1
|
Ding B, Meng W, Zang X, Lv Z. Metabolic characteristics of prostate cancer cells with high metastatic potential revealed by (S)-ethyl 1-(3-(4-chlorophenoxy)-2-hydroxypropyl)-3-(4-methoxyphenyl)-1H-pyrazole-5-carboxylate inhibition. J Pharm Biomed Anal 2025; 255:116611. [PMID: 39662125 DOI: 10.1016/j.jpba.2024.116611] [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: 08/23/2024] [Revised: 10/28/2024] [Accepted: 11/30/2024] [Indexed: 12/13/2024]
Abstract
A small molecule, (S)-ethyl 1-(3-(4-chlorophenoxy)-2-hydroxypropyl)-3-(4-methoxyphenyl)-1H-pyrazole-5-carboxylate (SEC), has been reported to be capable of suppressing metastasis of prostate cancer (PCa) cells. In this study, SEC was used to study the metabolic responses of PCa cell lines (LNCaP, PC3, and DU145) with different metastatic potential and the alterations of mTOR, p-mTOR, AMPK, and p-AMPK levels, when the PCa cells were inhibited. The ultra-high performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS)-based analysis showed that SEC induced the decreases of intracellular metabolites including glutamic acid, glutamine, and histidine (LNCaP); creatinine, citric acid/isocitric acid, and aspartic acid (PC3); and spermidine, S-hydroxymethylglutathione, LPE (20:3), and palmitic amide (DU145), and the increases of intracellular LPC (18:0) (LNCaP); tyrosine, pyroglutamic acid/pyrroline hydroxycarboxylic acid (PC3); and tyrosine, phenylalanine, phenylacetylglycine, spermine, histidine, and choline (DU145). SEC also caused the decrease of extracellular N1-acetylspermidine (LNCaP), erythronic acid/threonic acid (PC3 and DU145), and nicotinic acid/picolinic acid (DU145), and the increase of extracellular 5'-methylthioadenosine (DU145). High metastatic PC3 and DU145 cells exhibited changes in aromatic amino acid metabolism including tyrosine metabolism, phenylalanine, tyrosine, and tryptophan metabolism, and phenylalanine metabolism (PC3 and DU145), TCA cycle (PC3), arginine and proline metabolism, and glycerophospholipid metabolism (DU145), different from the low metastatic LNCaP cells, which had changes in alanine, aspartate, and glutamate metabolism, and arginine biosynthesis. In addition, the levels of p-mTOR and p-AMPK were shown to be obviously downregulated and upregulated, respectively, in high metastatic PC3 and DU145 cells upon SEC inhibition, while this behavior was not detected in LNCaP cells.
Collapse
Affiliation(s)
- Baoyan Ding
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China
| | - Wei Meng
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China
| | - Xiaoling Zang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China.
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China
| |
Collapse
|
2
|
Cong F, Huang J, Wu C, Zhong H, Qiu G, Luo T, Tang W. Integrin α6 and integrin β4 in exosomes promote lung metastasis of colorectal cancer. J Cancer Res Ther 2024; 20:2082-2093. [PMID: 39792419 DOI: 10.4103/jcrt.jcrt_230_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/23/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common cancers worldwide. The mechanisms underlying metastasis, which contributes to poor outcomes, remain elusive. METHODS We used the Cancer Genome Atlas dataset to compare mRNA expression patterns of integrin α6 (ITGA6) and integrin β4 (ITGB4) in patients with CRC. We measured ITGA6 and ITGB4 expression levels in highly metastatic (i.e., HCT116 and SW620) and lowly metastatic (i.e., SW480 and Caco2) CRC cell lines. Exosomes were isolated from cell culture media and characterized using western blotting and nanoparticle analyses. The role of exosomes in lung metastasis was investigated using xenograft experiments in mice models, which received CRC cell injection and were treated with exosomes. RESULTS ITGA6 and ITGB4 were significantly overexpressed in CRC tissues, and ITGA6 was associated with the American Joint Committee on Cancer (AJCC) stage and outcome. ITGA6 and ITGB4, as well as exosomal ITGA6 and ITGB4, were significantly more highly expressed in HCT116 and SW620 cells than in SW480 and Caco2 cells. The proliferation and tubulogenesis of vascular endothelial cells were markedly decreased by disruption of ITGA6 and ITGB4 but were markedly increased by ectopic expression of ITGA6 and ITGB4. Exosomal ITGA6 and ITGB4 promoted CRC metastasis to the lung in vivo. CONCLUSIONS Taken together, our findings suggested that exosomal ITGA6 and ITGB4 displayed organotropism to the lung and upregulated proliferation and tubulogenic capacities, which might help reduce lung metastasis from CRC. These findings provided new insights into the mechanisms of CRC metastasis and provided novel potential therapeutic targets.
Collapse
Affiliation(s)
- Fengyun Cong
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jiahao Huang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Changtao Wu
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Huage Zhong
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, China
| | - Guanhua Qiu
- Department of Ultrasound, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Tao Luo
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, China
| | - Weizhong Tang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, China
| |
Collapse
|
3
|
Seltmann K, Hettich B, Abele S, Gurri S, Mantella V, Leroux J, Werner S. Transport of CLCA2 to the nucleus by extracellular vesicles controls keratinocyte survival and migration. J Extracell Vesicles 2024; 13:e12430. [PMID: 38602325 PMCID: PMC11007793 DOI: 10.1002/jev2.12430] [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: 04/04/2023] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/12/2024] Open
Abstract
Chloride channel accessory 2 (CLCA2) is a transmembrane protein, which promotes adhesion of keratinocytes and their survival in response to hyperosmotic stress. Here we show that CLCA2 is transported to the nucleus of keratinocytes via extracellular vesicles. The nuclear localization is functionally relevant, since wild-type CLCA2, but not a mutant lacking the nuclear localization signal, suppressed migration of keratinocytes and protected them from hyperosmotic stress-induced cell death. In the nucleus, CLCA2 bound to and activated β-catenin, resulting in enhanced expression of Wnt target genes. Mass-spectrometry-based interaction screening and functional rescue studies identified RNA binding protein 3 as a key effector of nuclear CLCA2. This is of likely relevance in vivo because both proteins co-localize in the human epidermis. Together, these results identify an unexpected nuclear function of CLCA2 in keratinocytes under homeostatic and stress conditions and suggest a role of extracellular vesicles and their nuclear transport in the control of key cellular activities.
Collapse
Affiliation(s)
- Kristin Seltmann
- Institute of Molecular Health Sciences, Department of BiologySwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Britta Hettich
- Department of Chemistry and Applied BiosciencesInstitute of Pharmaceutical Sciences, ETH ZurichZurichSwitzerland
| | - Seraina Abele
- Institute of Molecular Health Sciences, Department of BiologySwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Selina Gurri
- Institute of Molecular Health Sciences, Department of BiologySwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Valeria Mantella
- Department of Chemistry and Applied BiosciencesInstitute of Pharmaceutical Sciences, ETH ZurichZurichSwitzerland
| | - Jean‐Christophe Leroux
- Department of Chemistry and Applied BiosciencesInstitute of Pharmaceutical Sciences, ETH ZurichZurichSwitzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of BiologySwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| |
Collapse
|
4
|
Hazra S, Dey S, Mandal BB, Ramachandran C. In Vitro Profiling of the Extracellular Matrix and Integrins Expressed by Human Corneal Endothelial Cells Cultured on Silk Fibroin-Based Matrices. ACS Biomater Sci Eng 2023; 9:2438-2451. [PMID: 37023465 DOI: 10.1021/acsbiomaterials.2c01566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Developing a scaffold for culturing human corneal endothelial (HCE) cells is crucial as an alternative cell therapeutic approach to bridge the growing gap between the demand and availability of healthy donor corneas for transplantation. Silk films are promising substrates for the culture of these cells; however, their tensile strength is several-fold greater than the native basement membrane which can possibly influence the dynamics of cell-matrix interaction and the extracellular matrix (ECM) secreted by the cells in long-term culture. In our current study, we assessed the secretion of ECM and the expression of integrins by the HCE cells on Philosamia ricini (PR) and Antheraea assamensis (AA) silk films and fibronectin-collagen (FNC)-coated plastic dishes to understand the cell-ECM interaction in long-term culture. The expression of ECM proteins (collagens 1, 4, 8, and 12, laminin, and fibronectin) on silk was comparable to that on the native tissue. The thicknesses of collagen 8 and laminin at 30 days on both PR (4.78 ± 0.55 and 5.53 ± 0.51 μm, respectively) and AA (4.66 ± 0.72 and 5.71 ± 0.61 μm, respectively) were comparable with those of the native tissue (4.4 ± 0.63 and 5.28 ± 0.72 μm, respectively). The integrin expression by the cells on the silk films was also comparable to that on the native tissue, except for α3 whose fluorescence intensity was significantly higher on PR (p ≤ 0.01) and AA (p ≤ 0.001), compared to that on the native tissue. This study shows that the higher tensile strength of the silk films does not alter the ECM secretion or cell phenotype in long-term culture, confirming the suitability of using this material for engineering the HCE cells for transplantation.
Collapse
Affiliation(s)
- Swatilekha Hazra
- Hyderabad Eye Research Foundation, LV Prasad Eye Institute, Hyderabad 500034, India
- Manipal Academy of Higher Education, Manipal 576104, India
| | - Souradeep Dey
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Biman B Mandal
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- Jyoti and Bhupat Mehta School of Health Sciences & Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | | |
Collapse
|
5
|
Ma F, Ma Y, Liu K, Gao J, Li S, Sun X, Li G. Resveratrol induces DNA damage-mediated cancer cell senescence through the DLC1-DYRK1A-EGFR axis. Food Funct 2023; 14:1484-1497. [PMID: 36651299 DOI: 10.1039/d2fo01188c] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inducing cell senescence is widely regarded as a potent tumor suppression mechanism. Resveratrol has attracted increasing attention for its capacity to prevent and suppress cancer. However, the mechanism of resveratrol on the induction of cancer cell senescence has not been well clarified. Our results showed that resveratrol inhibited cell viability and colony formation and promoted cell senescence along with augmentation of SA-β-gal activity and modulation of senescence-associated molecular markers p53, p21 and LaminB protein in breast and liver cancer cells. The underlying mechanism was that resveratrol increased ROS generation to enhance tumor suppressor gene DLC1 expression, and DLC1 further inhibited the DYRK1A-EGFR axis to trigger DNA damage accompanied by up-regulation of the DNA double strand break marker protein γH2AX and down-regulation of the DNA repair related proteins p-BRCA1 and RAD51, eventually leading to cancer cell senescence. Resveratrol also effectively inhibited the volume of transplanted tumor with increased SA-β-gal activity and DLC1 level in a chicken embryo allantoic membrane xenograft tumor model. This is the first report to investigate whether resveratrol induces DNA damage-mediated cancer cell senescence through the DLC1-DYRK1A-EGFR axis, which could provide a solid base for resveratrol's application in cancer prevention and clinical treatment as a food additive or adjuvant therapies.
Collapse
Affiliation(s)
- Fengqiu Ma
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Yan Ma
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Keke Liu
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Junying Gao
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Shasha Li
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Xiaowen Sun
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Guorong Li
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| |
Collapse
|
6
|
Ning J, Wang X, Li N, Cui X, Li N, Zhao B, Miao J, Lin Z. ZBM-H-induced activation of GRP78 ATPase promotes apoptosis via annexin A7 in A549 lung cancer cells. J Cell Biochem 2022; 123:798-806. [PMID: 35118704 DOI: 10.1002/jcb.30224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 12/18/2021] [Accepted: 01/18/2022] [Indexed: 12/17/2022]
Abstract
Hypochlorous acid (HOCl) is an essential signal for the regulation of cancer cell fate, including autophagy and apoptosis. HOCl regulated autophagy by affecting the oxidation modification of glucose-regulated protein 78 (GRP78) and the activity of GRP78 ATPase. The mechanism of GRP78 ATPase in cell apoptosis has however not yet been clarified. Here we reported that ZBM-H, as a probe of HOCl, was able to directly bind to GRP78 in the presence or absence of ATP. Following ZBM-H treatment, the interaction between GRP78 and annexin A7 (ANXA7) was promoted, and this was accompanied by increased phosphorylation of integrin β4 (ITGB4). In addition, ZBM-H enhanced the phosphorylation of ANXA7. ABO, an inhibitor of ANXA7, inhibited ZBM-H-induced ITGB4 phosphorylation and apoptosis, while ANXA7 activator SEC had opposite effect. Collectively, these data provide new evidence for the mechanism by which ZBM-H-induced activation of GRP78 ATPase regulates apoptosis of A549 lung cancer cells.
Collapse
Affiliation(s)
- Junya Ning
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China.,Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Xiaotan Wang
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Nan Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| | - Xiaoling Cui
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| | - Na Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| | - Baoxiang Zhao
- Institute of Organic Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Jinan, China
| | - Junying Miao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| | - Zhaomin Lin
- Central Research Laboratory, The Second Hospital, Shandong University, Jinan, China
| |
Collapse
|
7
|
Gwili N, Jones SJ, Amri WA, Carr IM, Harris S, Hogan BV, Hughes WE, Kim B, Langlands FE, Millican-Slater RA, Pramanik A, Thorne JL, Verghese ET, Wells G, Hamza M, Younis L, El Deeb NMF, Hughes TA. Transcriptome profiles of stem-like cells from primary breast cancers allow identification of ITGA7 as a predictive marker of chemotherapy response. Br J Cancer 2021; 125:983-993. [PMID: 34253873 PMCID: PMC8476506 DOI: 10.1038/s41416-021-01484-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/07/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Breast cancer stem cells (BCSCs) are drivers of therapy-resistance, therefore are responsible for poor survival. Molecular signatures of BCSCs from primary cancers remain undefined. Here, we identify the consistent transcriptome of primary BCSCs shared across breast cancer subtypes, and we examine the clinical relevance of ITGA7, one of the genes differentially expressed in BCSCs. METHODS Primary BCSCs were assessed using immunohistochemistry and fluorescently labelled using Aldefluor (n = 17). Transcriptomes of fluorescently sorted BCSCs and matched non-stem cancer cells were determined using RNA-seq (n = 6). ITGA7 expression was examined in breast cancers using immunohistochemistry (n = 305), and its functional role was tested using siRNA in breast cancer cells. RESULTS Proportions of BCSCs varied from 0 to 9.4%. 38 genes were significantly differentially expressed in BCSCs; genes were enriched for functions in vessel morphogenesis, motility, and metabolism. ITGA7 was found to be significantly downregulated in BCSCs, and low expression significantly correlated with reduced survival in patients treated with chemotherapy, and with chemoresistance in breast cancer cells in vitro. CONCLUSIONS This study is the first to define the molecular profile of BCSCs from a range of primary breast cancers. ITGA7 acts as a predictive marker for chemotherapy response, in accordance with its downregulation in BCSCs.
Collapse
Affiliation(s)
- Noha Gwili
- grid.9909.90000 0004 1936 8403School of Medicine, University of Leeds, Leeds, UK ,grid.7155.60000 0001 2260 6941Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Stacey J. Jones
- grid.9909.90000 0004 1936 8403School of Medicine, University of Leeds, Leeds, UK ,grid.415967.80000 0000 9965 1030Department of Breast Surgery, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Waleed Al Amri
- grid.416132.30000 0004 1772 5665Department of Histopathology and Cytopathology, The Royal Hospital, Muscat, Oman
| | - Ian M. Carr
- grid.9909.90000 0004 1936 8403School of Medicine, University of Leeds, Leeds, UK
| | - Sarah Harris
- grid.9909.90000 0004 1936 8403School of Physics and Astronomy, University of Leeds, Leeds, UK
| | - Brian V. Hogan
- grid.415967.80000 0000 9965 1030Department of Breast Surgery, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - William E. Hughes
- grid.414235.50000 0004 0619 2154Children’s Medical Research Institute, Westmead, NSW Australia ,grid.1005.40000 0004 4902 0432St. Vincent’s Clinical School, University of New South Wales, Sydney, Australia
| | - Baek Kim
- grid.415967.80000 0000 9965 1030Department of Breast Surgery, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Fiona E. Langlands
- Department of Breast Surgery, Bradford Teaching Hospitals NHS Trust, Bradford, UK
| | | | - Arindam Pramanik
- grid.9909.90000 0004 1936 8403School of Medicine, University of Leeds, Leeds, UK
| | - James L. Thorne
- grid.9909.90000 0004 1936 8403School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Eldo T. Verghese
- grid.443984.6Department of Histopathology, St. James’s University Hospital, Leeds, UK
| | - Geoff Wells
- grid.83440.3b0000000121901201School of Pharmacy, University College London, London, UK
| | - Mervat Hamza
- grid.7155.60000 0001 2260 6941Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Layla Younis
- grid.7155.60000 0001 2260 6941Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nevine M. F. El Deeb
- grid.7155.60000 0001 2260 6941Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Thomas A. Hughes
- grid.9909.90000 0004 1936 8403School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
8
|
Weisz A, Abadi U, Mausbach L, Gurwitz D, Ellis M, Ashur-Fabian O. Nuclear αvβ3 integrin expression, post translational modifications and regulation in hematological malignancies. Hematol Oncol 2021; 40:72-81. [PMID: 34534368 DOI: 10.1002/hon.2927] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/05/2021] [Accepted: 09/07/2021] [Indexed: 11/09/2022]
Abstract
αvβ3 integrin, a plasma membrane protein, is amply expressed on an array of tumors. We identified nuclear αvβ3 pool in ovarian cancer cells and were interested to explore this phenomenon in two rare and aggressive types of leukemia, T-cell acute lymphoblastic leukemia (T-ALL) and Mast cell leukemia (MCL) using Jurkat and HMC-1 cell lines, respectively. Moreover, we collected primary cells from patients with chronic lymphocytic leukemia (CLL, n = 11), the most common chronic adult leukemia and used human lymphoblastoid cell lines (LCL) generated from normal B cells. Nuclear αvβ3 integrin was assessed by Western blots, confocal microscopy, and the ImageStream technology which combines flow-cytometry with microscopy. We further examined post translational modifications (phosphorylation/glycosylation), nuclear trafficking regulation using inhibitors for MAPK (U0126) and PI3K (LY294002), as well as nuclear interactions by performing Co-immunoprecipitation (Co-IP). αvβ3 integrin was identified in all cell models within the nucleus and is N-glycosylated. In primary CLL cells the β3 integrin monomer is tyrosine Y759 phosphorylated, suggesting an active receptor conformation. MAPK and PI3K inhibition in Jurkat and CLL cells led to αvβ3 enhancement in the nucleus and a reduction in the membrane. The nuclear αvβ3 integrin interacts with ERK, Histone H3 and Lamin B1 in Jurkat, Histone H3 in CLL cells, but not in control LCL cells. To conclude, this observational study provides the identification of nuclear αvβ3 in hematological malignancies and lays the basis for novel cancer-relevant actions, which may be independent from the membrane functions.
Collapse
Affiliation(s)
- Avivit Weisz
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Uri Abadi
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lisa Mausbach
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel
| | - David Gurwitz
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Martin Ellis
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Ashur-Fabian
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
9
|
Huang R, Zheng Z, Xian S, Zhang J, Jia J, Song D, Yan P, Yin H, Hu P, Zhu X, Huang Z, Meng T, Zhang J. Identification of prognostic and bone metastatic alternative splicing signatures in bladder cancer. Bioengineered 2021; 12:5289-5304. [PMID: 34402716 PMCID: PMC8806927 DOI: 10.1080/21655979.2021.1964252] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bladder cancer (BLCA), originating from the epithelium of the urinary bladder, was the second most common malignancy in the urinary system with a high metastasis rate and poor post-metastasis prognosis. Alternative splicing events (ASEs) were regarded as important markers of tumor progression and prognosis, however, their roles in bladder cancer bone metastasis have not been recognized. In this study, we constructed a predictive model based on ASEs and explored the molecular mechanism of ASEs in BLCA bone metastasis, based on data from the Cancer Genome Atlas (TCGA) and TCGASpliceSeq databases. We proposed the hypothesis that the splicing events of ITGB4 was regulated by the splicing factor JUP, and this regulation might play a key role in BLCA bone metastasis through the glycosphingolipid biosynthesis ganglio series pathway.
Collapse
Affiliation(s)
- Runzhi Huang
- Key Laboratory Of Spine And Spinal Cord Injury Repair And Regeneration Tongji University, Ministry Of Education, Shanghai, China.,Division Of Spine, Department Of Orthopedics, Tongji Hospital Affiliated To Tongji University School Of Medicine, Shanghai, China
| | - Zixuan Zheng
- Tongji University School Of Medicine (Shanghai Pulmonary Hospital), Shanghai, China
| | - Shuyuan Xian
- Key Laboratory Of Spine And Spinal Cord Injury Repair And Regeneration Tongji University, Ministry Of Education, Shanghai, China.,Division Of Spine, Department Of Orthopedics, Tongji Hospital Affiliated To Tongji University School Of Medicine, Shanghai, China
| | - Jiayao Zhang
- School Of Mathematical Sciences Of Tongji University, Shanghai, China
| | - Jingyi Jia
- Tongji University School Of Medicine (Shanghai Pulmonary Hospital), Shanghai, China
| | - Dianwen Song
- Department Of Orthopedics, Shanghai General Hospital, School Of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Penghui Yan
- Department Of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huabin Yin
- Department Of Orthopedics, Shanghai General Hospital, School Of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Peng Hu
- Department Of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaolong Zhu
- Department Of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zongqiang Huang
- Department Of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tong Meng
- Department Of Orthopedics, Shanghai General Hospital, School Of Medicine, Shanghai Jiaotong University, Shanghai, China.,Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School Of Medicine, Shanghai, China
| | - Jie Zhang
- Key Laboratory Of Spine And Spinal Cord Injury Repair And Regeneration Tongji University, Ministry Of Education, Shanghai, China.,Division Of Spine, Department Of Orthopedics, Tongji Hospital Affiliated To Tongji University School Of Medicine, Shanghai, China
| |
Collapse
|
10
|
Yang H, Xu Z, Peng Y, Wang J, Xiang Y. Integrin β4 as a Potential Diagnostic and Therapeutic Tumor Marker. Biomolecules 2021; 11:biom11081197. [PMID: 34439865 PMCID: PMC8394641 DOI: 10.3390/biom11081197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 11/05/2022] Open
Abstract
Integrin β4 (ITGβ4) is a class of transmembrane adhesion molecules composed of hemidesmosomes (HDs). Its unique long intracellular domain provides intricate signal transduction functions. These signal transduction effects are especially prominent in tumors. Many recent studies have shown that integrin β4 is differentially expressed in various tumors, and it plays a vital role in tumor invasion, proliferation, epithelial–mesenchymal transition, and angiogenesis. Therefore, we categorize the research related to integrin β4, starting from its structure and function in tumor tissues, and provide a basic description. Based on its structure and function, we believe that integrin β4 can be used as a tumor marker. In clinical practice, it is described as a diagnostic marker for the targeted treatment of cancer and will be helpful in the clinical diagnosis and treatment of tumors.
Collapse
Affiliation(s)
- Haoyu Yang
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Zixuan Xu
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Yuqian Peng
- School of Basic Medical Science, Central South University, Changsha 410013, China; (H.Y.); (Z.X.); (Y.P.)
| | - Jiali Wang
- Xiang Ya School of Medicine, Central South University, Changsha 410013, China;
| | - Yang Xiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Correspondence: ; Tel.:+86-139-7312-8943
| |
Collapse
|
11
|
Inhibitors of the PI3K/Akt/mTOR Pathway in Prostate Cancer Chemoprevention and Intervention. Pharmaceutics 2021; 13:pharmaceutics13081195. [PMID: 34452154 PMCID: PMC8400324 DOI: 10.3390/pharmaceutics13081195] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/serine-threonine kinase (Akt)/mammalian target of the rapamycin (mTOR)-signaling pathway has been suggested to have connections with the malignant transformation, growth, proliferation, and metastasis of various cancers and solid tumors. Relevant connections between the PI3K/Akt/mTOR pathway, cell survival, and prostate cancer (PC) provide a great therapeutic target for PC prevention or treatment. Recent studies have focused on small-molecule mTOR inhibitors or their usage in coordination with other therapeutics for PC treatment that are currently undergoing clinical testing. In this study, the function of the PI3K/Akt/mTOR pathway, the consequence of its dysregulation, and the development of mTOR inhibitors, either as an individual substance or in combination with other agents, and their clinical implications are discussed. The rationale for targeting the PI3K/Akt/mTOR pathway, and specifically the application and potential utility of natural agents involved in PC treatment is described. In addition to the small-molecule mTOR inhibitors, there are evidence that several natural agents are able to target the PI3K/Akt/mTOR pathway in prostatic neoplasms. These natural mTOR inhibitors can interfere with the PI3K/Akt/mTOR pathway through multiple mechanisms; however, inhibition of Akt and suppression of mTOR 1 activity are two major therapeutic approaches. Combination therapy improves the efficacy of these inhibitors to either suppress the PC progression or circumvent the resistance by cancer cells.
Collapse
|
12
|
Grewal T, Rentero C, Enrich C, Wahba M, Raabe CA, Rescher U. Annexin Animal Models-From Fundamental Principles to Translational Research. Int J Mol Sci 2021; 22:ijms22073439. [PMID: 33810523 PMCID: PMC8037771 DOI: 10.3390/ijms22073439] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Routine manipulation of the mouse genome has become a landmark in biomedical research. Traits that are only associated with advanced developmental stages can now be investigated within a living organism, and the in vivo analysis of corresponding phenotypes and functions advances the translation into the clinical setting. The annexins, a family of closely related calcium (Ca2+)- and lipid-binding proteins, are found at various intra- and extracellular locations, and interact with a broad range of membrane lipids and proteins. Their impacts on cellular functions has been extensively assessed in vitro, yet annexin-deficient mouse models generally develop normally and do not display obvious phenotypes. Only in recent years, studies examining genetically modified annexin mouse models which were exposed to stress conditions mimicking human disease often revealed striking phenotypes. This review is the first comprehensive overview of annexin-related research using animal models and their exciting future use for relevant issues in biology and experimental medicine.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Mohamed Wahba
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Carsten A. Raabe
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
| | - Ursula Rescher
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| |
Collapse
|
13
|
Devereaux J, Dargahi N, Fraser S, Nurgali K, Kiatos D, Apostolopoulos V. Leucocyte-Rich Platelet-Rich Plasma Enhances Fibroblast and Extracellular Matrix Activity: Implications in Wound Healing. Int J Mol Sci 2020; 21:ijms21186519. [PMID: 32900003 PMCID: PMC7556022 DOI: 10.3390/ijms21186519] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Platelet-rich plasma (PRP) is an autologous blood product that contains a high concentration of platelets and leucocytes, which are fundamental fibroblast proliferation agents. Literature has emerged that offers contradictory findings about leucocytes within PRP. Herein, we elucidated the effects of highly concentrated leucocytes and platelets on human fibroblasts. Methods: Leucocyte-rich, PRP (LR-PRP) and leucocyte-poor, platelet-poor plasma (LP-PPP) were compared to identify their effects on human fibroblasts, including cell proliferation, wound healing and extracellular matrix and adhesion molecule gene expressions. Results: The LR-PRP exhibited 1422.00 ± 317.21 × 103 platelets/µL and 16.36 ± 2.08 × 103 white blood cells/µL whilst the LP-PPP demonstrated lower concentrations of 55.33 ± 10.13 × 103 platelets/µL and 0.8 ± 0.02 × 103 white blood cells/µL. LR-PRP enhanced fibroblast cell proliferation and cell migration, and demonstrated either upregulation or down-regulation gene expression profile of the extracellular matrix and adhesion molecules. Conclusion: LR-PRP has a continuous stimulatory anabolic and ergogenic effect on human fibroblast cells.
Collapse
Affiliation(s)
- Jeannie Devereaux
- College of Health and Biomedicine, Victoria University, Melbourne, VIC 3011, Australia;
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia; (N.D.); (S.F.); (K.N.)
- Correspondence: (J.D.); (V.A.); Tel.: +613-83958218 (J.D.); +613-99192025 (V.A.)
| | - Narges Dargahi
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia; (N.D.); (S.F.); (K.N.)
| | - Sarah Fraser
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia; (N.D.); (S.F.); (K.N.)
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia; (N.D.); (S.F.); (K.N.)
| | - Dimitrios Kiatos
- College of Health and Biomedicine, Victoria University, Melbourne, VIC 3011, Australia;
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia; (N.D.); (S.F.); (K.N.)
- Correspondence: (J.D.); (V.A.); Tel.: +613-83958218 (J.D.); +613-99192025 (V.A.)
| |
Collapse
|
14
|
Seraya-Bareket C, Weisz A, Shinderman-Maman E, Teper-Roth S, Stamler D, Arbib N, Kadan Y, Fishman A, Kidron D, Edelstein E, Ellis M, Ashur-Fabian O. The identification of nuclear αvβ3 integrin in ovarian cancer: non-paradigmal localization with cancer promoting actions. Oncogenesis 2020; 9:69. [PMID: 32728020 PMCID: PMC7391722 DOI: 10.1038/s41389-020-00254-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/09/2020] [Indexed: 12/25/2022] Open
Abstract
Nuclear translocation of transmembrane proteins was reported in high-grade serous ovarian cancer (HGSOC), a highly aggressive gynecological malignancy. Although the membrane receptor αvβ3 integrin is amply expressed in HGSOC and involved in disease progression, its nuclear localization was never demonstrated. Nuclear αvβ3 was explored in HGSOC cells (OVCAR3, KURAMOCHI, and JHOS4), nuclear localization signal (NLS) modified β3 OVCAR3, Chinese hamster ovaries (CHO-K1) and human embryonic kidney (HEK293) before/after transfections with β3/β1 integrins. We used the ImageStream technology, Western blots (WB), co immunoprecipitations (Co-IP), confocal immunofluorescence (IF) microscopy, flow cytometry for cell counts and cell cycle, wound healing assays and proteomics analyses. Fresh/archived tumor tissues were collected from nine HGSOC patients and normal ovarian and fallopian tube (FT) tissues from eight nononcological patients and assessed for nuclear αvβ3 by WB, confocal IF microscopy and immunohistochemistry (IHC). We identified nuclear αvβ3 in HGSOC cells and tissues, but not in normal ovaries and FTs. The nuclear integrin was Tyr 759 phosphorylated and functionally active. Nuclear αvβ3 enriched OVCAR3 cells demonstrated induced proliferation and oncogenic signaling, intact colony formation ability and inhibited migration. Proteomics analyses revealed a network of nuclear αvβ3-bound proteins, many of which with key cancer-relevant activities. Identification of atypical nuclear localization of the αvβ3 integrin in HGSOC challenges the prevalent conception that the setting in which this receptor exerts its pleiotropic actions is exclusively at the cell membrane. This discovery proposes αvβ3 moonlighting functions and may improve our understanding of the molecular basis of ovarian cancer pathogenesis.
Collapse
Affiliation(s)
- Chen Seraya-Bareket
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, 44821, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Avivit Weisz
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, 44821, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Elena Shinderman-Maman
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, 44821, Kfar-Saba, Israel.,Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Sharon Teper-Roth
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, 44821, Kfar-Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dina Stamler
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, 44821, Kfar-Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nissim Arbib
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Gynecologic Oncology Division, Department of Obstetrics and Gynecology, Meir Medical Center, 44821, Kfar Saba, Israel
| | - Yfat Kadan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Gynecologic Oncology Division, Department of Obstetrics and Gynecology, Meir Medical Center, 44821, Kfar Saba, Israel
| | - Ami Fishman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Gynecologic Oncology Division, Department of Obstetrics and Gynecology, Meir Medical Center, 44821, Kfar Saba, Israel
| | - Debora Kidron
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Pathology, Meir Medical Center, 44821, Kfar Saba, Israel
| | - Evgeny Edelstein
- Department of Pathology, Meir Medical Center, 44821, Kfar Saba, Israel
| | - Martin Ellis
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, 44821, Kfar-Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Ashur-Fabian
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, 44821, Kfar-Saba, Israel. .,Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
15
|
Meng X, Liu P, Wu Y, Liu X, Huang Y, Yu B, Han J, Jin H, Tan X. Integrin beta 4 (ITGB4) and its tyrosine-1510 phosphorylation promote pancreatic tumorigenesis and regulate the MEK1-ERK1/2 signaling pathway. Bosn J Basic Med Sci 2020; 20:106-116. [PMID: 31242404 PMCID: PMC7029197 DOI: 10.17305/bjbms.2019.4255] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer death, with a 5-year survival rate of only 1–4%. Integrin-mediated cell adhesion is critical for the initiation, progression, and metastasis of cancer. In this study we investigated the role of integrin β4 (ITGB4) and its phosphorylation at tyrosine Y1510 (p-ITGB4-Y1510) in the tumorigenesis of pancreatic cancer. We analyzed the expression of ITGB4 and p-ITGB4-Y1510 in pancreatic cancer tissue and cell lines using immunohistochemistry, Western blot, or semi-quantitative reverse transcription PCR. ITGB4 and p-ITGB4-Y1510 were highly expressed in pancreatic cancer (n = 176) compared with normal pancreatic tissue (n = 171). High p-ITGB4-Y1510 expression correlated with local invasion and distant metastasis of pancreatic cancer, and high ITGB4 was significantly associated with poor survival of patients. Inhibition of ITGB4 by siRNA significantly reduced migration and invasion of PC-1.0 and AsPC-1 cells. Overexpression of the mutant ITGB4-Y1510A (a mutation of tyrosine to alanine at 1510 position) in PC-1.0 and AsPC-1 cells not only blocked the ITGB4 phosphorylation at Y1510 but also suppressed the expression of ITGB4 (p < 0.05 vs. wild-type ITGB4). The transfection of PC-1.0 and AsPC-1 cells with ITGB4-Y1510A significantly decreased the level of p-mitogen-activated protein kinase kinase (MEK)1 (T292) and p-extracellular signal-regulated kinase (ERK)1/2 but did not affect the level of p-MEK1 (T386) and p-MEK2 (T394). Overall, our study showed that ITGB4 and its phosphorylated form promote cell migration and invasion in pancreatic cancer and that p-ITGB4-Y1510 regulates the downstream MEK1-ERK1/2 signaling cascades. Targeting ITGB4 or its phosphorylation at Y1510 may be a novel therapeutic option for pancreatic cancer.
Collapse
Affiliation(s)
- Xiangli Meng
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China
| | - Peng Liu
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China
| | - Yunhao Wu
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China
| | - Xinlu Liu
- Department of Anus and Intestine Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yinpeng Huang
- Minimally Invasive Area of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Boqiang Yu
- Department of General Surgery, Fushun Central Hospital, Fushun, China
| | - Jiahong Han
- Department of Surgery, Liaoning Electric Power Center Hospital, Shenyang, China.
| | - Haoyi Jin
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China.
| | - Xiaodong Tan
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China.
| |
Collapse
|
16
|
Ning J, Lin Z, Zhao X, Zhao B, Miao J. Inhibiting lysine 353 oxidation of GRP78 by a hypochlorous probe targeting endoplasmic reticulum promotes autophagy in cancer cells. Cell Death Dis 2019; 10:858. [PMID: 31719525 PMCID: PMC6851114 DOI: 10.1038/s41419-019-2095-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/08/2019] [Accepted: 10/25/2019] [Indexed: 12/18/2022]
Abstract
The level of hypochlorous acid (HOCl) in cancer cells is higher than that in non-cancer cells. HOCl is an essential signal for the regulation of cell fate and works mainly through the protein post-translational modifications in cancer cells. However, the mechanism of HOCl regulating autophagy has not been clarified. Here we reported that a HOCl probe named ZBM-H targeted endoplasmic reticulum and induced an intact autophagy flux in lung cancer cells. Furthermore, ZBM-H promoted the binding of GRP78 to AMPK and increased the phosphorylation of AMPK in a dose- and time-dependent manner. GRP78 knockdown inhibited ZBM-H-induced AMPK phosphorylation and ZBM-H-stimulated autophagy. In addition, mass spectrometry combined with point mutation experiments revealed that ZBM-H increased GRP78 activity by inhibiting HOCl-induced lysine 353 oxidation of GRP78. Following ZBM-H treatment in vitro and in vivo, cell growth was significantly inhibited while apoptosis was induced. Nevertheless, exogenous HOCl partially reversed ZBM-H-inhibited cell growth and ZBM-H-induced GRP78 activation. In brief, we found that an endoplasmic reticulum-targeted HOCl probe named ZBM-H, acting through attenuating HOCl-induced GRP78 oxidation, inhibited tumor cell survival by promoting autophagy and apoptosis. Overall, these data demonstrated a novel mechanism of hypochlorous acid regulating autophagy by promoting the oxidation modification of GRP78.
Collapse
Affiliation(s)
- Junya Ning
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, 266237, PR China
| | - Zhaomin Lin
- Central Research Laboratory, the Second Hospital, Shandong University, Jinan, 250033, PR China
| | - Xuan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, 266237, PR China
| | - Baoxiang Zhao
- Institute of Organic Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, PR China.
| | - Junying Miao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, 266237, PR China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, 250012, PR China.
| |
Collapse
|
17
|
Li J, Luo M, Ou H, Liu X, Kang X, Yin W. Integrin β4 promotes invasion and anoikis resistance of papillary thyroid carcinoma and is consistently overexpressed in lymphovascular tumor thrombus. J Cancer 2019; 10:6635-6648. [PMID: 31777592 PMCID: PMC6856897 DOI: 10.7150/jca.36125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/29/2019] [Indexed: 12/20/2022] Open
Abstract
Although the majority of papillary thyroid cancers (PTC) are indolent, a subset of PTCs behaves aggressively due to extensive invasion and distant metastasis. Integrin β4, a member of the integrin family, has been shown to enhance the progression in some malignancies; however, its role in PTC remains unclear. Here, we demonstrated that β4 overexpression was associated with extrathyroid extension, lymph node metastasis, high TNM stage, and poor overall survival based on The Cancer Genome Atlas cohort. Immunohistochemistry showed that β4 expression was significantly upregulated in the tumors with infiltrating growth pattern, as well as those with positive lymphovascular invasion. Moreover, β4 was invariably overexpressed in the lymphovascular tumor thrombi, which has not been reported before. After shRNA-induced knockdown of β4 in vitro, the migration, invasion and scratch repair ability of the tumor cells were significantly reduced. Furthermore, β4 reduction decreased anchorage-independent growth and increased anoikis. The bioinformatics analysis revealed that approximately 70 pathways were significantly dysregulated in the high β4 expression group. The MAPK pathway and propanoate metabolism were located in the network center of those pathways. Taken together, our results suggest that β4 could promote the tumor's aggressiveness by enhancing invasion and antagonizing anoikis. The upregulated expression of β4 in the tumor thrombi is intrinsically linked to its role in strengthening the anoikis resistance.
Collapse
Affiliation(s)
- Jian Li
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, 518036, China.,State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, 518055, China
| | - Minghua Luo
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, 518036, China
| | - Huiting Ou
- Department of Endocrinology, Shenzhen Second People's Hospital, Guangdong Province, 518035, China
| | - Xiaoling Liu
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, 518036, China
| | - Xueling Kang
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, 518036, China
| | - Weihua Yin
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, 518036, China
| |
Collapse
|
18
|
Mo HQ, Tian FJ, Li X, Zhang J, Ma XL, Zeng WH, Lin Y, Zhang Y. ANXA7 regulates trophoblast proliferation and apoptosis in preeclampsia. Am J Reprod Immunol 2019; 82:e13183. [PMID: 31446642 DOI: 10.1111/aji.13183] [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: 06/20/2019] [Revised: 08/06/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022] Open
Abstract
PROBLEM Preeclampsia (PE) is a unique gestational disorder leading to maternal and neonatal morbidity and mortality. AnnexinA7 (ANXA7) is a calcium-dependent phospholipid-binding protein that promotes membrane fusion during exocytosis. However, the function of ANXA7 in placental trophoblast is poorly understood. The present study aimed to investigate a possible association between ANXA7 and human trophoblast apoptosis. METHODS We collected human placental tissues from patients with PE and normal pregnant women to elucidate the expression level of ANXA7. The ANXA7-knockdown and ANXA7-overexpressing HTR8/SVneo cells were utilized for studying the function of ANXA7 in trophoblast. The proliferation and apoptosis levels of trophoblast were examined with Western blot assay, flow cytometry, Cell Counting Kit-8 assay, and immunohistochemistry. RESULTS ANXA7 expression was significantly lower in placentas from patients with PE patients compared with that in from normal pregnant controls. Knockdown of ANXA7 induced cell apoptosis and inhibited cell proliferation in HTR-8 via by downregulating BCL2 protein levels. Overexpression of ANXA7 reduced apoptosis and promoted HTR8 proliferation. Further analyses showed that ANXA7 knockdown inhibited the activation of the JAK1/STAT3 pathway in HTR-8 cells. CONCLUSION Our findings revealed a new regulatory pathway of ANXA7/JAK1/STAT3 in trophoblast apoptosis in preeclampsia, suggesting that ANXA7 is a potential therapeutic target for preeclampsia.
Collapse
Affiliation(s)
- Hui-Qin Mo
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fu-Ju Tian
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Li
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Zhang
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Ling Ma
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Hong Zeng
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Lin
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
ITGB4-mediated metabolic reprogramming of cancer-associated fibroblasts. Oncogene 2019; 39:664-676. [PMID: 31534187 DOI: 10.1038/s41388-019-1014-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/28/2019] [Accepted: 09/05/2019] [Indexed: 11/09/2022]
Abstract
Integrin beta 4 (ITGB4) overexpression in cancer cells contributes to cancer progression. However, the role of stromal ITGB4 expression in cancer progression remains poorly understood, despite stromal ITGB4 overexpression in malignant cancers. In our study, ITGB4-overexpressing triple negative breast cancer (TNBC) cells provided cancer-associated fibroblasts (CAFs) with ITGB4 proteins via exosomes, which induced BNIP3L-dependent mitophagy and lactate production in CAFs. In coculture assays, the ITGB4-induced mitophagy and glycolysis were suppressed in CAFs by knocking down ITGB4 or inhibiting exosome generation in MDA-MB-231, or blocking c-Jun or AMPK phosphorylation in CAFs. ITGB4-overexpressing CAF-conditioned medium promoted the proliferation, epithelial-to-mesenchymal transition, and invasion of breast cancer cells. In a co-transplant mouse model, MDA-MB-231 made a bigger tumor mass with CAFs than ITGB4 knockdown MDA-MB-231. Herein, we presented how TNBC-derived ITGB4 protein triggers glycolysis in CAFs via BNIP3L-dependent mitophagy and suggested the possibility that ITGB4-induced mitophagy could be targeted as a cancer therapy.
Collapse
|
20
|
Ni Y, Wang X, Yin X, Li Y, Liu X, Wang H, Liu X, Zhang J, Gao H, Shi B, Zhao S. Plectin protects podocytes from adriamycin-induced apoptosis and F-actin cytoskeletal disruption through the integrin α6β4/FAK/p38 MAPK pathway. J Cell Mol Med 2018; 22:5450-5467. [PMID: 30187999 PMCID: PMC6201223 DOI: 10.1111/jcmm.13816] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/16/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023] Open
Abstract
Podocyte injury is an early pathological change characteristic of various glomerular diseases, and apoptosis and F‐actin cytoskeletal disruption are typical features of podocyte injury. In this study, we found that adriamycin (ADR) treatment resulted in typical podocyte injury and repressed plectin expression. Restoring plectin expression protected against ADR‐induced podocyte injury whereas siRNA‐mediated plectin silencing produced similar effects as ADR‐induced podocyte injury, suggesting that plectin plays a key role in preventing podocyte injury. Further analysis showed that plectin repression induced significant integrin α6β4, focal adhesion kinase (FAK) and p38 MAPK phosphorylation. Mutating Y1494, a key tyrosine residue in the integrin β4 subunit, blocked FAK and p38 phosphorylation, thereby alleviating podocyte injury. Inhibitor studies demonstrated that FAK Y397 phosphorylation promoted p38 activation, resulting in podocyte apoptosis and F‐actin cytoskeletal disruption. In vivo studies showed that administration of ADR to rats resulted in significantly increased 24‐hour urine protein levels along with decreased plectin expression and activated integrin α6β4, FAK, and p38. Taken together, these findings indicated that plectin protects podocytes from ADR‐induced apoptosis and F‐actin cytoskeletal disruption by inhibiting integrin α6β4/FAK/p38 pathway activation and that plectin may be a therapeutic target for podocyte injury‐related glomerular diseases.
Collapse
Affiliation(s)
- Yongliang Ni
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Department of Urology, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Xin Wang
- Department of Urology, Tengzhou Central People's Hospital affiliated to Jining Medical College, Xintan Road 181, Tengzhou, China
| | - Xiaoxuan Yin
- Department of Traditional Chinese Medicine, Yankuang Group General Hospital, Zoucheng, China
| | - Yan Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xigao Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Haixin Wang
- Department of Urology, Yankuang Group General Hospital, Zoucheng, China
| | - Xiangjv Liu
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University
| | - Jun Zhang
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University
| | - Haiqing Gao
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shaohua Zhao
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University
| |
Collapse
|
21
|
SEC-induced activation of ANXA7 GTPase suppresses prostate cancer metastasis. Cancer Lett 2017; 416:11-23. [PMID: 29247827 DOI: 10.1016/j.canlet.2017.12.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 02/06/2023]
Abstract
Annexin A7 (ANXA7) is a suppressor of tumorigenesis and metastasis in prostate cancer. Activated ANXA7 GTPase promotes prostate cancer cell apoptosis. However, the role and underlying mechanism of ANXA7 GTPase in prostate cancer metastasis have not been established. RKIP is a metastatic suppressor and downregulated in prostate cancer metastases. The binding of RKIP and its target proteins could inhibit the activation of its interactive partners. However, the effect of RKIP on ANXA7 GTPase activation is not clear. Here, we report that activation of ANXA7 GTPase by a small molecule SEC ((S)-ethyl 1-(3-(4-chlorophenoxy)-2-hydroxypropyl)-3- (4-methoxyphenyl)-1H-pyrazole-5-carboxylate) effectively inhibited prostate cancer metastasis. Mechanistically, activated ANXA7 promoted AMPK phosphorylation, leading to decreased mTORC1 activity, suppressed STAT3 nuclear translocation, and downregulation of pro-metastatic genes, including CCL2, APLN, and IL6ST. Conversely, RKIP interacted with ANXA7 and impaired activation of ANXA7 GTPase by SEC and its downstream signaling pathway. Notably, SEC treatment suppressed metastasis of prostate cancer cells in in vivo orthotopic analysis. Together, our findings provide a novel insight into how metastasis of prostate cancer with low RKIP expression is suppressed by SEC-induced activation of ANXA7 GTPase via the AMPK/mTORC1/STAT3 signaling pathway.
Collapse
|
22
|
Clinical and therapeutic potential of protein kinase PKR in cancer and metabolism. Expert Rev Mol Med 2017; 19:e9. [PMID: 28724458 DOI: 10.1017/erm.2017.11] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The protein kinase R (PKR, also called EIF2AK2) is an interferon-inducible double-stranded RNA protein kinase with multiple effects on cells that plays an active part in the cellular response to numerous types of stress. PKR has been extensively studied and documented for its relevance as an antiviral agent and a cell growth regulator. Recently, the role of PKR related to metabolism, inflammatory processes, cancer and neurodegenerative diseases has gained interest. In this review, we summarise and discuss the involvement of PKR in several cancer signalling pathways and the dual role that this kinase plays in cancer disease. We emphasise the importance of PKR as a molecular target for both conventional chemotherapeutics and emerging treatments based on novel drugs, and its potential as a biomarker and therapeutic target for several pathologies. Finally, we discuss the impact that the recent knowledge regarding PKR involvement in metabolism has in our understanding of the complex processes of cancer and metabolism pathologies, highlighting the translational research establishing the clinical and therapeutic potential of this pleiotropic kinase.
Collapse
|
23
|
Zhao X, Dong W, Gao Y, Shin DS, Ye Q, Su L, Jiang F, Zhao B, Miao J. Novel indolyl-chalcone derivatives inhibit A549 lung cancer cell growth through activating Nrf-2/HO-1 and inducing apoptosis in vitro and in vivo. Sci Rep 2017. [PMID: 28634389 PMCID: PMC5478673 DOI: 10.1038/s41598-017-04411-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Increasing evidence indicates that Nrf-2, named the nuclear factor-erythroid 2-related factor, may perform anticancer function. In this study, a series of novel substituted phenyl- (3-methyl-1H-indol-2-yl)-prop-2-en-1-one (indolyl-chalcone) derivatives were synthesized and their effects on Nrf-2 activity were observed. We found that compounds 3a-3d and 6c elevated Nrf-2 activity. Then we evaluated their anticancer activities in vitro and in vivo by utilizing human lung cancer cell line A549. The in vitro results showed that among the compounds, 3d performed effectively anti-growth activity by inducing A549 lung cancer cell apoptosis and activating Nrf-2/HO-1 (heme oxygenase-1) pathway. In vivo, we proved that compound 3d inhibited the tumor growth effectively through inducing cell apoptosis without affecting CAM normal angiogenesis. These data suggest that our discovery of a novel Nrf-2 activator compound 3d would provide a new point of human lung cancer treatment.
Collapse
Affiliation(s)
- Xuan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, China
| | - WenLiang Dong
- Department of Chemistry, Changwon National University, Changwon, 51140, South Korea
| | - YuanDi Gao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, China
| | - Dong-Shoo Shin
- Department of Chemistry, Changwon National University, Changwon, 51140, South Korea
| | - Qing Ye
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Le Su
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, China
| | - Fan Jiang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - BaoXiang Zhao
- Institute of Organic Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China.
| | - JunYing Miao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, China. .,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
24
|
Vuong NQ, Goegan P, De Rose F, Breznan D, Thomson EM, O'Brien JS, Karthikeyan S, Williams A, Vincent R, Kumarathasan P. Responses of A549 human lung epithelial cells to cristobalite and α-quartz exposures assessed by toxicoproteomics and gene expression analysis. J Appl Toxicol 2016; 37:721-731. [PMID: 27917503 PMCID: PMC5434822 DOI: 10.1002/jat.3420] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/12/2016] [Accepted: 10/26/2016] [Indexed: 12/19/2022]
Abstract
In this study, we used cytotoxicity assays, proteomic and gene expression analyses to examine the difference in response of A549 cells to two silica particles that differ in physical properties, namely cristobalite (CR) and α‐quartz (Min‐U‐Sil 5, MI). Cytotoxicity assays such as lactate dehydrogenase release, 5‐bromo‐2′‐deoxyuridine incorporation and cellular ATP showed that both silica particles could cause cell death, decreased cell proliferation and metabolism in the A549 human lung epithelial cells. While cytotoxicity assays revealed little difference between CR and MI exposures, proteomic and gene expression analyses unveiled both similar and unique molecular changes in A549 cells. For instance, two‐dimensional gel electrophoresis data indicated that the expression of proteins in the cell death (e.g., ALDH1A1, HTRA2 and PRDX6) and cell proliferation (e.g., FSCN1, HNRNPAB and PGK1) pathways were significantly different between the two silica particles. Reverse transcription–polymerase chain reaction data provided additional evidence supporting the proteomic findings. Preliminary assessment of the physical differences between CR and MI suggested that the extent of surface interaction between particles and cells could explain some of the observed biological effects. However, the differential dose–response curves for some other genes and proteins suggest that other physical attributes of particulate matter can also contribute to particulate matter‐related cellular toxicity. Our results demonstrated that toxicoproteomic and gene expression analyses are sensitive in distinguishing subtle toxicity differences associated with silica particles of varying physical properties compared to traditional cytotoxicity endpoints. Copyright © 2016 Her Majesty the Queen in Right of Canada. Journal of Applied Toxicology published by John Wiley & Sons, Ltd. In this study, we used cytotoxicity assays, proteomic and gene expression analyses to examine the difference in response of A549 cells to two silica particles that differ in physical properties, namely cristobalite (CR) and α‐quartz (Min‐U‐Sil 5, MI). Cytotoxicity assays such as lactate dehydrogenase release, 5‐bromo‐2'‐deoxyuridine incorporation and cellular ATP showed that both silica particles could cause cell death, decreased cell proliferation and metabolism in the A549 human lung epithelial cells. While cytotoxicity assays revealed little difference between CR and MI exposures, proteomic and gene expression analyses unveiled both similar and unique molecular changes in A549 cells.
Collapse
Affiliation(s)
- Ngoc Q Vuong
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada.,Department of Biochemistry, Faculty of Science, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Patrick Goegan
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Francesco De Rose
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Dalibor Breznan
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Errol M Thomson
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Julie S O'Brien
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Subramanian Karthikeyan
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Andrew Williams
- Biostatistics Section, Population Studies Division, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Renaud Vincent
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada.,Department of Biochemistry, Faculty of Science, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Premkumari Kumarathasan
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| |
Collapse
|
25
|
Liu S, Wang Z, Miao J. Potential roles of annexin A7 GTPase in autophagy, senescence and apoptosis. RSC Adv 2016. [DOI: 10.1039/c6ra21736b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This review covers the roles of ANXA7 GTPase in orchestrating autophagy, senescence and apoptosis interactive networks in various cell types.
Collapse
Affiliation(s)
- ShuYan Liu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology
- School of Life Science
- Shandong University
- Jinan 250100
- China
| | - ZhaoYang Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology
- School of Life Science
- Shandong University
- Jinan 250100
- China
| | - JunYing Miao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology
- School of Life Science
- Shandong University
- Jinan 250100
- China
| |
Collapse
|