly before death because of lung maturation defects. NLK-/- lungs exhibited smaller and compressed alveoli and the mesenchyme remained thick and hyperplastic. This phenotype was caused by epithelial activation of vascular endothelial growth factor (VEGF) via recruitment of Lef1 to the promoter of VEGF. Elevated expression of VEGF and activation of the VEGF receptor through phosphorylation promoted an increase in the proliferation rate of epithelial and endothelial cells. In summary, our study identifies NLK as a novel signaling molecule for proper lung development through the interconnection between epithelial and endothelial cells during lung morphogenesis.
Collapse
Affiliation(s)
- Hengning Ke
- Molecular Tumor Pathology, Department of Laboratory Medicine, Lund University, Sweden
| | | | - Kristofer Ahlqvist
- Molecular Tumor Pathology, Department of Laboratory Medicine, Lund University, Sweden
| | - Michael J Seckl
- Department of Medical Oncology, Imperial College Healthcare NHS Trust and Imperial College London, London, UK
| | | | - Ramin Massoumi
- Molecular Tumor Pathology, Department of Laboratory Medicine, Lund University, Sweden
| |
Collapse
|
33
|
Guo L, Yilamu D, Sun L, Liu S, Ma F. Association among the expression of β-catenin, cyclin D1 and estrogen receptor-β in human breast cancer. Exp Ther Med 2015; 10:1423-1428. [PMID: 26622501 DOI: 10.3892/etm.2015.2657] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 05/15/2015] [Indexed: 11/06/2022] Open
Abstract
β-catenin, cyclin D1 and estrogen receptor (ER)-β are closely associated with the pathogenesis of breast cancer. In the present study, tissue samples were collected from 226 patients with breast cancer. Subsequently, immunohistochemical analysis was performed to detect the expression of β-catenin, cyclin D1 and ER-β, and the Kaplan-Meier method was used for survival analysis. The abnormal expression rate of β-catenin was 75.2%, while the cyclin D1 positive expression rate was 77.0% and the ER-β positive expression rate was 43.4%. In the tissue samples exhibiting abnormal expression of β-catenin, the positive expression rate of cyclin D1 (85.9%) was significantly higher compared with the samples that expressed β-catenin normally (50.0%). Furthermore, the positive expression rate of ER-β (35.7%) in the β-catenin normal expression tissues was significantly lower compared with that in the β-catenin abnormal expression tissues (45.9%). In the tissues with positive cyclin D1 expression, the positive expression rate of ER-β (48.4%) was significantly higher compared with the cyclin D1 negative expression samples (26.9%). In addition, patients with normal expression of β-catenin and positive expression of cyclin D1 exhibited longer tumor-free survival times. Therefore, an association exists among the abnormal expression of β-catenin and the positive expression of cyclin D1 and ER-β, which may contribute to the development of breast cancer.
Collapse
Affiliation(s)
- Liying Guo
- Department of Breast Cancer, Digestive and Vascular Center, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang 830054, P.R. China
| | - Dilimina Yilamu
- Department of Breast Cancer, Digestive and Vascular Center, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang 830054, P.R. China
| | - Liting Sun
- Department of Breast Cancer, Digestive and Vascular Center, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang 830054, P.R. China
| | - Sha Liu
- Department of Breast Cancer, Digestive and Vascular Center, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang 830054, P.R. China
| | - Fangjing Ma
- Department of Breast Cancer, Digestive and Vascular Center, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang 830054, P.R. China
| |
Collapse
|
34
|
Matthijs Blankesteijn W, Hermans KCM. Wnt signaling in atherosclerosis. Eur J Pharmacol 2015; 763:122-30. [PMID: 25987418 DOI: 10.1016/j.ejphar.2015.05.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/01/2015] [Indexed: 02/03/2023]
Abstract
Atherosclerosis is a disease of the vascular wall that forms the basis for a large spectrum of pathologies of various organs and tissues. Although massive research efforts in the last decades have yielded valuable information about its underlying molecular mechanisms, this has not led to a translation into effective therapeutic interventions that can stop the progression or even can induce regression of atherosclerosis. This underscores the importance of investigations on the involvement of novel signaling pathways in the development and progression of this condition. In this review we focus on the role of Wnt signaling in atherosclerosis. Experimental evidence is presented that Wnt signaling is involved in many aspects of the development and progression of vascular lesions including endothelial dysfunction, macrophage activation and the proliferation and migration of vascular smooth muscle cells. Subsequently, we will discuss the role of Wnt signaling in myocardial infarction and stroke, two common pathologies resulting from the progression of atherosclerotic lesions towards an unstable phenotype. Despite the fact that the published data sometimes are ambiguous or even conflicting, a picture is emerging that an attenuation of Wnt signaling is beneficial for the cardiovascular system that is compromised by atherosclerosis.
Collapse
Affiliation(s)
- W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht, Maastricht University, P.O. Box 616, 6200MD Maastricht, The Netherlands.
| | - Kevin C M Hermans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht, Maastricht University, P.O. Box 616, 6200MD Maastricht, The Netherlands
| |
Collapse
|
35
|
Silva JG, Corrales-Medina FF, Maher OM, Tannir N, Huh WW, Rytting ME, Subbiah V. Clinical next generation sequencing of pediatric-type malignancies in adult patients identifies novel somatic aberrations. Oncoscience 2015; 2:187-92. [PMID: 25859559 PMCID: PMC4381709 DOI: 10.18632/oncoscience.131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/18/2015] [Indexed: 12/20/2022] Open
Abstract
Pediatric malignancies in adults, in contrast to the same diseases in children are clinically more aggressive, resistant to chemotherapeutics, and carry a higher risk of relapse. Molecular profiling of tumor sample using next generation sequencing (NGS) has recently become clinically available. We report the results of targeted exome sequencing of six adult patients with pediatric-type malignancies : Wilms tumor(n=2), medulloblastoma(n=2), Ewing's sarcoma( n=1) and desmoplastic small round cell tumor (n=1) with a median age of 28.8 years. Detection of druggable somatic aberrations in tumors is feasible. However, identification of actionable target therapies in these rare adult patients with pediatric-type malignancies is challenging. Continuous efforts to establish a rare disease registry are warranted.
Collapse
Affiliation(s)
- Jorge Galvez Silva
- Division of Pediatrics, The University of Texas MD Anderson Children's Cancer Hospital, Houston, TX
| | - Fernando F Corrales-Medina
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Miami-Miller School of Medicine, Miami, FL
| | - Ossama M Maher
- Division of Pediatrics, The University of Texas MD Anderson Children's Cancer Hospital, Houston, TX
| | - Nizar Tannir
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Winston W Huh
- Division of Pediatrics, The University of Texas MD Anderson Children's Cancer Hospital, Houston, TX
| | - Michael E Rytting
- Division of Pediatrics, The University of Texas MD Anderson Children's Cancer Hospital, Houston, TX
| | - Vivek Subbiah
- Division of Pediatrics, The University of Texas MD Anderson Children's Cancer Hospital, Houston, TX ; Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
36
|
Manzo-Merino J, Contreras-Paredes A, Vázquez-Ulloa E, Rocha-Zavaleta L, Fuentes-Gonzalez AM, Lizano M. The Role of Signaling Pathways in Cervical Cancer and Molecular Therapeutic Targets. Arch Med Res 2014; 45:525-39. [DOI: 10.1016/j.arcmed.2014.10.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 10/29/2014] [Indexed: 12/24/2022]
|
37
|
Ye YL, Zhou CL, Pang Z, Zheng JJ. MicroRNA570 inhibits human colon cancer HT-29 cell proliferation via Wnt/β-catenin pathway. Shijie Huaren Xiaohua Zazhi 2014; 22:2439-2444. [DOI: 10.11569/wcjd.v22.i17.2439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate the effect of microRNA570 (miR-570) on the proliferation and biological properties of HT-29 cells and the possible mechanisms involved.
METHODS: An miR-570 mimic was transfected into human colon cancer HT-29 cells. Cell proliferation was studied by cell counting kit-8 assay (CCK-8). Total miRNA was extracted and the relative expression of miR-570 was quantified by real-time quantitative PCR. Western blot and real-time quantitative PCR were performed to analyze the expression of β-catenin in the cytoplasm and nucleus. The expression of c-myc, cyclinD1 and survivin was also detected by real-time quantitative PCR and Western blot.
RESULTS: After transfection with the miR-570 mimic, the proliferation ability of HT-29 cells decreased. The inhibitory effect was most significant at 72 h (53.23% ± 1.22%) (P < 0.05). The expression of miR-570 was up-regulated more than 20 times (20.01 ± 0.32) (P < 0.05). The mRNA and protein expression of β-catenin, c-myc, cyclinD1 and survivin was decreased in the miR-570 mimic group, compared with blank and negative control groups. Western blot results further confirmed that the protein expression of β-catenin in the nucleus decreased significantly, although its expression in the cytoplasm did not change significantly.
CONCLUSION: MiR-570 significantly inhibits the proliferation of HT-29 cells possibly by regulation of the Wnt/β-catenin signal pathway.
Collapse
|
38
|
Nakashima T, Ohkusa T, Okamoto Y, Yoshida M, Lee JK, Mizukami Y, Yano M. Rapid electrical stimulation causes alterations in cardiac intercellular junction proteins of cardiomyocytes. Am J Physiol Heart Circ Physiol 2014; 306:H1324-33. [DOI: 10.1152/ajpheart.00653.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The intercellular junctions contain two complexes, adhesion junctions (AJ) and connexin (Cx) gap junctions (GJs). GJs provide the pathway for intercellular current flow. AJs mediate normal mechanical coupling and play an important role in the stability of GJs. We investigated the effects of rapid electrical stimulation (RES) on cardiac intercellular junctions, especially β-catenin and Cx43 alterations. We also studied the effects of ANG II receptor blockade on intercellular junction remodeling. Neonatal rats were euthanized by decapitation, and cardiomyocytes were prepared, cultured, and subjected to RES. We used real-time PCR, western blot analysis, and immunohistochemical methods. Conduction properties were examined by an extracellular potential mapping system. Cx43 protein expression in cardiomyocytes was significantly increased after 60 min. β-Catenin expression in the total cell fraction was significantly increased after 30 min. The expression level of β-catenin in the nucleus, which functions as a T cell factor/lymphocyte enhancer binding factor transcriptional activator of Cx43 with its degradation regulated by glycogen synthase kinase-3β, was dramatically increased after 10 min. Conduction velocity was increased significantly by RES for 60 min. Olmesartan prevented most these effects of RES. We showed an increase of phosphorylated glycogen synthase kinase-3β, which is phosphorylated by activated MAPKs and inhibits β-catenin degradation, was attenuated by olmesartan. The changes in β-catenin precede Cx43 GJ remodeling and might play an important role in the formation and stability of GJs. Olmesartan might be a new upstream arrhythmia therapy by modulating intercellular junction remodeling through the β-catenin signaling pathway.
Collapse
Affiliation(s)
- Tadamitsu Nakashima
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Tomoko Ohkusa
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yoko Okamoto
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Masaaki Yoshida
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Jong-Kook Lee
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka, Japan; and
| | - Yoichi Mizukami
- Center for Gene Research, Yamaguchi University, Yamaguchi, Japan
| | - Masafumi Yano
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| |
Collapse
|
39
|
Gomez-Millan J, Perez L, Aroca I, Del Mar Delgado M, De Luque V, Román A, Torres E, Ramos S, Perez S, Bayo E, Medina JA. Preoperative chemoradiotherapy in rectal cancer induces changes in the expression of nuclear β-catenin: prognostic significance. BMC Cancer 2014; 14:192. [PMID: 24629143 PMCID: PMC3995577 DOI: 10.1186/1471-2407-14-192] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 03/07/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Preoperative chemoradiotherapy (CRT) is the cornerstone of treatment for locally advanced rectal cancer (LARC). Although high local control is achieved, overall rates of distant control remain suboptimal. Colorectal carcinogenesis is associated with critical alterations of the Wnt/β-catenin pathway involved in proliferation and survival. The aim of this study was to assess whether CRT induces changes in the expression of β-catenin/E-cadherin, and to determine whether these changes are associated with survival. METHODS The Immunohistochemical expression of nuclear β-catenin and membranous E-cadherin was prospectively analysed in tumour blocks from 98 stage II/III rectal cancer patients treated with preoperative CRT. Tumour samples were collected before and after CRT treatment. All patients were treated with pelvic RT (46-50 Gy in 2 Gy fractions) and 5-fluorouracil (5FU) intravenous infusion (225 mg/m2) or capecitabine (825 mg/m2) during RT treatment, followed by total mesorectal excision (TME). Disease-free survival (DFS) was analysed using the Kaplan-Meier method and a multivariate Cox regression model was employed for the Multivariate analysis. RESULTS CRT induced significant changes in the expression of nuclear β-catenin (49% of patients presented an increased expression after CRT, 17% a decreased expression and 34% no changes; p = 0.001). After a median follow-up of 25 months, patients that overexpressed nuclear β-catenin after CRT showed poor survival compared with patients that experienced a decrease in nuclear β-catenin expression (3-year DFS 92% vs. 43%, HR 0.17; 95% CI 0.03 to 0.8; p = 0.02). In the multivariate analysis for DFS, increased nuclear β-catenin expression after CRT almost reached the cut-off for significance (p = 0.06). CONCLUSIONS In our study, preoperative CRT for LARC induced significant changes in nuclear β-catenin expression, which had a major impact on survival. Finding a way to decrease CRT resistance would significantly improve LARC patient survival.
Collapse
Affiliation(s)
- Jaime Gomez-Millan
- Department of Radiation Oncology, University Hospital Virgen de la Victoria, Campus Teatinos s/n, Málaga, 29010, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Liu Y, Dong QZ, Wang S, Xu HT, Miao Y, Wang L, Wang EH. Kaiso interacts with p120-catenin to regulate β-catenin expression at the transcriptional level. PLoS One 2014; 9:e87537. [PMID: 24498333 PMCID: PMC3911973 DOI: 10.1371/journal.pone.0087537] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 12/30/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND We have reported that p120-catenin could regulate β-catenin transcription in lung cancer cells, but the specific mechanism is unclear. METHODS AND RESULTS In this study, bisulfite sequencing PCR showed that the β-catenin promoter region in SPC-A-1 and LTEP-a-2 lung cancer cell lines has Kaiso binding sites sequences and CpG islands which may combine with Kaiso. The demethylating reagent 5-Aza-2'-deoxycytidine significantly upregulated β-catenin mRNA expression in lung cancer cell lines, whereas expression was significantly reduced following transfection with Kaiso. However, the upregulation of β-catenin mRNA expression after treatment with 5-Aza-2'-deoxycytidine was not reduced by subsequent transfection with Kaiso cDNA. Chromatin immunoprecipitation showed that, in lung cancer cell lines, methylated CpG-dinucleotides sequences combined with Kaiso and the Kaiso binding sites sequence did not. The capacity of Kaiso to combine with p120-catenin isoforms was confirmed by immunoprecipitation. CONCLUSIONS Based on these results, we concluded that Kaiso participates in the regulation by p120ctn of β-catenin mRNA expression in the lung cancer cell lines.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, PR China
| | - Qian-Ze Dong
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, PR China
| | - Si Wang
- Department of Medical Microbiology and Parasitology, College of Basic Medical Sciences of China Medical University, Shenyang, PR China
| | - Hong-Tao Xu
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, PR China
| | - Yuan Miao
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, PR China
| | - Liang Wang
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, PR China
| | - En-Hua Wang
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, PR China
- * E-mail:
| |
Collapse
|
41
|
Yoshida GJ, Saya H. Inversed relationship between CD44 variant and c-Myc due to oxidative stress-induced canonical Wnt activation. Biochem Biophys Res Commun 2014; 443:622-7. [DOI: 10.1016/j.bbrc.2013.12.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 12/03/2013] [Indexed: 01/10/2023]
|
42
|
Brändstedt J, Wangefjord S, Borgquist S, Nodin B, Eberhard J, Manjer J, Jirström K. Influence of anthropometric factors on tumour biological characteristics of colorectal cancer in men and women: a cohort study. J Transl Med 2013; 11:293. [PMID: 24256736 PMCID: PMC3874681 DOI: 10.1186/1479-5876-11-293] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 11/12/2013] [Indexed: 12/22/2022] Open
Abstract
Background Obesity is a well established risk factor of colorectal cancer (CRC), but how body size influences risk of colorectal cancer defined by key molecular alterations remains unclear. In this study, we investigated the relationship between height, weight, body mass index (BMI), waist- and hip circumference, waist-hip ratio (WHR) and risk of CRC according to expression of beta-catenin, cyclin D1, p53 and microsatellite instability status of the tumours in men and women, respectively. Methods Immunohistochemical expression of beta-catenin, cyclin D1, p53 and MSI-screening status was assessed in tissue microarrays with tumours from 584 cases of incident CRC in the Malmö Diet and Cancer Study. Six anthropometric factors: height, weight, BMI, waist- and hip circumference, and WHR were categorized by quartiles of baseline measurements and relative risks of CRC according to expression of beta-catenin, cyclin D1, p53 and MSI status were calculated using multivariate Cox regression models. Results High height was associated with risk of cyclin D1 positive, and p53 negative CRC in women but not with any investigative molecular subsets of CRC in men. High weight was associated with beta-catenin positive, cyclin D1 positive, p53 negative and microsatellite stable (MSS) tumours in women, and with beta-catenin negative and p53 positive tumours in men. Increased hip circumference was associated with beta-catenin positive, p53 negative and MSS tumours in women and with beta-catenin negative, cyclin D1 positive, p53 positive and MSS tumours in men. In women, waist circumference and WHR were not associated with any molecular subsets of CRC. In men, both high WHR and high waist circumference were associated with beta-catenin positive, cyclin D1 positive and p53 positive tumours. WHR was also associated with p53 negative CRC, and waist circumference with MSS tumours. High BMI was associated with increased risk of beta-catenin positive and MSS CRC in women, and with beta-catenin positive, cyclin D1 positive and p53 positive tumours in men. Conclusions Findings from this large prospective cohort study indicate sex-related differences in the relationship between obesity and CRC risk according to key molecular characteristics, and provide further support of an influence of lifestyle factors on different molecular pathways of colorectal carcinogenesis.
Collapse
Affiliation(s)
- Jenny Brändstedt
- Department of Clinical Sciences, Division of Pathology, Lund University, Skåne University Hospital, Lund, Sweden.
| | | | | | | | | | | | | |
Collapse
|
43
|
Mei G, Xia L, Zhou J, Zhang Y, Tuo Y, Fu S, Zou Z, Wang Z, Jin D. Neuropeptide SP activates the WNT signal transduction pathway and enhances the proliferation of bone marrow stromal stem cells. Cell Biol Int 2013; 37:1225-32. [PMID: 23893958 DOI: 10.1002/cbin.10158] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 07/04/2013] [Indexed: 12/24/2022]
Abstract
Substance P (SP) mediates multiple activities in various cell types, such as proliferation, anti-apoptotic response, and inflammation. We have investigated the effects of SP, NK1 antagonist and DKK1 on proliferation of bone marrow stromal stem cells (BMSCs), as well as the underlying mechanism. Isolated BMSCs were exposed to SP (10(-8) M) (group A), SP + NK1 antagonist (1 µM) (group B), SP + DKK1 (0.2 µg/mL) (group C), or the same amount of PBS (group D). Expression of gene and protein of Wnt/β-catenin signalling was detected using quantitative PCR and western blotting. SP (10(-8) M) significantly enhanced the proliferation of BMSCs and the number of viable cells was reduced by treatment with NK1 antagonist (1 µM) or DKK1 (0.2 µg/mL). SP also significantly increased the expression of C-myc mRNA, Lef1, β-catenin protein and C-myc protein, but decreased the expression of Tcf7 and p-β-catenin protein compared to group D. These roles of SP were inhibited by the NK1 antagonist and DKK1. Expression of CyclinD1 and β-catenin mRNAs, however, was not significantly influenced by SP, NK1 antagonist and DKK1. These findings suggest that SP enhances BMSC proliferation via regulation of the Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Gang Mei
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Analysing the impact of nucleo-cytoplasmic shuttling of β-catenin and its antagonists APC, Axin and GSK3 on Wnt/β-catenin signalling. Cell Signal 2013; 25:2210-21. [PMID: 23872074 DOI: 10.1016/j.cellsig.2013.07.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 07/02/2013] [Accepted: 07/09/2013] [Indexed: 01/03/2023]
Abstract
The canonical Wnt signalling pathway plays a critical role in development and disease. The key player of the pathway is β-catenin. Its activity is mainly regulated by the destruction complex consisting of APC, Axin and GSK3. In the nucleus, the complex formation of β-catenin and TCF initiates target gene expression. Our study provides a comprehensive analysis of the role of nucleo-cytoplasmic shuttling of APC, Axin, and GSK3 and the inactivation of β-catenin by the destruction complex in Wnt/β-catenin signalling. We address the following questions: Can nucleo-cytoplasmic shuttling of APC, Axin and GSK3 increase the [β-catenin/TCF] concentration? And, how is the [β-catenin/TCF] concentration influenced by phosphorylation and subsequent degradation of nuclear β-catenin? Based on experimental findings, we develop a compartmental model and conduct several simulation experiments. Our analysis reveals the following key findings: 1) nucleo-cytoplasmic shuttling of β-catenin and its antagonists can yield a spatial separation between the said proteins, which results in a breakdown of β-catenin degradation, followed by an accumulation of β-catenin and hence leads to an increase of the [β-catenin/TCF] concentration. Our results strongly suggest that Wnt signalling can benefit from nucleo-cytoplasmic shuttling of APC, Axin and GSK3, although they are in general β-catenin antagonising proteins. 2) The total robustness of the [β-catenin/TCF] output is closely linked to its absolute concentration levels. We demonstrate that the compartmental separation of β-catenin and the destruction complex does not only lead to a maximization, but additionally to an increased robustness of [β-catenin/TCF] signalling against perturbations in the cellular environment. 3) A nuclear accumulation of the destruction complex renders the pathway robust against fluctuations in Wnt signalling and against changes in the compartmental distribution of β-catenin. 4) Elucidating the impact of destruction complex inhibition, we show that the [β-catenin/TCF] concentration is more effectively enhanced by inhibition of the kinase GSK3 rather than the binding of β-catenin to the destruction complex.
Collapse
|
45
|
Wada N, Hashinaga T, Otabe S, Yuan X, Kurita Y, Kakino S, Ohoki T, Nakayama H, Fukutani T, Tajiri Y, Yamada K. Selective modulation of Wnt ligands and their receptors in adipose tissue by chronic hyperadiponectinemia. PLoS One 2013; 8:e67712. [PMID: 23861788 PMCID: PMC3701684 DOI: 10.1371/journal.pone.0067712] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 05/22/2013] [Indexed: 11/23/2022] Open
Abstract
Background Adiponectin-transgenic mice had many small adipocytes in both subcutaneous and visceral adipose tissues, and showed higher sensitivity to insulin, longer life span, and reduced chronic inflammation. We hypothesized that adiponectin regulates Wnt signaling in adipocytes and thereby modulates adipocyte proliferation and chronic inflammation in adipose tissue. Materials and Methods We examined the expression of all Wnt ligands and their receptors and the activity of Wnt signaling pathways in visceral adipose tissue from wild-type mice and two lines of adiponectin-transgenic mice. The effects of adiponectin were also investigated in cultured 3T3-L1 cells. Results The Wnt5b, Wnt6, Frizzled 6 (Fzd6), and Fzd9 genes were up-regulated in both lines of transgenic mice, whereas Wnt1, Wnt2, Wnt5a, Wnt9b, Wnt10b, Wnt11, Fzd1, Fzd2, Fzd4, Fzd7, and the Fzd coreceptor low-density-lipoprotein receptor-related protein 6 (Lrp6) were reduced. There was no difference in total β-catenin levels in whole-cell extracts, non-phospho-β-catenin levels in nuclear extracts, or mRNA levels of β-catenin target genes, indicating that hyperadiponectinemia did not affect canonical Wnt signaling. In contrast, phosphorylated calcium/calmodulin-dependent kinase II (p-CaMKII) and phosphorylated Jun N-terminal kinase (p-JNK) were markedly reduced in adipose tissue from the transgenic mice. The adipose tissue of the transgenic mice consisted of many small cells and had increased expression of adiponectin, whereas cyclooxygenase-2 expression was reduced. Wnt5b expression was elevated in preadipocytes of the transgenic mice and decreased in diet-induced obese mice, suggesting a role in adipocyte differentiation. Some Wnt genes, Fzd genes, and p-CaMKII protein were down-regulated in 3T3-L1 cells cultured with a high concentration of adiponectin. Conclusion Chronic hyperadiponectinemia selectively modulated the expression of Wnt ligands, Fzd receptors and LRP coreceptors accompanied by the inhibition of the Wnt/Ca2+ and JNK signaling pathways, which may be involved in the altered adipocyte cellularity, endogenous adiponectin production, and anti-inflammatory action induced by hyperadiponectinemia.
Collapse
Affiliation(s)
- Nobuhiko Wada
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Toshihiko Hashinaga
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Shuichi Otabe
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Xiaohong Yuan
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Yayoi Kurita
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Satomi Kakino
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Tsuyoshi Ohoki
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hitomi Nakayama
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Tomoka Fukutani
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Yuji Tajiri
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Kentaro Yamada
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
- * E-mail:
| |
Collapse
|
46
|
Pyrvinium pamoate does not activate protein kinase CK1, but promotes Akt/PKB down-regulation and GSK3 activation. Biochem J 2013; 452:131-7. [PMID: 23438105 DOI: 10.1042/bj20121140] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
It has been reported that pyrvinium pamoate (PyrPam), an FDA (U.S. Food and Drug Administration)-approved anthelminthic drug, is a potent inhibitor of Wnt signalling by a mechanism which implies the direct activation of protein kinase CK1α. In the present paper, we provide data ruling out any direct stimulatory effect of PyrPam on CK1, by showing that the catalytic activity of CK1α and those of its isoforms δ and γ1 are not significantly affected by PyrPam when tested with the aid of specific peptide and protein substrates. Accordingly, cell treatment with PyrPam has no significant effect on the phosphorylation of β-catenin Ser(45). By contrast, the phosphorylation of β-catenin Thr(41) is increased upon cell treatment with PyrPam, through a mechanism that implies the upstream dephosphorylation of Akt/PKB (protein kinase B) and of GSK3 (glycogen synthase kinase 3). It can be concluded from the present study that PyrPam is not a bona fide activator of CK1, its perturbation of cell signalling pathways being mediated by a complex mechanism initiated by a fall in Akt phosphorylation whose down-regulation promotes reduced phosphorylation and activation of GSK3. Consistent with this, lysates of cells treated with PyrPam display enhanced protein phosphorylation which is unaffected by CK1 inhibition, while disappearing upon inhibition of GSK3. Our data are consistent with the observation that PyrPam ultimately inhibits Wnt signalling despite its lack of efficacy on CK1.
Collapse
|
47
|
Peng XL, Ji MY, Yang ZR, Song J, Dong WG. Tumor suppressor function of ezrin-radixin-moesin-binding phosphoprotein-50 through β-catenin/E-cadherin pathway in human hepatocellular cancer. World J Gastroenterol 2013; 19:1306-1313. [PMID: 23483729 PMCID: PMC3587489 DOI: 10.3748/wjg.v19.i8.1306] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/01/2013] [Accepted: 01/24/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine the effect and molecular mechanism of ezrin-radixin-moesin-binding phosphoprotein-50 (EBP50) in hepatocellular carcinoma (HCC).
METHODS: Three human HCC cell lines, i.e., SM-MC7721, HepG2 and Hep3B, were used. We transfected the Pbk-CMV-HA-EBP50 plasmid into SMMC7721 cells with Lipofectamine 2000 to overexpress EBP50. Western blotting were performed to determine the effects of the plasmid on EBP50 expression and to detect the expression of β-catenin and E-cadherin before and after the transfection of the plasmid into SMMC7721 cells. In vitro cell proliferation was assessed with a Cell Counting Kit-8 (CCK-8) assay. Cell cycle distribution was assessed with flow cytometry. Invasion and migration ability of before and after the transfection were determined with a transwell assay. Cell apoptosis was demonstrated with Annexin V-FITC. The effect of EBP50 overexpressing on tumor growth in vivo was performed with a xenograft tumor model in nude mice.
RESULTS: The transfection efficiency was confirmed with Western blotting (1.36 ± 0.07 vs 0.81 ± 0.09, P < 0.01). The CCK8 assay demonstrated that the growth of cells overexpressing EBP50 was significantly lower than control cells (P < 0.01). Cell cycle distribution showed there was a G0/G1 cell cycle arrest in cells overexpressing EBP50 (61.3% ± 3.1% vs 54.0% ± 2.4%, P < 0.05). The transwell assay showed that cell invasion and migration were significantly inhibited in cells overexpressing EBP50 compared with control cells (5.8 ± 0.8 vs 21.6 ± 1.3, P < 0.01). Annexin V-FITC revealed that apoptosis was significantly increased in cells overexpressing EBP50 compared with control cells (14.8% ± 2.7% vs 3.4% ± 1.3%, P < 0.05). The expression of β-catenin was downregulated and E-cadherin was upregulated in cells overexpressing EBP50 compared with control cells (0.28 ± 0.07 vs 0.56 ± 0.12, P < 0.05; 0.55 ± 0.08 vs 0.39 ± 0.07, P < 0.05). In vivo tumor growth assay confirmed that up-regulation of EBP50 could obviously slow the growth of HCC derived from SMMC7721 cells (28.9 ± 7.2 vs 70.1 ± 7.2, P < 0.01).
CONCLUSION: The overexpression of EBP50 could inhibit the growth of SMMC7721 cells and promote apoptosis by modulating β-catenin, E-cadherin. EBP50 may serve asa potential therapeutic target in HCC.
Collapse
|
48
|
Usongo M, Li X, Farookhi R. Activation of the canonical WNT signaling pathway promotes ovarian surface epithelial proliferation without inducing β-catenin/Tcf-mediated reporter expression. Dev Dyn 2013; 242:291-300. [PMID: 23239518 DOI: 10.1002/dvdy.23919] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 11/28/2012] [Accepted: 12/04/2012] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND In response to activation of the canonical WNT signaling pathway, β-catenin cooperates with Lef/Tcf (lymphoid enhancer factor/T-cell factor) transcription factors to drive expression of Wnt target genes. The canonical WNT signaling pathway is involved in development, wound repair, and tumorigenesis. Studies examining the involvement of the canonical WNT signaling pathway in the development of ovarian surface epithelium (OSE) and ovarian carcinogenesis, however, have recently begun to emerge. In this study, we investigated the modulation of β-catenin and β-catenin/Tcf-signaling activity within the OSE using responsive transgenic mice and examined the response of primary OSE cells and ovarian cancer cell lines to activation of the canonical WNT signaling pathway. RESULTS β-catenin was localized to the lateral membrane of the ovarian epithelium. Stimulation of primary OSE cells in vitro with LiCl or Wnt3a led to GSK-3β inhibition and stabilization of β-catenin but failed to induce β-catenin/Tcf-mediated lacZ expression. Furthermore, E-cadherin expression was downregulated and the proliferative potency of OSE cells increased. Of four ovarian cancers cell lines screened, only the HEY cell line demonstrated induction of luciferase reporter upon canonical WNT stimulation. CONCLUSIONS These observations suggest that in ovarian adenocarcinoma, dysregulated WNT signaling may not always be indicative of β-catenin/Tcf-mediated transcriptional activity.
Collapse
Affiliation(s)
- Macalister Usongo
- Department of Experimental Medicine, McGill University, Montreal, Canada.
| | | | | |
Collapse
|
49
|
Park EJ, Chung HJ, Park HJ, Kim GD, Ahn YH, Lee SK. Suppression of Src/ERK and GSK-3/β-catenin signaling by pinosylvin inhibits the growth of human colorectal cancer cells. Food Chem Toxicol 2013; 55:424-33. [PMID: 23333577 DOI: 10.1016/j.fct.2013.01.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 12/17/2012] [Accepted: 01/07/2013] [Indexed: 02/07/2023]
Abstract
Pinosylvin, a naturally occurring trans-stilbenoid mainly found in Pinus species, has exhibited a potential cancer chemopreventive activity. However, the growth inhibitory activity against cancer cells and the underlying molecular mechanisms remain to be elucidated. Therefore, the anti-proliferative activity of pinosylvin was investigated in human colorectal HCT 116 cancer cells. Pinosylvin inhibited the proliferation of HCT 116 cells by arresting transition of cell cycle from G1 to S phase along with the downregulation of cyclin D1, cyclin E, cyclin A, cyclin dependent kinase 2 (CDK2), CDK4, c-Myc, and retinoblastoma protein (pRb), and the upregulation of p21(WAF1/CIP1) and p53. Pinosylvin was also found to attenuate the activation of proteins involved in focal adhesion kinase (FAK)/c-Src/extracellular signal-regulated kinase (ERK) signaling, and phosphoinositide 3-kinase (PI3K)/Akt/ glycogen synthase kinase 3β (GSK-3β) signaling pathway. Subsequently, pinosylvin suppressed the nuclear translocation of β-catenin, one of downstream molecules of PI3K/Akt/GSK-3β signaling, and these events led to the sequential downregulation of β-catenin-mediated transcription of target genes including BMP4, ID2, survivin, cyclin D1, MMP7, and c-Myc. These findings demonstrate that the anti-proliferative activity of pinosylvin might be associated with the cell cycle arrest and downregulation of cell proliferation regulating signaling pathways in human colorectal cancer cells.
Collapse
Affiliation(s)
- Eun-Jung Park
- College of Pharmacy, Ewha Womans University, Seoul 120-750, South Korea
| | | | | | | | | | | |
Collapse
|
50
|
Fairbairn EA, Bonthius J, Cherr GN. Polycyclic aromatic hydrocarbons and dibutyl phthalate disrupt dorsal-ventral axis determination via the Wnt/β-catenin signaling pathway in zebrafish embryos. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2012; 124-125:188-196. [PMID: 22975441 DOI: 10.1016/j.aquatox.2012.08.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/20/2012] [Accepted: 08/21/2012] [Indexed: 06/01/2023]
Abstract
The canonical Wnt/β-catenin signaling pathway is critical during early teleost development for establishing the dorsal-ventral axis. Within this pathway, GSK-3β, a key regulatory kinase in the Wnt pathway, regulates β-catenin degradation and thus the ability of β-catenin to enter nuclei, where it can activate expression of genes that have been linked to the specification of the dorsal-ventral axis. In this study, we describe the morphological abnormalities that resulted in zebrafish embryos when axis determination was disrupted by environmental contaminants. These abnormalities were linked to abnormal nuclear accumulation of β-catenin. Furthermore, we demonstrated that the developmental abnormalities and altered nuclear β-catenin accumulation occurred when embryos were exposed to commercial GSK-3β inhibitors. Zebrafish embryos were exposed to commercially available GSK-3 inhibitors (GSK-3 Inhibitor IX and 1-azakenpaullone), or common environmental contaminants (dibutyl phthalate or the polycyclic aromatic hydrocarbons phenanthrene and fluorene) from the 2 to 8-cell stage through the mid-blastula transition (MBT). These embryos displayed morphological abnormalities at 12.5 h post-fertilization (hpf) that were comparable to embryos exposed to lithium chloride (LiCl) (300 mM LiCl for 10 min, prior to the MBT), a classic disruptor of embryonic axis determination. Whole-mount immunolabeling and laser scanning confocal microscopy were used to localize β-catenin. The commercial GSK-3 Inhibitors as well as LiCl, dibutyl phthalate, fluorene and phenanthrene all induced an increase in the levels of nuclear β-catenin throughout the embryo, indicating that the morphological abnormalities were a result of disruption of Wnt/β-catenin signaling during dorsal-ventral axis specification. The ability of environmental chemicals to directly or indirectly target GSK-3β was assessed. Using Western blot analysis, the ability of these chemicals to affect enzymatic inhibitory phosphorylation at serine 9 on GSK-3β was examined, but no change in the serine phosphorylation state of GSK-3β was detected in exposed embryos. Furthermore, polycyclic aromatic hydrocarbons and dibutyl phthalate had no direct effect on the in vitro kinase activity of GSK-3β. While developmental abnormalities resulting from these axis-disrupting contaminants were linked to β-catenin accumulation in nuclei, the details of the disruption of this signaling pathway remain unknown. Since phenanthrene and fluorene as well as other hydrocarbons have been shown to disrupt axial development in sea urchin embryos, and since axis determination and the Wnt/β-catenin signaling pathway are highly conserved, we propose that these environmental contaminants may impact embryo development through a similar mechanism across phyla.
Collapse
Affiliation(s)
- Elise A Fairbairn
- University of California Davis, Bodega Marine Laboratory, Bodega Bay, CA 94923, USA.
| | | | | |
Collapse
|