1
|
Wang Z, Hou R, Wang S, Chen M, Zheng D, Zhang Z, Bai L, Chang C, Zhou S. FGFBP1 promotes triple-negative breast cancer progression through the KLK10-AKT axis. Biochem Biophys Res Commun 2025; 763:151763. [PMID: 40233428 DOI: 10.1016/j.bbrc.2025.151763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Accepted: 04/04/2025] [Indexed: 04/17/2025]
Abstract
Triple-negative breast cancer (TNBC) is highly malignant, with rapid tumor growth and metastasis. Due to ER-, PR- and HER2-of TNBC, FGFR pathway play a pivotal role in the progression of TNBC. Its ligand FGFs is mostly released from the extracellular matrix by fibroblast growth factor binding protein 1 (FGFBP1). However, little is known about the role of FGFBP1 in TNBC. In this study, we found that overexpression of FGFBP1 significantly promoted the proliferation, migration and invasion of TNBC cells in vitro and in vivo and vice versa. Mechanistically, overexpression of FGFBP1 upregulated the expression of KLK10, thereby activating AKT, which led to proliferation, migration and invasion of TNBC cells. After knocking down FGFBP1, the expression of KLK10 was reduced and the AKT pathway was inhibited. In addition, knocking down KLK10 or inhibiting AKT pathway impaired the promotion effect of overexpression of FGFBP1 on the proliferation and invasion of TNBC cells. These results suggest that FGFBP1 may promote the proliferation, migration and invasion of TNBC cells through the KLK10-AKT axis. Targeting FGFBP1 may serve as a new therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Ziqi Wang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Xuhui District, Shanghai, 200032, China
| | - Ruoqing Hou
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Xuhui District, Shanghai, 200032, China
| | - Shiyu Wang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Xuhui District, Shanghai, 200032, China
| | - Min Chen
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Pudong New Area, Shanghai, 200127, China
| | - Dongdong Zheng
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Xuhui District, Shanghai, 200032, China
| | - Zhiming Zhang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Xuhui District, Shanghai, 200032, China
| | - Lu Bai
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Xuhui District, Shanghai, 200032, China
| | - Cai Chang
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Xuhui District, Shanghai, 200032, China.
| | - Shichong Zhou
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Xuhui District, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
2
|
Carlton M, Zang T, Parker TJ, Punyadeera C, Voisey J, Cuttle L. Salivary Proteome Is Altered in Children With Small Area Thermal Burns. Proteomics Clin Appl 2025; 19:e202300107. [PMID: 39895030 PMCID: PMC11895759 DOI: 10.1002/prca.202300107] [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: 08/30/2023] [Revised: 08/03/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Saliva is a child appropriate biofluid, but it has not previously been used to evaluate the systemic response to burn injury in children. The aim of this study was to investigate the salivary proteome of children with small area thermal skin burns relative to different burn characteristics (mechanism, time to re-epithelialization and risk of emotional distress). SWATH Mass Spectrometry was used to quantify the abundance of 742 proteins in the saliva of children with burns (n = 22) and healthy controls (n = 37). Eight proteins were differentially abundant in the saliva of children with burns compared to healthy children, and these were associated with immune processes, epidermal cell differentiation and transferrin receptor binding. Eleven proteins were differentially abundant in patients with burns of different mechanisms. Scald burns had an over-representation of immune/inflammatory response processes, and contact burns had an over-representation of cornification, intermediate filament assembly and cell death cellular processes. Four proteins were elevated in patients who were at high risk for emotional distress and 15 proteins were correlated with time to wound re-epithelialization. This pilot study proves that saliva can be used for paediatric biomarker discovery and can be used as a diagnostic and prognostic sample to investigate systemic changes in a paediatric burn cohort.
Collapse
Affiliation(s)
- Morgan Carlton
- School of Biomedical ScienceFaculty of HealthQueensland University of TechnologyBrisbaneQueenslandAustralia
- Centre for Children's Burn and Trauma ResearchCentre for Children's Health ResearchQueensland University of TechnologySouth BrisbaneQueenslandAustralia
| | - Tuo Zang
- School of Biomedical ScienceFaculty of HealthQueensland University of TechnologyBrisbaneQueenslandAustralia
- Centre for Children's Burn and Trauma ResearchCentre for Children's Health ResearchQueensland University of TechnologySouth BrisbaneQueenslandAustralia
| | - Tony J. Parker
- School of Biomedical ScienceFaculty of HealthQueensland University of TechnologyBrisbaneQueenslandAustralia
| | - Chamindie Punyadeera
- Saliva and Liquid Biopsy Translational Research TeamCentre for Biomedical TechnologiesSchool of Biomedical SciencesQueensland University of TechnologyKelvin GroveQueenslandAustralia
- The School of Environment and ScienceGriffith Institute for Drug Discovery (GRIDD)and Menzies Health Institute Queensland (MIHQ)Griffith UniversityNathanQueenslandAustralia
| | - Joanne Voisey
- Centre for Genomics and Personalised HealthSchool of Biomedical ScienceFaculty of HealthQueensland University of TechnologySouth BrisbaneQueenslandAustralia
| | - Leila Cuttle
- School of Biomedical ScienceFaculty of HealthQueensland University of TechnologyBrisbaneQueenslandAustralia
- Centre for Children's Burn and Trauma ResearchCentre for Children's Health ResearchQueensland University of TechnologySouth BrisbaneQueenslandAustralia
| |
Collapse
|
3
|
Wenta T, Nastaly P, Lipinska B, Manninen A. Remodeling of the extracellular matrix by serine proteases as a prerequisite for cancer initiation and progression. Matrix Biol 2024; 134:197-219. [PMID: 39500383 DOI: 10.1016/j.matbio.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024]
Abstract
The extracellular matrix (ECM) serves as a physical scaffold for tissues that is composed of structural proteins such as laminins, collagens, proteoglycans and fibronectin, forming a three dimensional network, and a wide variety of other matrix proteins with ECM-remodeling and signaling functions. The activity of ECM-associated signaling proteins is tightly regulated. Thus, the ECM serves as a reservoir for water and growth regulatory signals. The ECM architecture is dynamically modulated by multiple serine proteases that process both structural and signaling proteins to regulate physiological processes such as organogenesis and tissue homeostasis but they also contribute to pathological events, especially cancer progression. Here, we review the current literature regarding the role of ECM remodeling by serine proteases (KLKs, uPA, furin, HtrAs, granzymes, matriptase, hepsin) in tumorigenesis.
Collapse
Affiliation(s)
- Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland.
| | - Paulina Nastaly
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
4
|
Daneva GN, Tsiakanikas P, Adamopoulos PG, Scorilas A. Kallikrein-related peptidases: mechanistic understanding for potential therapeutic targeting in cancer. Expert Opin Ther Targets 2024; 28:875-894. [PMID: 39431595 DOI: 10.1080/14728222.2024.2415014] [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: 07/28/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Human kallikrein-related peptidases (KLKs) represent a subgroup of 15 serine endopeptidases involved in various physiological processes and pathologies, including cancer. AREAS COVERED This review aims to provide a comprehensive overview of the KLK family, highlighting their genomic structure, expression profiles and substrate specificity. We explore the role of KLKs in tumorigenesis, emphasizing their potential as biomarkers and therapeutic targets in cancer treatment. The dysregulated activity of KLKs has been linked to various malignancies, making them promising candidates for cancer diagnostics and therapy. EXPERT OPINION : Recent advancements in understanding the mechanistic pathways of KLK-related tumorigenesis offer new prospects for developing targeted cancer treatments. Expert opinion suggests that while significant progress has been made, further research is necessary to fully exploit KLKs' potential in clinical applications.
Collapse
Affiliation(s)
- Glykeria N Daneva
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Tsiakanikas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis G Adamopoulos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
5
|
Awad AM, Dabous E, Alalem M, Alalem N, Nasr ME, Elawdan KA, Nasr GM, Said W, El Khashab K, Basiouny MS, Guirgis AA, Khalil H. MicroRNA-141-regulated KLK10 and TNFSF-15 gene expression in hepatoblastoma cells as a novel mechanism in liver carcinogenesis. Sci Rep 2024; 14:13492. [PMID: 38866875 PMCID: PMC11169620 DOI: 10.1038/s41598-024-63223-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 05/27/2024] [Indexed: 06/14/2024] Open
Abstract
Liver cancer is one of the most pivotal global health problems, leading hepatocellular carcinoma (HCC) with a significant increase in cases worldwide. The role of non-coding-RNA in cancer proliferation and carcinogenesis has attracted much attention in the last decade; however, microRNAs (miRNAs), as non-coding RNA, are considered master mediators in various cancer progressions. Yet the role of miR-141 as a modulator for specific cellular processes in liver cancer cell proliferation is still unclear. This study identified the role of miR-141 and its potential functions in liver carcinogenesis. The level of miR-141 in HepG2 and HuH7 cells was assessed using quantitative real-time PCR (qRT-PCR) and compared with its expression in normal hepatocytes. A new miR-141 construct has been performed in a CMV promoter vector tagged with GFP. Using microarray analysis, we identified the potentially regulated genes by miR-141 in transfected HepG2 cells. The protein profile of the kallikrein-related peptidase 10 (KLK10) and tumor necrosis factor TNFSF-15 was investigated in HepG2 cells transfected with either an inhibitor, antagonist miR-141, or miR-141 overexpression vector using immunoblotting and flow cytometry assay. Finally, ELISA assay has been used to monitor the produced inflammatory cytokines from transfected HepG2 cells. Our findings showed that the expression of miR-141 significantly increased in HepG2 and HuH7 cells compared to the normal hepatocytes. Transfection of HepG2 cells with an inhibitor, antagonist miR-141, showed a significant reduction of HepG2 cell viability, unlike the transfection of miR-141 overexpression vector. The microarray data of HepG2 cells overexpressed miR-141 provided a hundred downregulated genes, including KLK10 and TNFSF-15. Furthermore, the expression profile of KLK10 and TNFSF-15 markedly depleted in HepG2 cells transfected with miR-141 overexpression accompanied by a decreasing level of interleukin 6 (IL-6) and tumor necrosis factor-alpha (TNF-α), indicating the role of miR-141 in HepG2 cell proliferation and programmed cell death. Interestingly, the experimental rats with liver cancer induced by Diethylnitrosamine injection further confirmed the upregulation of miR-141 level, IL-10, and TNF-α and the disturbance in KLK10 and TNFSF-15 gene expression compared with their expression in normal rats. The in-silico online tools, IntaRNA and miRWalk were used to confirm the direct interaction and potential binding sites between miR-141 and identified genes. Thus, the seeding regions of potential targeted sequences was cloned upstream of luciferase reporter gene in pGL3 control vector. Interestingly, the luciferase activities of constructed vectors were significantly decreased in HepG2 cells pre-transfected with miR-141 overexpression vector, while increasing in cells pre-transfected with miR-141 specific inhibitor. In summary, these data suggest the crucial role of miR-141 in liver cancer development via targeting KLK10 and TNFSF-15 and provide miR-141 as an attractive candidate in liver cancer treatment and protection.
Collapse
Affiliation(s)
- Ahmed M Awad
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, 32897, Sadat City, Egypt
- Department of Molecular Diagnosis, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Emad Dabous
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, 32897, Sadat City, Egypt
- Department of Molecular Diagnosis, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Mai Alalem
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, 32897, Sadat City, Egypt
- Department of Molecular Diagnosis, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Nedaa Alalem
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, 32897, Sadat City, Egypt
- Department of Molecular Diagnosis, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Mahmoud E Nasr
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, 32897, Sadat City, Egypt
- Department of Molecular Diagnosis, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Khaled A Elawdan
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, 32897, Sadat City, Egypt
- Department of Molecular Diagnosis, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Ghada M Nasr
- Molecular Diagnostics Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Walid Said
- Microbiology and Chemistry Department, Faculty of Science, Benha University, Benha, Egypt
| | - Kareem El Khashab
- Medical Laboratory Department, High Technology Institute of Applied Health Science, Badr Academy for Science and Technology, Badr City, Egypt
| | - Mohamed S Basiouny
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Adel A Guirgis
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, 32897, Sadat City, Egypt
- Department of Molecular Diagnosis, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Hany Khalil
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, 32897, Sadat City, Egypt.
- Department of Molecular Diagnosis, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt.
| |
Collapse
|
6
|
Cheng W, Song Q, Zhou A, Lin L, Zhao Y, Duan J, Zhou Z, Peng Y, Liu C, Zeng Y, Chen P. LRG1 promotes the apoptosis of pulmonary microvascular endothelial cells through KLK10 in chronic obstructive pulmonary disease. Tob Induc Dis 2024; 22:TID-22-72. [PMID: 38707515 PMCID: PMC11069109 DOI: 10.18332/tid/186404] [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: 03/02/2023] [Revised: 09/16/2023] [Accepted: 03/23/2024] [Indexed: 05/07/2024] Open
Abstract
INTRODUCTION Cigarette smoking is one of the most important causes of COPD and could induce the apoptosis of pulmonary microvascular endothelial cells (PMVECs). The conditional knockout of LRG1 from endothelial cells reduced emphysema in mice. However, the mechanism of the deletion of LRG1 from endothelial cells rescued by cigarette smoke (CS) induced emphysema remains unclear. This research aimed to demonstrate whether LRG1 promotes the apoptosis of PMVECs through KLK10 in COPD. METHODS Nineteen patients were divided into three groups: control non-COPD (n=7), smoker non-COPD (n=7), and COPD (n=5). The emphysema mouse model defined as the CS exposure group was induced by CS exposure plus cigarette smoke extract (CSE) intraperitoneal injection for 28 days. Primary PMVECs were isolated from the mouse by magnetic bead sorting method via CD31-Dynabeads. Apoptosis was detected by western blot and flow cytometry. RESULTS LRG1 was increased in lung tissue of COPD patients and CS exposure mice, and CSE-induced PMVECs apoptosis model. KLK10 was over-expressed in lung tissue of COPD patients and CS exposure mice, and CSE-induced PMVECs apoptosis model. LRG1 promoted apoptosis in PMVECs. LRG1 knockdown reversed CSE-induced apoptosis in PMVECs. The mRNA and protein expression of KLK10 were increased after over-expressed LRG1 in PMVECs isolated from mice. Similarly, both the mRNA and protein levels of KLK10 were decreased after LRG1 knockdown in PMVECs. The result of co-immunoprecipitation revealed a protein-protein interaction between LRG1 and KLK10 in PMVECs. KLK10 promoted apoptosis via the down-regulation of Bcl-2/Bax in PMVECs. KLK10 knockdown could reverse CSE-induced apoptosis in PMVECs. CONCLUSIONS LRG1 promotes apoptosis via up-regulation of KLK10 in PMVECs isolated from mice. KLK10 promotes apoptosis via the down-regulation of Bcl-2/Bax in PMVECs. There was a direct protein-protein interaction between LRG1 and KLK10 in PMVECs. Our novel findings provide insights into the understanding of LRG1/KLK10 function as a potential molecule in COPD.
Collapse
Affiliation(s)
- Wei Cheng
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Qing Song
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Aiyuan Zhou
- Department of Pulmonary and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Lin
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Yiyang Zhao
- Department of Diagnostic Ultrasound, Xiangya Hospital, Central South University, Changsha, China
| | - Jiaxi Duan
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zijing Zhou
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Yating Peng
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Cong Liu
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Yuqin Zeng
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| | - Ping Chen
- Department of Pulmonary and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
| |
Collapse
|
7
|
Wu K, Wu B, Yan K, Ding Q, Miao Z. KLK10 promotes the progression of KRAS mutant colorectal cancer via PAR1-PDK1-AKT signaling pathway. Cell Biol Int 2024; 48:440-449. [PMID: 38115179 DOI: 10.1002/cbin.12113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/21/2023]
Abstract
Kirsten rat sarcoma virus (KRAS) gene mutation is common in colorectal cancer (CRC) and is often predictive of treatment failure and poor prognosis. To understand the mechanism, we compared the transcriptome of CRC patients with wild-type and mutant KRAS and found that KRAS mutation is associated with the overexpression of a secreted serine protease, kallikrein-related peptidase 10 (KLK10). Moreover, using in vitro and in vivo models, we found that KLK10 overexpression favors the rapid growth and liver metastasis of KRAS mutant CRC and can also impair the efficacy of KRAS inhibitors, leading to drug resistance and poor survival. Further functional assays revealed that the oncogenic role of KLK10 is mediated by protease-activated receptor 1 (PAR1). KLK10 cleaves and activates PAR1, which further activates 3-phosphoinositide-dependent kinase 1 (PDK1)-AKT oncogenic pathway. Notably, suppressing PAR1-PDK1-AKT cascade via KLK10 knockdown can effectively inhibit CRC progression and improve the sensitivity to KRAS inhibitor, providing a promising therapeutic strategy. Taken together, our study showed that KLK10 promotes the progression of KRAS mutant CRC via activating PAR1-PDK1-AKT signaling pathway. These findings expanded our knowledge of CRC development, especially in the setting of KRAS mutation, and also provided novel targets for clinical intervention.
Collapse
Affiliation(s)
- Kun Wu
- Department of Abdominal Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Boyu Wu
- Department of General Surgery, Shangrao Municipal Hospital, Shangrao, Jiangxi, China
| | - Kangpeng Yan
- Department of Abdominal Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Qunhua Ding
- Department of Abdominal Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Zhiguo Miao
- Department of Abdominal Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Luo YC, Lv YL, He RX, Shi XX, Jiang T. Kallikrein-related peptidase 10 predicts prognosis and mediates tumor immunomodulation in colorectal cancer. Biochem Biophys Res Commun 2023; 689:149217. [PMID: 37972446 DOI: 10.1016/j.bbrc.2023.149217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
The incidence and mortality rates of colorectal cancer (CRC) have significantly increased in recent years. It has been shown that early diagnosis of CRC improves the five-year survival of patients compared to late diagnosis, as patients with stage I disease have a five-year survival rate as high as 90 %. Through bioinformatics analysis, we identified Kallikrein 10 (KLK10), a member of the Kallikrein family, as a reliable predictor of CRC progression, particularly in patients with early-stage CRC. Furthermore, single-cell analysis revealed that KLK10 was highly expressed in tumor and partial immune cells. Analysis of the biological functions of KLK10 using the Kyoto encyclopedia of genes and genomes and gene ontology indicated that KLK10 plays a role in the proliferation and differentiation of cancer cells, along with the maintenance of tumor function and immune regulation, explicitly by T cells and macrophages. EdU cell proliferation staining, plate clone formation assay, and cell scratch assay demonstrated that KLK10 inhibition by siRNA affected the proliferation and migration of CRC cells. Cell cycle detection by flow cytometry demonstrated that KLK10 inhibition led to cell cycle arrest in the G1 phase. In addition, the proportion of M1 and M2 macrophages in 45 tumor specimens was analyzed by immunohistochemistry, the proportion of CD4+ T cells and CD8+ T cells in plasma was identified by flow cytometry, and their correlation with KLK10 was analyzed. The effects of KLK10 on T cells and macrophages were verified in independent cell experiments. The results revealed that KLK10 also activates CD4+ T cells, mediating M2-type macrophage polarization.
Collapse
Affiliation(s)
- Yi-Chao Luo
- Hunan Hospital of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Yuan-Lin Lv
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ruo-Xu He
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiao-Xia Shi
- Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tao Jiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Titova OE, Brunius C, Warensjö Lemming E, Stattin K, Baron JA, Byberg L, Michaëlsson K, Larsson SC. Comprehensive analyses of circulating cardiometabolic proteins and objective measures of fat mass. Int J Obes (Lond) 2023; 47:1043-1049. [PMID: 37550405 PMCID: PMC10599989 DOI: 10.1038/s41366-023-01351-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/03/2023] [Accepted: 07/14/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND The underlying molecular pathways for the effect of excess fat mass on cardiometabolic diseases is not well understood. Since body mass index is a suboptimal measure of body fat content, we investigated the relationship of fat mass measured by dual-energy X-ray absorptiometry with circulating cardiometabolic proteins. METHODS We used data from a population-based cohort of 4950 Swedish women (55-85 years), divided into discovery and replication samples; 276 proteins were assessed with three Olink Proseek Multiplex panels. We used random forest to identify the most relevant biomarker candidates related to fat mass index (FMI), multivariable linear regression to further investigate the associations between FMI characteristics and circulating proteins adjusted for potential confounders, and principal component analysis (PCA) for the detection of common covariance patterns among the proteins. RESULTS Total FMI was associated with 66 proteins following adjustment for multiple testing in discovery and replication multivariable analyses. Five proteins not previously associated with body size were associated with either lower FMI (calsyntenin-2 (CLSTN2), kallikrein-10 (KLK10)), or higher FMI (scavenger receptor cysteine-rich domain-containing group B protein (SSC4D), trem-like transcript 2 protein (TLT-2), and interleukin-6 receptor subunit alpha (IL-6RA)). PCA provided an efficient summary of the main variation in FMI-related circulating proteins involved in glucose and lipid metabolism, appetite regulation, adipocyte differentiation, immune response and inflammation. Similar patterns were observed for regional fat mass measures. CONCLUSIONS This is the first large study showing associations between fat mass and circulating cardiometabolic proteins. Proteins not previously linked to body size are implicated in modulation of postsynaptic signals, inflammation, and carcinogenesis.
Collapse
Affiliation(s)
- Olga E Titova
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Carl Brunius
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Eva Warensjö Lemming
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Department of Food studies, nutrition and dietetics, Uppsala University, Uppsala, Sweden
| | - Karl Stattin
- Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - John A Baron
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Liisa Byberg
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Karl Michaëlsson
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susanna C Larsson
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
George N, Bhandari P, Shruptha P, Jayaram P, Chaudhari S, Satyamoorthy K. Multidimensional outlook on the pathophysiology of cervical cancer invasion and metastasis. Mol Cell Biochem 2023; 478:2581-2606. [PMID: 36905477 PMCID: PMC10006576 DOI: 10.1007/s11010-023-04686-3] [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: 07/14/2022] [Accepted: 02/20/2023] [Indexed: 03/12/2023]
Abstract
Cervical cancer being one of the primary causes of high mortality rates among women is an area of concern, especially with ineffective treatment strategies. Extensive studies are carried out to understand various aspects of cervical cancer initiation, development and progression; however, invasive cervical squamous cell carcinoma has poor outcomes. Moreover, the advanced stages of cervical cancer may involve lymphatic circulation with a high risk of tumor recurrence at distant metastatic sites. Dysregulation of the cervical microbiome by human papillomavirus (HPV) together with immune response modulation and the occurrence of novel mutations that trigger genomic instability causes malignant transformation at the cervix. In this review, we focus on the major risk factors as well as the functionally altered signaling pathways promoting the transformation of cervical intraepithelial neoplasia into invasive squamous cell carcinoma. We further elucidate genetic and epigenetic variations to highlight the complexity of causal factors of cervical cancer as well as the metastatic potential due to the changes in immune response, epigenetic regulation, DNA repair capacity, and cell cycle progression. Our bioinformatics analysis on metastatic and non-metastatic cervical cancer datasets identified various significantly and differentially expressed genes as well as the downregulation of potential tumor suppressor microRNA miR-28-5p. Thus, a comprehensive understanding of the genomic landscape in invasive and metastatic cervical cancer will help in stratifying the patient groups and designing potential therapeutic strategies.
Collapse
Affiliation(s)
- Neena George
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Planetarium Complex, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Poonam Bhandari
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Planetarium Complex, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Padival Shruptha
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Planetarium Complex, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Pradyumna Jayaram
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Planetarium Complex, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sima Chaudhari
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Planetarium Complex, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Planetarium Complex, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
11
|
Dragan M, Chen Z, Li Y, Le J, Sun P, Haensel D, Sureshchandra S, Pham A, Lu E, Pham KT, Verlande A, Vu R, Gutierrez G, Li W, Jang C, Masri S, Dai X. Ovol1/2 loss-induced epidermal defects elicit skin immune activation and alter global metabolism. EMBO Rep 2023; 24:e56214. [PMID: 37249012 PMCID: PMC10328084 DOI: 10.15252/embr.202256214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
Skin epidermis constitutes the outer permeability barrier that protects the body from dehydration, heat loss, and myriad external assaults. Mechanisms that maintain barrier integrity in constantly challenged adult skin and how epidermal dysregulation shapes the local immune microenvironment and whole-body metabolism remain poorly understood. Here, we demonstrate that inducible and simultaneous ablation of transcription factor-encoding Ovol1 and Ovol2 in adult epidermis results in barrier dysregulation through impacting epithelial-mesenchymal plasticity and inflammatory gene expression. We find that aberrant skin immune activation then ensues, featuring Langerhans cell mobilization and T cell responses, and leading to elevated levels of secreted inflammatory factors in circulation. Finally, we identify failure to gain body weight and accumulate body fat as long-term consequences of epidermal-specific Ovol1/2 loss and show that these global metabolic changes along with the skin barrier/immune defects are partially rescued by immunosuppressant dexamethasone. Collectively, our study reveals key regulators of adult barrier maintenance and suggests a causal connection between epidermal dysregulation and whole-body metabolism that is in part mediated through aberrant immune activation.
Collapse
Affiliation(s)
- Morgan Dragan
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
| | - Zeyu Chen
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- Present address:
Department of Dermatology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
- Present address:
Institute of PsoriasisTongji University School of MedicineShanghaiChina
| | - Yumei Li
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Johnny Le
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Peng Sun
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Daniel Haensel
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- Present address:
Program in Epithelial BiologyStanford University School of MedicineStanfordCAUSA
| | - Suhas Sureshchandra
- Department of Physiology and Biophysics, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Anh Pham
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Eddie Lu
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Katherine Thanh Pham
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Amandine Verlande
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Remy Vu
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
| | - Guadalupe Gutierrez
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Wei Li
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Cholsoon Jang
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Selma Masri
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Xing Dai
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
- Department of Dermatology, School of MedicineUniversity of CaliforniaIrvineCAUSA
| |
Collapse
|
12
|
Gil J, Marques-Pamies M, Valassi E, Serra G, Salinas I, Xifra G, Casano-Sancho P, Carrato C, Biagetti B, Sesmilo G, Marcos-Ruiz J, Rodriguez-Lloveras H, Rueda-Pujol A, Aulinas A, Blanco A, Hostalot C, Simó-Servat A, Muñoz F, Rico M, Ibáñez-Domínguez J, Cordero E, Webb SM, Jordà M, Puig-Domingo M. Molecular characterization of epithelial-mesenchymal transition and medical treatment related-genes in non-functioning pituitary neuroendocrine tumors. Front Endocrinol (Lausanne) 2023; 14:1129213. [PMID: 37033229 PMCID: PMC10074986 DOI: 10.3389/fendo.2023.1129213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/23/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Different medical therapies have been developed for pituitary adenomas. However, Non-Functioning Pituitary Neuroendocrine Tumors (NF-PitNET) have shown little response to them. Furthermore, epithelial-mesenchymal transition (EMT) has been linked to resistance to medical treatment in a significant number of tumors, including pituitary adenomas. Methods We aimed to evaluate the expression of EMT-related markers in 72 NF-PitNET and 16 non-tumoral pituitaries. To further explore the potential usefulness of medical treatment for NF-PitNET we assessed the expression of somatostatin receptors and dopamine-associated genes. Results We found that SNAI1, SNAI2, Vimentin, KLK10, PEBP1, Ki-67 and SSTR2 were associated with invasive NF-PitNET. Furthermore, we found that the EMT phenomenon was more common in NF-PitNET than in GH-secreting pituitary tumors. Interestingly, PEBP1 was overexpressed in recurrent NF-PitNET, and could predict growth recurrence with 100% sensitivity but only 43% specificity. In parallel with previously reported studies, SSTR3 is highly expressed in our NF-PitNET cohort. However, SSTR3 expression is highly heterogeneous among the different histological variants of NF-PitNET with very low levels in silent corticotroph adenomas. Conclusion NF-PitNET showed an enhanced EMT phenomenon. SSTR3 targeting could be a good therapeutic candidate in NF-PitNET except for silent corticotroph adenomas, which express very low levels of this receptor. In addition, PEBP1 could be an informative biomarker of tumor regrowth, useful for predictive medicine in NF-PitNET.
Collapse
Affiliation(s)
- Joan Gil
- Endocrine Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Endocrinology, Research Center for Pituitary Diseases, Hospital Sant Pau, IIB-SPau, Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación en Red de Enfermedades Raras, CIBERER, Unit 747, Instituto de Salud Carlos III, Madrid, Spain
| | - Montserrat Marques-Pamies
- Endocrine Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Endocrinology, Hospital Municipal de Badalona, Badalona, Catalonia, Spain
| | - Elena Valassi
- Endocrine Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Endocrinology, Research Center for Pituitary Diseases, Hospital Sant Pau, IIB-SPau, Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación en Red de Enfermedades Raras, CIBERER, Unit 747, Instituto de Salud Carlos III, Madrid, Spain
| | - Guillermo Serra
- Department of Endocrinology, Son Espases University Hospital, Palma de Mallorca, Spain
| | - Isabel Salinas
- Department of Endocrinology and Nutrition, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Gemma Xifra
- Department of Endocrinology, Josep Trueta University Hospital, Girona, Spain
| | - Paula Casano-Sancho
- Centro de Investigación en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Pediatric Endocrinology Unit, Institut de Recerca SJS 39-57, Hospital Sant Joan de Déu, University of Barcelona, Esplugues, Spain
| | - Cristina Carrato
- Department of Pathology, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Betina Biagetti
- Department of Endocrinology, University Hospital Vall d’Hebron, Barcelona, Spain
| | - Gemma Sesmilo
- Department of Endocrinology, Dexeus University Hospital, Barcelona, Spain
| | - Jennifer Marcos-Ruiz
- Endocrine Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | | | - Anna Rueda-Pujol
- Endocrine Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Anna Aulinas
- Department of Endocrinology, Research Center for Pituitary Diseases, Hospital Sant Pau, IIB-SPau, Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación en Red de Enfermedades Raras, CIBERER, Unit 747, Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Blanco
- Department of Neurosurgery, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Cristina Hostalot
- Department of Neurosurgery, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Andreu Simó-Servat
- Department of Endocrinology, Hospital Universitari Mútua de Terrassa, Terrassa, Spain
| | - Fernando Muñoz
- Department of Neurosurgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marta Rico
- Department of Neurosurgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Esteban Cordero
- Department of Neurosurgery, University Hospital Vall d’Hebron, Barcelona, Spain
| | - Susan M. Webb
- Department of Endocrinology, Research Center for Pituitary Diseases, Hospital Sant Pau, IIB-SPau, Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación en Red de Enfermedades Raras, CIBERER, Unit 747, Instituto de Salud Carlos III, Madrid, Spain
| | - Mireia Jordà
- Endocrine Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Manel Puig-Domingo
- Endocrine Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Centro de Investigación en Red de Enfermedades Raras, CIBERER, Unit 747, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology, Hospital Municipal de Badalona, Badalona, Catalonia, Spain
| |
Collapse
|
13
|
Li K, Kratzmann V, Dai M, Gatzke N, Rocic P, Bramlage P, Grisk O, Lubomirov LT, Hoffmeister M, Lauxmann MA, Ritter O, Buschmann E, Bader M, Persson AB, Buschmann I, Hillmeister P. Angiotensin receptor-neprilysin inhibitor improves coronary collateral perfusion. Front Cardiovasc Med 2023; 9:981333. [PMID: 36818914 PMCID: PMC9936066 DOI: 10.3389/fcvm.2022.981333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/09/2022] [Indexed: 02/05/2023] Open
Abstract
Background We investigated the pleiotropic effects of an angiotensin receptor-neprilysin inhibitor (ARNi) on collateral-dependent myocardial perfusion in a rat model of coronary arteriogenesis, and performed comprehensive analyses to uncover the underlying molecular mechanisms. Methods A rat model of coronary arteriogenesis was established by implanting an inflatable occluder on the left anterior descending coronary artery followed by a 7-day repetitive occlusion procedure (ROP). Coronary collateral perfusion was measured by using a myocardial particle infusion technique. The putative ARNi-induced pro-arteriogenic effects were further investigated and compared with an angiotensin-converting enzyme inhibitor (ACEi). Expression of the membrane receptors and key enzymes in the natriuretic peptide system (NPS), renin-angiotensin-aldosterone system (RAAS) and kallikrein-kinin system (KKS) were analyzed by quantitative polymerase chain reaction (qPCR) and immunoblot assay, respectively. Protein levels of pro-arteriogenic cytokines were measured by enzyme-linked immunosorbent assay, and mitochondrial DNA copy number was assessed by qPCR due to their roles in arteriogenesis. Furthermore, murine heart endothelial cells (MHEC5-T) were treated with a neprilysin inhibitor (NEPi) alone, or in combination with bradykinin receptor antagonists. MHEC5-T proliferation was analyzed by colorimetric assay. Results The in vivo study showed that ARNis markedly improved coronary collateral perfusion, regulated the gene expression of KKS, and increased the concentrations of relevant pro-arteriogenic cytokines. The in vitro study demonstrated that NEPis significantly promoted MHEC5-T proliferation, which was diminished by bradykinin receptor antagonists. Conclusion ARNis improve coronary collateral perfusion and exert pro-arteriogenic effects via the bradykinin receptor signaling pathway.
Collapse
Affiliation(s)
- Kangbo Li
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Victoria Kratzmann
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Mengjun Dai
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nora Gatzke
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Petra Rocic
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Sam Houston State University, Huntsville, TX, United States
| | - Peter Bramlage
- Institute for Pharmacology and Preventive Medicine, Cloppenburg, Germany
| | - Olaf Grisk
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Lubomir T. Lubomirov
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Meike Hoffmeister
- Institute of Biochemistry, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Martin A. Lauxmann
- Institute of Biochemistry, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Oliver Ritter
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
- Department for Cardiology, Center for Internal Medicine I, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Eva Buschmann
- Department of Cardiology, University Clinic Graz, Graz, Austria
| | - Michael Bader
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Anja Bondke Persson
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ivo Buschmann
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| | - Philipp Hillmeister
- Department for Angiology, Center for Internal Medicine I, Deutsches Angiologie Zentrum Brandenburg - Berlin, University Clinic Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
- Faculty of Health Sciences Brandenburg, Joint Faculty of the Brandenburg University of Technology Cottbus – Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Brandenburg an der Havel, Germany
| |
Collapse
|
14
|
Yuan Y, Song J, Wu Q. Aberrant gene expression pattern in the glycolysis-cholesterol synthesis axis is linked with immune infiltration and prognosis in prostate cancer: A bioinformatics analysis. Medicine (Baltimore) 2022; 101:e31416. [PMID: 36316896 PMCID: PMC9622640 DOI: 10.1097/md.0000000000031416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Aberrant lipid metabolism is an early event in tumorigenesis and has been found in a variety of tumor types, especially prostate cancer (PCa). Therefore, We hypothesize that PCa can be stratified into metabolic subgroups based on glycolytic and cholesterogenic related genes, and the different subgroups are closely related to the immune microenvironment. Bioinformatics analysis of genomic, transcriptomic, and clinical data from a comprehensive cohort of PCa patients was performed. Datasets included the Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) dataset, GSE70768, our previously published PCa cohort. The unsupervised cluster analysis was employed to stratify PCa samples based on the expression of metabolic-related genes. Four molecular subtypes were identified, named Glycolytic, Cholesterogenic, Mixed, and Quiescent. Each metabolic subtype has specific features. Among the 4 subtypes, the cholesterogenic subtype exhibited better median survival, whereas patients with high expression of glycolytic genes showed the shortest survival. The mitochondrial pyruvate carriers (MPC) 1 exhibited expression difference between PCa metabolic subgroups, but not for MPCs 2. Glycolytic subtypes had lower immune cell scores, while Cholesterogenic subgroups had higher immune cell scores. Our results demonstrated that metabolic classifications based on specific glycolytic and cholesterol-producing pathways provide new biological insights into previously established subtypes and may guide develop personalized therapies for unique tumor metabolism characteristics.
Collapse
Affiliation(s)
- Yiwen Yuan
- Guizhou Medical University, Guiyang, Guizhou, P.R. China
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, P.R. China
| | - Jukun Song
- Department of Oral and Maxillofacial Surgery, the Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, Guizhou, P.R. China
| | - Qinghua Wu
- Guizhou Medical University, Guiyang, Guizhou, P.R. China
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, P.R. China
- *Correspondence: Qinghua Wu, Guizhou Medical University, Guiyang, Guizhou, P.R. China (e-mail: )
| |
Collapse
|
15
|
Liu Y, Gong W, Preis S, Dorn J, Kiechle M, Reuning U, Magdolen V, Dreyer TF. A Pair of Prognostic Biomarkers in Triple-Negative Breast Cancer: KLK10 and KLK11 mRNA Expression. Life (Basel) 2022; 12:life12101517. [PMID: 36294951 PMCID: PMC9605449 DOI: 10.3390/life12101517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/27/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with poor patient prognosis and limited therapeutic options. A lack of prognostic biomarkers and therapeutic targets fuels the need for new approaches to tackle this severe disease. Extracellular matrix degradation, release, and modulation of the activity of growth factors/cytokines/chemokines, and the initiation of signaling pathways by extracellular proteolytic networks, have been identified as major processes in the carcinogenesis of breast cancer. Members of the kallikrein-related peptidase (KLK) family contribute to these tumor-relevant processes, and are associated with breast cancer progression and metastasis. In this study, the clinical relevance of mRNA expression of two members of this family, KLK10 and KLK11, has been evaluated in TNBC. For this, their expression levels were quantified in tumor tissue of a large, well-characterized patient cohort (n = 123) via qPCR. Although, in general, the overall expression of both factors are lower in tumor tissue of breast cancer patients (encompassing all subtypes) compared to normal tissue of healthy donors, in the TNBC subtype, expression is even increased. In our cohort, a significant, positive correlation between the expression levels of both KLKs was detected, indicating a coordinate expression mode of these proteases. Elevated KLK10 and KLK11 mRNA levels were associated with poor patient prognosis. Moreover, both factors were found to be independent of other established clinical factors such as age, lymph node status, or residual tumor mass, as determined by multivariable Cox regression analysis. Thus, both proteases, KLK10 and KLK11, may represent unfavorable prognostic factors for TNBC patients and, furthermore, appear as promising potential targets for therapy in TNBC.
Collapse
Affiliation(s)
- Yueyang Liu
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
- Department of Gynecology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 519041, China
| | - Weiwei Gong
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
- Department of Hematology/Oncology, Guangzhou Women and Children’s Medical Center, Guangzhou 519041, China
| | - Sarah Preis
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
| | - Julia Dorn
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
| | - Marion Kiechle
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
| | - Tobias F. Dreyer
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-4140-7408
| |
Collapse
|
16
|
Zhao J, Liang K, Zhong H, Liu S, Sun P, He R. A cold-water polysaccharide-protein complex from Grifola frondosa exhibited antiproliferative activity via mitochondrial apoptotic and Fas/FasL pathways in HepG2 cells. Int J Biol Macromol 2022; 218:1021-1032. [PMID: 35863663 DOI: 10.1016/j.ijbiomac.2022.07.098] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/30/2022] [Accepted: 07/12/2022] [Indexed: 12/26/2022]
Abstract
Grifola frondosa (G. frondosa) is widely known for its anti-tumor potential, which has been demonstrated by numerous scientific researches. In this study, two water soluble polysaccharide-protein complexes were extracted from G. frondosa at 4 °C (GFG-4) and 100 °C (GFG-100) and purified. Compared with GFG-100, GFG-4 had a higher protein content and molecular weight. The main monosaccharides of GFG-4 and GFG-100 were rhamnose, glucose, and galactose, with an approximate ratio of 3.00: 1.00: 0.86 and 2.85: 1.00: 0.94, respectively. The Fourier transform infrared spectra indicated that the two polysaccharide-protein complexes displayed characteristic functional groups of polysaccharides and proteins, and mainly contain pyranose ring with α-glycosidic linkage. Atomic force microscope images showed that both GFG-4 and GFG-100 exhibited straight chains, and GFG-4 possessed a relatively abundant fraction of branched chains. Intriguingly, GFG-4 showed a stronger antiproliferative activity against HepG2 cells than GFG-100. The mechanisms were further investigated by quantitative real-time PCR and western blot, it found that GFG-4 inhibited the proliferation of HepG2 cells mainly through the intrinsic activation of mitochondrial pathway and the Fas/FasL-mediated Caspase-8/-3 pathway. Conclusively, G. frondosa cold-water extracted polysaccharide-protein complexes could be used as a functional food for preventing or treating hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jiahui Zhao
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Kaiyue Liang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China; Bioactives and Functional Foods Research Center, China National Light Industry, Hangzhou 310014, China
| | - Shizhu Liu
- Zhejiang Fangge Pharmaceutical Co., Ltd., Qingyuan 323800, China
| | - Peilong Sun
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China; Key Laboratory of Food Macromolecular Resources Processing Technology Research, China National Light Industry, Hangzhou 310014, China; Zhejiang Fangge Pharmaceutical Co., Ltd., Qingyuan 323800, China.
| | - Rongjun He
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China; Bioactives and Functional Foods Research Center, China National Light Industry, Hangzhou 310014, China; Zhejiang Fangge Pharmaceutical Co., Ltd., Qingyuan 323800, China.
| |
Collapse
|
17
|
Buschhaus JM, Rajendran S, Humphries BA, Cutter AC, Muñiz AJ, Ciavattone NG, Buschhaus AM, Cañeque T, Nwosu ZC, Sahoo D, Bevoor AS, Shah YM, Lyssiotis CA, Ghosh P, Wicha MS, Rodriguez R, Luker GD. Effects of iron modulation on mesenchymal stem cell-induced drug resistance in estrogen receptor-positive breast cancer. Oncogene 2022; 41:3705-3718. [PMID: 35732800 PMCID: PMC9288981 DOI: 10.1038/s41388-022-02385-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/31/2022] [Accepted: 06/13/2022] [Indexed: 01/03/2023]
Abstract
Patients with estrogen receptor-positive (ER+) breast cancer, the most common subtype, remain at risk for lethal metastatic disease years after diagnosis. Recurrence arises partly because tumor cells in bone marrow become resistant to estrogen-targeted therapy. Here, we utilized a co-culture model of bone marrow mesenchymal stem cells (MSCs) and ER+ breast cancer cells to recapitulate interactions of cancer cells in bone marrow niches. ER+ breast cancer cells in direct contact with MSCs acquire cancer stem-like (CSC) phenotypes with increased resistance to standard antiestrogenic drugs. We confirmed that co-culture with MSCs increased labile iron in breast cancer cells, a phenotype associated with CSCs and disease progression. Clinically approved iron chelators and in-house lysosomal iron-targeting compounds restored sensitivity to antiestrogenic therapy. These findings establish iron modulation as a mechanism to reverse MSC-induced drug resistance and suggest iron modulation in combination with estrogen-targeted therapy as a promising, translatable strategy to treat ER+ breast cancer.
Collapse
Affiliation(s)
- Johanna M Buschhaus
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Shrila Rajendran
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Brock A Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Alyssa C Cutter
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ayşe J Muñiz
- Macromolecular Science and Engineering and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109-2200, USA
| | - Nicholas G Ciavattone
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Alexander M Buschhaus
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Tatiana Cañeque
- Institut Curie, Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, PSL Research University, Paris, France
| | - Zeribe C Nwosu
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Debashis Sahoo
- Pediatrics, and Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Avinash S Bevoor
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pradipta Ghosh
- Departments of Medicine and Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Max S Wicha
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Raphaël Rodriguez
- Institut Curie, Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, PSL Research University, Paris, France
| | - Gary D Luker
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA.
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
- Department of Microbiology and Immunology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
18
|
Kong P, Zhang L, Zhang Z, Feng K, Sang Y, Duan X, Liu C, Sun T, Tao Z, Liu W. Emerging Proteins in CRPC: Functional Roles and Clinical Implications. Front Oncol 2022; 12:873876. [PMID: 35756667 PMCID: PMC9226405 DOI: 10.3389/fonc.2022.873876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer (PCa) is the most common cancer in men in the western world, but the lack of specific and sensitive markers often leads to overtreatment of prostate cancer which eventually develops into castration-resistant prostate cancer (CRPC). Novel protein markers for diagnosis and management of CRPC will be promising. In this review, we systematically summarize and discuss the expression pattern of emerging proteins in tissue, cell lines, and serum when castration-sensitive prostate cancer (CSPC) progresses to CRPC; focus on the proteins involved in CRPC growth, invasion, metastasis, metabolism, and immune microenvironment; summarize the current understanding of the regulatory mechanisms of emerging proteins in CSPC progressed to CRPC at the molecular level; and finally summarize the clinical applications of emerging proteins as diagnostic marker, prognostic marker, predictive marker, and therapeutic marker.
Collapse
Affiliation(s)
- Piaoping Kong
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lingyu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengliang Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Kangle Feng
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yiwen Sang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuzhi Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chunhua Liu
- Department of Blood Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
19
|
Williams D, Mahmoud M, Liu R, Andueza A, Kumar S, Kang DW, Zhang J, Tamargo I, Villa-Roel N, Baek KI, Lee H, An Y, Zhang L, Tate EW, Bagchi P, Pohl J, Mosnier LO, Diamandis EP, Mihara K, Hollenberg MD, Dai Z, Jo H. Stable flow-induced expression of KLK10 inhibits endothelial inflammation and atherosclerosis. eLife 2022; 11:e72579. [PMID: 35014606 PMCID: PMC8806187 DOI: 10.7554/elife.72579] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/08/2022] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis preferentially occurs in arterial regions exposed to disturbed blood flow (d-flow), while regions exposed to stable flow (s-flow) are protected. The proatherogenic and atheroprotective effects of d-flow and s-flow are mediated in part by the global changes in endothelial cell (EC) gene expression, which regulates endothelial dysfunction, inflammation, and atherosclerosis. Previously, we identified kallikrein-related peptidase 10 (Klk10, a secreted serine protease) as a flow-sensitive gene in mouse arterial ECs, but its role in endothelial biology and atherosclerosis was unknown. Here, we show that KLK10 is upregulated under s-flow conditions and downregulated under d-flow conditions using in vivo mouse models and in vitro studies with cultured ECs. Single-cell RNA sequencing (scRNAseq) and scATAC sequencing (scATACseq) study using the partial carotid ligation mouse model showed flow-regulated Klk10 expression at the epigenomic and transcription levels. Functionally, KLK10 protected against d-flow-induced permeability dysfunction and inflammation in human artery ECs, as determined by NFκB activation, expression of vascular cell adhesion molecule 1 and intracellular adhesion molecule 1, and monocyte adhesion. Furthermore, treatment of mice in vivo with rKLK10 decreased arterial endothelial inflammation in d-flow regions. Additionally, rKLK10 injection or ultrasound-mediated transfection of Klk10-expressing plasmids inhibited atherosclerosis in Apoe-/- mice. Moreover, KLK10 expression was significantly reduced in human coronary arteries with advanced atherosclerotic plaques compared to those with less severe plaques. KLK10 is a flow-sensitive endothelial protein that serves as an anti-inflammatory, barrier-protective, and anti-atherogenic factor.
Collapse
Affiliation(s)
- Darian Williams
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
- Molecular and Systems Pharmacology Program, Emory UniversityAtlantaUnited States
| | - Marwa Mahmoud
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Renfa Liu
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
- Department of Biomedical Engineering, Peking UniversityBeijingChina
| | - Aitor Andueza
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Sandeep Kumar
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Dong-Won Kang
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Jiahui Zhang
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Ian Tamargo
- Molecular and Systems Pharmacology Program, Emory UniversityAtlantaUnited States
| | - Nicolas Villa-Roel
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | - Kyung-In Baek
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
| | | | | | - Leran Zhang
- Department of Chemistry, Imperial College LondonLondonUnited Kingdom
| | - Edward W Tate
- Department of Chemistry, Imperial College LondonLondonUnited Kingdom
| | - Pritha Bagchi
- Emory Integrated Proteomics Core, Emory UniversityAtlantaUnited States
| | - Jan Pohl
- Biotechnology Core Facility Branch, Centers for Disease Control and PreventionAtlantaUnited States
| | - Laurent O Mosnier
- Department of Molecular Medicine, Scripps Research InstituteSan DiegoUnited States
| | | | - Koichiro Mihara
- Department of Physiology and Pharmacology, University of CalgaryCalgaryCanada
| | - Morley D Hollenberg
- Department of Physiology and Pharmacology, University of CalgaryCalgaryCanada
| | - Zhifei Dai
- Department of Biomedical Engineering, Peking UniversityBeijingChina
| | - Hanjoong Jo
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of TechnologyAtlantaUnited States
- Molecular and Systems Pharmacology Program, Emory UniversityAtlantaUnited States
- Department of Medicine, Emory UniversityAtlantaUnited States
| |
Collapse
|
20
|
Zarei M, Sahebi Vaighan N, Ziai SA. Purinergic receptor ligands: the cytokine storm attenuators, potential therapeutic agents for the treatment of COVID-19. Immunopharmacol Immunotoxicol 2021; 43:633-643. [PMID: 34647511 PMCID: PMC8544669 DOI: 10.1080/08923973.2021.1988102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/25/2021] [Indexed: 12/13/2022]
Abstract
The coronavirus disease-19 (COVID-19), at first, was reported in Wuhan, China, and then rapidly became pandemic throughout the world. Cytokine storm syndrome (CSS) in COVID-19 patients is associated with high levels of cytokines and chemokines that cause multiple organ failure, systemic inflammation, and hemodynamic instabilities. Acute respiratory distress syndrome (ARDS), a common complication of COVID-19, is a consequence of cytokine storm. In this regard, several drugs have been being investigated to suppress this inflammatory condition. Purinergic signaling receptors comprising of P1 adenosine and P2 purinoceptors play a critical role in inflammation. Therefore, activation or inhibition of some subtypes of these kinds of receptors is most likely to be beneficial to attenuate cytokine storm. This article summarizes suggested therapeutic drugs with potential anti-inflammatory effects through purinergic receptors.
Collapse
Affiliation(s)
- Malek Zarei
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navideh Sahebi Vaighan
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Ziai
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Mirza S, Kalluchi A, Raza M, Saleem I, Mohapatra B, Pal D, Ouellette MM, Qiu F, Yu L, Lobanov A, Zheng ZM, Zhang Y, Alsaleem MA, Rakha EA, Band H, Rowley MJ, Band V. Ecdysoneless Protein Regulates Viral and Cellular mRNA Splicing to Promote Cervical Oncogenesis. Mol Cancer Res 2021; 20:305-318. [PMID: 34670863 DOI: 10.1158/1541-7786.mcr-21-0567] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/10/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022]
Abstract
High-risk human papillomaviruses (HPV), exemplified by HPV16/18, are causally linked to human cancers of the anogenital tract, skin, and upper aerodigestive tract. Previously, we identified Ecdysoneless (ECD) protein, the human homolog of the Drosophila ecdysoneless gene, as a novel HPV16 E6-interacting protein. Here, we show that ECD, through its C-terminal region, selectively binds to high-risk but not to low-risk HPV E6 proteins. We demonstrate that ECD is overexpressed in cervical and head and neck squamous cell carcinoma (HNSCC) cell lines as well as in tumor tissues. Using The Cancer Genome Atlas dataset, we show that ECD mRNA overexpression predicts shorter survival in patients with cervical and HNSCC. We demonstrate that ECD knockdown in cervical cancer cell lines led to impaired oncogenic behavior, and ECD co-overexpression with E7 immortalized primary human keratinocytes. RNA-sequencing analyses of SiHa cells upon ECD knockdown showed to aberrations in E6/E7 RNA splicing, as well as RNA splicing of several HPV oncogenesis-linked cellular genes, including splicing of components of mRNA splicing machinery itself. Taken together, our results support a novel role of ECD in viral and cellular mRNA splicing to support HPV-driven oncogenesis. IMPLICATIONS: This study links ECD overexpression to poor prognosis and shorter survival in HNSCC and cervical cancers and identifies a critical role of ECD in cervical oncogenesis through regulation of viral and cellular mRNA splicing.
Collapse
Affiliation(s)
- Sameer Mirza
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Achyuth Kalluchi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Mohsin Raza
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Irfana Saleem
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Bhopal Mohapatra
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Dhananjaya Pal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Michel M Ouellette
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Fang Qiu
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska
| | - Lulu Yu
- Tumor Virus RNA Biology Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Alexei Lobanov
- CCR Collaborative Bioinformatics Resource (CCBR), National Cancer Institute, Bethesda, Maryland
| | - Zhi-Ming Zheng
- Tumor Virus RNA Biology Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Ying Zhang
- Northshore University Health System, Chicago, Illinois
| | - Mansour A Alsaleem
- Department of Pathology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Department of Applied Medical Sciences, Onizah Community College, Qassim University, Qassim, Saudi Arabia
| | - Emad A Rakha
- Department of Pathology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Hamid Band
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska.
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
| | - M Jordan Rowley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska.
| | - Vimla Band
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska.
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
22
|
Hu J, Luo H, Qu Q, Liao X, Huang C, Chen J, Cai Z, Bao Y, Chen G, Li B, Cui W. Cell Membrane-Inspired Polymeric Vesicles for Combined Photothermal and Photodynamic Prostate Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:42511-42520. [PMID: 32897691 DOI: 10.1021/acsami.0c11636] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Photothermal therapy (PTT) and photodynamic therapy (PDT) have emerged as highly prospective therapeutic modalities in cancer therapy. Notwithstanding, a critical challenge still remains in the exploration of an effective strategy to maximize the synergistic efficacy of PTT and PDT due to low photoconversion efficiency. Herein, inspired by the phospholipid bimolecular structure of the cell membrane, bionic cell membrane polymeric vesicles with photothermal/photodynamic synergy for prostate cancer therapy at one wavelength's excitation are constructed in one step by the coordination of hexadecyl trimethyl ammonium bromide (CTAB) from the surface of hydrophobic gold nanorods (AuNRs) with indocyanine green (ICG) and polycaprolactone (PCL), achieving their self-assembly in aqueous solutions. Importantly, the aggregation of the assembly improves the stability of the vesicles, realizing the synergistic effect of PTT and PDT for prostate cancer therapy. After being assembled within polymeric vesicles, bifunctional photosensitizer ICG can generate reactive oxygen species (ROS) and photothermal effect under light treatment. Their ROS not only induce PDT efficacy but also destroy the integrity of the lysosomal membrane, promoting the translocation of ICG and another photosensitizer called gold nanorods (AuNRs) into the cytosol. Moreover, their photothermal effects produced by both photosensitizers are able to engender greater damage to the tumor cells because of the close distance with organelles. This structure manifests good cellular uptake, highly effective tumor accumulation, high photothermal conversion efficiency, and excellent properties of enhanced photobleaching resistance, which are beneficial to ICG-based fluorescence tumor imaging. Using the same near-infrared (NIR) wavelength for excitation, the AuNR/ICG vesicles can reduce the side effect rate of photodamage on the skin. In addition, by generating reactive oxygen species (ROS) and double photothermal effect, the vesicles under NIR excitation can promote the apoptosis of PC3 tumor cells. Taken together, the spontaneous self-assembled AuNR/ICG vesicles exhibit huge potential in advanced-stage prostate cancer therapy, especially for the prostate-specific membrane antigen (PSMA)-negative castration-resistant subtype.
Collapse
Affiliation(s)
- Jiajia Hu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
- Department of Pharmacology and Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, Connecticut 06511, United States
| | - Huanhuan Luo
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing 314000, P. R. China
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Qian Qu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Xiaofeng Liao
- Department of Pharmacology and Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, Connecticut 06511, United States
| | - Chenglong Huang
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing 314000, P. R. China
| | - Jiayi Chen
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing 314000, P. R. China
| | - Zhenhai Cai
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing 314000, P. R. China
| | - Yi Bao
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing 314000, P. R. China
| | - Gang Chen
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing 314000, P. R. China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Wenguo Cui
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| |
Collapse
|
23
|
Lin CL, Ying TH, Yang SF, Wang SW, Cheng SP, Lee JJ, Hsieh YH. Transcriptional Suppression of miR-7 by MTA2 Induces Sp1-Mediated KLK10 Expression and Metastasis of Cervical Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:699-710. [PMID: 32402941 PMCID: PMC7218230 DOI: 10.1016/j.omtn.2020.04.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/18/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
MTA2 is involved in tumor proliferation and metastasis. However, the role of MTA2 in cervical cancer thus far has not been identified. In this study, we report that elevated expression of MTA2 negatively correlates with Kallikrein-10 (KLK10) expression and poor prognosis of cervical cancer patients. Knockdown of MTA2 substantially inhibited tumor cell migration and invasion, and it enhanced KLK10 expression of the cervical cancer cells in vitro and in vivo. Functionally, shMTA2-mediated suppression of cell mobility was significantly restored by knockdown of KLK10. We also found that Sp1 (transcription factor specificity protein 1) is critical for shMTA2-induced transcriptional upregulation of KLK10 and subsequent biological functions. Furthermore, we found that the expression of miR-7 is elevated by MTA2 silencing and then by direct inhibition of Sp1 expression. Knockdown of Sp1 additively enhanced KLK10 expression in MTA2-knocked down cervical cancer cells, suggesting that the miR-7/Sp1 axis acts as an effector of MTA2 to impact KLK10 levels and mobility of cervical cancer cells. Taken together, our findings provide new insights into the physiological relationship between MTA2 and KLK10 via regulating the miR-7/Sp1 axis, and they provide a potential therapeutic target in cervical cancer.
Collapse
Affiliation(s)
- Chia-Liang Lin
- Institute of Biochemistry, Microbiology, and Immunology, Chung Shan Medical University, Taichung, Taiwan; Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, New Taipei City, Taiwan
| | - Tsung-Ho Ying
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Ping Cheng
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, New Taipei City, Taiwan; Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan; Department of Pharmacology, Taipei Medical University, Taipei, Taiwan
| | - Jie-Jen Lee
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, New Taipei City, Taiwan; Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan; Department of Pharmacology, Taipei Medical University, Taipei, Taiwan.
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
24
|
Moradi A, Srinivasan S, Clements J, Batra J. Beyond the biomarker role: prostate-specific antigen (PSA) in the prostate cancer microenvironment. Cancer Metastasis Rev 2020; 38:333-346. [PMID: 31659564 DOI: 10.1007/s10555-019-09815-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prostate-specific antigen (PSA) blood test is the accepted biomarker of tumor recurrence. PSA levels in serum correlate with disease progression, though its diagnostic accuracy is questionable. As a result, significant progress has been made in developing modified PSA tests such as PSA velocity, PSA density, 4Kscore, PSA glycoprofiling, Prostate Health Index, and the STHLM3 test. PSA, a serine protease, is secreted from the epithelial cells of the prostate. PSA has been suggested as a molecular target for prostate cancer therapy due to the fact that it is not only active in prostate tissue but also has a pivotal role on prostate cancer signaling pathways including proliferation, invasion, metastasis, angiogenesis, apoptosis, immune response, and tumor microenvironment regulation. Here, we summarize the current standing of PSA in prostate cancer progression as well as its utility in prostate cancer therapeutic approaches with an emphasis on the role of PSA in the tumor microenvironment.
Collapse
Affiliation(s)
- Afshin Moradi
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia. .,Translational Research Institute, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
25
|
Goettig P, Brandstetter H, Magdolen V. Surface loops of trypsin-like serine proteases as determinants of function. Biochimie 2019; 166:52-76. [PMID: 31505212 PMCID: PMC7615277 DOI: 10.1016/j.biochi.2019.09.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023]
Abstract
Trypsin and chymotrypsin-like serine proteases from family S1 (clan PA) constitute the largest protease group in humans and more generally in vertebrates. The prototypes chymotrypsin, trypsin and elastase represent simple digestive proteases in the gut, where they cleave nearly any protein. Multidomain trypsin-like proteases are key players in the tightly controlled blood coagulation and complement systems, as well as related proteases that are secreted from diverse immune cells. Some serine proteases are expressed in nearly all tissues and fluids of the human body, such as the human kallikreins and kallikrein-related peptidases with specialization for often unique substrates and accurate timing of activity. HtrA and membrane-anchored serine proteases fulfill important physiological tasks with emerging roles in cancer. The high diversity of all family members, which share the tandem β-barrel architecture of the chymotrypsin-fold in the catalytic domain, is conferred by the large differences of eight surface loops, surrounding the active site. The length of these loops alters with insertions and deletions, resulting in remarkably different three-dimensional arrangements. In addition, metal binding sites for Na+, Ca2+ and Zn2+ serve as regulatory elements, as do N-glycosylation sites. Depending on the individual tasks of the protease, the surface loops determine substrate specificity, control the turnover and allow regulation of activation, activity and degradation by other proteins, which are often serine proteases themselves. Most intriguingly, in some serine proteases, the surface loops interact as allosteric network, partially tuned by protein co-factors. Knowledge of these subtle and complicated molecular motions may allow nowadays for new and specific pharmaceutical or medical approaches.
Collapse
Affiliation(s)
- Peter Goettig
- Division of Structural Biology, Department of Biosciences, University of Salzburg, Billrothstrasse 11, 5020, Salzburg, Austria.
| | - Hans Brandstetter
- Division of Structural Biology, Department of Biosciences, University of Salzburg, Billrothstrasse 11, 5020, Salzburg, Austria
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, School of Medicine, Technical University of Munich, Ismaninger Strasse 22, 81675, München, Germany
| |
Collapse
|
26
|
NES1/KLK10 and hNIS gene therapy enhanced iodine-131 internal radiation in PC3 proliferation inhibition. Front Med 2019; 13:646-657. [DOI: 10.1007/s11684-018-0643-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 04/26/2018] [Indexed: 12/26/2022]
|
27
|
Schilling O, Biniossek ML, Mayer B, Elsässer B, Brandstetter H, Goettig P, Stenman UH, Koistinen H. Specificity profiling of human trypsin-isoenzymes. Biol Chem 2019; 399:997-1007. [PMID: 29883318 DOI: 10.1515/hsz-2018-0107] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/05/2018] [Indexed: 01/18/2023]
Abstract
In humans, three different trypsin-isoenzymes have been described. Of these, trypsin-3 appears to be functionally different from the others. In order to systematically study the specificity of the trypsin-isoenzymes, we utilized proteome-derived peptide libraries and quantitative proteomics. We found similar specificity profiles dominated by the well-characterized preference for cleavage after lysine and arginine. Especially, trypsin-1 slightly favored lysine over arginine in this position, while trypsin-3 did not discriminate between them. In the P1' position, which is the residue C-terminal to the cleavage site, we noticed a subtle enrichment of alanine and glycine for all three trypsins and for trypsin-3 there were additional minor P1' and P2' preferences for threonine and aspartic acid, respectively. These findings were confirmed by FRET peptide substrates showing different susceptibility to cleavage by different trypsins. The preference of trypsin-3 for aspartic acid in P2' is explained by salt bridge formation with the unique Arg193. This salt bridge enables and stabilizes a canonical oxyanion conformation by the amides of Ser195 and Arg193, thus manifesting a selective substrate-assisted catalysis. As trypsin-3 has been proposed to be a therapeutic target and marker for cancers, our results may aid the development of specific inhibitors for cancer therapy and diagnostic probes.
Collapse
Affiliation(s)
- Oliver Schilling
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, D-79104 Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Martin L Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany
| | - Bettina Mayer
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany
| | - Brigitta Elsässer
- Department of Biosciences, University of Salzburg, Billrothstr. 11, A-5020 Salzburg, Austria
| | - Hans Brandstetter
- Department of Biosciences, University of Salzburg, Billrothstr. 11, A-5020 Salzburg, Austria
| | - Peter Goettig
- Department of Biosciences, University of Salzburg, Billrothstr. 11, A-5020 Salzburg, Austria
| | - Ulf-Håkan Stenman
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Central Hospital, Haartmaninkatu 8, FI-00290 Helsinki, Finland
| | - Hannu Koistinen
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Central Hospital, Haartmaninkatu 8, FI-00290 Helsinki, Finland
| |
Collapse
|
28
|
Loessner D, Goettig P, Preis S, Felber J, Bronger H, Clements JA, Dorn J, Magdolen V. Kallikrein-related peptidases represent attractive therapeutic targets for ovarian cancer. Expert Opin Ther Targets 2018; 22:745-763. [PMID: 30114962 DOI: 10.1080/14728222.2018.1512587] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Aberrant levels of kallikrein-related peptidases (KLK1-15) have been linked to cancer cell proliferation, invasion and metastasis. In ovarian cancer, the KLK proteolytic network has a crucial role in the tissue and tumor microenvironment. Publically available ovarian cancer genome and expression data from multiple patient cohorts show an upregulation of most KLKs. Areas covered: Here, we review the expression levels of all 15 members of this family in normal and ovarian cancer tissues, categorizing them into highly and moderately or weakly expressed KLKs, and their association with patient prognosis and survival. We summarize their tumor-biological functions determined in cell-based assays and xenograft models, further highlighting their suitability as cancer biomarkers and attractive candidates for drug development. Finally, we discuss some different pharmaceutical approaches, including peptide-based and small molecule inhibitors, cyclic peptides, depsipeptides, engineered natural inhibitors, antibodies, RNA/DNA-based aptamers, prodrugs, miRNA and siRNA. Expert opinion: In light of the results from clinical and tumor-biological studies, together with the available pharmaceutical tools, we suggest KLK4, KLK5, KLK6 and possibly KLK7 as preferred targets for inhibition in ovarian cancer.
Collapse
Affiliation(s)
- Daniela Loessner
- a Barts Cancer Institute , Queen Mary University of London , London , UK.,b Institute of Health and Biomedical Innovation , Queensland University of Technology (QUT) , Brisbane , Australia
| | - Peter Goettig
- c Department of Biosciences , University of Salzburg , Salzburg , Austria
| | - Sarah Preis
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Johanna Felber
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Holger Bronger
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Judith A Clements
- b Institute of Health and Biomedical Innovation , Queensland University of Technology (QUT) , Brisbane , Australia.,e Australian Prostate Cancer Research Centre - Queensland , Queensland University of Technology (QUT), Translational Research Institute , Brisbane , Australia
| | - Julia Dorn
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Viktor Magdolen
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| |
Collapse
|
29
|
Geng X, Liu Y, Dreyer T, Bronger H, Drecoll E, Magdolen V, Dorn J. Elevated tumor tissue protein expression levels of kallikrein-related peptidases KLK10 and KLK11 are associated with a better prognosis in advanced high-grade serous ovarian cancer patients. Am J Cancer Res 2018; 8:1856-1864. [PMID: 30323977 PMCID: PMC6176182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 06/08/2023] Open
Abstract
Several members of the KLK family have been proposed to modulate various tumor-relevant processes. Previously, we have shown that in advanced high-grade serous ovarian cancer tissue high KLK11 mRNA levels were significantly associated with prolonged overall and progression-free patients' survival. Furthermore, KLK11 mRNA expression positively correlated with KLK10 mRNA. In the present study, we examined the prognostic value for both KLK10 and KLK11 on the protein expression level by immunohistochemistry (IHC). A cohort encompassing 159 patient tumor samples afflicted with advanced high-grade (FIGO III/IV) serous ovarian cancer, present on tissue microarrays (TMA), was analyzed. For estimation of KLK10 and KLK11 immunoreactivity, an automated digital IHC image analysis algorithm was selected to quantify the antibody staining intensity in the tissues via an immunoreactive score (IRS). In line with the results obtained by mRNA analysis, KLK10 protein expression values were significantly and positively correlated with KLK11 protein expression values. In Kaplan-Meier analyses, both elevated KLK10, KLK11, and the combination of KLK10 and KLK11 protein levels were significantly linked with prolonged overall survival (OS). The addition of KLK10, KLK11 or the KLK10+KLK11 combination IRS to the base model in multivariate Cox analysis demonstrated that high KLK11 and KLK10+KLK11 protein expression levels, apart from clinical parameters, remained favorable independent predictive markers for OS. In conclusion, in the present study, we have validated the coordinate expression of KLK10 and KLK11 in advanced high-grade serous ovarian cancer. Furthermore, both increased KLK10 and KLK11 protein expression is associated with favorable prognosis in this major ovarian cancer subtype. The combined KLK10+KLK11 marker performed even stronger than KLK10 or KLK11 alone.
Collapse
Affiliation(s)
- Xiaocong Geng
- Department of Obstetrics and Gynecology, Technische Universität MünchenMunich, Germany
| | - Yueyang Liu
- Department of Obstetrics and Gynecology, Technische Universität MünchenMunich, Germany
| | - Tobias Dreyer
- Department of Obstetrics and Gynecology, Technische Universität MünchenMunich, Germany
| | - Holger Bronger
- Department of Obstetrics and Gynecology, Technische Universität MünchenMunich, Germany
| | - Enken Drecoll
- Institute of Pathology, Technische Universität MünchenMunich, Germany
| | - Viktor Magdolen
- Department of Obstetrics and Gynecology, Technische Universität MünchenMunich, Germany
| | - Julia Dorn
- Department of Obstetrics and Gynecology, Technische Universität MünchenMunich, Germany
| |
Collapse
|
30
|
Xu LJ, Duan Y, Wang P, Yin HQ. MiR-199b-5p promotes tumor growth and metastasis in cervical cancer by down-regulating KLK10. Biochem Biophys Res Commun 2018; 503:556-563. [PMID: 29807015 DOI: 10.1016/j.bbrc.2018.05.165] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 05/25/2018] [Indexed: 12/12/2022]
Abstract
MiR-199 b-5p and kallikrein-related peptidase 10 (KLK10) are related to various disease processes and pathogenesis. However, little is known about the molecular mechanisms of miR-199 b-5p and KLK10 in human cervical cancer. In the present study, we found that miR-199 b-5p was highly expressed in cervical cancer tissues and cell lines, and was positively correlated with overall survival (OS) and progression-free survival (PFS), higher incidences of larger tumor sizes, late International Federation of Gynecology and Obstetrics (FIGO) stages and preoperative metastasis. Further, we found that transfecting miR-199 b-5p mimics into cervical cancer cells promoted tumor progression through enhancing the cell viability, migration, and suppressing apoptosis by using 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT), wound healing and flow cytometry analysis. Luciferase reporter assays indicated that miR-199 b-5p targeted the 3'-untranslated region (3'-UTR) of KLK10. Over-expressing KLK10 reversed the role of miR-199 b-5p in accelerating cervical cancer progression. Suppressing miR-199 b-5p expressions improved apoptosis and reduced the cell viability, while the process was reversed in KLK10-knockdown cervical cancer cells. In vivo analysis verified the effects of miR-199 b-5p on promoting cervical cancer progression, accompanied with reduced KLK10 expressions. In summary, we identified that miR-199 b-5p played as a tumor promoter in cervical cancer cell growth by targeting KLK10, and miR-199 b-5p might function as a novel biomarker for diagnosis or therapeutic targets of human cervical cancer.
Collapse
Affiliation(s)
- Li-Juan Xu
- Clinical Laboratory of the First Affiliated Hospital to Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou, Henan, 450052, China
| | - Yu Duan
- Clinical Laboratory of the First Affiliated Hospital to Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou, Henan, 450052, China
| | - Peng Wang
- Center for Tumor Biotherapy, The First Affiliated Hospital and College of Public Health and Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Hui-Qing Yin
- Clinical Laboratory of the First Affiliated Hospital to Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
31
|
Cao XY, Zhang XX, Yang MW, Hu LP, Jiang SH, Tian GA, Zhu LL, Li Q, Sun YW, Zhang ZG. Aberrant upregulation of KLK10 promotes metastasis via enhancement of EMT and FAK/SRC/ERK axis in PDAC. Biochem Biophys Res Commun 2018; 499:584-593. [PMID: 29621546 DOI: 10.1016/j.bbrc.2018.03.194] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 03/26/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic Ductal Adenocarcinoma (PADC) metastasis is the leading cause of morality of this severe malignant tumor. Proteases are key players in the degradation of extracellular matrix which promotes the cascade of tumor metastasis. As a kind of serine proteases, the kallikrein family performs vital function on the cancer proteolysis scene, which have been proved in diverse malignant tumors. However, the specific member of kallikrein family and its function in PDAC remain unexplored. In this study, by data mining of GEO datasets, we have identified KLK10 is upregulated gene in PDAC. We found that KLK10 was significantly overexpressed in tissues of pancreatic intraepithelial neoplasia (PanIN) and PDAC from Pdx1-Cre; LSL-KrasG12D/+ mice (KC) and Pdx1-Cre; LSL-KrasG12D/+; LSL-Trp53R172H/+ mice (KPC) by immunohistochemical analysis. Moreover, KLK10 is extremely elevated in the PDAC tissues, especially that from the PDAC patients with lymphatic and distant metastasis. Aberrant KLK10 expression is significantly correlated with poor prognosis and shorter survival by univariable and multivariable analysis. Functionally, knockdown of KLK10 observably inhibits invasion and metastatic phenotype of PDAC cells in vitro and metastasis in vivo. In addition, blockade of KLK10 attenuates epithelial-mesenchymal transition and activation of FAK-SRC-ERK signaling, which explains the mechanism of KLK10 in promoting metastasis. Collectively, KLK10 should be considered as a promising biomarker for diagnosis and potential target for therapy in PDAC.
Collapse
Affiliation(s)
- Xiao-Yan Cao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Xiao-Xin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Min-Wei Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, PR China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Shu-Heng Jiang
- Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Guang-Ang Tian
- Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Li-Li Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Qing Li
- Shanghai Medical College of Fudan University, Shanghai 200032, PR China.
| | - Yong-Wei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, PR China.
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| |
Collapse
|
32
|
El Sherbini MA, Mansour AA, Sallam MM, Shaban EA, Shehab ElDin ZA, El-Shalakany AH. KLK10 exon 3 unmethylated PCR product concentration: a new potential early diagnostic marker in ovarian cancer? - A pilot study. J Ovarian Res 2018; 11:32. [PMID: 29690914 PMCID: PMC5913797 DOI: 10.1186/s13048-018-0407-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/17/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND KLK10 exon 3 hypermethylation correlated to tumor-specific lack of KLK10 expression in cancer cell lines and primary tumors. In the present study we investigate the possible role of KLK10 exon 3 methylation in ovarian tumor diagnosis and prognosis. RESULTS Qualitative methylation-specific PCR (MSP) results did not show statistically significant differences in patient group samples (normal and tumor) where all samples were positive only for the unmethylated-specific PCR except for two malignant samples that were either doubly positive (serous carcinoma) or doubly negative (Sertoli-Leydig cell tumor) for the two MSP tests. However, KLK10 exon 3 unmethylated PCR product concentration (ng/μl) showed statistically significant differences in benign and malignant patient group samples; mean ± SD (n): tumor: 0.077 ± 0.035 (14) and 0.047 ± 0.021 (15), respectively, p-value = 0.011; and normal: 0.094 ± 0.039 (7) and 0.046 ± 0.027 (6), respectively, p-value = 0.031. Moreover, ROC curve analysis of KLK10 exon 3 unmethylated PCR product concentration in overall patient group samples showed good diagnostic ability (AUC = 0.778; p-value = 0.002). Patient survival (living and died) showed statistically significant difference according to preoperative serum CA125 concentration (U/ml); median (n): 101.25 (10) and 1252 (5), respectively, p-value = 0.037, but not KLK10 exon 3 unmethylated PCR product concentration (ng/μl) in overall malignant patient samples; mean ± SD (n): 0.042 ± 0.015 (14) and 0.055 ± 0.032 (7), p-value = 0.228. CONCLUSION To the best of our knowledge, this is the first report on KLK10 exon 3 unmethylated PCR product concentration as potential early epigenetic diagnostic marker in primary ovarian tumors. Taken into account the limitations in our study (small sample size and semi-quantitative PCR product analysis) further studies are strongly recommended.
Collapse
Affiliation(s)
- Mustafa A. El Sherbini
- Medical Biochemistry Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amal A. Mansour
- Medical Biochemistry Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maha M. Sallam
- Medical Biochemistry Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Emtiaz A. Shaban
- Medical Biochemistry Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Amr H. El-Shalakany
- Gynecologic Oncology Unit, Ain Shams University Maternity Hospital, Cairo, Egypt
| |
Collapse
|
33
|
Aref-Eshghi E, Schenkel LC, Ainsworth P, Lin H, Rodenhiser DI, Cutz JC, Sadikovic B. Genomic DNA Methylation-Derived Algorithm Enables Accurate Detection of Malignant Prostate Tissues. Front Oncol 2018; 8:100. [PMID: 29740534 PMCID: PMC5925605 DOI: 10.3389/fonc.2018.00100] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/21/2018] [Indexed: 01/27/2023] Open
Abstract
Introduction The current methodology involving diagnosis of prostate cancer (PCa) relies on the pathology examination of prostate needle biopsies, a method with high false negative rates partly due to temporospatial, molecular, and morphological heterogeneity of prostate adenocarcinoma. It is postulated that molecular markers have a potential to assign diagnosis to a considerable portion of undetected prostate tumors. This study examines the genome-wide DNA methylation changes in PCa in search of genomic markers for the development of a diagnostic algorithm for PCa screening. Methods Archival PCa and normal tissues were assessed using genomic DNA methylation arrays. Differentially methylated sites and regions (DMRs) were used for functional assessment, gene-set enrichment and protein interaction analyses, and examination of transcription factor-binding patterns. Raw signal intensity data were used for identification of recurrent copy number variations (CNVs). Non-redundant fully differentiating cytosine-phosphate-guanine sites (CpGs), which did not overlap CNV segments, were used in an L1 regularized logistic regression model (LASSO) to train a classification algorithm. Validation of this algorithm was performed using a large external cohort of benign and tumor prostate arrays. Results Approximately 6,000 probes and 600 genomic regions showed significant DNA methylation changes, primarily involving hypermethylation. Gene-set enrichment and protein interaction analyses found an overrepresentation of genes related to cell communications, neurogenesis, and proliferation. Motif enrichment analysis demonstrated enrichment of tumor suppressor-binding sites nearby DMRs. Several of these regions were also found to contain copy number amplifications. Using four non-redundant fully differentiating CpGs, we trained a classification model with 100% accuracy in discriminating tumors from benign samples. Validation of this algorithm using an external cohort of 234 tumors and 92 benign samples yielded 96% sensitivity and 98% specificity. The model was found to be highly sensitive to detect metastatic lesions in bone, lymph node, and soft tissue, while being specific enough to differentiate the benign hyperplasia of prostate from tumor. Conclusion A considerable component of PCa DNA methylation profile represent driver events potentially established/maintained by disruption of tumor suppressor activity. As few as four CpGs from this profile can be used for screening of PCa.
Collapse
Affiliation(s)
- Erfan Aref-Eshghi
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada.,Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences, London, ON, Canada
| | - Laila C Schenkel
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada.,Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences, London, ON, Canada
| | - Peter Ainsworth
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada.,Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences, London, ON, Canada
| | - Hanxin Lin
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada.,Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences, London, ON, Canada
| | - David I Rodenhiser
- Department of Pediatrics, Western University and Children's Health Research Institute, London, ON, Canada.,Department of Biochemistry, Western University and Children's Health Research Institute, London, ON, Canada.,Department of Oncology, Western University and Children's Health Research Institute, London, ON, Canada
| | - Jean-Claude Cutz
- Department of Pathology and Laboratory Medicine, McMaster University, Hamilton, ON, Canada
| | - Bekim Sadikovic
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada.,Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences, London, ON, Canada
| |
Collapse
|
34
|
Clinical relevance of kallikrein-related peptidase 9, 10, 11, and 15 mRNA expression in advanced high-grade serous ovarian cancer. PLoS One 2017; 12:e0186847. [PMID: 29095848 PMCID: PMC5667830 DOI: 10.1371/journal.pone.0186847] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 10/09/2017] [Indexed: 11/19/2022] Open
Abstract
KLK9, 10, 11, and 15 may represent potential cancer biomarkers for evaluating ovarian cancer prognosis. In the present study, we selected a homogeneous cohort including 139 patients of advanced high-grade serous ovarian cancer (FIGO stage III/IV) and assessed the mRNA levels of KLK9, 10, 11, and 15 in tumor tissue by quantitative PCR. No significant associations of KLK9, 10, 11, and 15 mRNA with established clinical parameters (residual tumor mass, ascitic fluid volume) were found. Pronounced correlations between KLK10/KLK11 (rs = 0.647) and between KLK9/KLK15 (rs = 0.716) mRNA, but not between other combinations, indicate coordinate expression of distinct pairs of peptidases. In univariate Cox regression analysis, elevated KLK11 mRNA levels were significantly linked with prolonged overall survival (OS; p = 0.021) and progression-free survival (PFS; p = 0.008). KLK15 mRNA levels showed a trend towards significance in case of OS (p = 0.06); KLK9 and KLK10 mRNA expression levels were not associated with patients' outcome. In multivariable Cox analysis, KLK11 mRNA expression levels, apart from residual tumor mass, remained an independent predictive marker for OS (p = 0.007) and PFS (p = 0.015). Here, elevated KLK15 mRNA expression levels turned out to be significantly related to prolonged OS (p = 0.025) as well. High KLK11 but not the other KLK mRNA levels can be considered as strong independent favorable prognostic factor in this major ovarian cancer subtype.
Collapse
|
35
|
Debela M, Magdolen V, Bode W, Brandstetter H, Goettig P. Structural basis for the Zn2+ inhibition of the zymogen-like kallikrein-related peptidase 10. Biol Chem 2017; 397:1251-1264. [PMID: 27611765 PMCID: PMC5551965 DOI: 10.1515/hsz-2016-0205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/04/2016] [Indexed: 12/18/2022]
Abstract
Although kallikrein-related peptidase 10 (KLK10) is expressed in a variety of human tissues and body fluids, knowledge of its physiological functions is fragmentary. Similarly, the pathophysiology of KLK10 in cancer is not well understood. In some cancer types, a role as tumor suppressor has been suggested, while in others elevated expression is associated with poor patient prognosis. Active human KLK10 exhibits a unique, three residue longer N-terminus with respect to other serine proteases and an extended 99-loop nearly as long as in tissue kallikrein KLK1. Crystal structures of recombinant ligand-free KLK10 and a Zn2+ bound form explain to some extent the mixed trypsin- and chymotrypsin-like substrate specificity. Zn2+-inhibition of KLK10 appears to be based on a unique mechanism, which involves direct binding and blocking of the catalytic triad. Since the disordered N-terminus and several loops adopt a zymogen-like conformation, the active protease conformation is very likely induced by interaction with the substrate, in particular at the S1 subsite and at the unusual Ser193 as part of the oxyanion hole. The KLK10 structures indicate that the N-terminus, the nearby 75-, 148-, and the 99-loops are connected in an allosteric network, which is present in other trypsin-like serine proteases with several variations.
Collapse
Affiliation(s)
| | - Viktor Magdolen
- Klinische Forschergruppe der Frauenklinik, Klinikum rechts der Isar der TU München, Ismaninger Str. 22, D-81675 München, Germany
| | - Wolfram Bode
- Max-Planck-Institut für Biochemie, Proteinase Research Group, Am Klopferspitz 18, D-82152 Martinsried, Germany
| | - Hans Brandstetter
- Division of Structural Biology, Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, A-5020 Salzburg, Austria
| | | |
Collapse
|