1
|
Yuan S, Yin Y, Wang K, Zhou H, Qian C. Tetraspanin-29 activates Notch signaling by interacting with ADAM10 to enhance its activity in colorectal cancer. Biochem Cell Biol 2022; 100:292-300. [PMID: 35968819 DOI: 10.1139/bcb-2021-0114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
ADAM10 acts upstream of Notch signaling and plays oncogenic roles in various cancers. Tetraspanin family proteins regulate ADAM10 trafficking and activity. Here, we aimed to investigate whether and how tetraspanin-29 modulates ADAM10 in colorectal cancer (CRC). We found that ADAM10 expression was upregulated in CRC tissues and this was cross-validated in the TCGA COAD data set. The ADAM10 protein level and its α-secretase activity were enhanced in CRC cell lines compared with control cell lines. Co-immunoprecipitation showed ADAM10 interacted with tetraspanin-29 in the LoVo cell line. Tetraspanin-29 knockdown reduced the cell surface trafficking and α-secretase activity of ADAM10. In addition, tetraspanin-29 knockdown inhibited Notch activity in a luciferase reporter assay and reduced the levels of cleaved Notch1 and Notch target genes such as HES2, c-MYC, and cyclin D3. Consistently, tetraspanin-29 overexpression increased cleaved Notch1 and this effect was blocked by ADAM10 inhibitors. The TCGA COAD data set confirmed the positive correlations of tetraspanin-29 with HES2, c-MYC, and cyclin D3. Thus, the tetraspanin-29/ADAM10/Notch pathway plays an important role in CRC.
Collapse
Affiliation(s)
- Shijie Yuan
- Xiehe Stem Cell Genetic Engineering Co. Ltd., East China Center, No. 988 Tianheng Road, Huzhou, Zhejiang 313000, China
| | - Yan Yin
- Department of Colon-rectal Surgery, Huzhou Maternity & Child Health Care Hospital, No. 2 East Street, Huzhou, Zhejiang 313000, China
| | - Kuansong Wang
- Department of Pathology, Xiangya Medical College, Central South University, No. 172 Tongzipo Road, Changsha, Hunan 410003, China
| | - Haiyan Zhou
- Department of Pathology, Xiangya Medical College, Central South University, No. 172 Tongzipo Road, Changsha, Hunan 410003, China
| | - Cheng Qian
- Department of Colon-rectal Surgery, Huzhou Maternity & Child Health Care Hospital, No. 2 East Street, Huzhou, Zhejiang 313000, China
| |
Collapse
|
2
|
ADAM10 is involved in the oncogenic process and chemo-resistance of triple-negative breast cancer via regulating Notch1 signaling pathway, CD44 and PrPc. Cancer Cell Int 2021; 21:32. [PMID: 33413403 PMCID: PMC7791678 DOI: 10.1186/s12935-020-01727-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 12/21/2020] [Indexed: 01/15/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC) is the most challenging breast cancer subtype to treat, because it is so aggressive with shorter survival. Chemotherapy remains the standard treatment due to the lack of specific and effective molecular targets. The aim of the present study is to investigate the potential roles of A Disintegrin and Metalloproteinase 10 (ADAM10) on TNBC cells and the effects of combining ADAM10 expression and neoadjuvant chemotherapy treatment (NACT) to improve the overall survival in breast cancer patients. Methods Using a series of breast cancer cell lines, we measured the expression of ADAM10 and its substrates by quantitative real-time PCR assay (qRT-PCR) and western blot analysis. Cell migration and invasion, cell proliferation, drug sensitivity assay, cell cycle and apoptosis were conducted in MDA-MB-231 cells cultured with ADAM10 siRNA. The effect of ADAM10 down-regulation by siRNA on its substrates was assessed by western blot analysis. We performed immunohistochemical staining for ADAM10 in clinical breast cancer tissues in 94 patients receiving NACT. Results The active form of ADAM10 was highly expressed in TNBC cell lines. Knockdown of ADAM10 in MDA-MB-231 cells led to a significant decrease in cell proliferation, migration, invasion and the IC50 value of paclitaxel and adriamycin, while induced cell cycle arrest and apoptosis. And these changes were correlated with down-regulation of Notch signaling, CD44 and cellular prion protein (PrPc). In clinical breast cancer cases, a high ADAM10 expression in pre-NACT samples was strongly associated with poorer response to NACT and shorter overall survival. Conclusions These data suggest the previously unrecognized roles of ADAM10 in contributing to the progression and chemo-resistance of TNBC.
Collapse
|
3
|
Xu Y, Lai Y, Cao L, Li Y, Chen G, Chen L, Weng H, Chen T, Wang L, Ye Y. Human umbilical cord mesenchymal stem cells-derived exosomal microRNA-451a represses epithelial-mesenchymal transition of hepatocellular carcinoma cells by inhibiting ADAM10. RNA Biol 2020; 18:1408-1423. [PMID: 33206588 DOI: 10.1080/15476286.2020.1851540] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Exosomes derived from human umbilical cord mesenchymal stem cells (hucMSCs) expressing microRNAs (miRNAs) have been highlighted in human cancers. However, the detailed molecular mechanism of hucMSCs-derived exosomal miR-451a on hepatocellular carcinoma (HCC) remains further investigation. Our study aims to explore the impact of exosomal miR-451a on the progression of HCC. Expression of miR-451a and a disintegrin and metalloprotease 10 (ADAM10) in HCC tissues and adjacent normal tissues were determined. The exosomes were extracted from hucMSCs and co-cultured with Hep3B and SMMC-7721 cell lines. After the treatment of relative exosomes or exosome inhibitor GW4869 in Hep3B and SMMC-7721 cells, the paclitaxel resistance and malignant phenotypes of HCC cells were measured. Moreover, the effect of hucMSCs-derived exosomes on the expression of miR-451a and ADAM10 in HCC cells was assessed. The targeting relationship between miR-451a and ADAM10 was verified to detect the impact of ADAM10-wild type and ADAM10-mutant type (MUT) on HCC cell processes. Low expression of miR-451a and high expression of ADAM10 indicated a poor prognosis of HCC patients. MiR-451a was up-regulated while ADAM10 was down-regulated in HCC cells after co-culture with HucMSC-derived exosomes. The exosomes elevated miR-451a and inhibited ADAM10 to suppress the paclitaxel resistance, cell cycle transition, proliferation, migration and invasion, and promote apoptosis of HCC cells. ADAM10 was verified to be a target gene of miR-451a. ADAM10-MUT promoted HCC process independent of miR-451a mimic. HucMSC-derived exosomal miR-451a could restrict the epithelial-mesenchymal transition of HCC cells by targeting ADAM10, which might provide new targets for HCC treatment.
Collapse
Affiliation(s)
- Yunxiuxiu Xu
- Department of Hepato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Lai
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linhui Cao
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanshan Li
- Department of Blood Transfusion, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guangcheng Chen
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Chen
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Intensive Care Unit, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hanqin Weng
- Department of Hepato-Billiary Surgery, Dongguan people's Hospital, Southern Medical University, Guangdong, China
| | - Tao Chen
- Department of Hepato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lingyun Wang
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yibiao Ye
- Department of Hepato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Rao CV, Asch AS, Carr DJJ, Yamada HY. "Amyloid-beta accumulation cycle" as a prevention and/or therapy target for Alzheimer's disease. Aging Cell 2020; 19:e13109. [PMID: 31981470 PMCID: PMC7059149 DOI: 10.1111/acel.13109] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/16/2019] [Accepted: 12/25/2019] [Indexed: 02/06/2023] Open
Abstract
The cell cycle and its regulators are validated targets for cancer drugs. Reagents that target cells in a specific cell cycle phase (e.g., antimitotics or DNA synthesis inhibitors/replication stress inducers) have demonstrated success as broad-spectrum anticancer drugs. Cyclin-dependent kinases (CDKs) are drivers of cell cycle transitions. A CDK inhibitor, flavopiridol/alvocidib, is an FDA-approved drug for acute myeloid leukemia. Alzheimer's disease (AD) is another serious issue in contemporary medicine. The cause of AD remains elusive, although a critical role of latent amyloid-beta accumulation has emerged. Existing AD drug research and development targets include amyloid, amyloid metabolism/catabolism, tau, inflammation, cholesterol, the cholinergic system, and other neurotransmitters. However, none have been validated as therapeutically effective targets. Recent reports from AD-omics and preclinical animal models provided data supporting the long-standing notion that cell cycle progression and/or mitosis may be a valid target for AD prevention and/or therapy. This review will summarize the recent developments in AD research: (a) Mitotic re-entry, leading to the "amyloid-beta accumulation cycle," may be a prerequisite for amyloid-beta accumulation and AD pathology development; (b) AD-associated pathogens can cause cell cycle errors; (c) thirteen among 37 human AD genetic risk genes may be functionally involved in the cell cycle and/or mitosis; and (d) preclinical AD mouse models treated with CDK inhibitor showed improvements in cognitive/behavioral symptoms. If the "amyloid-beta accumulation cycle is an AD drug target" concept is proven, repurposing of cancer drugs may emerge as a new, fast-track approach for AD management in the clinic setting.
Collapse
Affiliation(s)
- Chinthalapally V. Rao
- Center for Cancer Prevention and Drug DevelopmentDepartment of MedicineHematology/Oncology SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Adam S. Asch
- Stephenson Cancer CenterDepartment of MedicineHematology/Oncology SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Daniel J. J. Carr
- Department of OphthalmologyUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Hiroshi Y. Yamada
- Center for Cancer Prevention and Drug DevelopmentDepartment of MedicineHematology/Oncology SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| |
Collapse
|
5
|
Wang C, Li Q, Lv J, Sun X, Cao Y, Yu K, Miao C, Zhang ZS, Yao Z, Wang Q. Alpha-hemolysin of uropathogenic Escherichia coli induces GM-CSF-mediated acute kidney injury. Mucosal Immunol 2020; 13:22-33. [PMID: 31719643 PMCID: PMC6914670 DOI: 10.1038/s41385-019-0225-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 02/04/2023]
Abstract
Uropathogenic Escherichia coli (UPEC) is the leading cause of urinary tract infections (UTIs), inducing acute pyelonephritis and may result in permanent renal scarring and failure. Alpha-hemolysin (HlyA), a key UPEC toxin, causes serious tissue damage; however, the mechanism through which HlyA induces kidney injury remains unclear. In the present study, granulocyte-macrophage colony-stimulating factor (GM-CSF) secreted by renal epithelial cells was upregulated by HlyA in vitro and in vivo, which induced M1 macrophage accumulation in kidney, and ADAM10 was found involved in HlyA-induced GM-CSF. Macrophage elimination or GM-CSF neutralization protected against acute kidney injury in mice, and increased GM-CSF was detected in urine of patients infected by hlyA-positive UPEC. In addition, HlyA was found to promote UPEC invasion into renal epithelial cells by interacting with Nectin-2 in vitro. However, HlyA did not affect bacterial titers during acute kidney infections, and HlyA-induced invasion did not contribute to GM-CSF upregulation in vitro, which indicate that HlyA-induced GM-CSF is independent of bacteria invasion. The role of GM-CSF in HlyA-mediated kidney injury may lead to novel strategies to treat acute pyelonephritis.
Collapse
Affiliation(s)
- Changying Wang
- 0000 0000 9792 1228grid.265021.2Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070 Tianjin, China
| | - Qianqian Li
- 0000 0000 9792 1228grid.265021.2Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070 Tianjin, China
| | - Junqiang Lv
- 0000 0000 9792 1228grid.265021.2Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070 Tianjin, China
| | - Xuan Sun
- 0000 0000 9792 1228grid.265021.2Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070 Tianjin, China
| | - Yang Cao
- 0000 0004 1798 6160grid.412648.dDepartment of Clinical Laboratory, The Second Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Kaiyuan Yu
- 0000 0000 9792 1228grid.265021.2Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070 Tianjin, China
| | - Chunhui Miao
- 0000 0000 9792 1228grid.265021.2Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070 Tianjin, China
| | - Zhi-Song Zhang
- 0000 0000 9878 7032grid.216938.7State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Collaborative Innovation Center for Biotherapy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, 300350 Tianjin, China
| | - Zhi Yao
- 0000 0000 9792 1228grid.265021.2Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070 Tianjin, China ,0000 0000 9792 1228grid.265021.22011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, 300070 Tianjin, China
| | - Quan Wang
- 0000 0000 9792 1228grid.265021.2Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070 Tianjin, China
| |
Collapse
|
6
|
Park JH, Choi JY, Jo C, Koh YH. Involvement of ADAM10 in acrolein-induced astrocytic inflammation. Toxicol Lett 2019; 318:44-49. [PMID: 31639409 DOI: 10.1016/j.toxlet.2019.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/05/2019] [Accepted: 10/10/2019] [Indexed: 12/29/2022]
Abstract
Acrolein is a neurotoxin produced through lipid peroxidation in the brain affected by ischemic stroke, which results in neuronal cell injury and inflammation. However the mechanism underlying acrolein-induced brain inflammation remains unclear. Therefore we examined how acrolein leads to astrocytic inflammation. It was found that acrolein increased the levels of NLRP3 and cleaved caspase-1, which led to the maturation of interleukin-1β (IL-1β). ELISA assay results, which showed that acrolein increased the secreted IL-1β, further supported acrolein-induced astrocytic inflammation. Acrolein increased ADAM10 protein levels and the cleavage of N-cadherin. The ADAM10 inhibitor, GI 254023X blocked N-cadherin cleavage by acrolein, suggesting that ADAM10 is an upstream of N-cadherin. Furthermore, we found that acrolein activated p38 MAPK and NF-κB p65, while pretreatment with p38 MAPK inhibitor, SB203580 and GI 254023X inhibited NF-κB p65 activation and NLRP3 inflammasome. This suggests that p38 MAPK mediates the activation of NF-κB p65, which is associated with NLRP3 expression. Finally, we showed that acrolein induced cell toxicity and decrease of EAAT1 expression, suggesting that acrolein may induce a loss of glutamate uptake function. In conclusion, we demonstrate that acrolein induces astrocytic inflammation through NLRP3 inflammasome, which is regulated by ADAM10 and attributed to p38 MAPK-activated NF-κB p65 activity.
Collapse
Affiliation(s)
- Jung Hyun Park
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2(i)-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Ji-Young Choi
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2(i)-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Chulman Jo
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2(i)-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Young Ho Koh
- Division of Brain Diseases, Center for Biomedical Sciences, Korea National Institute of Health, 187 Osongsaengmyeong2(i)-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea.
| |
Collapse
|
7
|
Yao Y, Sun W, Sun Q, Jing B, Liu S, Liu X, Shen G, Chen R, Wang H. Platelet-Derived Exosomal MicroRNA-25-3p Inhibits Coronary Vascular Endothelial Cell Inflammation Through Adam10 via the NF-κB Signaling Pathway in ApoE -/- Mice. Front Immunol 2019; 10:2205. [PMID: 31632389 PMCID: PMC6783608 DOI: 10.3389/fimmu.2019.02205] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 08/30/2019] [Indexed: 12/15/2022] Open
Abstract
Introduction: Coronary artery disease originates from the blockage of the inner walls of the coronary arteries due to a plaque buildup. Accumulating studies have highlighted the role of microRNAs (miRs) delivered by exosomes in the progression of coronary artery disease. Thus, the current study was to elucidate the role and mechanism by which miR-25-3p influences oxidized low density lipoprotein (ox-LDL)-induced coronary vascular endothelial cell (CVEC) inflammation. Methods: Primarily isolated CVECs were treated with ox-LDL to induce inflammation. Atherosclerosis models were induced in ApoE-/- mice and the peripheral blood platelet exosomes (PLT-Exo) were extracted and induced by thrombin, followed by co-culture with CVECs. The relationship between miR-25-3p and A disintegrin and metalloprotease 10 (Adam10) as well as the involvement of the NF-κB signaling pathway was evaluated. In order to evaluate the effect of PLT-Exo containing miR-25-3p on ox-LDL-induced CVEC inflammation, lipid accumulation and fibrosis, miR-25-3p mimic/inhibitor (in vitro), miR-25-3p agomir (in vivo), and si-Adam10 were delivered. Results: MiR-25-3p was expressed poorly in ox-LDL-induced CVECs and vascular tissues but exhibited high levels of expression in thrombin-induced PLT-Exo of atherosclerosis models of ApoE-/- mice. CVECs endocytosed PLT-Exo upregulated the miR-25-3p expression. Adam10 was identified as a target gene of miR-25-3p. The thrombin-induced activated PLT-Exo carrying miR-25-3p reduced Adam10 expression to inhibit ox-LDL-induced CVEC inflammation and lipid deposition through downregulating levels of α-smooth muscle actin, Collagen I a1, Collagen III a1, triglycerides, total cholesterol, interleukin (IL)-1β, IL-6, and tumor necrosis factor-α. Furthermore, the NF-κB signaling pathway participated in the inhibitory effect of PLT-Exo carrying miR-25-3p. Conclusion: Collectively, PLT-Exo overexpressing miR-25-3p attenuates ox-LDL-induced CVEC inflammation in ApoE-/- mouse models of atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Haiyang Wang
- Department of Vascular and Interventional Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
8
|
Cytoplasmic fragment of CD147 generated by regulated intramembrane proteolysis contributes to HCC by promoting autophagy. Cell Death Dis 2017; 8:e2925. [PMID: 28703811 PMCID: PMC5550841 DOI: 10.1038/cddis.2017.251] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/17/2017] [Accepted: 04/26/2017] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal and prevalent cancers worldwide. CD147 (EMMPRIN or basigin) is a leading gene relating to hepatocarcinogenesis and metastasis, and is detected in transmembrane, exosome or circulating forms in HCC patients. The endosome recycling of CD147 further enhances the function of this oncoprotein from a dynamic perspective. However, previous studies about CD147 mainly focused on one separate form, and little attention has been paid to how the different forms of tumor-derived CD147 changes. Moreover, uncovering the roles of the residual C-terminal portion of CD147 after shedding is inevitable to fully understand CD147 promoting tumor progression. In this study, we discovered that under low-cholesterol condition, CD147 endocytosis is inhibited but its shedding mediated by ADAM10 is enhanced. Further procession of residual CD147 in the lysosome produces nuclear-localized CD147-ICD (intracellular domain of CD147), which contributes to autophagy through NF-κB–TRAIL–caspase8–ATG3 axis. As autophagy endows cancer cells with increased adaptability to chemotherapy, and HAb 18 (a specific antibody targeting CD147) inhibits CD147 shedding and sequential CD147-ICD enhances autophagy, we found the combination of HAb 18 and cisplatin exhibited marked antitumor efficiency.
Collapse
|
9
|
Kahl BS, Dreyling M, Gordon LI, Quintanilla-Martinez L, Sotomayor EM. Recent advances and future directions in mantle cell lymphoma research: report of the 2016 mantle cell lymphoma consortium workshop. Leuk Lymphoma 2017; 58:1561-1569. [PMID: 28140709 DOI: 10.1080/10428194.2017.1283036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin lymphoma typically associated with the t(11;14) chromosomal translocation, resulting in overexpression of cyclin D1. Although MCL is associated with clinical heterogeneity, outcomes are generally poor and no standard treatment has been established. However, the recent approval of ibrutinib provides a new therapeutic option. Moreover, recent clinical trials have provided new perspectives on the relative efficacy and safety of various approaches for both transplant-eligible and transplant-ineligible patients. Multiple novel strategies are being evaluated in the treatment of MCL, including both targeted agents and cellular immunotherapies. At the Lymphoma Research Foundation's 12th MCL Workshop, researchers gathered to discuss research findings, clinical trial results, and future directions related to MCL, its biology, and its treatment. This manuscript, which includes a summary of each presentation, aims to review recent findings in MCL research and highlight potential areas for future study.
Collapse
Affiliation(s)
- Brad S Kahl
- a Department of Medicine, Washington University in Saint Louis , St. Louis , MO , USA
| | - Martin Dreyling
- b Department of Medicine, University of Munich-Grosshadern , Munich , Germany
| | - Leo I Gordon
- c Department of Medicine, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center , Chicago , IL , USA
| | | | - Eduardo M Sotomayor
- e Department of Medicine, George Washington University , Washington DC , USA
| |
Collapse
|
10
|
SINKOVICS JOSEPHG. The cnidarian origin of the proto-oncogenes NF-κB/STAT and WNT-like oncogenic pathway drives the ctenophores (Review). Int J Oncol 2015; 47:1211-29. [PMID: 26239915 PMCID: PMC4583530 DOI: 10.3892/ijo.2015.3102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/26/2015] [Indexed: 01/09/2023] Open
Abstract
The cell survival pathways of the diploblastic early multicellular eukaryotic hosts contain and operate the molecular machinery resembling those of malignantly transformed individual cells of highly advanced multicellular hosts (including Homo). In the present review, the STAT/NF-κB pathway of the cnidarian Nematostella vectensis is compared with that of human tumors (malignant lymphomas, including Reed-Sternberg cells) pointing out similarities, including possible viral initiation in both cases. In the ctenophore genome and proteome, β-catenin gains intranuclear advantages due to a physiologically weak destructive complex in the cytoplasm, and lack of natural inhibitors (the dickkopfs). Thus, a scenario similar to what tumor cells initiate and achieve is presented through several constitutive loss-of-function type mutations in the destructive complex and in the elimination of inhibitors. Vice versa, malignantly transformed individual cells of advanced multicellular hosts assume pheno-genotypic resemblance to cells of unicellular or early multicellular hosts, and presumably to their ancient predecessors, by returning to the semblance of immortality and to the resumption of the state of high degree of resistance to physicochemical insults. Human leukemogenic and oncogenic pathways are presented for comparisons. The supreme bioengineers RNA/DNA complex encoded both the malignantly transformed immortal cell and the human cerebral cortex. The former generates molecules for the immortality of cellular life in the Universe. The latter invents the inhibitors of the process in order to gain control over it.
Collapse
Affiliation(s)
- JOSEPH G. SINKOVICS
- St. Joseph Hospital's Cancer Institute Affiliated with the H.L. Moffitt Comprehensive Cancer Center; Department of Molecular Medicine, The University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
11
|
Wu C, Molavi O, Zhang H, Gupta N, Alshareef A, Bone KM, Gopal K, Wu F, Lewis JT, Douglas DN, Kneteman NM, Lai R. STAT1 is phosphorylated and downregulated by the oncogenic tyrosine kinase NPM-ALK in ALK-positive anaplastic large-cell lymphoma. Blood 2015; 126:336-45. [PMID: 25921060 DOI: 10.1182/blood-2014-10-603738] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 04/24/2015] [Indexed: 02/05/2023] Open
Abstract
The tumorigenicity of most cases of ALK-positive anaplastic large-cell lymphoma (ALK+ ALCL) is driven by the oncogenic fusion protein NPM-ALK in a STAT3-dependent manner. Because it has been shown that STAT3 can be inhibited by STAT1 in some experimental models, we hypothesized that the STAT1 signaling pathway is defective in ALK+ ALCL, thereby leaving the STAT3 signaling unchecked. Compared with normal T cells, ALK+ ALCL tumors consistently expressed a low level of STAT1. Inhibition of the ubiquitin-proteasome pathway appreciably increased STAT1 expression in ALK+ ALCL cells. Furthermore, we found evidence that NPM-ALK binds to and phosphorylates STAT1, thereby promoting its proteasomal degradation in a STAT3-dependent manner. If restored, STAT1 is functionally intact in ALK+ ALCL cells, because it effectively upregulated interferon-γ, induced apoptosis/cell-cycle arrest, potentiated the inhibitory effects of doxorubicin, and suppressed tumor growth in vivo. STAT1 interfered with the STAT3 signaling by decreasing STAT3 transcriptional activity/DNA binding and its homodimerization. The importance of the STAT1/STAT3 functional interaction was further highlighted by the observation that short interfering RNA knockdown of STAT1 significantly decreased apoptosis induced by STAT3 inhibition. Thus, STAT1 is a tumor suppressor in ALK+ ALCL. Phosphorylation and downregulation of STAT1 by NPM-ALK represent other mechanisms by which this oncogenic tyrosine kinase promotes tumorigenesis.
Collapse
MESH Headings
- Anaplastic Lymphoma Kinase
- Animals
- Apoptosis
- Blotting, Western
- Case-Control Studies
- Cell Proliferation
- Cell Transformation, Neoplastic
- Down-Regulation
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Interferon-gamma
- Lymphoma, Large-Cell, Anaplastic/genetics
- Lymphoma, Large-Cell, Anaplastic/metabolism
- Lymphoma, Large-Cell, Anaplastic/pathology
- Mice
- Mice, SCID
- Phosphorylation
- Proteasome Endopeptidase Complex/metabolism
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- RNA, Small Interfering/genetics
- Receptor Protein-Tyrosine Kinases/metabolism
- STAT1 Transcription Factor/antagonists & inhibitors
- STAT1 Transcription Factor/genetics
- STAT1 Transcription Factor/metabolism
- STAT3 Transcription Factor/antagonists & inhibitors
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Tumor Cells, Cultured
- Ubiquitin/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chengsheng Wu
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Ommoleila Molavi
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada; Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, East Azerbaijan Province, Iran
| | - Haifeng Zhang
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Nidhi Gupta
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Abdulraheem Alshareef
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Kathleen M Bone
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Keshav Gopal
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Fang Wu
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | | | | | | | - Raymond Lai
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada; Department of Oncology, University of Alberta, Edmonton, AB, Canada; and DynaLIFE Dx Medical Laboratories, Edmonton, AB, Canada
| |
Collapse
|
12
|
Zingoni A, Cecere F, Vulpis E, Fionda C, Molfetta R, Soriani A, Petrucci MT, Ricciardi MR, Fuerst D, Amendola MG, Mytilineos J, Cerboni C, Paolini R, Cippitelli M, Santoni A. Genotoxic Stress Induces Senescence-Associated ADAM10-Dependent Release of NKG2D MIC Ligands in Multiple Myeloma Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:736-48. [PMID: 26071561 DOI: 10.4049/jimmunol.1402643] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 05/06/2015] [Indexed: 01/10/2023]
Abstract
Genotoxic stress can promote antitumor NK cell responses by upregulating the surface expression of activating ligands on cancer cells. Moreover, a number of studies suggested a role for soluble NK group 2D ligands in the impairment of NK cell tumor recognition and killing. We investigated whether genotoxic stress could promote the release of NK group 2D ligands (MHC class I-related chain [MIC]A and MICB), as well as the molecular mechanisms underlying this event in human multiple myeloma (MM) cells. Our results show that genotoxic agents used in the therapy of MM (i.e., doxorubicin and melphalan) selectively affect the shedding of MIC molecules that are sensitive to proteolytic cleavage, whereas the release of the short MICA*008 allele, which is frequent in the white population, is not perturbed. In addition, we found that a disintegrin and metalloproteinase 10 expression is upregulated upon chemotherapeutic treatment both in patient-derived CD138(+)/CD38(+) plasma cells and in several MM cell lines, and we demonstrate a crucial role for this sheddase in the proteolytic cleavage of MIC by means of silencing and pharmacological inhibition. Interestingly, the drug-induced upregulation of a disintegrin and metalloproteinase 10 on MM cells is associated with a senescent phenotype and requires generation of reactive oxygen species. Moreover, the combined use of chemotherapeutic drugs and metalloproteinase inhibitors enhances NK cell-mediated recognition of MM cells, preserving MIC molecules on the cell surface and suggesting that targeting of metalloproteinases in conjunction with chemotherapy could be exploited for NK cell-based immunotherapeutic approaches, thus contributing to avoid the escape of malignant cells from stress-elicited immune responses.
Collapse
Affiliation(s)
- Alessandra Zingoni
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy;
| | - Francesca Cecere
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Elisabetta Vulpis
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Maria Teresa Petrucci
- Department of Cellular Biotechnology and Hematology, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Maria Rosaria Ricciardi
- Department of Cellular Biotechnology and Hematology, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Daniel Fuerst
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm of the German Red Cross Blood Transfusion Service, Baden Wuerttemberg-Hessen, 89081 Ulm, Germany; Institute of Transfusion Medicine, University of Ulm, 89081 Ulm, Germany; and
| | | | - Joannis Mytilineos
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm of the German Red Cross Blood Transfusion Service, Baden Wuerttemberg-Hessen, 89081 Ulm, Germany; Institute of Transfusion Medicine, University of Ulm, 89081 Ulm, Germany; and
| | - Cristina Cerboni
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy
| | - Angela Santoni
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome 00161, Italy; Institute Pasteur-Fondazione Cenci Bolognetti, "Sapienza" University of Rome, 00161 Rome, Italy
| |
Collapse
|
13
|
NPM-ALK mediates phosphorylation of MSH2 at tyrosine 238, creating a functional deficiency in MSH2 and the loss of mismatch repair. Blood Cancer J 2015; 5:e311. [PMID: 25978431 PMCID: PMC4476014 DOI: 10.1038/bcj.2015.35] [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: 10/19/2014] [Revised: 03/16/2015] [Accepted: 04/07/2015] [Indexed: 12/22/2022] Open
Abstract
The vast majority of anaplastic lymphoma kinase-positive anaplastic large cell lymphoma (ALK+ALCL) tumors express the characteristic oncogenic fusion protein NPM-ALK, which mediates tumorigenesis by exerting its constitutive tyrosine kinase activity on various substrates. We recently identified MSH2, a protein central to DNA mismatch repair (MMR), as a novel binding partner and phosphorylation substrate of NPM-ALK. Here, using liquid chromatography–mass spectrometry, we report for the first time that MSH2 is phosphorylated by NPM-ALK at a specific residue, tyrosine 238. Using GP293 cells transfected with NPM-ALK, we confirmed that the MSH2Y238F mutant is not tyrosine phosphorylated. Furthermore, transfection of MSH2Y238F into these cells substantially decreased the tyrosine phosphorylation of endogenous MSH2. Importantly, gene transfection of MSH2Y238F abrogated the binding of NPM-ALK with endogenous MSH2, re-established the dimerization of MSH2:MSH6 and restored the sensitivity to DNA mismatch-inducing drugs, indicative of MMR return. Parallel findings were observed in two ALK+ALCL cell lines, Karpas 299 and SUP-M2. In addition, we found that enforced expression of MSH2Y238F into ALK+ALCL cells alone was sufficient to induce spontaneous apoptosis. In conclusion, our findings have identified NPM-ALK-induced phosphorylation of MSH2 at Y238 as a crucial event in suppressing MMR. Our studies have provided novel insights into the mechanism by which oncogenic tyrosine kinases disrupt MMR.
Collapse
|
14
|
Ebsen H, Lettau M, Kabelitz D, Janssen O. Subcellular localization and activation of ADAM proteases in the context of FasL shedding in T lymphocytes. Mol Immunol 2015; 65:416-28. [PMID: 25745808 DOI: 10.1016/j.molimm.2015.02.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/20/2015] [Accepted: 02/08/2015] [Indexed: 10/23/2022]
Abstract
The "A Disintegrin And Metalloproteinases" (ADAMs) form a subgroup of the metzincin endopeptidases. Proteolytically active members of this protein family act as sheddases and govern key processes in development and inflammation by regulating cell surface expression and release of cytokines, growth factors, adhesion molecules and their receptors. In T lymphocytes, ADAM10 sheds the death factor Fas Ligand (FasL) and thereby regulates T cell activation, death and effector function. Although FasL shedding by ADAM10 was confirmed in several studies, its regulation is still poorly defined. We recently reported that ADAM10 is highly abundant on T cells whereas its close relative ADAM17 is expressed at low levels and transiently appears at the cell surface upon stimulation. Since FasL is also stored intracellularly and brought to the plasma membrane upon stimulation, we addressed where the death factor gets exposed to ADAM proteases. We report for the first time that both ADAM10 and ADAM17 are associated with FasL-containing secretory lysosomes. Moreover, we demonstrate that TCR/CD3/CD28-stimulation induces a partial positioning of both proteases and FasL to lipid rafts and only the activation-induced raft-positioning results in FasL processing. TCR/CD3/CD28-induced FasL proteolysis is markedly affected by reducing both ADAM10 and ADAM17 protein levels, indicating that in human T cells also ADAM17 is implicated in FasL processing. Since FasL shedding is affected by cholesterol depletion and by inhibition of Src kinases or palmitoylation, we conclude that it requires mobilization and co-positioning of ADAM proteases in lipid raft-like platforms associated with an activation of raft-associated Src-family kinases.
Collapse
Affiliation(s)
- Henriette Ebsen
- University of Kiel, Institute of Immunology, University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3 Bldg 17, D-24105 Kiel, Germany
| | - Marcus Lettau
- University of Kiel, Institute of Immunology, University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3 Bldg 17, D-24105 Kiel, Germany
| | - Dieter Kabelitz
- University of Kiel, Institute of Immunology, University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3 Bldg 17, D-24105 Kiel, Germany
| | - Ottmar Janssen
- University of Kiel, Institute of Immunology, University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3 Bldg 17, D-24105 Kiel, Germany.
| |
Collapse
|
15
|
Qu M, Qiu BO, Xiong W, Chen D, Wu A. Expression of a-disintegrin and metalloproteinase 10 correlates with grade of malignancy in human glioma. Oncol Lett 2015; 9:2157-2162. [PMID: 26137031 DOI: 10.3892/ol.2015.2993] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 02/05/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to determine the expression of a-disintegrin and metalloproteinase 10 (ADAM10) in human glioma tissues from surgical specimens and discuss its possible significance in glioma biology. A total of 43 glioma specimens obtained from patients between 2007 and 2010 were collected and a series of assays were performed. Of these, 22 cases were low-grade gliomas, while 21 cases were high-grade gliomas. In addition, 20 cases of meningioma were used as the control group. Reverse transcription-polymerase chain reaction (RT-PCR), western blot analysis and immunohistochemistry were used to determine the mRNA and protein expression levels of ADAM10. Besides the quantitative analysis, histological observations were also performed to localize ADAM10 expression in glioma cells. The RT-PCR and western blot analysis results demonstrated increased ADAM10 expression in the low-grade glioma samples compared with the control (P<0.05), while ADAM10 expression was further increased in the high-grade glioma samples (P<0.01 vs. control; P<0.05 vs. low-grade glioma), indicating that the mRNA and protein expression levels of ADAM10 were malignancy-dependent. The immunohistochemical analysis revealed that the ADAM10 protein was located on both the tumor cell membrane and blood vessel walls within tumor tissues. In conclusion, these results indicated that ADAM10 expression correlates with the grade of malignancy in human glioma from surgical specimens. In addition, the fact that ADAM10 protein was expressed on cell membranes and blood vessel walls within tumor tissues, indicates that its expression may be associated with invasive tumor growth and peritumoral edema formation.
Collapse
Affiliation(s)
- Min Qu
- Department of Neurosurgery, The Central Hospital of Dalian City, Dalian, Liaoning 116033, P.R. China
| | - B O Qiu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Wende Xiong
- Department of Neurosurgery, The Central Hospital of Dalian City, Dalian, Liaoning 116033, P.R. China
| | - Dong Chen
- Department of Neurosurgery, The Central Hospital of Dalian City, Dalian, Liaoning 116033, P.R. China
| | - Anhua Wu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
16
|
Zhang Y, Molavi O, Su M, Lai R. The clinical and biological significance of STAT1 in esophageal squamous cell carcinoma. BMC Cancer 2014; 14:791. [PMID: 25355139 PMCID: PMC4233059 DOI: 10.1186/1471-2407-14-791] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/13/2014] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Loss of STAT1 (Signal Transducer and Activator of Transcription-1) has been implicated in the pathobiology of a number of cancer types. Nonetheless, the biological and clinical significance of STAT1 in esophageal squamous cell carcinomas (ESCC) has not been comprehensively studied. METHODS Using immunohistochemistry, we detected the STAT1 expression in a cohort of ESCC patients; In-vitro experiments, we used enforced gene transfection of STAT1C into two STAT1- weak/negative ESCC cell lines and siRNA knockdown of STAT1 in two STAT1-strong ESCC cell lines to detect STAT1 function in ESCC. RESULTS We found that the expression of STAT1 was heterogeneous in ESCC, with 64 (49.0%) strongly positive cases, 59 (45.0%) weakly positive cases and 8 (6.1%) negative cases. STAT1 expression inversely correlated with the depth of tumor invasion and tumor size (p=0.047 and p=0.029, respectively, Chi square). Furthermore, patients with STAT1-strong/weak tumors had a significantly longer survival compared to those with STAT1-negative tumors (33.6 months versus 13.1 months, p=0.019). In patients carrying tumors of aggressive cytology (n=50), those with STAT1-strong tumors survived significantly longer than those with STAT1-weak/negative tumors (34.6 months versus 20.5 months, p=0.011). Our in-vitro experiments revealed that STAT1 is proapoptotic and inhibitory to cell-cycle progression and colony formation. Lastly, we found evidence that STAT1 signaling in ESCC cells down-regulated the expression and/or activity of NF-κB and STAT3, both of which are known to have oncogenic potential. CONCLUSION To conclude, our findings suggest that STAT1 is a tumor suppressor in ESCC. Loss of STAT1, which is frequent in ESCC, contributes to the pathogenesis of these tumors.
Collapse
Affiliation(s)
- Ying Zhang
- />Department of Pathology, Shantou University Medical College, 22 Xinling Road, Shantou, 515031 Guangdong Province China
- />Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta Canada
| | - Ommoleila Molavi
- />Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta Canada
| | - Min Su
- />Department of Pathology, Shantou University Medical College, 22 Xinling Road, Shantou, 515031 Guangdong Province China
| | - Raymond Lai
- />Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta Canada
- />Department of Oncology, University of Alberta, Edmonton, Alberta Canada
- />DynaLIFEDX Medical Laboratories, Edmonton, Alberta Canada
- />Department of Laboratory Medicine and Pathology, University of Alberta, 5142 Katz Group Centre for Pharmacy and Health Research, Edmonton, Alberta T6G 2E1 Canada
| |
Collapse
|
17
|
Shao Y, Sha XY, Bai YX, Quan F, Wu SL. Effect of A disintegrin and metalloproteinase 10 gene silencing on the proliferation, invasion and migration of the human tongue squamous cell carcinoma cell line TCA8113. Mol Med Rep 2014; 11:212-8. [PMID: 25333745 PMCID: PMC4237091 DOI: 10.3892/mmr.2014.2717] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 09/18/2014] [Indexed: 01/05/2023] Open
Abstract
The present study aimed to investigate the effect of A disintegrin and metalloproteinase 10 (ADAM10) gene silencing on the proliferation, migration and invasion of the human tongue squamous cell carcinoma cell line TCA8113. RNA interference was used to knock down the expression of ADAM10 in the TCA8113 cell line and the proliferation, migration and invasive ability of the treated cells were observed in vitro. The expression levels of epidermal growth factor receptor (EGFR) and E-cadherin in the treated cells were determined by western blot analysis. The proliferation, migration and invasion abilities of cells in the ADAM10 siRNA-treated group were significantly lower than those in the control groups (P<0.05). In addition, compared with the control groups, the expression levels of EGFR and E-cadherin in the ADAM10 siRNA-treated cells were significantly decreased (P<0.05) and increased (P<0.05), respectively. These results suggested that ADAM10 is important in regulating the proliferation, invasion and migration of the human tongue squamous cell carcinoma cell line TCA8113 and that the mechanism may, at least in part, be associated with the upregulation of EGFR and the downregulation of E-cadherin.
Collapse
Affiliation(s)
- Yuan Shao
- Department of Otorhinolaryngology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiao-Ying Sha
- The Sixth Hepatic Disease Ward, The Affiliated Xi'an Eighth Hospital, Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yan-Xia Bai
- Department of Otorhinolaryngology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Fang Quan
- Department of Otorhinolaryngology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Sheng-Li Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
18
|
Chen YJ, Lin HC, Chen KC, Lin SR, Cheng TL, Chang LS. Taiwan cobra phospholipase A2 suppresses ERK-mediated ADAM17 maturation, thus reducing secreted TNF-α production in human leukemia U937 cells. Toxicon 2014; 86:79-88. [PMID: 24874889 DOI: 10.1016/j.toxicon.2014.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/09/2014] [Accepted: 05/12/2014] [Indexed: 11/17/2022]
Abstract
The goal of this study was to explore the signaling pathway regulating the processing of proADAM17 into ADAM17 in Taiwan cobra phospholipase A2 (PLA2)-treated human leukemia U937 cells. PLA2 induced reactive oxygen species (ROS)-elicited p38 MAPK activation and ERK inactivation in U937 cells. Catalytically inactive bromophenacylated PLA2 (BPB-PLA2) and PLA2 mutants evoked Ca(2+)-mediated p38 MAPK activation, and the level of phosphorylated ERK remained unchanged. PLA2 treatment reduced mature ADAM17 expression and secreted TNF-α (sTNF-α) production. Co-treatment of SB202190 (p38 MAPK inhibitor) and catalytically inactive PLA2 increased ERK phosphorylation, ADAM17 maturation and sTNF-α production. Nevertheless, mRNA levels of ADAM17 and TNF-α were insignificantly altered after PLA2 and SB202190/BPB-PLA2 treatment. ADAM17 activity assay and knock-down of ADAM17 revealed that ADAM17 was involved in sTNF-α production. Restoration of ERK activation increased the processing of proADAM17 into ADAM17 in PLA2-treated cells, while inactivation of ERK reduced ADAM17 maturation in untreated and SB202190/BPB-PLA2-treated cells. Removal of cell surface heparan sulfate abrogated PLA2 and SB202190/BPB-PLA2 effect on ADAM17 maturation. Taken together, the present data reveal that PLA2 suppresses ERK-mediated ADAM17 maturation, thus reducing sTNF-α production in U937 cells. Moreover, the binding with heparan sulfate is crucial for the PLA2 effect.
Collapse
Affiliation(s)
- Ying-Jung Chen
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Hui-Chen Lin
- Department of Nutrition Room, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan
| | - Ku-Chung Chen
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Shinne-Ren Lin
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
19
|
Faber TW, Pullen NA, Fernando JFA, Kolawole EM, McLeod JJA, Taruselli M, Williams KL, Rivera KO, Barnstein BO, Conrad DH, Ryan JJ. ADAM10 is required for SCF-induced mast cell migration. Cell Immunol 2014; 290:80-8. [PMID: 24950026 DOI: 10.1016/j.cellimm.2014.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/28/2014] [Accepted: 05/07/2014] [Indexed: 11/29/2022]
Abstract
A Disintegrin and Metalloproteinase (ADAM)-10 plays critical roles in neuronal migration and distribution. Recently, ADAM10 deletion was shown to disrupt myelopoiesis. We found that inducible deletion of ADAM10 using Mx1-driven Cre recombinase for a period of three weeks resulted in mast cell hyperplasia in the skin, intestine and spleen. Mast cells express surface ADAM10 in vitro and in vivo, at high levels compared to other immune cells tested. ADAM10 is important for mast cell migration, since ADAM10-deficiency reduced c-Kit-mediated migration. As with some mast cell proteases, ADAM10 expression could be altered by the cytokine microenvironment, being inhibited by IL-10 or TGFβ1, but not by several other T cell-derived cytokines. Collectively these data show that the ADAM10 protease is an important factor in mast cell migration and tissue distribution, and can be manipulated by environmental cues.
Collapse
Affiliation(s)
- Travis W Faber
- Department of Biology, Virginia Commonwealth University (VCU), Richmond, VA 23284-2012, United States
| | - Nicholas A Pullen
- Department of Biology, Virginia Commonwealth University (VCU), Richmond, VA 23284-2012, United States
| | - Josephine F A Fernando
- Department of Biology, Virginia Commonwealth University (VCU), Richmond, VA 23284-2012, United States
| | | | - Jamie J A McLeod
- Department of Biology, Virginia Commonwealth University (VCU), Richmond, VA 23284-2012, United States
| | - Marcela Taruselli
- Department of Biology, Virginia Commonwealth University (VCU), Richmond, VA 23284-2012, United States
| | - Kathryn L Williams
- Department of Biology, Virginia Commonwealth University (VCU), Richmond, VA 23284-2012, United States
| | - Kevin O Rivera
- Department of Biology, Virginia Commonwealth University (VCU), Richmond, VA 23284-2012, United States
| | - Brian O Barnstein
- Department of Biology, Virginia Commonwealth University (VCU), Richmond, VA 23284-2012, United States
| | - Daniel H Conrad
- Department of Microbiology and Immunology, Virginia Commonwealth University (VCU), Richmond, VA 23284-2012, United States
| | - John J Ryan
- Department of Biology, Virginia Commonwealth University (VCU), Richmond, VA 23284-2012, United States.
| |
Collapse
|
20
|
Chen YJ, Chang LS. Hydroquinone-induced miR-122 down-regulation elicits ADAM17 up-regulation, leading to increased soluble TNF-α production in human leukemia cells with expressed Bcr/Abl. Biochem Pharmacol 2013; 86:620-31. [PMID: 23791922 DOI: 10.1016/j.bcp.2013.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Revised: 06/12/2013] [Accepted: 06/12/2013] [Indexed: 01/10/2023]
Abstract
Studies on HQ-treated human leukemia K562 (Bcr/Abl-positive) cells were conducted to address the hydroquinone (HQ) mechanism that promotes soluble TNF-α (sTNF-α) production. HQ post-translationally down-regulated cell surface TNF-α expression increases the release of sTNF-α into K562 cell culture medium. Meanwhile, HQ increased ADAM17 mRNA stability, leading to ADAM17 up-regulation in HQ-treated cells. Knock-down of ADAM17 abrogated HQ-induced sTNF-α secretion. HQ-evoked miR-122 down-regulation was proven to promote ADAM17 mRNA stability and up-regulate ADAM17 expression. HQ-induced p38 MAPK and JNK activation were responsible for suppression of miR-122 promoter luciferase activity and miR-122 expression. Activation of p38 MAPK and JNK elicited phosphorylation of c-Jun, ATF-2 and c-Fos, and knock-down of c-Jun, ATF-2 and c-Fos restored miR-122 expression in HQ-treated cells. Chromatin immunoprecipitating and DNA affinity purification assay revealed c-Jun, ATF-2 and c-Fos binding to the miR-122 gene promoter region. Moreover, HQ-induced sTNF-α production in Bcr/Abl-positive leukemia cell lines KU812 and MEG-01 was also connected with miR-122 down-regulation and ADAM17 up-regulation, while HQ was unable to affect miR-122 and ADAM-17 expression on Bcr/Abl-negative leukemia U937 cells. Taken together, our data indicate that HQ induces down-regulation of miR-122 expression, leading to ADAM17 up-regulation and ADAM17-mediated TNF-α shedding. Consequently, HQ treatment increases the production of sTNF-α in leukemia cells with expressed Bcr/Abl.
Collapse
Affiliation(s)
- Ying-Jung Chen
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | | |
Collapse
|
21
|
Prox J, Willenbrock M, Weber S, Lehmann T, Schmidt-Arras D, Schwanbeck R, Saftig P, Schwake M. Tetraspanin15 regulates cellular trafficking and activity of the ectodomain sheddase ADAM10. Cell Mol Life Sci 2012; 69:2919-32. [PMID: 22446748 PMCID: PMC11114675 DOI: 10.1007/s00018-012-0960-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 02/18/2012] [Accepted: 03/08/2012] [Indexed: 10/28/2022]
Abstract
A disintegrin and metalloproteinase10 (ADAM10) has been implicated as a major sheddase responsible for the ectodomain shedding of a number of important surface molecules including the amyloid precursor protein and cadherins. Despite a well-documented role of ADAM10 in health and disease, little is known about the regulation of this protease. To address this issue we conducted a split-ubiquitin yeast two-hybrid screen to identify membrane proteins that interact with ADAM10. The yeast experiments and co-immunoprecipitation studies in mammalian cell lines revealed tetraspanin15 (TSPAN15) to specifically associate with ADAM10. Overexpression of TSPAN15 or RNAi-mediated knockdown of TSPAN15 led to significant changes in the maturation process and surface expression of ADAM10. Expression of an endoplasmic reticulum (ER) retention mutant of TSPAN15 demonstrated an interaction with ADAM10 already in the ER. Pulse-chase experiments confirmed that TSPAN15 accelerates the ER-exit of the ADAM10-TSPAN15 complex and stabilizes the active form of ADAM10 at the cell surface. Importantly, TSPAN15 also showed the ability to mediate the regulation of ADAM10 protease activity exemplified by an increased shedding of N-cadherin and the amyloid precursor protein. In conclusion, our data show that TSPAN15 is a central modulator of ADAM10-mediated ectodomain shedding. Therapeutic manipulation of its expression levels may be an additional approach to specifically regulate the activity of the amyloid precursor protein alpha-secretase ADAM10.
Collapse
Affiliation(s)
- Johannes Prox
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, 24098 Kiel, Germany
| | - Michael Willenbrock
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, 24098 Kiel, Germany
| | - Silvio Weber
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, 24098 Kiel, Germany
| | - Tobias Lehmann
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, 24098 Kiel, Germany
| | - Dirk Schmidt-Arras
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, 24098 Kiel, Germany
| | - Ralf Schwanbeck
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, 24098 Kiel, Germany
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, 24098 Kiel, Germany
| | - Michael Schwake
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, 24098 Kiel, Germany
| |
Collapse
|
22
|
Richards FM, Tape CJ, Jodrell DI, Murphy G. Anti-tumour effects of a specific anti-ADAM17 antibody in an ovarian cancer model in vivo. PLoS One 2012; 7:e40597. [PMID: 22792380 PMCID: PMC3394719 DOI: 10.1371/journal.pone.0040597] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 06/11/2012] [Indexed: 11/19/2022] Open
Abstract
ADAM 17 (TNF-α converting enzyme, TACE) is a potential target for cancer therapy, but the small molecule inhibitors reported to date are not specific to this ADAM family member. This membrane-bound metalloproteinase is responsible for ectodomain shedding of pathologically significant substrates including TNF-α and EGFR ligands. The aim of this study was to evaluate the pharmacokinetics, pharmacodynamics and anti-tumour efficacy of the first specific inhibitor, an anti-human ADAM17 IgG antibody, clone D1(A12). We used intraperitoneal xenografts of the human ovarian cancer cell line IGROV1-Luc in Balb/c nude mice, chosen because it was previously reported that growth of these xenografts is inhibited by knock-down of TNF-α. In vitro, 200 nM D1(A12) inhibited shedding of ADAM17 substrates TNF-α, TNFR1-α, TGF-α, amphiregulin (AREG), HB-EGF and IL-6Rα, from IGROV1-Luc cells, (4.7 nM IC(50) for TNF-α shedding). In IGROV1-Luc xenografts in vivo, D1(A12) IgG showed pharmacokinetic properties suitable for efficacy studies, with a single i.p. dose of 10 mg/kg D1(A12) sufficient to maintain IgG plasma and ascites fluid concentrations above 100 nM for more than 7 days. The plasma half life was 8.6 days. Next, an efficacy study was performed, dosing D1(A12) or anti-human TNF-α antibody infliximab at 10 mg/kg q7d, quantifying IGROV1-Luc tumour burden by bioluminescence. D1(A12) IgG showed a significant reduction in tumour growth (p = 0.005), 56% of vehicle control. Surprisingly, D1(A12) did not reduce the concentration of circulating human TNF-α, suggesting that another enzyme may compensate for inhibition of ADAM17 in vivo (but not in vitro). However, D1(A12) did show clear pharmacodynamic effects in the mice, with significant inhibition of shedding from tumour of ADAM17 substrates TNFR1-α, AREG, and TGF-α (4-15-fold reductions, p<0.0001 for all three). Thus, D1(A12) has anti-ADAM17 activity in vivo, inhibits shedding of EGFR ligands and has potential for use in EGF ligand-dependent tumours.
Collapse
Affiliation(s)
- Frances M Richards
- Pharmacology & Drug Development Group, Cancer Research UK Cambridge Research Institute, and Department of Oncology, University of Cambridge, Cambridge, United Kingdom.
| | | | | | | |
Collapse
|
23
|
Liu WH, Chang LS. Suppression of Akt/Foxp3-mediated miR-183 expression blocks Sp1-mediated ADAM17 expression and TNFα-mediated NFκB activation in piceatannol-treated human leukemia U937 cells. Biochem Pharmacol 2012; 84:670-80. [PMID: 22705645 DOI: 10.1016/j.bcp.2012.06.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 06/07/2012] [Accepted: 06/07/2012] [Indexed: 12/14/2022]
Abstract
To address the mechanism of piceatannol in inhibiting TNFα-mediated pathway, studies on piceatannol-treated human leukemia U937 cells were conducted. Piceatannol treatment reduced TNFα shedding and NFκB activation and decreased the release of soluble TNFα into the culture medium of U937 cells. Moreover, ADAM17 expression was down-regulated in piceatannol-treated cells. Over-expression of ADAM17 abrogated the ability of piceatannol to suppress TNFα-mediated NFκB activation. Piceatannol-evoked β-TrCP up-regulation promoted Sp1 degradation, thus reducing transcriptional level of ADAM17 gene in U937 cells. Piceatannol treatment induced p38 MAPK phosphorylation but inactivation of Akt and ERK. In contrast to p38 MAPK inhibitor or restoration of ERK activation, transfection of constitutive active Akt abolished the effect of piceatannol on β-TrCP, Sp1 and ADAM17 expression. Piceatannol-elicited down-regulation of miR-183 expression was found to cause β-TrCP up-regulation. Inactivation of Akt resulted in Foxp3 down-regulation and reduced miR-183 expression in piceatannol-treated cells. Knock-down of Foxp3 and chromatin immunoprecipitating revealed that Foxp3 genetically regulated transcription of miR-183 gene. Taken together, our data indicate that suppression of Akt/Foxp3-mediated miR-183 expression blocks Sp1-mediated ADAM17 expression in piceatannol-treated U937 cells. Consequently, piceatannol suppresses TNFα shedding, leading to inhibition of TNFα/NFκB pathway.
Collapse
Affiliation(s)
- Wen-Hsin Liu
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | | |
Collapse
|
24
|
Physiological functions of the amyloid precursor protein secretases ADAM10, BACE1, and Presenilin. Exp Brain Res 2011; 217:331-41. [DOI: 10.1007/s00221-011-2952-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 11/07/2011] [Indexed: 12/16/2022]
|