1
|
Zeng J, Wang C, Ruge F, Ji EK, Martin TA, Sanders AJ, Jia S, Hao C, Jiang WG. EPLIN, a prospective oncogenic molecule with contribution to growth, migration and drug resistance in pancreatic cancer. Sci Rep 2024; 14:30850. [PMID: 39730634 DOI: 10.1038/s41598-024-81485-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024] Open
Abstract
Most pancreatic cancer patients are diagnosed at advanced stages, with poor survival rates and drug resistance making pancreatic cancer one of the highest causes of cancer death in the UK. Understanding the underlying mechanism behind its carcinogenesis, metastasis and drug resistance has become an essential task for researchers. We have discovered that a well-established tumour suppressor, EPLIN, has an oncogenic rather than suppressive role in pancreatic cancer. Notably, upregulation of EPLIN was observed in pancreatic cancer samples compared to normal samples at RNA and protein levels. Moreover, the presence of EPLIN resulted in poor clinical outcomes in patients. We also report that inhibition of EPLIN led to reduced cellular growth and migration in pancreatic cancer cells. EPLIN regulates expression and phosphorylation levels of several key players in MAPK and PIK3CA-AKT signalling pathways, as well as key contributors of EMT. Furthermore, EPLIN mediates the inhibitory ability PIK3 kinases, MEK and ERK inhibitors have on cell migration. EPLIN was also found to have an impact on pancreatic cancer cells response to chemotherapeutic and EGFR/HER2 targeted therapeutic agents, namely gemcitabine, fluorouracil (5FU) and neratinib (Nerlynx).
Collapse
Affiliation(s)
- Jianyuan Zeng
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK
| | - Cai Wang
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK
| | - Fiona Ruge
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK
| | - Edison Ke Ji
- Gastrointestinal Cancer Centre, Peking University Cancer Hospital, Peking University, Fucheng Road, Haidian District, Beijing, China
| | - Tracey A Martin
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK
| | - Andrew J Sanders
- School of Education and Science, University of Gloucestershire, Francis Close Hall, Swindon Road, Cheltenham, GL50 4AZ, UK
| | - Shuqin Jia
- Gastrointestinal Cancer Centre, Peking University Cancer Hospital, Peking University, Fucheng Road, Haidian District, Beijing, China
| | - Chunyi Hao
- Gastrointestinal Cancer Centre, Peking University Cancer Hospital, Peking University, Fucheng Road, Haidian District, Beijing, China
| | - Wen G Jiang
- School of Medicine, Cardiff University, Henry Wellcome Building, Cardiff, CF14 4XN, UK.
| |
Collapse
|
2
|
Karimi F, Dinarvand N, Sabaghan M, Azadbakht O, Ataee S, Kharazinejad E, Moazamfard M. Diabetes and ovarian cancer: risk factors, molecular mechanisms and impact on prognosis. Endocrine 2024; 83:1-9. [PMID: 37552417 DOI: 10.1007/s12020-023-03477-6] [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: 01/06/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND AND AIM Diabetes mellitus has been linked to a lower rate of cancer survival and an increase in the incidence of most malignancies. Investigations showed that diabetes might affect ovarian cancer (OC) prognosis and survival. Based on the current information, this study intends to review the risk factors, molecular pathways, and impact of diabetes on OC. METHODS The data was derived from online databases, including Web of Science, PubMed, and Scopus. The inclusion criteria were original studies, which included the risk factors, molecular mechanisms, and impact of diabetes on OC. The effect of different antidiabetic drugs was also discussed in this manuscript. All of the clinical, in vivo, and in vitro studies were included in the present study. RESULTS The diagnosis of diabetes mellitus negatively affects the survival and prognosis in OC cases. The epidemiologic data shows that the risk of OC increases in patients with diabetes mellitus compared to the healthy population. Insulin-like growth factors family was raised in diabetic patients, which target several mechanisms, including targeting oxidative stress, angiogenesis, and tumor markers. Antidiabetic drugs such as metformin, sitagliptin, and rosiglitazone have a promising effect on elongation of survival and enhancement of prognosis in OC patients. CONCLUSIONS Diabetes mellitus is a significant risk factor for OC in women, and it negatively impacts survival and prognosis. Molecular mechanisms such as IGF family, oxidative stress, and inflammatory cytokines have been identified to explain this relationship. Antidiabetic drugs like metformin, sitagliptin, and rosiglitazone have shown promise in improving survival and prognosis of OC patients.
Collapse
Affiliation(s)
- Farzaneh Karimi
- Department of Physiology, Behbahan Faculty of Medical Sciences, Behbahan, Iran.
| | - Negar Dinarvand
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Sabaghan
- Department of Parasiotology, Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Omid Azadbakht
- Department of Radiology Technology, Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Shima Ataee
- Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Ebrahim Kharazinejad
- Department of Anatomy, Faculty of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| | - Mostafa Moazamfard
- Instructor of Operating Room, Behbahan Faculty of Medical Sciences, Behbahan, Iran.
| |
Collapse
|
3
|
Church FC. Suggestions on leading an academic research laboratory group. Open Life Sci 2022; 17:599-609. [PMID: 35800075 PMCID: PMC9202531 DOI: 10.1515/biol-2022-0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/24/2022] [Accepted: 03/25/2022] [Indexed: 11/15/2022] Open
Abstract
This commentary is about running an academic research laboratory group, including some reflections, memories, and tips on effectively managing such a group of scientists focused on one’s research. The author’s academic career has spanned from 1982 to 2022, including postdoctoral research associate through the rank of professor with tenure. Currently, the author is in the final year of 3 years of phased retirement. One must be willing to work hard at running a research laboratory. Also, stay focused on funding the laboratory tasks and publishing one’s work. Recruit the best people possible with advice from the collective laboratory group. Laboratory group members felt more like they were a part of a collective family than simply employees; however, what works best for the researcher is what matters. Several other points to discuss will include managing university roles, recruiting laboratory personnel, getting recognition, dealing with intellectual property rights, and publishing work. In closing, there are many more positives than negatives to leading a research laboratory group. Finally, one cannot replace the unforgettable memories and the legacy of a research laboratory group.
Collapse
Affiliation(s)
- Frank C. Church
- Department of Pathology and Laboratory Medicine, The University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
4
|
Nohawica M, Errachid A, Wyganowska-Swiatkowska M. Adipose-PAS interactions in the context of its localised bio-engineering potential (Review). Biomed Rep 2021; 15:70. [PMID: 34276988 PMCID: PMC8278035 DOI: 10.3892/br.2021.1446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/05/2021] [Indexed: 11/24/2022] Open
Abstract
Adipocytes are a known source of stem cells. They are easy to harvest, and are a suitable candidate for autogenous grafts. Adipose derived stem cells (ADSCs) have multiple target tissues which they can differentiate into, including bone and cartilage. In adipose tissue, ADSCs are able to differentiate, as well as providing energy and a supply of cytokines/hormones to manage the hypoxic and lipid/hormone saturated adipose environment. The plasminogen activation system (PAS) controls the majority of proteolytic activities in both adipose and wound healing environments, allowing for rapid cellular migration and tissue remodelling. While the primary activation pathway for PAS occurs through the urokinase plasminogen activator (uPA), which is highly expressed by endothelial cells, its function is not limited to enabling revascularisation. Proteolytic activity is dependent on protease activation, localisation, recycling mechanisms and substrate availability. uPA and uPA activated plasminogen allows pluripotent cells to arrive to new local environments and fulfil the niche demands. However, overstimulation, the acquisition of a migratory phenotype and constant protein turnover can be unconducive to the formation of structured hard and soft tissues. To maintain a suitable healing pattern, the proteolytic activity stimulated by uPA is modulated by plasminogen activator inhibitor 1. Depending on the physiological settings, different parts of the remodelling mechanism are activated with varying results. Utilising the differences within each microenvironment to recreate a desired niche is a valid therapeutic bio-engineering approach. By controlling the rate of protein turnover combined with a receptive stem cell lineage, such as ADSC, a novel avenue on the therapeutic opportunities may be identified, which can overcome limitations, such as scarcity of stem cells, low angiogenic potential or poor host tissue adaptation.
Collapse
Affiliation(s)
- Michal Nohawica
- Chair and Department of Dental Surgery and Periodontology, Poznan University of Medicinal Sciences, Poznan, Greater Poland 60-812, Poland
| | - Abdelmounaim Errachid
- Chair and Department of Dental Surgery and Periodontology, Poznan University of Medicinal Sciences, Poznan, Greater Poland 60-812, Poland
- Earth and Life Institute, University Catholique of Louvain, Louvain-la-Neuve B-1348, Belgium
| | - Marzena Wyganowska-Swiatkowska
- Chair and Department of Dental Surgery and Periodontology, Poznan University of Medicinal Sciences, Poznan, Greater Poland 60-812, Poland
| |
Collapse
|
5
|
Li L, Wang Y, Yu X, Bao Y, An L, Wei X, Yu W, Liu B, Li J, Yang J, Xia Y, Liu G, Cao F, Zhang X, Zhao D. Bone marrow mesenchymal stem cell-derived exosomes promote plasminogen activator inhibitor 1 expression in vascular cells in the local microenvironment during rabbit osteonecrosis of the femoral head. Stem Cell Res Ther 2020; 11:480. [PMID: 33176873 PMCID: PMC7656701 DOI: 10.1186/s13287-020-01991-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Background Nontraumatic osteonecrosis of the femoral head (NONFH) is a highly disabling orthopedic disease in young individuals. Plasminogen activator inhibitor 1 (PAI-1) has been reported to be positively associated with NONFH. We aimed to investigate the dysregulating PAI-1 in bone marrow mesenchymal stem cells (BMMSCs) and vascular cells in rabbit steroid-induced NONFH. Methods To verify the hypothesis that BMMSCs could promote thrombus formation in a paracrine manner, we collected exosomes from glucocorticoid-treated BMMSCs (GB-Exo) to determine their regulatory effects on vascular cells. microRNA sequencing was conducted to find potential regulators in GB-Exo. Utilizing gain-of-function and knockdown approaches, we testified the regulatory effect of microRNA in exosomes. Results The expression of PAI-1 was significantly increased in the local microenvironment of the femoral head in the ONFH model. GB-Exo promoted PAI-1 expression in vascular smooth muscle cells and vascular endothelial cells. We also revealed that miR-451-5p in GB-Exo plays a crucial role for the elevated PAI-1. Moreover, we identified miR-133b-3p and tested its role as a potential inhibitor of PAI-1. Conclusions This study provided considerable evidence for BMMSC exosomal miR-mediated upregulation of the fibrinolytic regulator PAI-1 in vascular cells. The disruption of coagulation and low fibrinolysis in the femoral head will eventually lead to a disturbance in the microcirculation of NONFH. We believe that our findings could be of great significance for guiding clinical trials in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-020-01991-2.
Collapse
Affiliation(s)
- Lu Li
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China.,Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Yikai Wang
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China.,Medical College of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Xiaobing Yu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Yongming Bao
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, People's Republic of China
| | - Lijia An
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, People's Republic of China
| | - Xiaowei Wei
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Weiting Yu
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Baoyi Liu
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China.,Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Junlei Li
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Jiahui Yang
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Yan Xia
- Department of Pathology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Ge Liu
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Fang Cao
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Xiuzhi Zhang
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Dewei Zhao
- National-Local Joint Engineering Laboratory for the Development of Orthopedic Implant Materials, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China. .,Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China.
| |
Collapse
|
6
|
Sulfated polysaccharide of Sepiella Maindroni ink inhibits the migration, invasion and matrix metalloproteinase-2 expression through suppressing EGFR-mediated p38/MAPK and PI3K/Akt/mTOR signaling pathways in SKOV-3 cells. Int J Biol Macromol 2018; 107:349-362. [DOI: 10.1016/j.ijbiomac.2017.08.178] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/22/2017] [Accepted: 08/31/2017] [Indexed: 12/20/2022]
|
7
|
Guo X, Guo N, Zhao J, Cai Y. Active targeting co-delivery system based on hollow mesoporous silica nanoparticles for antitumor therapy in ovarian cancer stem-like cells. Oncol Rep 2017; 38:1442-1450. [PMID: 28731164 PMCID: PMC5549023 DOI: 10.3892/or.2017.5829] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/21/2017] [Indexed: 01/01/2023] Open
Abstract
The combination of nanocarriers and chemotherapy drugs can release the chemotherapy drugs to the tumor tissue, which can enhance the antitumor effect and reduce the adverse reactions at the same time. In this study, a co-delivery system based on hollow mesoporous silica nanoparticles (HMSN) was developed and characterized. We also investigated the in vitro effect of this system on CD117+CD44+A2780 cell line. HMSN was selected as the nanocarrier, with -COOH modified on the surface and doxorubicin (DOX), NVP-AEW 541 (NVP) loaded inside. IGF‑1R was chosen as the drug target, apoptosis rate and expression of cyclin B1, Bax, Bcl-xl, p-Akt were used to evaluate the antitumor effect of HMSN‑COOH@DOX fluorescence NVP. The HMSN co-delivery system was successfully synthesized with encapsulation efficiency of 37% (DOX) and 44% (NVP), and high PH-sensitive property was observed. The apoptosis rate of CD117+CD44+A2780 ovarian cancer stem-like cells treated by HMSN co-delivery system were almost 3 times higher than those of the free drugs group. The expression of Bax was significantly increased while Bcl-xl, and p-Akt reduced (P≤0.05). These data indicate that the co-delivery system demonstrated a high efficiency in promoting apoptosis in ovarian cancer stem-like cells by targeting IGF‑1R, but further study is still needed.
Collapse
Affiliation(s)
- Xin Guo
- Medical School, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Nan Guo
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jianwen Zhao
- Medical School, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yunlang Cai
- Department of Gynaecology and Obstetrics, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
8
|
Birtolo C, Go VLW, Ptasznik A, Eibl G, Pandol SJ. Phosphatidylinositol 3-Kinase: A Link Between Inflammation and Pancreatic Cancer. Pancreas 2016; 45:21-31. [PMID: 26658038 PMCID: PMC4859755 DOI: 10.1097/mpa.0000000000000531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Even though a strong association between inflammation and cancer has been widely accepted, the underlying precise molecular mechanisms are still largely unknown. A complex signaling network between tumor and stromal cells is responsible for the infiltration of inflammatory cells into the cancer microenvironment. Tumor stromal cells such as pancreatic stellate cells (PSCs) and immune cells create a microenvironment that protects cancer cells through a complex interaction, ultimately facilitating their local proliferation and their migration to different sites. Furthermore, PSCs have multiple functions related to local immunity, angiogenesis, inflammation, and fibrosis. Recently, many studies have shown that members of the phosphoinositol-3-phosphate kinase (PI3K) family are activated in tumor cells, PSCs, and tumor-infiltrating inflammatory cells to promote cancer growth. Proinflammatory cytokines and chemokines secreted by immune cells and fibroblasts within the tumor environment can activate the PI3K pathway both in cancer and inflammatory cells. In this review, we focus on the central role of the PI3K pathway in regulating the cross talk between immune/stromal cells and cancer cells. Understanding the role of the PI3K pathway in the development of chronic pancreatitis and cancer is crucial for the discovery of novel and efficacious treatment options.
Collapse
Affiliation(s)
- Chiara Birtolo
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA,Department of Internal Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Italy
| | - Vay Liang W. Go
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Andrzej Ptasznik
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Stephen J. Pandol
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA,VA Greater Los Angeles Health Care System, Los Angeles, CA
| |
Collapse
|
9
|
Bourgeois DL, Kabarowski KA, Porubsky VL, Kreeger PK. High-grade serous ovarian cancer cell lines exhibit heterogeneous responses to growth factor stimulation. Cancer Cell Int 2015; 15:112. [PMID: 26648788 PMCID: PMC4672525 DOI: 10.1186/s12935-015-0263-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/26/2015] [Indexed: 02/04/2023] Open
Abstract
Background The factors driving the onset and progression of ovarian cancer are not well understood. Recent reports have identified cell lines that are representative of the genomic pattern of high-grade serous ovarian cancer (HGSOC), in which greater than 90 % of tumors have a mutation in TP53. However, many of these representative cell lines have not been widely used so it is unclear if these cell lines capture the variability that is characteristic of the disease. Methods We investigated six TP53-mutant HGSOC cell lines (Caov3, Caov4, OV90, OVCA432, OVCAR3, and OVCAR4) for migration, MMP2 expression, proliferation, and VEGF secretion, behaviors that play critical roles in tumor progression. In addition to comparing baseline variation between the cell lines, we determined how these behaviors changed in response to four growth factors implicated in ovarian cancer progression: HB-EGF, NRG1β, IGF1, and HGF. Results Baseline levels of each behavior varied across the cell lines and this variation was comparable to that seen in tumors. All four growth factors impacted cell proliferation or VEGF secretion, and HB-EGF, NRG1β, and HGF impacted wound closure or MMP2 expression in at least two cell lines. Growth factor-induced responses demonstrated substantial heterogeneity, with cell lines sensitive to all four growth factors, a subset of the growth factors, or none of the growth factors, depending on the response of interest. Principal component analysis demonstrated that the data clustered together based on cell line rather than growth factor identity, suggesting that response is dependent on intrinsic qualities of the tumor cell rather than the growth factor. Conclusions Significant variation was seen among the cell lines, consistent with the heterogeneity of HGSOC. Electronic supplementary material The online version of this article (doi:10.1186/s12935-015-0263-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Danielle L Bourgeois
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705 USA
| | - Karl A Kabarowski
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705 USA
| | - Veronica L Porubsky
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705 USA
| | - Pamela K Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705 USA
| |
Collapse
|
10
|
Ibrahim MA, Abou-Seri SM, Hanna MM, Abdalla MM, El Sayed NA. Design, synthesis and biological evaluation of novel condensed pyrrolo[1,2-c]pyrimidines featuring morpholine moiety as PI3Kα inhibitors. Eur J Med Chem 2015; 99:1-13. [DOI: 10.1016/j.ejmech.2015.05.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 03/30/2015] [Accepted: 05/22/2015] [Indexed: 11/25/2022]
|
11
|
Modulation of urokinase plasminogen activator system by poly(ADP-ribose)polymerase-1 inhibition. Cytotechnology 2014; 68:783-94. [PMID: 25471275 DOI: 10.1007/s10616-014-9829-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 11/21/2014] [Indexed: 10/24/2022] Open
Abstract
The urokinase plasminogen activator (uPA) system is a complex regulator of extracellular proteolysis which is involved in various physiological and pathological processes. The major components of this system are the serine protease uPA, two inhibitors PAI-1 and PAI-2, and the receptor uPAR. It has been previously shown by several groups that the uPA system has an important role in cancer progression and therefore its possible prognostic and therapeutic value has been evaluated. The aim of this study is to tackle the role of poly(ADP-ribosyl)ation in the induction of uPA activity in a glioblastoma cell line, A1235. This cell line is sensitive to alkylation damage and is a model for drug treatment. The components of the uPA system and the level of DNA damage were analyzed after alkylation agent treatment in combination with poly(ADP-ribose)polymerase-1 (PARP-1) inhibition. Here we show that the increase in uPA activity results from the net balance change between uPA and its inhibitor at mRNA level. Further, PARP-1 inhibition exerts its influence on uPA activity through DNA damage increase. Involvement of several signaling pathways, as well as cell specific regulation influencing the uPA system are discussed.
Collapse
|
12
|
Kamato D, Rostam MA, Piva TJ, Babaahmadi Rezaei H, Getachew R, Thach L, Bernard R, Zheng W, Little PJ, Osman N. Transforming growth factor β-mediated site-specific Smad linker region phosphorylation in vascular endothelial cells. J Pharm Pharmacol 2014; 66:1722-33. [DOI: 10.1111/jphp.12298] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 06/29/2014] [Indexed: 01/20/2023]
Abstract
Abstract
Objectives
Transforming growth factor (TGF)-β regulates the function of vascular endothelial cells and may be involved in endothelial dysfunction. The canonical TGF-β pathway involves TGF-β receptor-mediated carboxy-terminal phosphorylation of Smad2; however, TGF-β signalling also activates numerous serine/threonine kinases that phosphorylate Smad2 in its linker region. The expression of phosphorylated Smad linker proteins were determined following TGF-β stimulation in the absence and presence of different serine/threonine kinase inhibitors in vascular endothelial cells.
Methods
Proteins were quantified by Western blotting using specific antibodies to individual phosphorylated Smad2 linker region residues.
Key findings
TGF-β mediated the phosphorylation of all four Smad2 linker region residues of interest. Erk and Jnk specifically phosphorylate Ser245 while all mitogen-activated protein kinases phosphorylate Ser250 and Ser255. Thr220 and Ser245 are phosphorylated by phosphoinositide 3 kinase (PI3K), while Ser255 was phosphorylated by the PI3K/Akt pathway. CDK and GSK-3 were shown to phosphorylate Thr220 and Ser245. TGF-β also mediated plasminogen activator inhibitor-1 gene expression that was attenuated by p38 and CDK inhibitors.
Conclusions
TGF-β-mediated phosphorylation of individual serine/threonine sites in the linker region of Smad2 occurs in a highly specific manner by kinases. These phosphorylations provide an opportunity to further understand a therapeutically targeted and very specific signalling pathway in vascular endothelial cells.
Collapse
Affiliation(s)
- Danielle Kamato
- Discipline of Pharmacy, School of Medical Sciences and Diabetes Complications Group, Health Innovations Research Institute, RMIT University, Bundoora, Vic, Australia
| | - Muhamad Ashraf Rostam
- Discipline of Pharmacy, School of Medical Sciences and Diabetes Complications Group, Health Innovations Research Institute, RMIT University, Bundoora, Vic, Australia
| | - Terence J Piva
- Discipline of Cell Biology, School of Medical Sciences, RMIT University, Bundoora, Vic, Australia
| | - Hossein Babaahmadi Rezaei
- Discipline of Pharmacy, School of Medical Sciences and Diabetes Complications Group, Health Innovations Research Institute, RMIT University, Bundoora, Vic, Australia
- Department of Clinical Biochemistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Robel Getachew
- Discipline of Pharmacy, School of Medical Sciences and Diabetes Complications Group, Health Innovations Research Institute, RMIT University, Bundoora, Vic, Australia
| | - Lyna Thach
- Discipline of Pharmacy, School of Medical Sciences and Diabetes Complications Group, Health Innovations Research Institute, RMIT University, Bundoora, Vic, Australia
| | - Rebekah Bernard
- Discipline of Pharmacy, School of Medical Sciences and Diabetes Complications Group, Health Innovations Research Institute, RMIT University, Bundoora, Vic, Australia
| | - Wenhua Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Peter J Little
- Discipline of Pharmacy, School of Medical Sciences and Diabetes Complications Group, Health Innovations Research Institute, RMIT University, Bundoora, Vic, Australia
- Departments of Medicine, Nursing and Health Sciences and Immunology, Monash University School of Medicine, Central and Eastern Clinical School, Alfred Health, Prahran, Vic, Australia
| | - Narin Osman
- Discipline of Pharmacy, School of Medical Sciences and Diabetes Complications Group, Health Innovations Research Institute, RMIT University, Bundoora, Vic, Australia
- Departments of Medicine, Nursing and Health Sciences and Immunology, Monash University School of Medicine, Central and Eastern Clinical School, Alfred Health, Prahran, Vic, Australia
| |
Collapse
|
13
|
Zhang Y, Zhan Y, Zhang D, Dai B, Ma W, Qi J, Liu R, He L. Eupolyphaga sinensis walker displays inhibition on hepatocellular carcinoma through regulating cell growth and metastasis signaling. Sci Rep 2014; 4:5518. [PMID: 24980220 PMCID: PMC4076680 DOI: 10.1038/srep05518] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/13/2014] [Indexed: 11/09/2022] Open
Abstract
Tumor growth and metastasis are responsible for most cancer patients' deaths. Here, we report that eupolyphaga sinensis walker has an essential role in resisting hepatocellular carcinoma growth and metastasis. Compared with proliferation, colony formation, transwell assay and transplantable tumor in nude mouse in vitro and vivo, eupolyphaga sinensis walker extract (ESWE) showed good inhibition on the SMMC-7721 cell growth and metastasis. Using genome-wide microarray analysis, we found the down-regulated growth and metastasis factors, and selected down-regulated genes were confirmed by real-time PCR. Knockdown of a checkpoint PKCβ by siRNA significantly attenuated tumor inhibition and metastasis effects of ESWE. Moreover, our results indicate ESWE inhibits HCC growth by not only downregulating the signaling of PKCβ, Akt, m-TOR, Erk1/2, MEK-2, Raf and JNK-1, but also increasing cyclin D1 protein levels and decreasing amount of cyclin E, cyclin B1 and cdc2 of the cycle proteins. At the same time, ESWE reduced MMP2, MMP9 and CXCR4, PLG, NFκB and P53 activities. Overall, our studies demonstrate that ESWE is a key factor in growth and metastasis signaling inhibitor targeting the PKC, AKT, MAPK signaling and related metastasis signaling, having potential in cancer therapy.
Collapse
Affiliation(s)
- Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Yingzhuan Zhan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Dongdong Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Bingling Dai
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Weina Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Junpeng Qi
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Rui Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Langchong He
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| |
Collapse
|
14
|
Yang Z, Bach LA. Differential Effects of Insulin-Like Growth Factor Binding Protein-6 (IGFBP-6) on Migration of Two Ovarian Cancer Cell Lines. Front Endocrinol (Lausanne) 2014; 5:231. [PMID: 25601855 PMCID: PMC4283657 DOI: 10.3389/fendo.2014.00231] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/14/2014] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION IGFBP-6 inhibits angiogenesis as well as proliferation and survival of rhabdomyosarcoma cells. However, it promotes migration of these cells in an IGF-independent manner. The IGF system is implicated in ovarian cancer, so we studied the effects of IGFBP-6 in ovarian cancer cells. METHODS The effects of wild type (wt) and a non-IGF-binding mutant (m) of IGFBP-6 on migration of HEY and SKOV3 ovarian cancer cells, which, respectively, represent aggressive and transitional cancers, were studied. ERK and JNK phosphorylation were measured by Western blotting. RESULTS IGF-II, wt-, and mIGFBP-6 each promoted SKOV3 cell migration by 77-98% (p < 0.01). In contrast, IGF-II also increased HEY cell migration to 155 ± 13% of control (p < 0.001), but wt-IGFBP-6 and mIGFBP-6 decreased migration to 62 ± 5 and 66 ± 3%, respectively (p < 0.001). In these cells, coincubation of IGF-II with wt but not mIGFBP-6 increased migration. MAP kinase pathways are involved in IGFBP-6-induced rhabdomyosarcoma cell migration, so activation of these pathways was studied in HEY and SKOV3 cells. Wt and mIGFBP-6 increased ERK phosphorylation by 62-99% in both cell lines (p < 0.05). Wt-IGFBP-6 also increased JNK phosphorylation by 139-153% in both cell lines (p < 0.05), but the effect of mIGFBP-6 was less clear. ERK and JNK inhibitors partially inhibited the migratory effects of wt and mIGFBP-6 in SKOV3 cells, whereas the ERK inhibitor partially restored wt and mIGFBP-6-induced inhibition of HEY cell migration. The JNK inhibitor had a lesser effect on the actions of wtIGFBP-6 and no effect on the actions of mIGFBP-6 in HEY cells. CONCLUSION IGFBP-6 has opposing effects on migration of HEY and SKOV3 ovarian cancer cells, but activates MAP kinase pathways in both. Delineating the pathways underlying the differential effects on migration will increase our understanding of ovarian cancer metastasis and shed new light on the IGF-independent effects of IGFBP-6.
Collapse
Affiliation(s)
- Zhiyong Yang
- Department of Medicine (Alfred), Monash University, Prahran, VIC, Australia
| | - Leon A. Bach
- Department of Medicine (Alfred), Monash University, Prahran, VIC, Australia
- Department of Endocrinology and Diabetes, Alfred Hospital, Melbourne, VIC, Australia
- *Correspondence: Leon A. Bach, Department of Endocrinology and Diabetes, Alfred Hospital, Commercial Road, Melbourne, VIC 3004, Australia e-mail:
| |
Collapse
|
15
|
Wilson AJ, Liu AY, Roland J, Adebayo OB, Fletcher SA, Slaughter JC, Saskowski J, Crispens MA, Jones HW, James S, Fadare O, Khabele D. TR3 modulates platinum resistance in ovarian cancer. Cancer Res 2013; 73:4758-69. [PMID: 23720056 DOI: 10.1158/0008-5472.can-12-4560] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In metastatic ovarian cancer, resistance to platinum chemotherapy is common. Although the orphan nuclear receptor TR3 (nur77/NR4A1) is implicated in mediating chemotherapy-induced apoptosis in cancer cells, its role in ovarian cancer has not been determined. In an ovarian cancer tissue microarray, TR3 protein expression was elevated in stage I tumors, but downregulated in a significant subset of metastatic tumors. Moreover, TR3 expression was significantly lower in platinum-resistant tumors in patients with metastatic disease, and low TR3 staining was associated with poorer overall and progression-free survival. We have identified a direct role for TR3 in cisplatin-induced apoptosis in ovarian cancer cells. Nucleus-to-cytoplasm translocation of TR3 was observed in cisplatin-sensitive (OVCAR8, OVCAR3, and A2780PAR) but not cisplatin-resistant (NCI/ADR-RES and A2780CP20) ovarian cancer cells. Immunofluorescent analyses showed clear overlap between TR3 and mitochondrial Hsp60 in cisplatin-treated cells, which was associated with cytochrome c release. Ovarian cancer cells with stable shRNA- or transient siRNA-mediated TR3 downregulation displayed substantial reduction in cisplatin effects on apoptotic markers and cell growth in vitro and in vivo. Mechanistic studies showed that the cisplatin-induced cytoplasmic TR3 translocation required for apoptosis induction was regulated by JNK activation and inhibition of Akt. Finally, cisplatin resistance was partially overcome by ectopic TR3 overexpression and by treatment with the JNK activator anisomycin and Akt pathway inhibitor, wortmannin. Our results suggest that disruption of TR3 activity, via downregulation or nuclear sequestration, likely contributes to platinum resistance in ovarian cancer. Moreover, we have described a treatment strategy aimed at overcoming platinum resistance by targeting TR3.
Collapse
Affiliation(s)
- Andrew J Wilson
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, B1100 Medical Center North, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Li Y, Jiang Y, Wan Y, Zhang L, Tang W, Ma J, Wu S, Cheng W. Medroxyprogestogen enhances apoptosis of SKOV-3 cells via inhibition of the PI3K/Akt signaling pathway. J Biomed Res 2013; 27:43-50. [PMID: 23554793 PMCID: PMC3596754 DOI: 10.7555/jbr.27.20120051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 10/07/2011] [Accepted: 05/30/2012] [Indexed: 01/28/2023] Open
Abstract
We sought to assess the effect of progestin on the apoptosis of epithelial ovarian cancer cell line SKOV-3 and via regulation of phosphorylation signaling in. Epithelial ovarian cancer cell line SKOV-3 was treated with medroxyprogestogen, phosphatidylinositol 3-kinase inhibitor LY294002 and vehicle control. Akt, phospho-Akt, Bcl-2 and phospho-Bad proteins were examined by immunoblotting assays. Medroxyprogestogen-induced apoptosis was assessed by MTT assays and Annexin V apoptosis assay. We found no significant difference in Akt and Bad expression in both the medroxyprogestogen groups and the control group. The levels of phospho-Akt, Bcl-2 and phospho-Bad were decreased in all the medroxyprogestogen groups and significantly decreased in the high dose mitogen-activated protein (MAP) group (10 µmol/L). Viability of SKOV-3 was reduced and apparent apoptosis of SKOV-3 cells was observed with increased doses of MAP. The findings suggest that medroxyprogestogen can induce SKOV-3 cell apoptosis by inhibiting Akt phosphorylation.
Collapse
Affiliation(s)
- Yan Li
- Department of Gynecology and Obstetrics, the First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, China;
| | - Yi Jiang
- Department of Gynecology, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Yicong Wan
- Department of Gynecology and Obstetrics, the First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, China;
| | - Lin Zhang
- Department of Gynecology and Obstetrics, the First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, China;
| | - Weiwei Tang
- Department of Gynecology and Obstetrics, the First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, China;
| | - Jingjing Ma
- Department of Gynecology and Obstetrics, the First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, China;
| | - Shan Wu
- Department of Gynecology and Obstetrics, the First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, China;
| | - Wenjun Cheng
- Department of Gynecology, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
17
|
Oh SH, Kang JH, Kyu Woo J, Lee OH, Kim ES, Lee HY. A multiplicity of anti-invasive effects of farnesyl transferase inhibitor SCH66336 in human head and neck cancer. Int J Cancer 2012; 131:537-47. [PMID: 22113431 PMCID: PMC3374925 DOI: 10.1002/ijc.26373] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 06/28/2011] [Indexed: 11/10/2022]
Abstract
Metastasis is a critical event in the progression of head and neck squamous cell carcinoma (HNSCC) and closely correlates with clinical outcome. We previously showed that the farnesyl transferase inhibitor SCH66336 has antitumor activities in HNSCC by inducing the secretion of insulin-like growth factor binding protein 3 (IGFBP-3), which in turn inhibits tumor growth and angiogenesis. In our study, we found that SCH66336 at a sublethal dose for HNSCC inhibited the migration and invasion of HNSCC cells. The inhibitory effect of SCH66336 was associated with the blockade of the IGF-1 receptor (IGF-1R) pathway via suppressing IGF-1R itself and Akt expression. Consistent with previous work, induction of IGFBP-3 by SCH66336 also contributed in part to the anti-invasive effect. SCH66336 treatment also reduced the expression and activity of the urokinase-type plasminogen activator (uPA) and matrix metalloproteinase 2 (MMP-2), both important regulators of tumor metastasis. The effect of SCH66336 on uPA activity was inhibited partly by knockdown of IGFBP-3 using small interfering RNA. The inhibitory effect of SCH66336 on migration or invasion was attenuated partly or completely by knockdown of IGFBP-3, Akt or IGF-1R expression, respectively. Our results demonstrate that the IGF-1R pathway plays a major role in the proliferation, migration and invasion of HNSCC cells, suggesting that therapeutic obstruction of the IGF-1R pathway would be a useful approach to treating patients with HNSCC.
Collapse
Affiliation(s)
- Seung Hyun Oh
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | |
Collapse
|
18
|
Carter JC, Church FC. Mature breast adipocytes promote breast cancer cell motility. Exp Mol Pathol 2012; 92:312-7. [PMID: 22445926 DOI: 10.1016/j.yexmp.2012.03.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 03/08/2012] [Indexed: 12/13/2022]
Abstract
Adipocytes express substances involved in both normal physiology and pathological processes. One such adipocyte protein is the Serpin (serine protease inhibitor) plasminogen activator inhibitor-1 (PAI-1). PAI-1 functions to inhibit urokinase type plasminogen activator (uPA) though PAI-1 itself is also implicated in breast cancer progression. While the role of adipocytes in breast cancer development is not fully understood, obesity is a known risk factor associated with breast cancer. Thus, we characterized adipocytes from breast and omental tissues for PAI-1 and uPA, and the influence of adipocytes on breast cancer cell motility. Using preadipocyte cells from breast and omental adipose tissue, we differentiated each site into mature adipocytes. PAI-1 protein was found in breast adipocytes>omental preadipocytes>omental adipocytes>breast preadipocytes. Interestingly, uPA protein was not detected in any of these cell types. We then incubated breast adipocyte conditioned media (Adip-CM) and preadipocyte conditioned media (PreAdip-CM) on both normal (MCF-10A) and malignant (MCF-10CA1) breast epithelial cell lines. Adip-CM, but not PreAdip-CM, (a) increased cell motility in both MCF-10A and MCF-10CA1 cells; (b) increased cell-associated uPA activity in both cell lines; (c) increased phosphorylated-Akt levels in MCF-10CA1 cells; and (d) gene array profiles show altered expression of several genes associated with cancer adhesion, metastasis and signaling. Our results suggest that mature breast adipocytes are capable of altering the epithelial cell phenotype, producing a more motile cell type and further provide a potential link between obesity and risk of breast cancer.
Collapse
Affiliation(s)
- Jennifer C Carter
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
19
|
Peroxisome Proliferator-Activated Receptor-γ Ligands Alter Breast Cancer Cell Motility through Modulation of the Plasminogen Activator System. JOURNAL OF ONCOLOGY 2011; 2011:594258. [PMID: 22131991 PMCID: PMC3205730 DOI: 10.1155/2011/594258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 07/12/2011] [Accepted: 07/15/2011] [Indexed: 11/29/2022]
Abstract
We investigated peroxisome proliferator-activated receptor-γ (PPAR-γ) ligands effect on cell motility and the plasminogen activator system using normal MCF-10A and malignant MCF-10CA1 cell lines. Ciglitazone reduced both wound-induced migration and chemotaxis. However, the effect was not reversed with pretreatment of cells with the PPAR-γ-specific antagonist GW9662. Immunoblot analysis of conditioned media showed ciglitazone decreased plasminogen activator inhibitor-1 (PAI-1) in both cell lines; this effect was also unaltered by PPAR-γ antagonism. Alternatively, treatment with the ω-6 fatty acid arachidonic acid (ArA), but not the ω-3 fatty acid docosahexanoic acid, increased both MCF-10A cell migration and cell surface uPA activity. Pretreatment with a PPAR-γ antagonist reversed these effects, suggesting that ArA mediates its effect on cell motility and uPA activity through PPAR-γ activation. Collectively, the data suggest PPAR-γ ligands have a differential effect on normal and malignant cell migration and the plasminogen activation system, resulting from PPAR-γ-dependent and PPAR-γ-independent effects.
Collapse
|
20
|
Boldt HB, Conover CA. Overexpression of pregnancy-associated plasma protein-A in ovarian cancer cells promotes tumor growth in vivo. Endocrinology 2011; 152:1470-8. [PMID: 21303951 DOI: 10.1210/en.2010-1095] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) is an important regulatory component of the IGF system. Through proteolysis of inhibitory IGF binding proteins (IGFBPs), PAPP-A acts as a positive modulator of local IGF signaling in a variety of biological systems. A role of IGF in the progression of several common forms of human cancer is now emerging, and therapeutic intervention of IGF receptor signaling is currently being explored. However, little is known about the activities of other components of the IGF system in relation to cancer. We hypothesized that PAPP-A acts to enhance tumor growth in vivo. To test this hypothesis, we overexpressed wild-type PAPP-A or a mutant PAPP-A with markedly reduced IGFBP protease activity in SKOV3 cells, a human ovarian carcinoma cell line with low tumorigenic potential. In vitro, SKOV3 clones with elevated PAPP-A expression (PAPP-A-1, PAPP-A-28) showed accelerated anchorage-independent growth in soft agar assays compared to clones overexpressing mutant PAPP-A (E483Q-1, E483Q-5) and vector controls. PAPP-A-28, with the highest PAPP-A expression and IGFBP proteolytic activity, also had markedly increased cell invasion through Matrigel. In vivo, we found significantly accelerated tumor growth rates of PAPP-A-overexpressing SKOV3 clones compared with mutant PAPP-A and controls. Investigation of angiogenesis indicated that overexpression of PAPP-A favored development of mature tumor vasculature and that tumor precursors of PAPP-A-28 in particular had a significantly higher degree of vascularization months before obvious tumor development. In conclusion, our data show that PAPP-A proteolytic activity enhances the tumorigenic potential of ovarian cancer cells and establish a novel tumor growth-promoting role of PAPP-A.
Collapse
Affiliation(s)
- Henning B Boldt
- Division of Endocrinology, Metabolism, and Nutrition, Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | |
Collapse
|
21
|
Wolff C, Malinowsky K, Berg D, Schragner K, Schuster T, Walch A, Bronger H, Höfler H, Becker KF. Signalling networks associated with urokinase-type plasminogen activator (uPA) and its inhibitor PAI-1 in breast cancer tissues: new insights from protein microarray analysis. J Pathol 2010; 223:54-63. [PMID: 21125664 DOI: 10.1002/path.2791] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 09/14/2010] [Accepted: 09/18/2010] [Indexed: 12/29/2022]
Abstract
The urokinase-type plasminogen activator (uPA) and the main uPA inhibitor PAI-1 play important roles in cell migration and invasion in both physiological and pathological contexts. Both factors are clinically applicable predictive markers in node-negative breast cancer patients that are used to stratify patients for adjuvant chemotherapy. In addition to their classical functions in plasmin regulation, both factors are key components in cancer-related cell signalling. Such signalling cascades are well described in cell culture systems, but a better understanding of uPA- and PAI-1-associated signalling networks in clinical tissues is needed. We examined the expression of uPA, PAI-1, and 21 signalling molecules in 201 primary breast cancer tissues using protein microarrays. Expression of uPA was significantly correlated with the expression of ERK and Stat3, while expression of PAI-1 was correlated with the uPA receptor and Akt activation, presumably via integrin and HER-receptor signalling. Analysis of uPA expression did not reveal any significant correlation with staging, grading or age of the patients. The PAI-1 expression was correlated with nodal stage. Network monitoring for uPA and PAI-1 in breast cancer reveals interactions with main signalling cascades and extends the findings from cell culture experiments. Our results reveal possible mechanisms underlying cancer development.
Collapse
Affiliation(s)
- Claudia Wolff
- Institute of Pathology, Technische Universität München, Munich, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Carter JC, Campbell RA, Gibbons JA, Gramling MW, Wolberg AS, Church FC. Enhanced cell-associated plasminogen activator pathway but not coagulation pathway activity contributes to motility in metastatic breast cancer cells. J Thromb Haemost 2010; 8:1323-32. [PMID: 20180817 DOI: 10.1111/j.1538-7836.2010.03825.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Activation of tumor cell-associated coagulation and plasminogen activator pathways occurs in malignant disease processes, including breast cancer, and may promote metastatic activity. OBJECTIVES/METHODS To compare the coagulation and plasminogen activator pathways of normal and metastatic cells, we examined two cell lines from the MCF-10 family of breast cells: near-normal immortalized MCF-10A cells, and metastatic MCF-10CA1 cells. RESULTS MCF-10CA1 cell motility was significantly increased as compared with that of MCF-10A cells. The two cell types supported similar rates of factor Xa generation, plasma thrombin generation, and fibrin formation. MCF-10A cells produced a stable fibrin network, whereas MCF-10CA1 cells lysed the surrounding fibrin network within 24 h of network formation. Importantly, fibrin located proximal to (within 10 microm) the MCF-10CA1 cell surface lysed substantially faster than fibrin located 100 microm from the surface. MCF-10CA1 cells supported significantly increased plasmin generation rates as compared with MCF-10A cells, providing a mechanism for the increased fibrinolytic activity of these cells towards the fibrin network. Metastatic MCF-10CA1 cells had increased expression (mRNA and protein) levels of urokinase plasminogen activator (u-PA) and decreased levels of plasminogen activator inhibitor-1 as compared with MCF-10A cells. Blocking u-PA activity with the active site-directed protease inhibitor amiloride substantially decreased MCF-10CA1 cell motility. Phosphorylated Akt levels were elevated in MCF-10CA1 cells, which partially explains the increased u-PA expression. CONCLUSIONS These results suggest that the tumor-associated plasminogen activator pathway, not the coagulation pathway, is a key distinguishing feature between metastatic MCF10-CA1 cells and normal MCF-10A cells.
Collapse
Affiliation(s)
- J C Carter
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| | | | | | | | | | | |
Collapse
|
23
|
Appledorn DM, Dao KHT, O'Reilly S, Maher VM, McCormick JJ. Rac1 and Cdc42 are regulators of HRasV12-transformation and angiogenic factors in human fibroblasts. BMC Cancer 2010; 10:13. [PMID: 20067638 PMCID: PMC2826294 DOI: 10.1186/1471-2407-10-13] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Accepted: 01/12/2010] [Indexed: 01/12/2023] Open
Abstract
Background The activities of Rac1 and Cdc42 are essential for HRas-induced transformation of rodent fibroblasts. What is more, expression of constitutively activated mutants of Rac1 and/or Cdc42 is sufficient for their malignant transformation. The role for these two Rho GTPases in HRas-mediated transformation of human fibroblasts has not been studied. Here we evaluated the contribution of Rac1 and Cdc42 to maintaining HRas-induced transformation of human fibroblasts, and determined the ability of constitutively activated mutants of Rac1 or Cdc42 to induce malignant transformation of a human fibroblast cell strain. Methods Under the control of a tetracycline regulatable promoter, dominant negative mutants of Rac1 and Cdc42 were expressed in a human HRas-transformed, tumor derived fibroblast cell line. These cells were used to determine the roles of Rac1 and/or Cdc42 proteins in maintaining HRas-induced transformed phenotypes. Similarly, constitutively active mutants were expressed in a non-transformed human fibroblast cell strain to evaluate their potential to induce malignant transformation. Affymetrix GeneChip arrays were used for transcriptome analyses, and observed expression differences were subsequently validated using protein assays. Results Expression of dominant negative Rac1 and/or Cdc42 significantly altered transformed phenotypes of HRas malignantly transformed human fibroblasts. In contrast, expression of constitutively active mutants of Rac1 or Cdc42 was not sufficient to induce malignant transformation. Microarray analysis revealed that the expression of 29 genes was dependent on Rac1 and Cdc42, many of which are known to play a role in cancer. The dependence of two such genes, uPA and VEGF was further validated in both normoxic and hypoxic conditions. Conclusion(s) The results presented here indicate that expression of both Rac1 and Cdc42 is necessary for maintaining several transformed phenotypes in oncogenic HRas transformed human cells, including their ability to form tumors in athymic mice. Our data also indicate that expression of either activated Rac1 or Cdc42 alone is not sufficient for malignant transformation of human fibroblasts, although each is required for specific transformed phenotypes. Furthermore, our study elucidates that the expression of several highly significant cancer related genes require the activities of Rac1 and/or Cdc42 which may also play a critical role in cellular transformation.
Collapse
Affiliation(s)
- Daniel M Appledorn
- Carcinogenesis Laboratory, Michigan State University, East Lansing, Michigan, 48824-1302, USA
| | | | | | | | | |
Collapse
|
24
|
Beauchamp MC, Yasmeen A, Knafo A, Gotlieb WH. Targeting insulin and insulin-like growth factor pathways in epithelial ovarian cancer. JOURNAL OF ONCOLOGY 2010; 2010:257058. [PMID: 20069126 PMCID: PMC2804114 DOI: 10.1155/2010/257058] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Accepted: 10/09/2009] [Indexed: 01/07/2023]
Abstract
Ovarian cancer is the most lethal of all gynecological malignancies, due in part to the diagnosis at an advanced stage caused by the lack of specific signs and symptoms and the absence of reliable tests for screening and early detection. Most patients will respond initially to treatment but about 70% of them will suffer a recurrence. Therefore, new therapeutic modalities are urgently needed to overcome chemoresistance observed in ovarian cancer patients. Evidence accumulates suggesting that the insulin/insulin growth factor (IGF) pathways could act as a good therapeutic target in several cancers, including ovarian cancer. In this paper, we will focus on the role of insulin/IGF in ovarian cancer tumorigenesis and treatment.
Collapse
Affiliation(s)
- Marie-Claude Beauchamp
- Division of Gynecologic Oncology, Jewish General Hospital, McGill University, Montreal, QC, Canada H3T 1E2
- Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada H3T 1E2
| | - Amber Yasmeen
- Division of Gynecologic Oncology, Jewish General Hospital, McGill University, Montreal, QC, Canada H3T 1E2
- Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada H3T 1E2
| | - Ariane Knafo
- Division of Gynecologic Oncology, Jewish General Hospital, McGill University, Montreal, QC, Canada H3T 1E2
- Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada H3T 1E2
| | - Walter H. Gotlieb
- Division of Gynecologic Oncology, Jewish General Hospital, McGill University, Montreal, QC, Canada H3T 1E2
- Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada H3T 1E2
| |
Collapse
|
25
|
Agis H, Kandler B, Fischer MB, Watzek G, Gruber R. Activated platelets increase fibrinolysis of mesenchymal progenitor cells. J Orthop Res 2009; 27:972-80. [PMID: 19030175 DOI: 10.1002/jor.20819] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Bone regeneration is initiated by the formation of a blood clot. Activated platelets within this fibrin-rich matrix release signaling molecules that can attract mesenchymal progenitor cells. To gain insight into the cellular mechanism by which activated platelets can support the immigration of mesenchymal progenitors, we have tested the hypothesis that platelet-released signaling molecules increase the capacity of bone marrow stromal cells (BMSC) to activate plasminogen. We report herein that platelet-released supernatants (PRS) elevate total urokinase-type plasminogen activator (uPA) and total plasminogen activator inhibitor-1 (PAI-1) levels in BMSC, as assessed by immunoassay. Quantitative polymerase chain reaction showed an upregulation of uPA, uPA receptor, and PAI-1. Zymography and kinetic analysis based on casein hydrolysis revealed enhanced activity of cell-associated uPA upon exposure of BMSC to PRS. Inhibiting c-Jun N-terminal kinase (JNK) and phosphatidylinositol 3-kinase (PI3K) signaling reduced uPA production and decreased plasminogen activation. Corresponding Western blot analysis showed increased phosphorylation of JNK and AKT in BMSC treated with PRS. These results suggest that activated platelets can enhance the plasminogen activation capacity of mesenchymal progenitors through the stimulation of uPA production, requiring JNK and PI3K/AKT signaling. By this mechanism platelets may contribute to the organization of the blood clot during bone regeneration.
Collapse
Affiliation(s)
- Hermann Agis
- Department of Oral Surgery, Medical University of Vienna, Währingerstrasse 25a, 1090 Vienna, Austria
| | | | | | | | | |
Collapse
|
26
|
Plasminogen activator inhibitor 1 protects fibrosarcoma cells from etoposide-induced apoptosis through activation of the PI3K/Akt cell survival pathway. Neoplasia 2008; 10:1083-91. [PMID: 18813358 DOI: 10.1593/neo.08486] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 06/26/2008] [Accepted: 06/27/2008] [Indexed: 11/18/2022] Open
Abstract
High levels of plasminogen activator inhibitor (PAI-1) in tumors are associated with poor prognosis in several cancer types, and the reason for this association is not fully understood. Plasminogen activator inhibitor 1 has been suggested to contribute to tumor growth by protecting cancer cells from apoptosis, and we have previously shown that wild type murine fibrosarcoma cells are significantly more resistant to apoptosis induced by chemotherapy than PAI-1-deficient fibrosarcoma cells. Here, we further investigated the molecular mechanisms underlying the antiapoptotic function of PAI-1 focusing on the phosphatidylinositol 3-phosphate kinase (PI3K)/Akt cell survival pathway. We demonstrate that the activation level of the Akt cell survival pathway is reduced in PAI-1-deficient cells. Inhibition of either PI3K or Akt by synthetic inhibitors sensitized the wild type but not the PAI-1-deficient cells to etoposide-induced cell death. More importantly, reintroduction of PAI-1 expression in PAI-1-deficient cells induced an increase in Akt activity and protection against etoposide-induced apoptosis. Concordantly, silencing of PAI-1 by RNA interference in wild type fibrosarcoma cells decreased the level of active Akt, and this was accompanied by a sensitization of the cells to etoposide-induced cell death. Altogether, our data suggest that PAI-1 influences sensitivity to etoposide-induced apoptosis through the PI3K/Akt cell survival pathway by acting upstream of PI3K and Akt. This points to PAI-1 as a possible therapeutic target in cancer diseases where PAI-1 inhibits chemotherapy-induced apoptosis.
Collapse
|
27
|
Liu P, Xu B, Li J, Lu H. LY294002 inhibits leukemia cell invasion and migration through early growth response gene 1 induction independent of phosphatidylinositol 3-kinase-Akt pathway. Biochem Biophys Res Commun 2008; 377:187-90. [DOI: 10.1016/j.bbrc.2008.09.094] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 09/23/2008] [Indexed: 01/04/2023]
|
28
|
Impact of statins on modulation by insulin of expression of plasminogen activator inhibitor type-1. Coron Artery Dis 2008; 19:355-61. [DOI: 10.1097/mca.0b013e328300dbe3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
29
|
Beaulieu LM, Whitley BR, Wiesner TF, Rehault SM, Palmieri D, Elkahloun AG, Church FC. Breast cancer and metabolic syndrome linked through the plasminogen activator inhibitor-1 cycle. Bioessays 2007; 29:1029-38. [PMID: 17876797 PMCID: PMC4046619 DOI: 10.1002/bies.20640] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is a physiological inhibitor of urokinase (uPA), a serine protease known to promote cell migration and invasion. Intuitively, increased levels of PAI-1 should be beneficial in downregulating uPA activity, particularly in cancer. By contrast, in vivo, increased levels of PAI-1 are associated with a poor prognosis in breast cancer. This phenomenon is termed the "PAI-1 paradox". Many factors are responsible for the upregulation of PAI-1 in the tumor microenvironment. We hypothesize that there is a breast cancer predisposition to a more aggressive stage when PAI-1 is upregulated as a consequence of Metabolic Syndrome (MetS). MetS exerts a detrimental effect on the breast tumor microenvironment that supports cancer invasion. People with MetS have an increased risk of coronary heart disease, stroke, peripheral vascular disease and hyperinsulinemia. Recently, MetS has also been identified as a risk factor for breast cancer. We hypothesize the existence of the "PAI-1 cycle". Sustained by MetS, adipocytokines alter PAI-1 expression to promote angiogenesis, tumor-cell migration and procoagulant microparticle formation from endothelial cells, which generates thrombin and further propagates PAI-1 synthesis. All of these factors culminate in a chemotherapy-resistant breast tumor microenvironment. The PAI-1 cycle may partly explain the PAI-1 paradox. In this hypothesis paper, we will discuss further how MetS upregulates PAI-1 and how an increased level of PAI-1 can be linked to a poor prognosis.
Collapse
Affiliation(s)
- Lea M. Beaulieu
- Departments of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599-7035
| | - Brandi R. Whitley
- Departments of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599-7035
| | - Theodore F. Wiesner
- Departments of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7035
| | - Sophie M. Rehault
- Departments of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599-7035
| | - Diane Palmieri
- Departments of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599-7035
| | - Abdel G. Elkahloun
- NHGRI-NIH Genome Technology Branch, National Institute of Health, Bethesda, MD 20892
| | - Frank C. Church
- Departments of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599-7035
| |
Collapse
|
30
|
Fu RX, Yang YM, Zhang CY. Effect of Genistein on the expression of Akt and phosphorylated Akts in human gastric carcinoma cell line. Shijie Huaren Xiaohua Zazhi 2007; 15:1055-1059. [DOI: 10.11569/wcjd.v15.i10.1055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of Genestein on the expression of Akt and its phosphorylated proteins (p-Akts) in human gastric carcinoma cell line SGC-7901.
METHODS: SGC-7901 cells were exposed to 0、5、10、20、40 and 80 μmol/L Genistein for 24 h. The effect of Genistein on cell proliferation was measured by methyl thiazolyl tetrazolium (MTT) assay, and the apoptosis of SGC-7901 cells was evaluated by Hoechst 33342 staining. The expression of Akt and p-Akts (Ser473 and Thr308) were measured by Western blot.
RESULTS: The inhibitory rate was 6.85% ± 3.71%, 13.19% ± 1.90%, 20.94% ± 1.83%, 29.58% ± 1.19% and 41.75% ± 1.92%, respectively, as SGC-7901 cells were exposed to 5, 10, 20, 40 and 80 μmol/L Genistein for 24 h. Typical morphological changes of apoptosis were induced by Genistein at the concentrations (20-80 μmol/L). Genistein had no marked influences on Akt expression. Both Ser473 and Thr308 were expressed in SGC-7901 cells 24 h after Genistein treatment, and they were decreased with the increasing of Genistein concentrations. The weakest expression appeared as 80 μmol/L Genistein was used.
CONCLUSION: The anti-cancer action of Genistein may be associated with the decreased expression of p-Akt (Ser473) and p-Akt (Thr308).
Collapse
|