1
|
Moore LL, Qu D, Chandrekesan P, Pitts K, May R, Anderson BE, Brown M, Houchen CW. A Novel D-peptide modulates DCLK1 Gelsolin interactions, reducing PDAC tumor growth. RESEARCH SQUARE 2025:rs.3.rs-6099914. [PMID: 40162222 PMCID: PMC11952641 DOI: 10.21203/rs.3.rs-6099914/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
What drives inflammation-associated tumorigenesis and progression in pancreatic ductal adenocarcinoma (PDAC)? Doublecortin-like kinase 1 (DCLK1) is a central driver of inflammation-associated tumorigenesis, with elevated expression linked to worse clinical outcomes. Isoform 4, which lacks microtubule-binding domains but contains a unique extracellular domain (ECD), plays a pivotal role in tumor progression. We identified novel D-peptides that selectively target this ECD, significantly suppressing PDAC cell proliferation in vitro and tumor growth in xenograft models without inducing cell death. In silico modeling and binding assays revealed DCLK1 isoform 4 interacts with pro-tumorigenic proteins like plasma gelsolin (pGSN), with D-peptides modulating these interactions. These findings underscore DCLK1's non-kinase functions as a therapeutic target and highlight novel avenues for developing precision treatments aimed at halting cancer progression and improving patient outcomes.
Collapse
Affiliation(s)
| | - Dongfeng Qu
- University of Oklahoma Health Sciences Center
| | | | | | - Randal May
- University of Oklahoma Health Sciences Center
| | | | - Milton Brown
- Macon & Joan Brock Virginia Health Sciences at Old Dominion University
| | | |
Collapse
|
2
|
Liu W, Yin H, Xie Z, Fang F, Chu J, Yang L, Huang L, Tu S, Cai H, Wu Z, Wei A, Liu C, Hong Y, Tian X, Cheng Y, Pan J, Wang N, Zhang K. FYB1-targeted modulation of CAPG promotes AML progression. Mol Cell Biochem 2025; 480:985-999. [PMID: 38700746 PMCID: PMC11836086 DOI: 10.1007/s11010-024-04992-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/17/2024] [Indexed: 02/19/2025]
Abstract
Acute myeloid leukemia (AML) is a rare and heterogeneous disease. Over the past few decades, patient prognosis has improved with continuous improvements in treatment, but outcomes for some patients with primary drug resistance or relapse after treatment remain poor. Additional therapies to improve outcomes for these patients are urgently needed. FYB1 expression differs substantially between AML tissues and normal tissues. High FYB1 expression is correlated with poorer overall survival (OS), indicating that FYB1 may regulate AML progression. Therefore, understanding the effect of FYB1 on AML could improve the success rate of therapeutic approaches and prognosis for patients with AML. In this study, through analysis of large databases and both in vivo and in vitro experiments, we assessed the expression and role of FYB1 in AML and the relationship of FYB with patient prognosis. Downstream targets of the FYB1 gene were analyzed by RNA-seq. Database mining and in vitro experiments were used to further clarify the effect of the downstream target gelsolin-like actin-capping protein (CAPG) on AML cells and its relationship with patient prognosis. FYB1 expression was significantly higher in AML tissue and corresponded with a poor prognosis. FYB1 knockdown inhibited AML cell proliferation, promoted cell apoptosis, reduced cell adhesion capability and significantly reduced the tumor formation rate in mice. In addition, FYB1 knockdown induced a notable decrease in CAPG expression. The suppression of CAPG significantly inhibited cell proliferation and increased cell apoptosis. The conclusions of this study underscore the pivotal role of the FYB1/CAPG axis in promoting AML. We propose that the FYB1/CAPG axis could serve as a new thread in the development of therapeutic strategies for AML.
Collapse
Affiliation(s)
- Wenyuan Liu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Hongli Yin
- Institute of Pediatric Research, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou City, 215003, China
| | - Zhiwei Xie
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou City, 215003, China
| | - Jinhua Chu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Linhai Yang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Lingling Huang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Songji Tu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Huaju Cai
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Zhengyu Wu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Anbang Wei
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Chengzhu Liu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Yi Hong
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Xiaotong Tian
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China
| | - Yan Cheng
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China.
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou City, 215003, China.
| | - Ningling Wang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China.
| | - Kunlong Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei City, 230601, Anhui Province, China.
| |
Collapse
|
3
|
Li M, Liu Z, Cao X, Xiao W, Wang S, Zhao C, Zhao Y, Xie Y. [Gly14]-Humanin ameliorates high glucose-induced endothelial senescence via SIRT6. Sci Rep 2024; 14:30924. [PMID: 39730568 DOI: 10.1038/s41598-024-81878-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
High glucose (HG) induced endothelial senescence is related to endothelial dysfunction and cardiovascular complications in diabetic patients. Humanin, a member of mitochondrial derived peptides (MDPs), is thought to contribute to aging-related cardiovascular protection. The goal of the study is to explore the pathogenesis of HG-induced endothelial senescence and potential anti-senescent effects of Humanin. Human umbilical vein endothelial cells (HUVECs) were exposed to glucose to induce senescence, determined by β-galactosidase staining and the expressions of p21, p53, and p16. A clinically relevant dose of HG (15 mM, HG) induced endothelial senescence after 72 h incubation without elevated apoptosis. HG-induced senescence was attributed to the induction of reactive oxygen species (ROS) caused by SIRT6 downregulation, as ROS inhibitor N-acetyl cysteine blocked HG-induced senescence, while inactivation of SIRT6 increased ROS levels and promoted senescence. Strikingly. pretreatment with [Gly14]-Humanin (HNG) antagonized the downregulation of SIRT6 in response to HG and alleviated ROS production and cell senescence. HG-induced reduction of SIRT6 results in ROS overproduction and endothelial senescence. Humanin protects against HG-induced endothelial senescence via SIRT6. This study provides new directions for biological products related to Humanin to be a potential candidate for the prevention of vascular aging in diabetes.
Collapse
Affiliation(s)
- Muqin Li
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Department of Endocrinology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, 222061, JiangSu, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215004, China
| | - Zhihua Liu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xueqin Cao
- Department of Endocrinology, The Fourth Affiliated Hospital of Soochow University, Chongwen Road No. 9, Suzhou, 215000, Jiangsu, China
| | - Wenjin Xiao
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Shurong Wang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Chengyuan Zhao
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Department of endocrinology, Taizhou school of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Ying Zhao
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Soochow Medical College of Soochow University, Suzhou, 215123, China.
| | - Ying Xie
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| |
Collapse
|
4
|
Fernandez MK, Sinha M, Kühnemuth R, Renz M. Repeated FRAP of the actin-binding protein CapG in the cell nucleus-a functional assay for EGF signaling in the single live breast cancer cell. Sci Rep 2024; 14:23159. [PMID: 39369027 PMCID: PMC11455965 DOI: 10.1038/s41598-024-73887-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 09/23/2024] [Indexed: 10/07/2024] Open
Abstract
Compartmentalization and differential distribution of proteins within a cell maintain cellular function and viability. CapG is a gelsolin-related actin-binding protein that distributes in steady state diffusively throughout cytoplasm and cell nucleus. To detect changes in CapG's nucleocytoplasmic shuttling in response to external stimuli on the single cell level, we established repeated FRAP experiments of one and the same breast cancer cell. With this experimental set up, we found that ATP-depletion reversibly decreased CapG's shuttling into the cell nucleus. The addition of epidermal growth factor (EGF) increased CapG's nuclear shuttling within minutes. Serum-starvation doubled the number of breast cancer cells from 40 to 80% displaying increased CapG shuttling in response to EGF. Testing five different potential CapG phosphorylation sites, we found that serine 70 mediates the increase in CapG's nuclear shuttling triggered by EGF. Thus, repeated FRAP of CapG in the cell nucleus can be used as functional readout of signaling cascades in the same single live breast cancer cell.
Collapse
Affiliation(s)
| | - M Sinha
- Stanford University, Stanford, USA
| | - R Kühnemuth
- Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - M Renz
- Department of Gynecology With Center for Oncological Surgery, Charité - Universitätsmedizin Berlin, Mittelallee 9, 13353, Berlin, Germany.
| |
Collapse
|
5
|
Hsieh CC, Li TW, Li CC, Chen SH, Wei YL, Chiang NJ, Shen CH. DKK1 as a chemoresistant protein modulates oxaliplatin responses in colorectal cancer. Oncogenesis 2024; 13:34. [PMID: 39333078 PMCID: PMC11436992 DOI: 10.1038/s41389-024-00537-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
Oxaliplatin is effective against colorectal cancer (CRC), but resistance hampers treatment. We found upregulated Dickkopf-1 (DKK1, a secreted protein) in oxaliplatin-resistant (OR) CRC cell lines and DKK1 levels increased by more than 2-fold in approximately 50% of oxaliplatin-resistant CRC tumors. DKK1 activates AKT via cytoskeleton-associated protein 4 (CKAP4, a DKK1 receptor), modulating oxaliplatin responses in vitro and in vivo. The leucine zipper (LZ) domain of CKAP4 and cysteine-rich domain 1 (CRD1) of secreted DKK1 are crucial for their interaction and AKT signaling. By utilizing the LZ protein, we disrupted DKK1 signaling, enhancing oxaliplatin sensitivity in OR CRC cells and xenograft tumors. This suggests that DKK1 as a chemoresistant factor in CRC via AKT activation. Targeting DKK1 with the LZ protein offers a promising therapeutic strategy for oxaliplatin-resistant CRC with high DKK1 levels. This study sheds light on oxaliplatin resistance mechanisms and proposes an innovative intervention for managing this challenge.
Collapse
Affiliation(s)
- Chi-Che Hsieh
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 704, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Ting-Wei Li
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 704, Taiwan
- Department of Life Sciences, National Cheng Kung University, Tainan, 704, Taiwan
| | - Chun-Chun Li
- Department of Life Sciences, National Cheng Kung University, Tainan, 704, Taiwan
| | - Shang-Hung Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 704, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan
| | - You-Lin Wei
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 704, Taiwan
| | - Nai-Jung Chiang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| | - Che-Hung Shen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 704, Taiwan.
- Doctoral Program in Tissue Engineering and Regenerative Medicine, Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
6
|
Long Y, Wu J, Shen Y, Gan C, Zhang C, Wang G, Jing J, Zhang C, Pan W. CAPG is a novel biomarker for early gastric cancer and is involved in the Wnt/β-catenin signaling pathway. Cell Death Discov 2024; 10:15. [PMID: 38191512 PMCID: PMC10774411 DOI: 10.1038/s41420-023-01767-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 01/10/2024] Open
Abstract
Past studies have shown that the Gelsolin-like actin-capping protein (CAPG) regulates cell migration and proliferation and is strongly associated with tumor progression. We present the first study of the mechanism of action of CAPG in early gastric cancer (EGC). We demonstrate that CAPG expression is upregulated in gastric cancer (GC) especially EGC. CAPG promotes GC proliferation, migration, invasion, and metastasis in vivo and in vitro. More importantly, CAPG plays a role in GC by involving the Wnt/β-catenin signaling pathway. Our findings suggest that CAPG may function as a novel biomarker for EGC.
Collapse
Affiliation(s)
- Yan Long
- Cancer Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, Zhejiang, China
| | - JiaQi Wu
- Department of Gastroenterology, Affiliated Hospital of Hangzhou Normal University, 310015, Hangzhou, China
| | - Yu Shen
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenxiao Gan
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuandong Zhang
- The Medical College of QingDao University, Qingdao, Shandong, China
| | - Gang Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Jiyong Jing
- Department of Medical Education and Simulation Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
| | - Chenjing Zhang
- Cancer Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, Zhejiang, China.
| | - Wensheng Pan
- Cancer Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Halász H, Szatmári Z, Kovács K, Koppán M, Papp S, Szabó-Meleg E, Szatmári D. Changes of Ex Vivo Cervical Epithelial Cells Due to Electroporation with JMY. Int J Mol Sci 2023; 24:16863. [PMID: 38069185 PMCID: PMC10706833 DOI: 10.3390/ijms242316863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
The ionic environment within the nucleoplasm might diverge from the conditions found in the cytoplasm, potentially playing a role in the cellular stress response. As a result, it is conceivable that interactions of nuclear actin and actin-binding proteins (ABPs) with apoptosis factors may differ in the nucleoplasm and cytoplasm. The primary intracellular stress response is Ca2+ influx. The junctional mediating and regulating Y protein (JMY) is an actin-binding protein and has the capability to interact with the apoptosis factor p53 in a Ca2+-dependent manner, forming complexes that play a regulatory role in cytoskeletal remodelling and motility. JMY's presence is observed in both the cytoplasm and nucleoplasm. Here, we show that ex vivo ectocervical squamous cells subjected to electroporation with JMY protein exhibited varying morphological alterations. Specifically, the highly differentiated superficial and intermediate cells displayed reduced nuclear size. In inflamed samples, nuclear enlargement and simultaneous cytoplasmic reduction were observable and showed signs of apoptotic processes. In contrast, the less differentiated parabasal and metaplastic cells showed increased cytoplasmic activity and the formation of membrane protrusions. Surprisingly, in severe inflammation, vaginosis or ASC-US (Atypical Squamous Cells of Undetermined Significance), JMY appears to influence only the nuclear and perinuclear irregularities of differentiated cells, and cytoplasmic abnormalities still existed after the electroporation. Our observations can provide an appropriate basis for the exploration of the relationship between cytopathologically relevant morphological changes of epithelial cells and the function of ABPs. This is particularly important since ABPs are considered potential diagnostic and therapeutic biomarkers for both cancers and chronic inflammation.
Collapse
Affiliation(s)
- Henriett Halász
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| | | | - Krisztina Kovács
- Department of Pathology, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | | | - Szilárd Papp
- DaVinci Clinics, 7635 Pécs, Hungary; (M.K.); (S.P.)
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| | - Dávid Szatmári
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| |
Collapse
|
8
|
Alsofyani AA, Nedjadi T. Gelsolin, an Actin-Binding Protein: Bioinformatic Analysis and Functional Significance in Urothelial Bladder Carcinoma. Int J Mol Sci 2023; 24:15763. [PMID: 37958747 PMCID: PMC10647509 DOI: 10.3390/ijms242115763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 11/15/2023] Open
Abstract
The involvement of the actin-regulatory protein, gelsolin (GSN), in neoplastic transformation has been reported in different cancers including bladder cancer. However, the exact mechanism by which GSN influences bladder cancer development is not well understood. Here, we sought to reveal the functional significance of GSN in bladder cancer by undertaking a comprehensive bioinformatic analysis of TCGA datasets and through the assessment of multiple biological functions. GSN expression was knocked down in bladder cancer cell lines with two siRNA isoforms targeting GSN. Proliferation, migration, cell cycle and apoptosis assays were carried out. GSN expression, enrichment analysis, protein-protein interaction and immune infiltration analysis were verified through online TCGA tools. The data indicated that GSN expression is associated with bladder cancer proliferation, migration and enhanced cell apoptosis through regulation of NF-κB expression. GSN expression correlated with various inflammatory cells and may influence the immunity of the tumor microenvironment. Computational analysis identified several interacting partners which are associated with cancer progression and patient outcome. The present results demonstrate that GSN plays an important role in bladder cancer pathogenesis and may serve as a potential biomarker and therapeutic target for cancer therapy.
Collapse
Affiliation(s)
| | - Taoufik Nedjadi
- King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Jeddah 21423, Saudi Arabia;
| |
Collapse
|
9
|
Zhao Y, Ma R, Wang C, Hu R, Wu W, Sun X, Chen B, Zhang W, Chen Y, Zhou J, Yuan P. CAPG interference induces apoptosis and ferroptosis in colorectal cancer cells through the P53 pathway. Mol Cell Probes 2023; 71:101919. [PMID: 37468079 DOI: 10.1016/j.mcp.2023.101919] [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: 03/15/2023] [Revised: 06/21/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
PURPOSE Given the high incidence and mortality rates of colorectal cancer (CRC) and the inadequacy of existing treatments for many patients, this study aimed to explore the potential of Capping Actin Protein (CAPG), a protein involved in actin-related movements, as a novel therapeutic target for CRC. METHODS Bioinformatic analysis of gene expression was conducted using the UALCAN website. Cell proliferation was measured using the CCK-8 kit. Cell cycle, apoptosis, and ferroptosis were analyzed using flow cytometry. Tumorigenesis was evaluated by the subcutaneous inoculation of CRC cells into BALB/c nude female mice. Differentially expressed genes and signaling pathways were identified using RNA sequencing. RESULTS CAPG was significantly overexpressed in human CRC tissues and its upregulation was correlated with poor overall survival. CAPG knockdown led to notable inhibition of CRC cells in vitro and in vivo. Interference with CAPG blocked the cell cycle at the G1 phase and triggered apoptosis and ferroptosis by upregulating the P53 pathway in CRC cells. CONCLUSION CRC patients with higher CAPG levels have a poorer prognosis. CAPG inhibits apoptosis and ferroptosis, while promoting CRC cell proliferation by repressing the P53 pathway. Our study suggests that CAPG may be a potential therapeutic target for CRC prognosis and treatment.
Collapse
Affiliation(s)
- Yingying Zhao
- Guangdong Institute of Gastroenterology, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rui Ma
- Guangdong Institute of Gastroenterology, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chuyue Wang
- Guangdong Institute of Gastroenterology, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rong Hu
- Guangdong Institute of Gastroenterology, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weili Wu
- Guangdong Institute of Gastroenterology, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiang Sun
- Department of Medical Informatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Baotao Chen
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | | | - You Chen
- Guangdong Institute of Gastroenterology, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiajian Zhou
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Ping Yuan
- Guangdong Institute of Gastroenterology, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
10
|
Ma Q, Zhao M, Long B, Li H. Super-enhancer-associated gene CAPG promotes AML progression. Commun Biol 2023; 6:622. [PMID: 37296281 PMCID: PMC10256737 DOI: 10.1038/s42003-023-04973-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Acute myeloid leukemia is the most common acute leukemia in adults, the barrier of refractory and drug resistance has yet to be conquered in the clinical. Abnormal gene expression and epigenetic changes play an important role in pathogenesis and treatment. A super-enhancer is an epigenetic modifier that promotes pro-tumor genes and drug resistance by activating oncogene transcription. Multi-omics integrative analysis identifies the super-enhancer-associated gene CAPG and its high expression level was correlated with poor prognosis in AML. CAPG is a cytoskeleton protein but has an unclear function in AML. Here we show the molecular function of CAPG in regulating NF-κB signaling pathway by proteomic and epigenomic analysis. Knockdown of Capg in the AML murine model resulted in exhausted AML cells and prolonged survival of AML mice. In conclusion, SEs-associated gene CAPG can contributes to AML progression through NF-κB.
Collapse
Affiliation(s)
- Qian Ma
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, China
| | - Minyi Zhao
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Bing Long
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haixia Li
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, China.
| |
Collapse
|
11
|
Hartl L, Maarschalkerweerd PAF, Butler JM, Manz XD, Thijssen VLJL, Bijlsma MF, Duitman J, Spek CA. C/EBPδ Suppresses Motility-Associated Gene Signatures and Reduces PDAC Cell Migration. Cells 2022; 11:3334. [PMID: 36359732 PMCID: PMC9655908 DOI: 10.3390/cells11213334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 10/26/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is among the most aggressive human cancers and occurs globally at an increasing incidence. Metastases are the primary cause of cancer-related death and, in the majority of cases, PDAC is accompanied by metastatic disease at the time of diagnosis, making it a particularly lethal cancer. Regrettably, to date, no curative treatment has been developed for patients with metastatic disease, resulting in a 5-year survival rate of only 11%. We previously found that the protein expression of the transcription factor CCAAT/Enhancer-Binding Protein Delta (C/EBPδ) negatively correlates with lymph node involvement in PDAC patients. To better comprehend the etiology of metastatic PDAC, we explored the role of C/EBPδ at different steps of the metastatic cascade, using established in vitro models. We found that C/EBPδ has a major impact on cell motility, an important prerequisite for tumor cells to leave the primary tumor and to reach distant sites. Our data suggest that C/EBPδ induces downstream pathways that modulate actin cytoskeleton dynamics to reduce cell migration and to induce a more epithelial-like cellular phenotype. Understanding the mechanisms dictating epithelial and mesenchymal features holds great promise for improving the treatment of PDAC.
Collapse
Affiliation(s)
- Leonie Hartl
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| | - Pien A. F. Maarschalkerweerd
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Joe M. Butler
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Xue D. Manz
- Department of Pulmonary Medicine, Amsterdam UMC Location VU University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Victor L. J. L. Thijssen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam UMC Location VU University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Maarten F. Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| | - JanWillem Duitman
- Department of Pulmonary Medicine, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity, Inflammatory Diseases, 1105 AZ Amsterdam, The Netherlands
| | - C. Arnold Spek
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
12
|
Differential Intracellular Protein Distribution in Cancer and Normal Cells-Beta-Catenin and CapG in Gynecologic Malignancies. Cancers (Basel) 2022; 14:cancers14194788. [PMID: 36230711 PMCID: PMC9561979 DOI: 10.3390/cancers14194788] [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: 08/31/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 12/05/2022] Open
Abstract
Simple Summary The distribution and mobility of proteins inside the living cell can be used to differentiate cancer from normal cells. This review highlights differential protein distribution of two exemplary proteins, beta-catenin and CapG, and their role in gynecologic cancers. Recognizing differential protein distribution in cancer cells may have diagnostic and therapeutic implications. Abstract It is well-established that cancer and normal cells can be differentiated based on the altered sequence and expression of specific proteins. There are only a few examples, however, showing that cancer and normal cells can be differentiated based on the altered distribution of proteins within intracellular compartments. Here, we review available data on shifts in the intracellular distribution of two proteins, the membrane associated beta-catenin and the actin-binding protein CapG. Both proteins show altered distributions in cancer cells compared to normal cells. These changes are noted (i) in steady state and thus can be visualized by immunohistochemistry—beta-catenin shifts from the plasma membrane to the cell nucleus in cancer cells; and (ii) in the dynamic distribution that can only be revealed using the tools of quantitative live cell microscopy—CapG shuttles faster into the cell nucleus of cancer cells. Both proteins may play a role as prognosticators in gynecologic malignancies: beta-catenin in endometrial cancer and CapG in breast and ovarian cancer. Thus, both proteins may serve as examples of altered intracellular protein distribution in cancer and normal cells.
Collapse
|
13
|
The p53 and Calcium Regulated Actin Rearrangement in Model Cells. Int J Mol Sci 2022; 23:ijms23169078. [PMID: 36012344 PMCID: PMC9408879 DOI: 10.3390/ijms23169078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Long-term cellular stress maintains high intracellular Ca2+ concentrations which ultimately initiates apoptosis. Our interest is focused on how the gelsolin (GSN) and junctional mediating and regulating Y protein (JMY) play important roles in stress response. Both of these proteins can bind p53 and actin. We investigated using in vitro fluorescence spectroscopy and found that the p53 competes with actin in GSN to inhibit p53–JMY complex formation. A high Ca2+ level initializes p53 dimerization; the dimer competes with actin on JMY, which can lead to p53–JMY cotransport into the nucleus. Here we investigated how the motility and division rate of HeLa cells changes due to low-voltage electroporation of GSN or JMY in scratching assays. We revealed that JMY inhibits their motion, but that it can accelerate the cell division. GSN treatment slows down cell division but does not affect cell motility. HeLa cells fully recovered the gap 20 h after the electroporation with JMY and then started to release from the glass slides. Taken together, our in vitro results indicate that GSN and JMY may play an important role in the cellular stress response.
Collapse
|
14
|
Hsieh CH, Wang YC. Emerging roles of plasma gelsolin in tumorigenesis and modulating the tumor microenvironment. Kaohsiung J Med Sci 2022; 38:819-825. [PMID: 35942641 DOI: 10.1002/kjm2.12578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 02/06/2023] Open
Abstract
The protein expression of gelsolin, an actin scavenger controlling cytoskeletal remodeling, cell morphology, differentiation, movement, and apoptosis, has been found to be significantly decreased in several pathological conditions including neurodegenerative diseases, inflammatory disorders, and cancers. Its extracellular isoform, called plasma gelsolin (pGSN), is one of the most abundant plasma proteins in the circulation, and has emerged as a novel diagnostic biomarker for early disease detection. Current evidence reveals that gelsolin can function as either an oncoprotein or a tumor suppressor depending on the carcinoma type. Interestingly, recent studies have shown that pGSN is also involved in immunomodulation, revealing the multifunctional roles of pGSN in tumor progression. In this review, we discuss the current knowledge focusing on the roles of gelsolin in inflammation and wound healing, cancers, and tumor microenvironment. Future prospects of pGSN related studies and clinical application are also addressed.
Collapse
Affiliation(s)
- Chih-Hsiung Hsieh
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ching Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
15
|
He H, Liyanarachchi S, Li W, Comiskey DF, Yan P, Bundschuh R, Turkoglu AM, Brock P, Ringel MD, de la Chapelle A. Transcriptome analysis discloses dysregulated genes in normal appearing tumor-adjacent thyroid tissues from patients with papillary thyroid carcinoma. Sci Rep 2021; 11:14126. [PMID: 34238982 PMCID: PMC8266864 DOI: 10.1038/s41598-021-93526-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/22/2021] [Indexed: 01/10/2023] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer. The molecular characteristics of histologically normal appearing tissue adjacent to the tumor (NAT) from PTC patients are not well characterized. The aim of this study was to characterize the global gene expression profile of NAT and compare it with those of normal and tumor thyroid tissues. We performed total RNA sequencing with fresh frozen thyroid tissues from a cohort of three categories of samples including NAT, normal thyroid (N), and PTC tumor (T). Transcriptome analysis shows that NAT presents a unique gene expression profile, which was not associated with sex or the presence of lymphocytic thyroiditis. Among the differentially expressed genes (DEGs) of NAT vs N, 256 coding genes and 5 noncoding genes have been reported as cancer genes involved in cell proliferation, apoptosis, and/or tumorigenesis. Bioinformatics analysis with Ingenuity Pathway Analysis software revealed that “Cancer, Organismal Injury and Abnormalities, Cellular Response to Therapeutics, and Cellular Movement” were major dysregulated pathways in the NAT tissues. This study provides improved insight into the complexity of gene expression changes in the thyroid glands of patients with PTC.
Collapse
Affiliation(s)
- Huiling He
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA.,The Ohio State University Comprehensive Cancer Center, The Ohio State University, McCampbell Hall South Room 565, 1581 Dodd Drive, Columbus, OH, 43210, USA
| | - Sandya Liyanarachchi
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA.,The Ohio State University Comprehensive Cancer Center, The Ohio State University, McCampbell Hall South Room 565, 1581 Dodd Drive, Columbus, OH, 43210, USA
| | - Wei Li
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA.,The Ohio State University Comprehensive Cancer Center, The Ohio State University, McCampbell Hall South Room 565, 1581 Dodd Drive, Columbus, OH, 43210, USA
| | - Daniel F Comiskey
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA.,The Ohio State University Comprehensive Cancer Center, The Ohio State University, McCampbell Hall South Room 565, 1581 Dodd Drive, Columbus, OH, 43210, USA
| | - Pearlly Yan
- Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA.,The Ohio State University Comprehensive Cancer Center, The Ohio State University, McCampbell Hall South Room 565, 1581 Dodd Drive, Columbus, OH, 43210, USA
| | - Ralf Bundschuh
- Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA.,Department of Physics, The Ohio State University, Columbus, OH, 43210, USA.,Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
| | - Altan M Turkoglu
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA
| | - Pamela Brock
- Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA.,The Ohio State University Comprehensive Cancer Center, The Ohio State University, McCampbell Hall South Room 565, 1581 Dodd Drive, Columbus, OH, 43210, USA
| | - Matthew D Ringel
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA. .,The Ohio State University Comprehensive Cancer Center, The Ohio State University, McCampbell Hall South Room 565, 1581 Dodd Drive, Columbus, OH, 43210, USA.
| | - Albert de la Chapelle
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA.,The Ohio State University Comprehensive Cancer Center, The Ohio State University, McCampbell Hall South Room 565, 1581 Dodd Drive, Columbus, OH, 43210, USA
| |
Collapse
|
16
|
Yang C, Sun Y, Ouyang X, Li J, Zhu Z, Yu R, Wang L, Jia L, Ding G, Wang Y, Jiang F. Pain May Promote Tumor Progression via Substance P-Dependent Modulation of Toll-like Receptor-4. PAIN MEDICINE 2021; 21:3443-3450. [PMID: 32914185 DOI: 10.1093/pm/pnaa265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND In a previous study, persistent pain was suggested to be a risk factor for tumor patients. However, the mechanism underlying this phenomenon is still unclear. Substance P (SP), a pain-related neuropeptide secreted by the neural system and the immune system, plays an important role in the induction and maintenance of persistent pain. METHODS In this study, in order to explore whether SP participates in the influence of pain on tumor progression, the serum samples of lung cancer and breast cancer patients were collected and tested. An elevated expression of SP was found in patients with pain. RESULTS Cell pharmacological experiments revealed that SP can upregulate the expression of Toll-like receptor-4 (TLR-4) in tumor cells and increase the proliferation, migration, and invasive activity of tumor cells. As high expression of TLR-4 has the ability to enhance the biological activity of tumor cells, TLR-4 is thought to be involved in SP-induced tumor proliferation, migration, and invasion. Treatment of tumor cells with Aprepitant, a specific blocker of the NK-1 receptor, could reduce the expression of TLR-4 and reduce the proliferation, invasion, and migration activities of tumor cells; further proof of the influence of SP on TLR-4 expression depends on the NK-1 receptor located in tumor cells. CONCLUSIONS Based on the results above, we proposed a possible mechanism underlying pain affecting tumor progression: The presence of pain increases the content of SP in patients' blood, and elevated SP increases the expression of tumor TLR-4 by acting on the NK-1 receptor, which ultimately affects the biological activity of the tumor.
Collapse
Affiliation(s)
- Chao Yang
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Yunheng Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyan Ouyang
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Jing Li
- Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhen Zhu
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Ruihua Yu
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Li Wang
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Lin Jia
- Shanghai International Medical Center, Shanghai, China
| | - Gang Ding
- Shanghai International Medical Center, Shanghai, China
| | - Yaosheng Wang
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Feng Jiang
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| |
Collapse
|
17
|
Wang M, Ma X, Zhou K, Mao H, Liu J, Xiong X, Zhao X, Narva S, Tanaka Y, Wu Y, Guo C, Sugiyama H, Zhang W. Discovery of Pyrrole-imidazole Polyamides as PD-L1 Expression Inhibitors and Their Anticancer Activity via Immune and Nonimmune Pathways. J Med Chem 2021; 64:6021-6036. [PMID: 33949196 DOI: 10.1021/acs.jmedchem.1c00120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In recent years, PD-1 immune checkpoint inhibitors based on monoclonal antibodies have revolutionized cancer therapy, but there still exist unresolved issues, such as the high cost, the relatively low response rates, and so on, compared with small-molecule drugs. Herein a type of pyrrole-imidazole (Py-Im) polyamide as a small-molecule DNA binder was designed and synthesized, which could competitively bind to the same double-stranded DNA stretch in the PD-L1 promoter region as the STAT3 binding site and thus downregulate PD-L1 expression. It was demonstrated that the Py-Im polyamides directly caused apoptosis in tumor cells and retarded cell migration in the absence of T cells through inhibiting the Akt/caspase-3 pathway. Also, in a coculture system, they enhanced the T-cell-mediated killing of tumor cells by the reversal of immune escape. Because such polyamides induced antitumor effects via both immune and nonimmune pathways, they could be further developed as promising PD-L1 gene-targeting antitumor drugs.
Collapse
Affiliation(s)
- Ming Wang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.,Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xudong Ma
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.,Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Kang Zhou
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.,Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Huijuan Mao
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.,Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jiachun Liu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.,Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xuqiong Xiong
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.,Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xiaoyin Zhao
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.,Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Suresh Narva
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.,Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yoshimasa Tanaka
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Yanling Wu
- Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Chuanxin Guo
- Department of Chemistry, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - Hiroshi Sugiyama
- Department of Chemistry, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.,Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
18
|
Cancer stem cell transcriptome landscape reveals biomarkers driving breast carcinoma heterogeneity. Breast Cancer Res Treat 2021; 186:89-98. [PMID: 33389402 DOI: 10.1007/s10549-020-06045-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/02/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Breast carcinomas are heterogeneous diseases with distinct clinical outcomes and cancer stem cell (CSC) percentages. Exploring breast carcinoma stem cell landscape could help understand the heterogeneity of such cancers with profound clinical relevance. METHODS We conducted transcriptional profiling of CSCs and non-stem cancer cells isolated from three triple-negative breast carcinoma cell lines, analyzed the CSC transcriptome landscape that drives breast carcinoma heterogeneity through differentially expressed gene identification, gene ontology (GO) and pathway enrichment analyses as well as network construction, and experimentally validated the network hub gene. RESULTS We identified a CSC feature panel consisting of 122 and 381 over-represented and under-expressed genes capable of differentiating breast carcinoma subtypes. We also underpinned the prominent roles of the PI3K-AKT pathway in empowering carcinoma cells with uncontrolled proliferative and migrative abilities that ultimately foster cancer stemness, and revealed the potential promotive roles of ATP6V1B1 on breast carcinoma stemness through functional in vitro studies. CONCLUSIONS Our study contributes in identifying a CSC feature panel for breast carcinomas that drives breast carcinoma heterogeneity at the transcriptional level, which provides a reservoir for diagnostic marker and/or therapeutic target identification once experimentally validated as demonstrated by ATP6V1B1.
Collapse
|
19
|
Chen ZF, Huang ZH, Chen SJ, Jiang YD, Qin ZK, Zheng SB, Chen T. Oncogenic potential of macrophage‑capping protein in clear cell renal cell carcinoma. Mol Med Rep 2020; 23:80. [PMID: 33236143 PMCID: PMC7716403 DOI: 10.3892/mmr.2020.11718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Macrophage-capping protein (CapG) is a newly characterized oncogene involved in several types of cancer. However, the expression patterns and biological mechanisms of CapG in clear cell renal cell carcinoma (ccRCC) are unclear. The present study aimed to investigate the roles of CapG in the prognosis, proliferation and metastasis of ccRCC. In the present study, the expression of CapG was analyzed by western blotting in 24 paired ccRCC and adjacent normal tissue samples. Another 152 tissue samples from 152 patients with ccRCC were examined by immunohistochemistry. Compared with normal tissue, CapG expression was significantly increased in ccRCC tissue, and high CapG expression was associated with advanced tumor stage, histological grade, lymph node metastasis, and poor overall survival. Moreover, CapG was an independent predictor of survival. Lentivirus-mediated CapG knockdown significantly inhibited 786-O cell proliferation, migration, and invasion, induced cell cycle arrest at the G2/M phase, and increased apoptosis in vitro. Microarray analysis indicated that RAC, CDC42 and ERK/MAPK signaling were disrupted by CapG knockdown in 786-O cells. In conclusion, the present findings indicate that CapG plays an oncogenic role in ccRCC and may represent a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Zhuang-Fei Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ze-Hai Huang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Shi-Jun Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yao-Dong Jiang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zi-Ke Qin
- Department of Urology, Cancer Center, Sun Yat‑Sen University and State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong 510060, P.R. China
| | - Shao-Bin Zheng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Tong Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
20
|
Meleady P, Abdul Rahman R, Henry M, Moriarty M, Clynes M. Proteomic analysis of pancreatic ductal adenocarcinoma. Expert Rev Proteomics 2020; 17:453-467. [PMID: 32755290 DOI: 10.1080/14789450.2020.1803743] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC), which represents approximately 80% of all pancreatic cancers, is a highly aggressive malignant disease and one of the most lethal among all cancers. Overall, the 5-year survival rate among all pancreatic cancer patients is less than 9%; these rates have shown little change over the past 30 years. A more comprehensive understanding of the molecular mechanisms underlying this complex disease is crucial to the development of new diagnostic tools for early detection and disease monitoring, as well as to identify new and more effective therapeutics to improve patient outcomes. AREA COVERED We summarize recent advances in proteomic strategies and mass spectrometry to identify new biomarkers for early detection and monitoring of disease progression, predict response to therapy, and to identify novel proteins that have the potential to be 'druggable' therapeutic targets. An overview of proteomic studies that have been conducted to further our mechanistic understanding of metastasis and chemotherapy resistance in PDAC disease progression will also be discussed. EXPERT COMMENTARY The results from these PDAC proteomic studies on a variety of PDAC sample types (e.g., blood, tissue, cell lines, exosomes, etc.) provide great promise of having a significant clinical impact and improving patient outcomes.
Collapse
Affiliation(s)
- Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University , Dublin, Ireland
| | - Rozana Abdul Rahman
- St. Vincent's University Hospital , Dublin, Ireland.,St. Luke's Hospital , Dublin, Ireland
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University , Dublin, Ireland
| | - Michael Moriarty
- National Institute for Cellular Biotechnology, Dublin City University , Dublin, Ireland.,St. Luke's Hospital , Dublin, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University , Dublin, Ireland
| |
Collapse
|
21
|
Histoepigenetic analysis of the mesothelin network within pancreatic ductal adenocarcinoma cells reveals regulation of retinoic acid receptor gamma and AKT by mesothelin. Oncogenesis 2020; 9:62. [PMID: 32616712 PMCID: PMC7332500 DOI: 10.1038/s41389-020-00245-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 12/26/2022] Open
Abstract
To enable computational analysis of regulatory networks within the cancer cell in its natural tumor microenvironment, we develop a two-stage histoepigenetic analysis method. The first stage involves iterative computational deconvolution to estimate sample-specific cancer-cell intrinsic expression of a gene of interest. The second stage places the gene within a network module. We validate the method in simulation experiments, show improved performance relative to differential expression analysis from bulk samples, and apply it to illuminate the role of the mesothelin (MSLN) network in pancreatic ductal adenocarcinoma (PDAC). The network analysis and subsequent experimental validation in a panel of PDAC cell lines suggests AKT activation by MSLN through two known activators, retinoic acid receptor gamma (RARG) and tyrosine kinase non receptor 2 (TNK2). Taken together, these results demonstrate the potential of histoepigenetic analysis to reveal cancer-cell specific molecular interactions directly from patient tumor profiles.
Collapse
|
22
|
Weigand M, Degroote RL, Amann B, Renner S, Wolf E, Hauck SM, Deeg CA. Proteome profile of neutrophils from a transgenic diabetic pig model shows distinct changes. J Proteomics 2020; 224:103843. [PMID: 32470542 DOI: 10.1016/j.jprot.2020.103843] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/13/2020] [Accepted: 05/23/2020] [Indexed: 12/12/2022]
Abstract
INSC94Y transgenic pigs develop a stable diabetic phenotype early after birth and therefore allow studying the influence of hyperglycemia on primary immune cells in an early stage of diabetes mellitus in vivo. Since immune response is altered in diabetes mellitus, with deviant neutrophil function discussed as one of the possible causes in humans and mouse models, we investigated these immune cells in INSC94Y transgenic pigs and wild type controls at protein level. A total of 2371 proteins were quantified by label-free LC-MS/MS. Subsequent differential proteome analysis of transgenic animals and controls revealed clear differences in protein abundances, indicating a deviant behavior of granulocytes in the diabetic state. Interestingly, abundance of myosin regulatory light chain 9 (MLC-2C) was increased 5-fold in cells of diabetic pigs. MLC-2C directly affects cell contractility by regulating myosin ATPase activity, can act as transcription factor and was also associated with inflammation. It might contribute to impaired neutrophil cell adhesion, migration and phagocytosis. Our study provides novel insights into proteome changes in neutrophils from a large animal model for permanent neonatal diabetes mellitus and points to dysregulation of neutrophil function even in an early stage of this disease. Data are available via ProteomeXchange with identifier PXD017274. SIGNIFICANCE: Our studies provide novel basic information about the neutrophil proteome of pigs and contribute to a better understanding of molecular mechanisms involved in altered immune cell function in an early stage diabetes. We demonstrate proteins that are dysregulated in neutrophils from a transgenic diabetic pig and have not been described in this context so far. The data presented here are highly relevant for veterinary medicine and have translational quality for diabetes in humans.
Collapse
Affiliation(s)
- Maria Weigand
- Department of Veterinary Sciences, LMU, Munich, Germany
| | | | - Barbara Amann
- Department of Veterinary Sciences, LMU, Munich, Germany
| | - Simone Renner
- Gene Center and Department of Veterinary Sciences, LMU, Munich, Germany; Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Eckhard Wolf
- Gene Center and Department of Veterinary Sciences, LMU, Munich, Germany; Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU, Munich, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Germany
| | | |
Collapse
|
23
|
Ni M, Zhao Y, Zhang WJ, Jiang YJ, Fu H, Huang F, Li DJ, Shen FM. microRNA-802 accelerates hepatocellular carcinoma growth by targeting RUNX3. J Cell Physiol 2020; 235:7128-7135. [PMID: 32003017 DOI: 10.1002/jcp.29611] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/22/2020] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. Prognosis is often unfavorable. In this study, the effects of microRNA-802 (miR-802) on HCC progression were assessed in vivo and in vitro. miR-802 was found to be significantly upregulated in HCC tumor tissue compared to paired adjacent nontumor tissue. In vitro, transfection with a miR-802 mimic accelerated SMMC-7721 cellular proliferation, increased accumulation of the cell-cycle S-phase cell populations, as well as cell migration. In vivo injection of a miR-802 agomir promoted HCC proliferation in nude mice. Targets of miR-802 were predicted by miRWalk, miRanda, RNA22, and Targetscan. By luciferase reporter assay RUNX3 was identified as a direct target of miR-802. As judged by western blot analysis, RUNX3 was upregulated when miR-802 was inhibited. These data demonstrate increased miR-802 expression in patients with HCC and that miR-802 overexpression promotes tumor cell growth, in a RUNX3-dependent manner.
Collapse
Affiliation(s)
- Min Ni
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yi Zhao
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Wen-Jing Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yu-Jie Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Hui Fu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Fang Huang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
24
|
Gelsolin Promotes Cancer Progression by Regulating Epithelial-Mesenchymal Transition in Hepatocellular Carcinoma and Correlates with a Poor Prognosis. JOURNAL OF ONCOLOGY 2020; 2020:1980368. [PMID: 32377190 PMCID: PMC7199561 DOI: 10.1155/2020/1980368] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 11/03/2019] [Accepted: 11/09/2019] [Indexed: 12/19/2022]
Abstract
Gelsolin (GSN), a cytoskeletal protein, is frequently overexpressed in different cancers and promotes cell motility. The biological function of GSN in hepatocellular carcinoma (HCC) and its mechanism remain unclear. The expression of GSN was assessed in a cohort of 188 HCC patients. The effects of GSN on the migration and invasion of tumour cells were examined. Then, the role of GSN in tumour growth in vivo was determined by using a cancer metastasis assay. The possible mechanism by which GSN promotes HCC progression was explored. As a result, GSN was overexpressed in HCC tissues. High GSN expression was significantly correlated with late Edmondson grade, encapsulation, and multiple tumours. Patients with high GSN expression had worse overall survival (OS) and disease-free survival (DFS) than those with low GSN expression. GSN expression was identified as an independent risk factor in both OS (hazard risk (HR) = 1.620, 95% confidence interval (CI) = 1.105–2.373, P < 0.001) and DFS (HR = 1.744, 95% CI = 1.205–2.523, P=0.003). Moreover, GSN knockdown significantly inhibited the migration and invasion of HCC tumour cells, while GSN overexpression attenuated these effects by regulating epithelial-mesenchymal transition (EMT) In conclusion, GSN promotes cancer progression and is associated with a poor prognosis in HCC patients. GSN promotes HCC progression by regulating EMT.
Collapse
|
25
|
Lang Z, Chen Y, Zhu H, Sun Y, Zhang H, Huang J, Zou Z. Prognostic and clinicopathological significance of CapG in various cancers: Evidence from a meta-analysis. Pathol Res Pract 2019; 215:152683. [PMID: 31685300 DOI: 10.1016/j.prp.2019.152683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 09/23/2019] [Accepted: 10/06/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The gelsolin-like actin-capping protein (CapG) is an actin-binding protein in the gelsolin superfamily. Increasing evidence indicates that CapG is highly expressed in various types of cancer. However, the role of CapG in malignant tumors is still controversial. Therefore, we conducted a meta-analysis to assess the prognostic value and clinicopathological significance of CapG in malignant tumors. METHOD We searched for eligible studies in the PubMed, Web of Science, Embase, and Cochrane databases. Stata SE12.0 software was used for quantitative meta-analysis. The hazard ratios (HRs) and odds ratios (ORs) with 95% CI were pooled to assess the relationship between CapG expression and overall survival (OS), as well as clinicopathological parameters. RESULTS Sixteen studies with a total of 1987 cancer patients were included in this meta-analysis. The results showed that higher CapG expression was statistically correlated with shorter OS (HR 1.70, 95% CI 1.43-1.97, P < 0.001), positive lymph node metastasis (OR 1.91, 95% CI 1.19-3.09, P = 0.008), advanced TNM stage (OR 1.87, 95% CI 1.17-3.00, P = 0.009), advanced T-primary stage (OR 2.54, 95% CI 1.08-6.00, P = 0.033) and male sex (OR 1.77, 95% CI 1.23-2.56, P = 0.002). However, no significant correlation was observed between increased CapG expression and advanced age, larger tumor size, differentiation, or advanced histopathologic grading (P > 0.05). CONCLUSIONS High CapG expression is associated with a poor prognosis and worse clinicopathological parameters in various cancers. CapG is a potential prognostic biomarker and a possible clinicopathological predictive factor for various cancers.
Collapse
Affiliation(s)
- Zhiquan Lang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China; Nanchang University, Nanchang, Jiangxi, PR China
| | - Yuting Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China; Nanchang University, Nanchang, Jiangxi, PR China
| | - Hanyan Zhu
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China; Nanchang University, Nanchang, Jiangxi, PR China
| | - Yuting Sun
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China; Nanchang University, Nanchang, Jiangxi, PR China
| | - Hao Zhang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China; Nanchang University, Nanchang, Jiangxi, PR China
| | - Junfu Huang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China; Nanchang University, Nanchang, Jiangxi, PR China
| | - Zhenhong Zou
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, PR China.
| |
Collapse
|
26
|
He S, Guo H, Zhao T, Meng Y, Chen R, Ren J, Pan L, Fan G, Jiang M, Qin G, Zhu Y, Gao X. A Defined Combination of Four Active Principles From the Danhong Injection Is Necessary and Sufficient to Accelerate EPC-Mediated Vascular Repair and Local Angiogenesis. Front Pharmacol 2019; 10:1080. [PMID: 31607924 PMCID: PMC6767990 DOI: 10.3389/fphar.2019.01080] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 08/26/2019] [Indexed: 12/21/2022] Open
Abstract
Many compounds in Chinese medicine formulae, including Danhong injection (DHI) formulae, are capable of stimulating angiogenesis and promoting vascular repair, but their chemical basis and action mechanisms remain poorly defined. The aim of this study is to determine the minimal native chemical composition of DHI for the pro-angiogenesis activity and to evaluate its contribution from local endothelial cells (ECs) and bone marrow-derived endothelial progenitor cells (EPCs). Our study demonstrated that the action of DHI in accelerating the recovery of hindlimb blood flow in a ischemic rat model was attributable to its local CXCR4-mediated pro-angiogenesis activity in mature endothelial cells, as well as to its ability to promote the proliferation, migration, adhesion, and angiogenesis of EPCs via integrated activation of SDF-1α/CXCR4, VEGF/KDR, and eNOS/MMP-9 signal pathways. Combination experiments narrowed down the angiogenic activity into a few components in DHI. Reconstitution experiment defined that a combination of tanshinol, protocatechuic aldehyde, salvianolic acid B, and salvianolic acid C in their native proportion was necessary and sufficient for DHI's angiogenic activity. Compared with the full DHI, the minimal reconstituted four active principles had the same effects in promoting tube formation in vitro, improving perfusion and recovery of ischemic limb, and enhancing angiogenesis in ischemic mice post-hindlimb ischemia in vivo.
Collapse
Affiliation(s)
- Shuang He
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of Traditional Chinese Medicine, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Hao Guo
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tiechan Zhao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of Traditional Chinese Medicine, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Yanzhi Meng
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of Traditional Chinese Medicine, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Rongrong Chen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of Traditional Chinese Medicine, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Jie Ren
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of Traditional Chinese Medicine, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Lanlan Pan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of Traditional Chinese Medicine, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Guanwei Fan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, and Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Miaomiao Jiang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of Traditional Chinese Medicine, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Gangjian Qin
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine & School of Engineering, The University of Alabama at Birmingham (UAB), Birmingham, AL, United States
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of Traditional Chinese Medicine, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, and Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
27
|
Pan B, Zhang T, Yang W, Liu Y, Chen Y, Zhou Z, Tang Y, Zeng J, Liu Y, Zhao C, Guo Y. SNX3 suppresses the migration and invasion of colorectal cancer cells by reversing epithelial-to-mesenchymal transition via the β-catenin pathway. Oncol Lett 2019; 18:5332-5340. [PMID: 31612043 PMCID: PMC6781754 DOI: 10.3892/ol.2019.10860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/10/2019] [Indexed: 01/05/2023] Open
Abstract
The Wnt/β-catenin signaling pathway is a well-studied pathway that drives the carcinogenesis and metastasis of colorectal cancer (CRC). The secretion of Wnt proteins is essential for the continuous activation of Wnt/β-catenin signaling in CRC. The secretion of Drosophila wingless, which is homologous to the human Wnt protein, is mediated by sorting nexin 3 (SNX3) in Drosophila; however, the role of SNX3 in CRC remains unknown. In the present study it was demonstrated that SNX3 reduced the migratory and invasive ability of HCT116 human CRC cells, and reversed epithelial-mesenchymal transition (EMT). Conversely, in the HT29 CRC cell line, which endogenously expresses high levels of SNX3, short hairpin RNA or siRNA-mediated knockdown of SNX3 induced EMT, and enhanced cell migration and invasion. In addition, upregulation of SNX3 significantly inhibited metastasis of HCT116 cells to the lungs of mice. These SNX3-mediated effects were associated with downregulation of β-catenin. Taken together, by downregulating β-catenin, SNX3 may mediate EMT and reverse CRC metastasis.
Collapse
Affiliation(s)
- Biran Pan
- Medical Research Center, The Third People's Hospital of Chengdu, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610036, P.R. China
| | - Tongtong Zhang
- Medical Research Center, The Third People's Hospital of Chengdu, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610036, P.R. China
| | - Wei Yang
- Medical Research Center, The Third People's Hospital of Chengdu, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610036, P.R. China
| | - Yanjun Liu
- Department of Gastrointestinal Surgery, The Third People's Hospital of Chengdu, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610036, P.R. China
| | - Yuning Chen
- Clinical Laboratory Department, Xindu People's Hospital, Chengdu, Sichuan 610500, P.R. China
| | - Zheng Zhou
- Medical Research Center, The Third People's Hospital of Chengdu, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610036, P.R. China
| | - Yan Tang
- Department of Pathology, The Third People's Hospital of Chengdu, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610036, P.R. China
| | - Jiawei Zeng
- Clinical Laboratory Department, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Yilun Liu
- Clinical Laboratory Department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Cong Zhao
- Department of Digestive Diseases, The Third People's Hospital of Chengdu, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610036, P.R. China
| | - Yuanbiao Guo
- Medical Research Center, The Third People's Hospital of Chengdu, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610036, P.R. China
| |
Collapse
|
28
|
Huang X, Gu R, Li J, Yang N, Cheng Z, Si W, Chen P, Huang W, Dong X. Diketopyrrolopyrrole-Au(I) as singlet oxygen generator for enhanced tumor photodynamic and photothermal therapy. Sci China Chem 2019. [DOI: 10.1007/s11426-019-9531-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
29
|
Zhou K, Liu J, Xiong X, Cheng M, Hu X, Narva S, Zhao X, Wu Y, Zhang W. Design, synthesis of 4,5-diazafluorene derivatives and their anticancer activity via targeting telomeric DNA G-quadruplex. Eur J Med Chem 2019; 178:484-499. [PMID: 31202994 DOI: 10.1016/j.ejmech.2019.06.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/02/2019] [Accepted: 06/03/2019] [Indexed: 01/19/2023]
Abstract
In our work, 19 novel 4,5-diazafluorene derivatives (11a-d, 12a-d, 13a-d, 14a-c, 15c, 16a-c) bearing a 1,3-disubstituted pyrazol/thioxothiazolidinone or thioxothiazolidinone-oxadiazole moieties were designed, synthesized, preliminarily explored for their antitumor activities and in vitro mechanism. All compounds showed different values of antiproliferative activity against A549, AGS, HepG2 and MCF-7 cell lines through CCK-8. Especially, the compound 14c exhibited the strongest activity and best selectivity against A549 cells with an IC50 1.13 μM and an SI value of 7.01 relative to MRC-5 cells, which was better than cisplatin (SI = 1.80) as a positive control. Experimental results at extracellular level demonstrated that compounds 14a-c could strongly interact with the G-quadruplex(es) formed in a 26 nt telomeric G-rich DNA, in particular, the 14c exhibits quite strong binding affinity with an association equilibrium constant (KA) of 7.04(±0.16) × 107 M-1 and more than 1000-fold specificity to G4-DNA over ds-DNA and Mut-DNA at the compound/G4-DNA ratio of 1:1. Further trap assay ascertained that compounds 14a-c owned strong inhibitory ability of telomerase activity in A549 cells, suggesting that these compounds have great possibility to target telomeric G-quadruplexes and consequently indirectly inhibit the telomerase activity. In addition, it is worthy of note that the remarkable inhibitory effects of 14a-c on the mobility of tested cancer cells were observed by wound healing assays. Furthermore, molecular docking and UV-Vis spectral results unclose the rationale for the interaction of compounds with such G-quadruplex(es). These results indicate that the growth and metastasis inhibition of cancer cells mediated by these 4,5-diazafluorene derivatives possibly result from their interaction with telomeric G-quadruplexes, suggesting that 4,5-diazafluorene derivatives, especially 14c, possess potential as anticancer drugs.
Collapse
Affiliation(s)
- Kang Zhou
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Jiachun Liu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xuqiong Xiong
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Mei Cheng
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiaolin Hu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Suresh Narva
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiaoyin Zhao
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yanling Wu
- Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310051, China.
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Lab of Chemical Biology and Molecular Drug Design, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
30
|
Zhaojie L, Yuchen L, Miao C, Yacun C, Shayi W, Anbang H, Xinhui L, Meng Z, Peipei W, Hongbing M, Feng W, Zhiming C, Xinyuan G. Gelsolin-like actin-capping protein has prognostic value and promotes tumorigenesis and epithelial-mesenchymal transition via the Hippo signaling pathway in human bladder cancer. Ther Adv Med Oncol 2019; 11:1758835919841235. [PMID: 31068979 PMCID: PMC6492362 DOI: 10.1177/1758835919841235] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 02/27/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Transitional cell carcinoma (TCC) of the bladder, the major histologic subtype of bladder cancer, is increasing in incidence and mortality, which requires the identification of effective biomarkers. Actin-regulating proteins have recently been proposed as important antitumor druggable targets. As a gelsolin-family actin-modulating protein, CAPG (gelsolin-like actin-capping protein) generated great interest due to its crucial effects in various biological and physiological processes; however, the role and mechanism of CAPG in TCCs remain unknown. Materials and methods: Bioinformatic analysis and immunohistochemistry of clinical specimens were performed to detect the expression level of CAPG. Both in vitro and in vivo assays were used to determine the oncogenic effect of CAPG in TCCs. Male 4–5-week-old BALB/c nude mice were used for in vivo tumorigenesis assays, while SCID mice were used for in vivo metastatic assays. Affymetrix microarray was used to identify the underlying molecular mechanism. Western blot and immunofluorescence were used to validate the expression and localization of proteins. Results: CAPG was frequently upregulated in TCCs and associated with clinical aggressiveness and worse prognosis. Functional assays demonstrated that CAPG could contribute to the tumorigenesis, metastasis and epithelial-mesenchymal transition (EMT) of TCCs both in vitro and in vivo. A novel mechanism that CAPG promoted TCC development via inactivating the Hippo pathway, leading to a nucleus translocation of Yes-associated protein was suggested. Conclusions: The current study identified CAPG as a novel and critical oncogene in TCCs, supporting the pursuit of CAPG as a potential target for TCC intervention.
Collapse
Affiliation(s)
- Lyu Zhaojie
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Liu Yuchen
- Key Laboratory of Medical Reprogramming Technology, Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Chen Miao
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chen Yacun
- Department of Pathology, The Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wu Shayi
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - He Anbang
- Department of Urology, Peking University First Hospital, The Institute of Urology, Peking University, National Urological Cancer Center, Beijing, China
| | - Liao Xinhui
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, ChinaDepartment of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhang Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wu Peipei
- Department of Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mei Hongbing
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Wang Feng
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Cai Zhiming
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 518035 Shenzhen, China
| | - Guan Xinyuan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong. Room L10-56, 10/F, Laboratory Block 21 Sassoon Road, Hong Kong State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, China
| |
Collapse
|
31
|
Stock K, Borrink R, Mikesch JH, Hansmeier A, Rehkämper J, Trautmann M, Wardelmann E, Hartmann W, Sperveslage J, Steinestel K. Overexpression and Tyr421-phosphorylation of cortactin is induced by three-dimensional spheroid culturing and contributes to migration and invasion of pancreatic ductal adenocarcinoma (PDAC) cells. Cancer Cell Int 2019; 19:77. [PMID: 30976201 PMCID: PMC6441202 DOI: 10.1186/s12935-019-0798-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 03/23/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The nucleation-promoting factor cortactin is expressed and promotes tumor progression and metastasis in various cancers. However, little is known about the biological role of cortactin in the progression of pancreatic ductal adenocarcinoma (PDAC). METHODS Cortactin and phosphorylated cortactin (Y421) were investigated immunohistochemically in 66 PDAC tumor specimens. To examine the functional role of cortactin in PDAC, we modulated cortactin expression by establishing two cortactin knockout cell lines (Panc-1 and BxPC-3) with CRISPR/Cas9 technique. Cortactin knockout was verified by immunoblotting and immunofluorescence microscopy and functional effects were determined by cell migration and invasion assays. A proteomic screening approach was performed to elucidate potential binding partners of cortactin. RESULTS Immunohistochemically, we observed higher cortactin expression and Tyr421-phosphorylation in PDAC metastases compared to primary tumor tissues. In PDAC cell lines Panc-1 and BxPC-3, knockdown of cortactin impaired migration and invasion, while cell proliferation was not affected. Three-dimensional spheroid culturing as a model for collective cell migration enhanced cortactin expression and Tyr421-phosphorylation. The activation of cortactin as well as the migratory capacity of PDAC cells could significantly be reduced by dasatinib, a Src family kinase inhibitor. Finally, we identified gelsolin as a novel protein interaction partner of cortactin in PDAC. CONCLUSION Our data provides evidence that cohesive cell migration induces cortactin expression and phosphorylation as a prerequisite for the gain of an invasive, pro-migratory phenotype in PDAC that can effectively be targeted with dasatinib.
Collapse
Affiliation(s)
- Katharina Stock
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Rebekka Borrink
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | | | - Anna Hansmeier
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Jan Rehkämper
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Marcel Trautmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Wolfgang Hartmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
- Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Jan Sperveslage
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Konrad Steinestel
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
- Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, Ulm, Germany
| |
Collapse
|
32
|
Wang S, Zhou H, Wu D, Ni H, Chen Z, Chen C, Xiang Y, Dai K, Chen X, Li X. MicroRNA let-7a regulates angiogenesis by targeting TGFBR3 mRNA. J Cell Mol Med 2018; 23:556-567. [PMID: 30467960 PMCID: PMC6307798 DOI: 10.1111/jcmm.13960] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/16/2018] [Accepted: 09/21/2018] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis has a great impact on human health, owing to its participation in development, wound healing and the pathogenesis of several diseases. It has been reported that let-7a is a tumour suppressor, but whether it plays a role in angiogenesis is unclear. Here we showed that let-7a, a microRNA conserved in vertebrates, regulated angiogenesis by concomitantly down-regulating TGFBR3. Overexpression of let-7a or knockdown of TGFBR3 in cell culture inhibited the tube formation and reduced migration rate. Moreover, xenograft experiments showed that overexpression of let-7a or knockdown of TGFBR3 had smaller tumour size. Downstream genes, such as VEGFC and MMP9, were also down-regulated in let-7a overexpression or TGFBR3 knockdown groups. Therefore, our results revealed a novel mechanism that let-7a regulate angiogenesis through post-transcriptional regulation of TGFBR3.
Collapse
Affiliation(s)
- Shao Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huandong Zhou
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dazhou Wu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huajing Ni
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongliang Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengshui Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Youqun Xiang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kezhi Dai
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China.,The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoming Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xi Li
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China.,The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
33
|
Plasma Gelsolin: Indicator of Inflammation and Its Potential as a Diagnostic Tool and Therapeutic Target. Int J Mol Sci 2018; 19:ijms19092516. [PMID: 30149613 PMCID: PMC6164782 DOI: 10.3390/ijms19092516] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/14/2018] [Accepted: 08/18/2018] [Indexed: 12/19/2022] Open
Abstract
Gelsolin, an actin-depolymerizing protein expressed both in extracellular fluids and in the cytoplasm of a majority of human cells, has been recently implicated in a variety of both physiological and pathological processes. Its extracellular isoform, called plasma gelsolin (pGSN), is present in blood, cerebrospinal fluid, milk, urine, and other extracellular fluids. This isoform has been recognized as a potential biomarker of inflammatory-associated medical conditions, allowing for the prediction of illness severity, recovery, efficacy of treatment, and clinical outcome. A compelling number of animal studies also demonstrate a broad spectrum of beneficial effects mediated by gelsolin, suggesting therapeutic utility for extracellular recombinant gelsolin. In the review, we summarize the current data related to the potential of pGSN as an inflammatory predictor and therapeutic target, discuss gelsolin-mediated mechanisms of action, and highlight recent progress in the clinical use of pGSN.
Collapse
|
34
|
Fu Q, Yang F, Zhao J, Yang X, Xiang T, Huai G, Zhang J, Wei L, Deng S, Yang H. Bioinformatical identification of key pathways and genes in human hepatocellular carcinoma after CSN5 depletion. Cell Signal 2018; 49:79-86. [PMID: 29885455 DOI: 10.1016/j.cellsig.2018.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/31/2018] [Accepted: 06/05/2018] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer. It has been previously reported that CSN5 depletion is an effective method in human HCC. In the current study, we aimed to uncover gene signatures and key pathways during HCC. Gene expression profiles of GSE26485 were downloaded from GEO database. Totally, 101 differentially expressed genes (DEGs) were up-regulated and 146 ones were down-regulated. Biological processes (BP) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) analysis showed that the DEGs were mainly enriched in regulation of cell growth, oxidation-reduction process, mitotic cytokinesis, negative regulation of macroautophagy, endosome organization, lysosome, biosynthesis of antibiotics, small cell lung cancer and glutathione metabolism and so on (P < 0.05). Protein-protein interaction (PPI) network, Kaplan-Meier, log-rank method, western blot, immunohistochemistry and encyclopedia of DNA elements (ENCODE) analysis showed that CSN5 depletion took effects through down-regulation of SMAD5-related pathways which include EXO1, CENPA and NCAPG, resulting in the inactivation of H3K4me3 and H3K36me3. Those genes represent the promising targets for therapeutic intervention in HCC patients.
Collapse
Affiliation(s)
- Qiang Fu
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China
| | - Fan Yang
- Women and Children Health Care Center of Luoyang, Luoyang 471000, Henan province, China
| | - Ji Zhao
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China
| | - Xingxing Yang
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China
| | - Tengxiao Xiang
- People's Hospital of Changshou Chongqing, Chongqing 401220, China
| | - Guoli Huai
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China
| | - Jiashu Zhang
- Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China; North Sichuan Medical College, Nanchong 637100, Sichuan province, China
| | - Liang Wei
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China
| | - Shaoping Deng
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China; North Sichuan Medical College, Nanchong 637100, Sichuan province, China; Human Islet Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston 02114, MA, USA.
| | - Hongji Yang
- Organ Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan province, China; Organ Transplantation translational medicine Key laboratory of Sichuan province,Chengdu, Sichuan 610072, China.
| |
Collapse
|
35
|
Huang S, Chi Y, Qin Y, Wang Z, Xiu B, Su Y, Guo R, Guo L, Sun H, Zeng C, Zhou S, Hu X, Liu S, Shao Z, Wu Z, Jin W, Wu J. CAPG enhances breast cancer metastasis by competing with PRMT5 to modulate STC-1 transcription. Theranostics 2018; 8:2549-2564. [PMID: 29721098 PMCID: PMC5928908 DOI: 10.7150/thno.22523] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/22/2018] [Indexed: 11/20/2022] Open
Abstract
Macrophage-capping protein (CAPG) has been shown to promote cancer cell metastasis, although the mechanism remains poorly understood. Methods: Breast cancer (BC) tissue microarray was used to test the role of CAPG in the prognosis of BC patients. Xenograft mice model was used to validate the metastasis promotion role of CAPG in vivo. Gene expression array, chromatin immunoprecipitation and luciferase report assay were performed to search for the target genes of CAPG. Protein immunoprecipitation, MS/MS analysis, tissue microarray and histone methyltransferase assay were used to explore the mechanism of CAPG regulating stanniocalcin 1 (STC-1) transcription. Results: We demonstrate a novel mechanism by which CAPG enhances BC metastasis via promoting the transcription of the pro-metastatic gene STC-1, contributing to increased metastasis in BC. Mechanistically, CAPG competes with the transcriptional repressor arginine methyltransferase 5 (PRMT5) for binding to the STC-1 promoter, leading to reduced histone H4R3 methylation and enhanced STC-1 transcription. Our study also indicates that both CAPG and PRMT5 are independent prognostic factors for BC patient survival. High CAPG level is associated with poor survival, while high PRMT5 expression favors a better prognosis in BC patients. Conclusion: Our findings identify a novel role of CAPG in the promotion of BC metastasis by epigenetically enhancing STC-1 transcription.
Collapse
|
36
|
Xing W, Gao W, Su H, Wang S, Zhang J, Mao G, Yan J. Salidroside influences the cellular cross-talk of human fetal lung diploid fibroblasts: A proteomic approach. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 58:114-121. [PMID: 29329019 DOI: 10.1016/j.etap.2018.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/04/2018] [Indexed: 06/07/2023]
Abstract
Senescence is a complex multiple factor proces, which is still poorly understood. The purpose of this study was to find the proteome of cultured human fetal lung diploid fibroblasts (2BS) of different population doubling (PD), as well as the altered proteome induced by salidroside (SAL) in 2BS cells. Proteins were identified by two-dimensional electrophoresis (2-DE) combining matrix-assisted laser desorption/ionization-time and flight mass spectrometry (MAL DI-TOF/MS). As a result, we found 16 proteins with two-fold variations in senescent cells or after SAL treatment, some being reduced such as reticulocalbin-1, heat shock protein beta-6, elongation factor 1-delta, F-actin-capping protein subunit alpha-1, and chloride intracellular channel 1. In contrast, 40S ribosomal protein SA, proteasome subunit alpha type-5, and zinc finger BED domain-containing protein 5 increased with cell age. Furthermore, heat shock protein beta-6, Zinc finger BED domain-containing protein 5 was increased in PD30 cells after 10 μM SAL treatment, whereas, elongation factor 1-delta, 6-phosphogluconolactonase, Nucleoside diphosphate kinase A, F-actin-capping protein subunit alpha-1, Probable ATP-dependent RNA helicase DDX41, Chloride intracellular channel 1, and Peroxiredoxin-6 were increased in PD50 cells after 10 μM SAL treatment. Some of these proteins were involved in the protein synthetic and degradative pathways, which emphasizes the metabolic disorder or functional impairment of cell senescence. Moreover, these proteins could be candidate biomarkers for evaluating the SAL anti-senescence effect.
Collapse
Affiliation(s)
- Wenmin Xing
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China
| | - Wenyan Gao
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, 310013, China
| | - Huili Su
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China
| | - Sanying Wang
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China
| | - Jing Zhang
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China.
| | - Jing Yan
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310013, China.
| |
Collapse
|
37
|
Yuan X, Wang W, Li J, Zheng P, Dong P, Chen L, Zhou Y, Xie G, Xu D, Liu Y, Shen L. Gelsolin suppresses gastric cancer metastasis through inhibition of PKR-p38 signaling. Oncotarget 2018; 7:53459-53470. [PMID: 27419625 PMCID: PMC5288199 DOI: 10.18632/oncotarget.10557] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/05/2016] [Indexed: 01/22/2023] Open
Abstract
The biological function of gelsolin in gastric cancer and its mechanism remained undefined. Here, we demonstrated that gelsolin was down-regulated in human gastric cancer tissues, and lower tumorous gelsolin significantly correlated with gastric cancer metastasis. Functionally, gelsolin suppressed the migration of gastric cancer cells in vitro and inhibited lung metastasis in vivo. In mechanism, gelsolin decreased epithelial–mesenchymal transition (EMT) inducing cytoskeleton remolding through inhibition of p38 signaling to suppress the migration of gastric cancer cell. Moreover, gelsolin bound to and decreased the phosphorylation of PKR, and then inhibited p38 signaling pathway. Finally, similar to the gastric cancer cell lines, PKR-p38 signaling pathway proteins tend to be activated and correlated with low expression of gelsolin in clinical gastric cancer tissues. Altogether, these results highlight the importance of gelsolin in suppression of gastric cancer metastasis through inhibition of PKR-p38 signaling pathway.
Collapse
Affiliation(s)
- Xiangliang Yuan
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Weiwei Wang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Junhua Li
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Peiming Zheng
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ping Dong
- Department of Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lei Chen
- Department of Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yunlan Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Guohua Xie
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Dakang Xu
- MIMR-PHI Institute of Medical Research, Monash University, Clayton, Victoria 3168, Australia.,Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 311121, China
| | - Yingbin Liu
- Department of Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
38
|
Gelsolin-Cu/ZnSOD interaction alters intracellular reactive oxygen species levels to promote cancer cell invasion. Oncotarget 2018; 7:52832-52848. [PMID: 27391159 PMCID: PMC5288152 DOI: 10.18632/oncotarget.10451] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 06/28/2016] [Indexed: 12/13/2022] Open
Abstract
The actin-binding protein, gelsolin, is a well known regulator of cancer cell invasion. However, the mechanisms by which gelsolin promotes invasion are not well established. As reactive oxygen species (ROS) have been shown to promote cancer cell invasion, we investigated on the hypothesis that gelsolin-induced changes in ROS levels may mediate the invasive capacity of colon cancer cells. Herein, we show that increased gelsolin enhances the invasive capacity of colon cancer cells, and this is mediated via gelsolin's effects in elevating intracellular superoxide (O2.-) levels. We also provide evidence for a novel physical interaction between gelsolin and Cu/ZnSOD, that inhibits the enzymatic activity of Cu/ZnSOD, thereby resulting in a sustained elevation of intracellular O2.-. Using microarray data of human colorectal cancer tissues from Gene Omnibus, we found that gelsolin gene expression positively correlates with urokinase plasminogen activator (uPA), an important matrix-degrading protease invovled in cancer invasion. Consistent with the in vivo evidence, we show that increased levels of O2.- induced by gelsolin overexpression triggers the secretion of uPA. We further observed reduction in invasion and intracellular O2.- levels in colon cancer cells, as a consequence of gelsolin knockdown using two different siRNAs. In these cells, concurrent repression of Cu/ZnSOD restored intracellular O2.- levels and rescued invasive capacity. Our study therefore identified gelsolin as a novel regulator of intracellular O2.- in cancer cells via interacting with Cu/ZnSOD and inhibiting its enzymatic activity. Taken together, these findings provide insight into a novel function of gelsolin in promoting tumor invasion by directly impacting the cellular redox milieu.
Collapse
|
39
|
Yun DP, Wang YQ, Meng DL, Ji YY, Chen JX, Chen HY, Lu DR. Actin-capping protein CapG is associated with prognosis, proliferation and metastasis in human glioma. Oncol Rep 2018; 39:1011-1022. [PMID: 29399702 PMCID: PMC5802022 DOI: 10.3892/or.2018.6225] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/02/2018] [Indexed: 12/20/2022] Open
Abstract
Glioma is the most aggressive and malignant primary brain tumor in adults. In the present study, we identified a vital oncoprotein, capping actin protein, gelsolin-like (CapG), and investigated its roles in the prognosis, proliferation and metastasis in glioma. The mRNA and protein levels of CapG were significantly increased in human glioma, and higher CapG expression was an independent prognostic factor for predicting unfavorable prognosis. The expression level of CapG was found to be associated with several common molecular features of glioblastoma (GBM; WHO grade IV glioma) in The Cancer Genome Atlas (TCGA) cohort. When analyzing the prognosis of GBM patients according to these molecular features, we observed that the prognostic value of CapG was affected by amplification of CDK6 or EGFR. However, overexpression of CapG markedly promoted cell growth in vitro, while depletion of CapG significantly inhibited cell proliferation by blocking the cell cycle in G1/S transition. Moreover, CapG manipulation in glioma cell lines U87 and U251 showed CapG-dependent cellular migration and invasiveness. These data suggest that CapG may serve as a prognostic biomarker with potentially important therapeutic implications for glioma.
Collapse
Affiliation(s)
- Da-Peng Yun
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Yu-Qi Wang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - De-Long Meng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yuan-Yuan Ji
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Ju-Xiang Chen
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Hong-Yan Chen
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Da-Ru Lu
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| |
Collapse
|
40
|
Wu W, Chen J, Ding Q, Yang S, Wang J, Yu H, Lin J. Function of the macrophage-capping protein in colorectal carcinoma. Oncol Lett 2017; 14:5549-5555. [PMID: 29113183 PMCID: PMC5656019 DOI: 10.3892/ol.2017.6888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 07/14/2017] [Indexed: 12/13/2022] Open
Abstract
To investigate the role of macrophage-capping protein (CapG) in the development and progression of colorectal carcinoma (CRC), immunohistochemistry (IHC), Kaplan-Meier survival analysis, wound healing and Transwell migration assays were performed. The IHC results demonstrated that CapG was relatively highly expressed in CRC tissue compared with non-tumor tissue (P<0.001), and that the expression of CapG was significantly associated with the tumor site, differentiation, lymph node metastasis and clinical stage (P=0.021, P=0.036, P=0.012 and P=0.009, respectively). Wound healing and Transwell migration assays demonstrated that the reduction of CapG expression in a CRC cell line by RNA interference was associated with significantly impaired motility (P<0.001). Kaplan-Meier survival analysis revealed that the expression of CapG in tumor samples was not significantly associated with disease-free survival time. In conclusion, CapG was overexpressed in CRC and was associated with tumor progression; therefore, it may be a useful prognostic biomarker and therapeutic target in CRC.
Collapse
Affiliation(s)
- Wei Wu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jingdi Chen
- 73rd Contingent, 95969 Troops, The Airborne Force of Chinese PLA, Wuhan, Hubei 430300, P.R. China
| | - Qianshan Ding
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Sheng Yang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jianping Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Honggang Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jun Lin
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
41
|
Yang C, Li H, Li H, Wang YF, Meng L, Yang YX. Mechanism study of 5-alkylresorcinols-induced colon cancer cell apoptosis in vitro. Shijie Huaren Xiaohua Zazhi 2017; 25:2621-2630. [DOI: 10.11569/wcjd.v25.i29.2621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the mechanism of 5-alkylresorcinols (5ARs)-induced apoptosis by detecting the effect on BCL2, Bax, PARP1 and Caspase3 expression patterns in colon cancer cell lines.
METHODS After HT29 and HCT 116 cells were treated with different concentrations of 5ARs, cell morphological changes were observed by phase-contrast microscopy, cell proliferation and apoptosis phenotypes were evaluated by Cell Counting Kit-8 (CCK8) and Annexin V-FITC/PI flow cytometric assays, respectively, and the protein levels of BCL2, Bax, PARP1 and Caspase3 were detected by Western blotting assays.
RESULTS CCK8 assays indicated that the proliferation of HT29 and HCT 116 cells treated with different concentrations of 5ARs was significantly decreased (P < 0.05), and treatment with 5ARs increased the apoptosis of HT29 and HCT 116 cells. 5ARs suppressed BCL2 expression and elevated the levels of Bax, PARP1, Caspase3 and the ratio of BAX to BCL2 in colon cancer cells.
CONCLUSION 5ARs can induce the apoptosis of HT29 and HCT 116 cells in vitro, possibly by enhancing BAX, PARP1 and Caspase3 expression and elevating the ratio of BAX to BCL2 in colon cancer cells.
Collapse
Affiliation(s)
- Chun Yang
- Department of Anal-Colorectal Surgery, the General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Heng Li
- Department of Anal-Colorectal Surgery, the General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Hai Li
- Department of Anal-Colorectal Surgery, the General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yu-Feng Wang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Meng
- Department of Anal-Colorectal Surgery, the General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yin-Xue Yang
- Department of Anal-Colorectal Surgery, the General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
42
|
Dehghan-Nayeri N, Eshghi P, Pour KG, Rezaei-Tavirani M, Omrani MD, Gharehbaghian A. Differential expression pattern of protein markers for predicting chemosensitivity of dexamethasone-based chemotherapy of B cell acute lymphoblastic leukemia. Cancer Chemother Pharmacol 2017; 80:177-185. [PMID: 28585036 DOI: 10.1007/s00280-017-3347-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/29/2017] [Indexed: 02/06/2023]
Abstract
Dexamethasone is considered as a direct chemotherapeutic agent in the treatment of pediatric acute lymphoblastic leukemia (ALL). Beside the advantages of the drug, some problems arising from the dose-related side effects are challenging issues during the treatment. Accordingly, the classification of patients to dexamethasone sensitive and resistance groups can help to select optimizing the therapeutic dose with the lowest adverse effects particularly in sensitive cases. For this purpose, we investigated inhibited proliferation and induced cytotoxicity in NALM-6 cells, as sensitive cells, after dexamethasone treatment. In addition, comparative protein expression analysis using the 2DE-MALDI-TOF MS technique was performed to identify the specific altered proteins. In addition, we evaluated mRNA expression levels of the identified proteins in bone-marrow samples from pediatric ALL patients using the real-time q-PCR method. Eventually, proteomic analysis revealed a combination of biomarkers, including capping proteins (CAPZA1 and CAPZB), chloride channel (CLIC1), purine nucleoside phosphorylase (PNP), and proteasome activator (PSME1), in response to the dexamethasone treatment. In addition, our results indicated low expression of identified proteins at both the mRNA and protein expression levels after drug treatment. Moreover, quantitative real-time PCR data analysis indicated that independent of the molecular subtypes of the leukemia, CAPZA1, CAPZB, CLIC1, and PNP expression levels were lower in ALL samples than normal samples, although PSME1 expression level was higher in ALL samples than normal samples. Furthermore, the expression level of all proteins (except PSME1) was different between high-risk and standard-risk patients that suggesting the prognostic value of them. In conclusion, our study suggests a panel of biomarkers comprising CAPZA1, CAPZB, CLIC1, PNP, and PSME1 as early diagnosis and treatment evaluation markers that may differentiate cancer cells which are presumably to benefit from dexamethasone-based chemotherapy and may facilitate the prediction of clinical outcome.
Collapse
MESH Headings
- Antineoplastic Agents, Hormonal/administration & dosage
- Antineoplastic Agents, Hormonal/pharmacology
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Child
- Child, Preschool
- Dexamethasone/administration & dosage
- Dexamethasone/pharmacology
- Drug Resistance, Neoplasm
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Infant
- Male
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Prognosis
- Proteomics
- RNA, Messenger/metabolism
- Real-Time Polymerase Chain Reaction
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
Collapse
Affiliation(s)
- Nasrin Dehghan-Nayeri
- Proteomics Research Center, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peyman Eshghi
- Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kourosh Goudarzi Pour
- Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Gharehbaghian
- Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
43
|
Papala A, Sylvester M, Dyballa-Rukes N, Metzger S, D'Haese J. Isolation and characterization of human CapG expressed and post-translationally modified in Pichia pastoris. Protein Expr Purif 2017; 134:25-37. [DOI: 10.1016/j.pep.2017.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/08/2017] [Accepted: 03/18/2017] [Indexed: 12/13/2022]
|
44
|
Osbun JW, Tatman PD, Kaur S, Parada C, Busald T, Gonzalez-Cuyar L, Shi M, Born DE, Zhang J, Ferreira M. Comparative Proteomic Profiling Using Two-Dimensional Gel Electrophoresis and Identification via LC-MS/MS Reveals Novel Protein Biomarkers to Identify Aggressive Subtypes of WHO Grade I Meningioma. J Neurol Surg B Skull Base 2017; 78:371-379. [PMID: 28875114 DOI: 10.1055/s-0037-1601889] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 03/03/2017] [Indexed: 12/26/2022] Open
Abstract
Background Meningomas represent the most common primary intracranial tumor. The majority are benign World Health Organization (WHO) Grade I lesions, but a subset of these behave in an aggressive manner. Protein biomarkers are needed to distinguish aggressive from benign Grade I lesions. Materials and Methods Pooled protein lysates were derived from five clinically aggressive Grade I and five typically benign WHO Grade I tumors snap frozen at the time of surgery. Proteins were separated in each group using two-dimensional gel electrophoresis (2DGE) and protein spots of interest were identified using liquid chromatography-mass spectrometry (LC-MS). Potential biomarker candidates were validated using western blot assays in individual tumor samples and by tissue microarray (TMA). Results Seven candidate biomarkers were obtained from the 2DGE and validated via western blot and TMA. Biomarker validation data allowed for the creation of predictive models using binary logistical regression that correctly identified 85.9% of aggressive tumors within the larger cohort of Grade I meningioma. Conclusion Simple protein separation by 2DGE and identification of candidate biomarkers by LC-MS allowed for the identification of seven candidate biomarkers that when used in predictive models accurately distinguish aggressive from benign behavior in WHO Grade I meningioma.
Collapse
Affiliation(s)
- Joshua W Osbun
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States
| | - Philip D Tatman
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States
| | - Sumanpreet Kaur
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States
| | - Carolina Parada
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States
| | - Tina Busald
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States
| | - Luis Gonzalez-Cuyar
- Department of Neuropathology, University of Washington, Seattle, Washington, United States
| | - Min Shi
- Department of Neuropathology, University of Washington, Seattle, Washington, United States
| | - Donald E Born
- Department of Neuropathology, Stanford University, Stanford, California, United States
| | - Jing Zhang
- Department of Neuropathology, University of Washington, Seattle, Washington, United States
| | - Manuel Ferreira
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States
| |
Collapse
|
45
|
Fluctuation of ROS regulates proliferation and mediates inhibition of migration by reducing the interaction between DLC1 and CAV-1 in breast cancer cells. In Vitro Cell Dev Biol Anim 2017; 53:354-362. [PMID: 28130753 DOI: 10.1007/s11626-016-0123-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/14/2016] [Indexed: 12/30/2022]
Abstract
The aim of our present study was to elucidate the effects of up-regulation and down-regulation of intracellular reactive oxygen species (ROS) level on proliferation, migration, and related molecular mechanism. Breast cancer cells were treated by catalase or H2O2. MTT, colony formation assay, and Hoechst/PI staining were used to evaluate proliferation and apoptosis. The level of intracellular ROS was measured by dichlorodihydrofluorescein diacetate probes. The ability of migration was detected by wound healing. Western blotting and coimmunoprecipitation (co-IP) were used to determine the expression of DLC1 and CAV-1 and their interaction. Our data indicated that up-regulation of intracellular ROS induced by H2O2 significantly inhibited proliferation and induced apoptosis accompanying G1 cell cycle arrest and elevated expression of p53. For cell migration, either up-regulation or down-regulation of ROS induced migration inhibition with reduction of interaction between DLC1 and CAV-1. Our results suggested that up-regulation of intracellular ROS inhibited proliferation by promoting expression of p53 and induced G1 cycle arrest and apoptosis. Fluctuation of ROS inhibited migration through reducing the interaction between DLC1 and CAV-1.
Collapse
|
46
|
Chen H, Wang T, Li K, He Q, Hou X, Yang R, Wang B. Effects of surface modification of quantum dots on viability and migration of triple-negative breast cancer cells. J Colloid Interface Sci 2017; 485:51-58. [DOI: 10.1016/j.jcis.2016.09.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/10/2016] [Accepted: 09/12/2016] [Indexed: 11/26/2022]
|
47
|
Xu X, Hu H, Wang X, Ye W, Su H, Hu Y, Dong L, Zhang R, Ying K. Involvement of CapG in proliferation and apoptosis of pulmonary arterial smooth muscle cells and in hypoxia-induced pulmonary hypertension rat model. Exp Lung Res 2016; 42:142-53. [PMID: 27093378 DOI: 10.3109/01902148.2016.1160304] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Actin-binding protein capping protein gelsolin-like (CapG) was preferentially expressed in human pulmonary arterial smooth muscle cells (PASMCs) under hypoxia, and reduced CapG expression was accompanied by impaired migration ability in vitro. We intended to investigate the effects of CapG on rat PASMCs and hypoxia-induced pulmonary hypertension (HPH) rat model. MATERIALS AND METHODS We investigated the effect of RNA interference-medicated down-regulation of CapG expression in rat PASMCs as well as in HPH rat model. The proliferation, apoptosis and cell cycle of PASMCs were evaluated. The HPH rat model was established by intratracheal instillation of lentiviral vector and subsequent hypoxia exposure for four weeks. Right ventricular systolic pressure, right ventricular hypertrophy and the percentage of medial wall thickness were measured to evaluate the development of HPH. RESULTS Knock-down CapG in PASMCs resulted in decreased proliferation, increased apoptosis and induced cell cycle inhibition. Down-regulation of CapG expression locally could attenuate pulmonary hypertension, pulmonary vascular remodeling and right ventricular hypertrophy in HPH rat model. CONCLUSIONS Our study indicated that CapG participated in the pathogenesis of pulmonary vascular remodeling in HPH rats, which was probably mediated by promoting the proliferation and inhibiting the apoptosis of PASMCs. We proposed CapG modulating protective effects of pulmonary hypertension.
Collapse
Affiliation(s)
- Xiaoling Xu
- a Department of Respiratory Medicine , Sir Run Run Shaw Hospital , Zhejiang University School of Medicine , Hangzhou, Zhejiang , China
| | - Huihui Hu
- a Department of Respiratory Medicine , Sir Run Run Shaw Hospital , Zhejiang University School of Medicine , Hangzhou, Zhejiang , China
| | - Xiaohua Wang
- a Department of Respiratory Medicine , Sir Run Run Shaw Hospital , Zhejiang University School of Medicine , Hangzhou, Zhejiang , China
| | - Wu Ye
- a Department of Respiratory Medicine , Sir Run Run Shaw Hospital , Zhejiang University School of Medicine , Hangzhou, Zhejiang , China
| | - Hua Su
- a Department of Respiratory Medicine , Sir Run Run Shaw Hospital , Zhejiang University School of Medicine , Hangzhou, Zhejiang , China
| | - Yanjie Hu
- a Department of Respiratory Medicine , Sir Run Run Shaw Hospital , Zhejiang University School of Medicine , Hangzhou, Zhejiang , China
| | - Liangliang Dong
- a Department of Respiratory Medicine , Sir Run Run Shaw Hospital , Zhejiang University School of Medicine , Hangzhou, Zhejiang , China
| | - Ruifeng Zhang
- a Department of Respiratory Medicine , Sir Run Run Shaw Hospital , Zhejiang University School of Medicine , Hangzhou, Zhejiang , China
| | - Kejing Ying
- a Department of Respiratory Medicine , Sir Run Run Shaw Hospital , Zhejiang University School of Medicine , Hangzhou, Zhejiang , China
| |
Collapse
|
48
|
Coleman O, Henry M, McVey G, Clynes M, Moriarty M, Meleady P. Proteomic strategies in the search for novel pancreatic cancer biomarkers and drug targets: recent advances and clinical impact. Expert Rev Proteomics 2016; 13:383-94. [PMID: 26985644 DOI: 10.1586/14789450.2016.1167601] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers; despite a low incidence rate it is the fourth leading cause of cancer-related death in the world. Improvement of the diagnosis, prognosis and treatment remains the main focus of pancreatic cancer research. Rapid developments in proteomic technologies has improved our understanding of the pancreatic cancer proteome. Here, the authors summarise the recent proteomic strategies undertaken in the search for: novel biomarkers for early diagnosis, pancreatic cancer-specific proteins which may be used for novel targeted therapies and proteins which may be useful for monitoring disease progression post-therapy. Recent advances and findings discussed here provide great promise of having a significant clinical impact and improving the outcome of patients with this malignancy.
Collapse
Affiliation(s)
- Orla Coleman
- a Department of Proteomics, National Institute for Cellular Biotechnology , Dublin City University , Glasnevin , Dublin 9 , Ireland
| | - Michael Henry
- a Department of Proteomics, National Institute for Cellular Biotechnology , Dublin City University , Glasnevin , Dublin 9 , Ireland
| | - Gerard McVey
- b St. Lukes Hospital , Rathgar , Dublin 6 , Ireland
| | - Martin Clynes
- a Department of Proteomics, National Institute for Cellular Biotechnology , Dublin City University , Glasnevin , Dublin 9 , Ireland
| | - Michael Moriarty
- a Department of Proteomics, National Institute for Cellular Biotechnology , Dublin City University , Glasnevin , Dublin 9 , Ireland.,b St. Lukes Hospital , Rathgar , Dublin 6 , Ireland
| | - Paula Meleady
- a Department of Proteomics, National Institute for Cellular Biotechnology , Dublin City University , Glasnevin , Dublin 9 , Ireland
| |
Collapse
|
49
|
Li T, Hong X, Zhao J, Teng Y, Zheng J, Chen H, Chen H, Li H. Gelsolin-like actin-capping protein is associated with patient prognosis, cellular apoptosis and proliferation in prostate cancer. Biomark Med 2016; 10:1251-1260. [PMID: 27924630 DOI: 10.2217/bmm-2016-0186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
AIM To investigate the role of gelsolin-like actin-capping protein (CapG) in prostate cancer (PCa). MATERIALS & METHODS CapG expression and its correlation with clinicopathological characters and patient prognosis were analyzed in 76 cases of PCa by immunohistochemistry and qRT-PCR. Then, the influence of CapG downregulation on cell apoptosis and proliferation were assessed. RESULTS CapG expression in PCa was significantly higher compared with those in matched adjacent noncancerous prostate tissues, and significantly correlated with clinicopathological characters. Survival analysis indicated that CapG could be an independent prognostic factor in PCa. Moreover, CapG depletion significantly affected cellular proliferation and apoptosis by regulating Caspase 6/Caspase 9/Bcl-2/p-Akt/Akt signaling pathway. CONCLUSION CapG, as a potential biomarker in PCa, is associated with patient prognosis, cellular apoptosis and proliferation.
Collapse
Affiliation(s)
- Tieqiu Li
- Department of Urology, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Xiuqin Hong
- Institute of Gerontology, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Jie Zhao
- Department of Pathology, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Yili Teng
- Department of Urology, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Jue Zheng
- Department of Urology, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Hao Chen
- Department of Urology, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Henggui Chen
- Department of Urology, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Huahua Li
- Department of Geriatric, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| |
Collapse
|
50
|
Kukoamine A inhibits human glioblastoma cell growth and migration through apoptosis induction and epithelial-mesenchymal transition attenuation. Sci Rep 2016; 6:36543. [PMID: 27824118 PMCID: PMC5099904 DOI: 10.1038/srep36543] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 10/18/2016] [Indexed: 02/05/2023] Open
Abstract
Cortex lycii radicis is the dried root bark of Lycium chinense, a traditional Chinese herb used in multiple ailments. The crude extract of Cortex lycii radicis has growth inhibition effect on GBM cells. Kukoamine A (KuA) is a spermine alkaloid derived from it. KuA possesses antioxidant, anti-inflammatory activities, but its anticancer activity is unknown. In this study, the growth and migration inhibition effect of KuA on human GBM cells and the possible mechanism of its activity were investigated. After KuA treatment, proliferation and colony formation of GBM cells were decreased significantly; apoptotic cells were increased; the cell cycle was arrested G0/G1 phase; the migration and invasion were decreased, the growth of tumors initiated from GBM cells was inhibited significantly; the expressions of 5-Lipoxygenase (5-LOX) were decreased, apoptotic proteins, Bax and caspase-3 were increased, and antiapoptotic protein Bcl-2 was decreased significantly; The expressions of CCAAT/enhancer binding protein β (C/EBPβ), N-cadherin, vimentin, twist and snail+slug were decreased significantly, while the expression of E-cadherin was increased significantly in KuA treated GBM cells and tumor tissues. KuA inhibited human glioblastoma cell growth and migration in vitro and in vivo through apoptosis induction and epithelial-mesenchymal transition attenuation by downregulating expressions of 5-LOX and C/EBPβ.
Collapse
|