1
|
Afratis NA, Riley BT, Chandler PG, Buckle AM, Sagi I. Allosteric Anti-KLK4 Antibody Development for Targeted Anti-cancer Effects in Ovarian Carcinoma. J Mol Biol 2025:169101. [PMID: 40154917 DOI: 10.1016/j.jmb.2025.169101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Kallikrein-related peptidases (KLKs) have garnered significant interest in the field of biomedical research due to their diverse roles in various physiological and pathological processes. However, the structurally conserved active site of the KLK family presents challenges for the development of specific inhibitors. Given the pro-tumorigenic roles KLKs play in various cancers, identifying new avenues for specific inhibition is therefore vital. Here, we introduce a novel approach to target a distinct KLK4 sequence by a unique immunization approach for monoclonal antibody generation, targeting loop 3, a region of high sequence and structural diversity as a candidate for allosteric control of KLK4 activity. Immunisation produced an antibody capable of interacting with both KLK4 and loop 3 with high affinity, which inhibited KLK4 proteolytic activity, and hindered proliferation and migration in ovarian cancer cell lines. Encouragingly, its potential application extends to preclinical models characterized by KLK4 overexpression. Our findings underscore the promise of this novel approach to addressing the challenges of specifically inhibiting ubiquitous serine proteases, with particular relevance to targeting KLK4, a protease instrumental in the progression of ovarian carcinoma and other cancer types.
Collapse
Affiliation(s)
- Nikolaos A Afratis
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel; Department of Agricultural Development, Agri-food & Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, Psachna 34400 Evia, Greece
| | - Blake T Riley
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Peter G Chandler
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Ashley M Buckle
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; San Diego Biomedical Research Institute, 3525 John Hopkins Ct, Suite 200, San Diego, CA 92121, USA
| | - Irit Sagi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel
| |
Collapse
|
2
|
Zheng G, Yan Z, Zou J, Zou X, Chai K, Zhang G. AR and YAP crosstalk: impacts on therapeutic strategies in prostate cancer. Front Oncol 2025; 15:1520808. [PMID: 39963114 PMCID: PMC11830605 DOI: 10.3389/fonc.2025.1520808] [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: 10/31/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Prostate cancer ranks as one of the most common types of cancer affecting men worldwide, and its progression is shaped by a diverse array of influencing factors. The AR signaling pathway plays a pivotal role in the pathogenesis of prostate cancer. While existing anti-androgen treatments show initial efficacy, they ultimately do not succeed in halting the advancement to CRPC. Recent studies have identified alterations in the Hippo-YAP signaling pathway within prostate cancer, highlighting intricate crosstalk with the AR signaling pathway. In this review, we examine the interactions and underlying mechanisms between AR and YAP, the key molecules in these two signaling pathways. AR regulates the stability and function of YAP by modulating its transcription, translation, and phosphorylation status, while YAP exerts both promotional and inhibitory regulatory effects on AR. Based on these findings, this paper investigates their significant roles in the onset, progression, and therapeutic resistance of prostate cancer, and discusses the clinical potential of YAP in prostate cancer treatment.
Collapse
Affiliation(s)
- Guansong Zheng
- First Clinical College, Gannan Medical University, Ganzhou, China
| | - Zhaojie Yan
- First Clinical College, Gannan Medical University, Ganzhou, China
| | - Junrong Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, Gannan Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, China
| | - Xiaofeng Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Keqiang Chai
- Department of Urology, Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, China
| | - Guoxi Zhang
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
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
|
Jin C, Liao S, Lu G, Geng BD, Ye Z, Xu J, Ge G, Yang D. Cellular senescence in metastatic prostate cancer: A therapeutic opportunity or challenge (Review). Mol Med Rep 2024; 30:162. [PMID: 38994760 PMCID: PMC11258599 DOI: 10.3892/mmr.2024.13286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
The treatment of patients with metastatic prostate cancer (PCa) is considered to be a long‑standing challenge. Conventional treatments for metastatic PCa, such as radical prostatectomy, radiotherapy and androgen receptor‑targeted therapy, induce senescence of PCa cells to a certain extent. While senescent cells can impede tumor growth through the restriction of cell proliferation and increasing immune clearance, the senescent microenvironment may concurrently stimulate the secretion of a senescence‑associated secretory phenotype and diminish immune cell function, which promotes PCa recurrence and metastasis. Resistance to established therapies is the primary obstacle in treating metastatic PCa as it can lead to progression towards an incurable state of disease. Therefore, understanding the molecular mechanisms that underly the progression of PCa is crucial for the development of novel therapeutic approaches. The present study reviews the phenomenon of treatment‑induced senescence in PCa, the dual role of senescence in PCa treatments and the mechanisms through which senescence promotes PCa metastasis. Furthermore, the present review discusses potential therapeutic strategies to target the aforementioned processes with the aim of providing insights into the evolving therapeutic landscape for the treatment of metastatic PCa.
Collapse
Affiliation(s)
- Cen Jin
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
- Medical Imaging School, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - Sijian Liao
- Clinical Medicine School, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - Guoliang Lu
- Department of Pediatrics, Anshun People's Hospital, Anshun, Guizhou 561000, P.R. China
| | - Bill D. Geng
- School of Natural Science, University of Texas at Austin, Austin, TX 78712, USA
| | - Zi Ye
- Clinical Medicine School, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - Jianwei Xu
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - Guo Ge
- Department of Human Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - Dan Yang
- Department of Surgery, Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| |
Collapse
|
6
|
Chou A, Sioson L, Sheen A, Ahadi MS, Gill AJ. Up-regulation of ALK is associated with altered Wnt/beta-catenin pathway in adult pancreatoblastoma. Pathology 2023; 55:129-133. [PMID: 35794050 DOI: 10.1016/j.pathol.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/21/2022] [Accepted: 04/07/2022] [Indexed: 01/24/2023]
Affiliation(s)
- Angela Chou
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia; NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia.
| | - Loretta Sioson
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Amy Sheen
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Mahsa S Ahadi
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia; NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Anthony J Gill
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia; NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia
| |
Collapse
|
7
|
SU CHENYING, HUANG GWOCHE, CHEN ICHENG, CHEN PEIYU, CHEN YUJEN, FANG HSUWEI. Distinct Expression of Surface and Genetic Biomarkers in Prostate Cancer Cell Lines. In Vivo 2023; 37:242-246. [PMID: 36593027 PMCID: PMC9843794 DOI: 10.21873/invivo.13073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND/AIM Surface biomarkers, such as CD44 and CD133, have been demonstrated to be expressed in prostate cancer cells, and our previous study has shown that prostate cancer cell lines could be divided into three groups according to the single and combined expression pattern of CD44 and 133. In order to refine prognostication in prostate cancer cells, we further investigated genetic biomarkers, prostate cancer antigen 3 (PCA3), kallikrein 4 (KLK4), and KLK9 in different prostate cancer cell lines. MATERIALS AND METHODS CWR22Rv1, PC3, and DU145 cell lines were cultured until 95% confluence. The single expression of CD44 or CD133 and their combined expression were analyzed by flow cytometry, and gene expression of b-actin, PCA3, KLK4, and KLK9 was analyzed by real-time polymerase chain reaction. RESULTS The single expression of CD133 was less than 4% in all cell lines examined. PC3 and DU145 cells displayed a high expression of CD44 (>91%), whereas CWR22Rv1 was the only cell line that demonstrated a high co-expression of both CD44 and CD133 (>91%). In addition, PC3 and DU145 displayed low expression of PCA3, KLK4, and KLK9 when compared with their own b-actin expression. In contrast, CWR22Rva showed high expression of PCA3 and KLK4 although KLK9 expression was also low. CONCLUSION Both surface and genetic biomarkers should be validated for a more accurate prognosis in prostate cancer.
Collapse
Affiliation(s)
- CHEN-YING SU
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan, R.O.C
| | - GWO-CHE HUANG
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei, Taiwan, R.O.C
| | - I-CHENG CHEN
- Accelerator for Happiness and Health Industry, National Taipei University of Technology, Taipei, Taiwan, R.O.C
| | - PEI-YU CHEN
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan, R.O.C
| | - YU-JEN CHEN
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei, Taiwan, R.O.C
| | - HSU-WEI FANG
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan, R.O.C.,Institute of Biomedical Engineering and Nanomedicine, National Health Research Institute, Zhunan, Taiwan, R.O.C
| |
Collapse
|
8
|
EPA Modulates KLK Genes via miR-378: A Potential Therapy in Prostate Cancer. Cancers (Basel) 2022; 14:cancers14112813. [PMID: 35681793 PMCID: PMC9179265 DOI: 10.3390/cancers14112813] [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: 10/30/2021] [Revised: 12/09/2021] [Accepted: 04/28/2022] [Indexed: 01/27/2023] Open
Abstract
It is known that miRNA-378a-3p (miR-378) could be induced by eicosapentaenoic acid (EPA), an omega-3 fatty acid. Herein, we first demonstrated how miR-378 exerts anti-prostate cancer (PCa) actions by influencing multiple target genes, including KLK2, KLK4, KLK6, and KLK14, which are implicated in PCa development, cell proliferation, and cell survival. Furthermore, these genes also correlate with androgen and mTOR signaling transduction, and are considered pivotal pathways for the onset and progression of PCa. In total, four PCa cell lines and eight pairing tissues (tumor vs. normal) from clinical PCa patients were included in the current study. The results showed high significance after EPA induced tumor cells containing higher expression levels of miR-378, and led the PCa cells having low cell viabilities, and they progressed to apoptosis when compared with normal prostate cells (p < 0.001). The findings indicated that EPA might become a potential therapy for PCa, especially because it is derived from the components of natural fish oil; it may prove to be a great help for solving the problem of castration-resistant prostate cancer (CRPC).
Collapse
|
9
|
Transcriptome profiling and proteomic validation reveals targets of the androgen receptor signaling in the BT-474 breast cancer cell line. Clin Proteomics 2022; 19:14. [PMID: 35568821 PMCID: PMC9107748 DOI: 10.1186/s12014-022-09352-2] [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: 10/23/2021] [Accepted: 04/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Accumulating evidence suggests that the androgen receptor (AR) and its endogenous ligands influence disease progression in breast cancer (BCa). However, AR-mediated changes in BCa differ among the various BCa subtypes according to their hormone receptor profile [i.e., presence/absence of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2, (HER2)]. Thus, we explored the androgen-regulated transcriptomic changes in the ER+PR+HER2+ BCa cell line, BT-474, and compared them with PR-mediated changes. METHODS We performed RNA sequencing analysis in treated BT-474 cells with dihydrotestosterone (DHT) and progesterone. Validation of the top ten differentially androgen-regulated genes and a number of other genes found in enriched signaling pathways was performed by qRT-PCR in BT-474 and other BCa cell lines. In addition, a parallel reaction monitoring targeted proteomic approach was developed to verify selected transcripts at the protein level. RESULTS In total 19,450 transcripts were detected, of which 224 were differentially regulated after DHT treatment. The increased expression of two well-known androgen-regulated genes, KLK2 (p < 0.05) and KLK3 (p < 0.001), confirmed the successful androgen stimulation in BT-474 cells. The transcription factor, ZBTB16, was the most highly upregulated gene, with ~ 1000-fold change (p < 0.001). Pathway enrichment analysis revealed downregulation of the DNA replication processes (p < 0.05) and upregulation of the androgen signaling and fatty acid metabolism pathways (p < 0.05). Changes related to progesterone treatment showed opposite effects in gene expression than DHT treatment. Similar expression profiles were observed among other BCa cell lines expressing high levels of AR (ZR75.1 and MBA-MB-453). The parallel reaction monitoring targeted proteomic analysis further confirmed that altered protein expression (KLK3, ALOX15B) in the supernatant and cell lysate of DHT-treated BT-474 cells, compared to control cells. DISCUSSION Our findings suggest that AR modulates the metabolism of BT-474 cells by affecting the expression of a large number of genes and proteins. Based on further pathway analysis, we suggest that androgen receptor acts as a tumor suppressor in the BT-474 cells.
Collapse
|
10
|
Srinivasan S, Kryza T, Batra J, Clements J. Remodelling of the tumour microenvironment by the kallikrein-related peptidases. Nat Rev Cancer 2022; 22:223-238. [PMID: 35102281 DOI: 10.1038/s41568-021-00436-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 02/07/2023]
Abstract
Kallikrein-related peptidases (KLKs) are critical regulators of the tumour microenvironment. KLKs are proteolytic enzymes regulating multiple functions of bioactive molecules including hormones and growth factors, membrane receptors and the extracellular matrix architecture involved in cancer progression and metastasis. Perturbations of the proteolytic cascade generated by these peptidases, and their downstream signalling actions, underlie tumour emergence or blockade of tumour growth. Recent studies have also revealed their role in tumour immune suppression and resistance to cancer therapy. Here, we present an overview of the complex biology of the KLK family and its context-dependent nature in cancer, and discuss the different therapeutic strategies available to potentially target these proteases.
Collapse
Affiliation(s)
- Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Thomas Kryza
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Mater Research Institute, The University of Queensland, Woolloongabba, Brisbane, Queensland, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Centre for Genomics and Personalised Medicine, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia.
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
11
|
Construction of Glycolytic Regulator Gene Signature to Predict the Prognosis and Tumor Immune Cell Infiltration Levels for Prostate Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9273559. [PMID: 35242214 PMCID: PMC8888065 DOI: 10.1155/2022/9273559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/17/2022] [Indexed: 12/28/2022]
Abstract
Prostate cancer (PCa) is the commonly generated noncutaneous neoplasm among men worldwide. Glycolysis had been validated to promote cancer progression. However, the clinical significance of glycolytic regulators in PCa was not well understood. Here, we discovered that glycolytic regulators were dysregulated in PCa samples using GSE8511, GSE6919, and GEPIA. By detecting the expression of these regulators in PCa samples, we found that SLC2A1, SLC2A3, HK2, PFKFB2, TPI1, PKM2, and LDHA had higher expression in PCa compared with normal tissues. Moreover, both higher expression of TPI1, ALDOA, ENO1, LDHA, and PKM and lower expression of LDHB and HK2 were significantly related to shorter progression-free survival time in PCa. Of note, an 8 gene-based risk score was further constructed and confirmed to have a good performance in predicting progression-free survival (PFS) time in PCa. The signature risk score significantly correlated with NK cell, neutrophil cell, macrophage M2 cell, and myeloid dendritic cell infiltration levels in PCa. After bioinformatics analysis, our data suggested glycolytic regulators participated in the regulation of multiple nonmetabolic biological processes, such as RNA transport, biosynthesis of antibiotics, and cell cycle. We recapitulate that the glycolytic regulator signature was a prospective indicator for prognosis and immune cell infiltration levels in PCa.
Collapse
|
12
|
Kumar A, Kasikci Y, Badredine A, Azzag K, Quintyn Ranty ML, Zaidi F, Aragou N, Mazerolles C, Malavaud B, Mendoza-Parra MA, Vandel L, Gronemeyer H. Patient-matched analysis identifies deregulated networks in prostate cancer to guide personalized therapeutic intervention. Am J Cancer Res 2021; 11:5299-5318. [PMID: 34873462 PMCID: PMC8640800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/12/2021] [Indexed: 06/13/2023] Open
Abstract
Prostate cancer (PrCa) is the second most common malignancy in men. More than 50% of advanced prostate cancers display the TMPRSS2-ERG fusion. Despite extensive cancer genome/transcriptome data, little is known about the impact of mutations and altered transcription on regulatory networks in the PrCa of individual patients. Using patient-matched normal and tumor samples, we established somatic variations and differential transcriptome profiles of primary ERG-positive prostate cancers. Integration of protein-protein interaction and gene-regulatory network databases defined highly diverse patient-specific network alterations. Different components of a given regulatory pathway were altered by novel and known mutations and/or aberrant gene expression, including deregulated ERG targets, and were validated by using a novel in silico methodology. Consequently, different sets of pathways were altered in each individual PrCa. In a given PrCa, several deregulated pathways share common factors, predicting synergistic effects on cancer progression. Our integrated analysis provides a paradigm to identify druggable key deregulated factors within regulatory networks to guide personalized therapies.
Collapse
Affiliation(s)
- Akinchan Kumar
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics and CancerIllkirch, France
- Centre National de la Recherche Scientifique, UMR7104Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258Illkirch, France
- Université de StrasbourgIllkirch, France
- Equipe Labellisée Ligue Contre le Cancer
| | - Yasenya Kasikci
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics and CancerIllkirch, France
- Centre National de la Recherche Scientifique, UMR7104Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258Illkirch, France
- Université de StrasbourgIllkirch, France
- Equipe Labellisée Ligue Contre le Cancer
| | - Alaa Badredine
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics and CancerIllkirch, France
- Centre National de la Recherche Scientifique, UMR7104Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258Illkirch, France
- Université de StrasbourgIllkirch, France
- Equipe Labellisée Ligue Contre le Cancer
- CNRS UMR8199-EGID Building, Lille University-Faculty of Medicine Henri-WarembourgLille, France
| | - Karim Azzag
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPSFrance
- Lillehei Heart Institute, Department of Medicine, University of MinnesotaMinneapolis, MN, USA
| | - Marie L Quintyn Ranty
- Institut Universitaire du Cancer Toulouse-Oncopole (IUCT-O)Toulouse, France
- Pathology Department, CHUCaen, France
| | - Falek Zaidi
- Institut Universitaire du Cancer Toulouse-Oncopole (IUCT-O)Toulouse, France
| | - Nathalie Aragou
- Institut Universitaire du Cancer Toulouse-Oncopole (IUCT-O)Toulouse, France
| | | | - Bernard Malavaud
- Institut Universitaire du Cancer Toulouse-Oncopole (IUCT-O)Toulouse, France
| | - Marco A Mendoza-Parra
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics and CancerIllkirch, France
- Centre National de la Recherche Scientifique, UMR7104Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258Illkirch, France
- Université de StrasbourgIllkirch, France
- Equipe Labellisée Ligue Contre le Cancer
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Université Evry-val-d’Essonne, University Paris-SaclayÉvry, France
| | - Laurence Vandel
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPSFrance
- Université Clermont Auvergne, CNRS, Inserm, GReDClermont-Ferrand, France
| | - Hinrich Gronemeyer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics and CancerIllkirch, France
- Centre National de la Recherche Scientifique, UMR7104Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258Illkirch, France
- Université de StrasbourgIllkirch, France
- Equipe Labellisée Ligue Contre le Cancer
| |
Collapse
|
13
|
Xie J, Kusnadi EP, Furic L, Selth LA. Regulation of mRNA Translation by Hormone Receptors in Breast and Prostate Cancer. Cancers (Basel) 2021; 13:3254. [PMID: 34209750 PMCID: PMC8268847 DOI: 10.3390/cancers13133254] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Breast and prostate cancer are the second and third leading causes of death amongst all cancer types, respectively. Pathogenesis of these malignancies is characterised by dysregulation of sex hormone signalling pathways, mediated by the estrogen receptor-α (ER) in breast cancer and androgen receptor (AR) in prostate cancer. ER and AR are transcription factors whose aberrant function drives oncogenic transcriptional programs to promote cancer growth and progression. While ER/AR are known to stimulate cell growth and survival by modulating gene transcription, emerging findings indicate that their effects in neoplasia are also mediated by dysregulation of protein synthesis (i.e., mRNA translation). This suggests that ER/AR can coordinately perturb both transcriptional and translational programs, resulting in the establishment of proteomes that promote malignancy. In this review, we will discuss relatively understudied aspects of ER and AR activity in regulating protein synthesis as well as the potential of targeting mRNA translation in breast and prostate cancer.
Collapse
Affiliation(s)
- Jianling Xie
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Eric P Kusnadi
- Translational Prostate Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Luc Furic
- Translational Prostate Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Luke A Selth
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
- Freemasons Centre for Male Health and Wellbeing, Flinders University, Bedford Park, SA 5042, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
14
|
Pällmann N, Deng K, Livgård M, Tesikova M, Jin Y, Frengen NS, Kahraman N, Mokhlis HM, Ozpolat B, Kildal W, Danielsen HE, Fazli L, Rennie PS, Banerjee PP, Üren A, Jin Y, Kuzu OF, Saatcioglu F. Stress-Mediated Reprogramming of Prostate Cancer One-Carbon Cycle Drives Disease Progression. Cancer Res 2021; 81:4066-4078. [PMID: 34183356 DOI: 10.1158/0008-5472.can-20-3956] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/01/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022]
Abstract
One-carbon (1C) metabolism has a key role in metabolic programming with both mitochondrial (m1C) and cytoplasmic (c1C) components. Here we show that activating transcription factor 4 (ATF4) exclusively activates gene expression involved in m1C, but not the c1C cycle in prostate cancer cells. This includes activation of methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) expression, the central player in the m1C cycle. Consistent with the key role of m1C cycle in prostate cancer, MTHFD2 knockdown inhibited prostate cancer cell growth, prostatosphere formation, and growth of patient-derived xenograft organoids. In addition, therapeutic silencing of MTHFD2 by systemically administered nanoliposomal siRNA profoundly inhibited tumor growth in preclinical prostate cancer mouse models. Consistently, MTHFD2 expression is significantly increased in human prostate cancer, and a gene expression signature based on the m1C cycle has significant prognostic value. Furthermore, MTHFD2 expression is coordinately regulated by ATF4 and the oncoprotein c-MYC, which has been implicated in prostate cancer. These data suggest that the m1C cycle is essential for prostate cancer progression and may serve as a novel biomarker and therapeutic target. SIGNIFICANCE: These findings demonstrate that the mitochondrial, but not cytoplasmic, one-carbon cycle has a key role in prostate cancer cell growth and survival and may serve as a biomarker and/or therapeutic target.
Collapse
Affiliation(s)
- Nora Pällmann
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Ke Deng
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Marte Livgård
- Department of Biosciences, University of Oslo, Oslo, Norway.,Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Martina Tesikova
- Department of Mathematics and Science, University of South-Eastern Norway, Borre, Norway
| | - Yixin Jin
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | - Nermin Kahraman
- Gynecological Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Hamada M Mokhlis
- Gynecological Oncology, MD Anderson Cancer Center, Houston, Texas.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Bulent Ozpolat
- Gynecological Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Wanja Kildal
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Havard Emil Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, University of Oslo, Oslo, Norway.,Department of Informatics, University of Oslo, Oslo, Norway.,Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK
| | - Ladan Fazli
- The Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Paul S Rennie
- The Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Partha P Banerjee
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Aykut Üren
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Yang Jin
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Omer F Kuzu
- Department of Biosciences, University of Oslo, Oslo, Norway.
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway. .,Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
15
|
Gong W, Zhu C, Liu Y, Muckenhuber A, Bronger H, Scorilas A, Kiechle M, Dorn J, Magdolen V, Dreyer T. Elevated levels of both microRNA 378 (miR-378) and kallikrein-related peptidase 4 (KLK4) mRNA are associated with an unfavorable prognosis in triple-negative breast cancer. Am J Transl Res 2021; 13:1594-1606. [PMID: 33841682 PMCID: PMC8014413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/08/2021] [Indexed: 06/12/2023]
Abstract
Triple-negative breast cancer (TNBC) patients have the worst outcome among all breast cancer subtypes. In oral squamous carcinoma cells, miR-378 was reported to target the mRNA of kallikrein-related peptidase 4 (KLK4), resulting in inhibition of cell proliferation, migration and invasion, induction of apoptosis, and reduction of tumor growth in vivo. Similarly, a miR-378/KLK4 axis has been proposed in prostate cancer. Here, we analyzed the correlation between miR-378 and KLK4 mRNA expression and determined the prognostic impact of both factors in TNBC. miR-378 and KLK4 mRNA expression levels were determined by quantitative PCR in tumor tissue of TNBC patients (n=103) and correlated with clinical parameters and patients' survival. There was no significant correlation between miR-378 and KLK4 mRNA expression. In univariate Cox regression analysis, elevated miR-378 expression was significantly associated with shortened disease-free survival (DFS, P=0.047) and overall survival (OS, P=0.031), high KLK4 mRNA levels were linked to a worse DFS (P=0.033). Combination of KLK4 mRNA and miR-378 (KLK4+miR-378, low/low versus high and/or high) allowed even better discrimination between favorable and unfavorable prognosis (DFS, P=0.008; OS, P=0.025). In multivariable analysis, miR-378 and KLK4+miR-378 expression remained independent predictive factors for DFS (P=0.014, P=0.010, respectively) and OS (P=0.016, P=0.049, respectively), while KLK4 mRNA only showed a trend towards significance for DFS (P=0.061). Our findings suggest that in TNBC there is no significant impact of miR-378 on KLK4 expression. Both factors, miR-378 and, to a lesser extent, KLK4 mRNA represent unfavorable prognostic markers in TNBC patients.
Collapse
Affiliation(s)
- Weiwei Gong
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of MunichGermany
- Department of Hematology-Oncology, Guangzhou Women and Children’s Medical CenterGuangzhou, People’s Republic of China
| | - Caixia Zhu
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of MunichGermany
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen UniversityGuangzhou, People’s Republic of China
| | - Yueyang Liu
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of MunichGermany
- Department of Gynecology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou, People’s Republic of China
| | | | - Holger Bronger
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of MunichGermany
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, University of AthensGreece
| | - Marion Kiechle
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of MunichGermany
| | - Julia Dorn
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of MunichGermany
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of MunichGermany
| | - Tobias Dreyer
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of MunichGermany
| |
Collapse
|
16
|
Zhang J, Yuan J, Li Z, Fu C, Xu M, Yang J, Jiang X, Zhou B, Ye X, Xu C. Exploring and exploiting plant cyclic peptides for drug discovery and development. Med Res Rev 2021; 41:3096-3117. [PMID: 33599316 DOI: 10.1002/med.21792] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 01/10/2021] [Accepted: 01/31/2021] [Indexed: 01/07/2023]
Abstract
Ever since the discovery of insulin, natural peptides have become an important resource for therapeutic development. Decades of research has led to the discovery of a long list of peptide drugs with broad applications in clinics, from antibiotics to hypertension treatment to pain management. Many of these US FDA-approved peptide drugs are derived from microorganisms and animals. By contrast, the great potential of plant cyclic peptides as therapeutics remains largely unexplored. These macrocyclic peptides typically have rigid structures, good bioavailability and membrane permeability, making them appealing candidates for drug development and engineering. In this review, we introduce the three major classes of plant cyclic peptides and summarize their potential medical applications. We discuss how we can leverage the genome information of many different plants to quickly search for new cyclic peptides and how we can take advantage of the insights gained from their biosynthetic pathways to transform the process of production and drug development. These recent developments have provided a new angle for exploring and exploiting plant cyclic peptides, and we believe that many more peptide drugs derived from plants are about to come.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.,Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, Guangdong, China
| | - Jimin Yuan
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Zhijie Li
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Chunjin Fu
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Menglong Xu
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - Jing Yang
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Xin Jiang
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Boping Zhou
- Department of Infectious Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Xiufeng Ye
- Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Chengchao Xu
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.,Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| |
Collapse
|
17
|
Tse BWC, Kryza T, Yeh MC, Dong Y, Sokolowski KA, Walpole C, Dreyer T, Felber J, Harris J, Magdolen V, Russell PJ, Clements JA. KLK4 Induces Anti-Tumor Effects in Human Xenograft Mouse Models of Orthotopic and Metastatic Prostate Cancer. Cancers (Basel) 2020; 12:cancers12123501. [PMID: 33255452 PMCID: PMC7761350 DOI: 10.3390/cancers12123501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/05/2022] Open
Abstract
Simple Summary The serine protease kallikrein-related peptidase 4 (KLK4) has been reported to potentially play a role in the progression of prostate cancer and other cancer types. However, most of these reports have been limited to in vitro studies. In vivo cancer models offer greater complexity to mimic the characteristics of cancer growth and metastasis in humans. In this study, we used in vivo models of prostate cancer and demonstrated that KLK4 can strongly inhibit the growth of primary prostate tumors as well as bone metastases. To our knowledge, this is the first report of an anti-tumor effect of KLK4 in prostate cancer in vivo. Abstract Recent reports have suggested the role of kallikrein-related peptidase 4 (KLK4) to be that of remodeling the tumor microenvironment in many cancers, including prostate cancer. Notably, these studies have suggested a pro-tumorigenic role for KLK4, especially in prostate cancer. However, these have been primarily in vitro studies, with limited in vivo studies performed to date. Herein, we employed an orthotopic inoculation xenograft model to mimic the growth of primary tumors, and an intracardiac injection to induce metastatic dissemination to determine the in vivo tumorigenic effects of KLK4 overexpressed in PC3 prostate cancer cells. Notably, we found that these KLK4-expressing cells gave rise to smaller localized tumors and decreased metastases than the parent PC-3 cells. To our knowledge, this is the first report of an anti-tumorigenic effect of KLK4, particularly in prostate cancer. These findings also provide a cautionary tale of the need for in vivo analyses to substantiate in vitro experimental data.
Collapse
Affiliation(s)
- Brian W.-C. Tse
- Preclinical Imaging Facility, Translational Research Institute, Brisbane 4102, Australia;
- Australian Prostate Cancer Research Centre—Queensland, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane 4102, Australia; (T.K.); (M.-C.Y.); (Y.D.); (C.W.); (P.J.R.); (J.A.C.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4102, Australia;
- Correspondence:
| | - Thomas Kryza
- Australian Prostate Cancer Research Centre—Queensland, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane 4102, Australia; (T.K.); (M.-C.Y.); (Y.D.); (C.W.); (P.J.R.); (J.A.C.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4102, Australia;
- Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane 4102, Australia
| | - Mei-Chun Yeh
- Australian Prostate Cancer Research Centre—Queensland, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane 4102, Australia; (T.K.); (M.-C.Y.); (Y.D.); (C.W.); (P.J.R.); (J.A.C.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4102, Australia;
| | - Ying Dong
- Australian Prostate Cancer Research Centre—Queensland, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane 4102, Australia; (T.K.); (M.-C.Y.); (Y.D.); (C.W.); (P.J.R.); (J.A.C.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4102, Australia;
| | - Kamil A. Sokolowski
- Preclinical Imaging Facility, Translational Research Institute, Brisbane 4102, Australia;
| | - Carina Walpole
- Australian Prostate Cancer Research Centre—Queensland, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane 4102, Australia; (T.K.); (M.-C.Y.); (Y.D.); (C.W.); (P.J.R.); (J.A.C.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4102, Australia;
- Translational Research Institute, Mater Research Institute—The University of Queensland, Brisbane 4102, Australia
| | - Tobias Dreyer
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany; (T.D.); (J.F.); (V.M.)
| | - Johanna Felber
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany; (T.D.); (J.F.); (V.M.)
| | - Jonathan Harris
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4102, Australia;
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany; (T.D.); (J.F.); (V.M.)
| | - Pamela J. Russell
- Australian Prostate Cancer Research Centre—Queensland, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane 4102, Australia; (T.K.); (M.-C.Y.); (Y.D.); (C.W.); (P.J.R.); (J.A.C.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4102, Australia;
| | - Judith A. Clements
- Australian Prostate Cancer Research Centre—Queensland, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane 4102, Australia; (T.K.); (M.-C.Y.); (Y.D.); (C.W.); (P.J.R.); (J.A.C.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane 4102, Australia;
| |
Collapse
|
18
|
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
|
19
|
Advances of Zinc Signaling Studies in Prostate Cancer. Int J Mol Sci 2020; 21:ijms21020667. [PMID: 31963946 PMCID: PMC7014440 DOI: 10.3390/ijms21020667] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/16/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common cancers and the second leading cause of cancer-related death among men worldwide. Despite progresses in early diagnosis and therapeutic strategies, prognosis for patients with advanced PCa remains poor. Noteworthily, a unique feature of healthy prostate is its highest level of zinc content among all soft tissues in the human body, which dramatically decreases during prostate tumorigenesis. To date, several reviews have suggested antitumor activities of zinc and its potential as a therapeutic strategy of PCa. However, an overview about the role of zinc and its signaling in PCa is needed. Here, we review literature related to the content, biological function, compounds and clinical application of zinc in PCa. We first summarize zinc content in prostate tissue and sera of PCa patients with their clinical relevance. We then elaborate biological functions of zinc signaling in PCa on three main aspects, including cell proliferation, death and tumor metastasis. Finally, we discuss clinical applications of zinc-containing compounds and proteins involved in PCa signaling pathways. Based on currently available studies, we conclude that zinc plays a tumor suppressive role and can serve as a biomarker in PCa diagnosis and therapies.
Collapse
|
20
|
Constitutively bound CTCF sites maintain 3D chromatin architecture and long-range epigenetically regulated domains. Nat Commun 2020; 11:54. [PMID: 31911579 PMCID: PMC6946690 DOI: 10.1038/s41467-019-13753-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 11/25/2019] [Indexed: 12/19/2022] Open
Abstract
The architectural protein CTCF is a mediator of chromatin conformation, but how CTCF binding to DNA is orchestrated to maintain long-range gene expression is poorly understood. Here we perform RNAi knockdown to reduce CTCF levels and reveal a shared subset of CTCF-bound sites are robustly resistant to protein depletion. The ‘persistent’ CTCF sites are enriched at domain boundaries and chromatin loops constitutive to all cell types. CRISPR-Cas9 deletion of 2 persistent CTCF sites at the boundary between a long-range epigenetically active (LREA) and silenced (LRES) region, within the Kallikrein (KLK) locus, results in concordant activation of all 8 KLK genes within the LRES region. CTCF genome-wide depletion results in alteration in Topologically Associating Domain (TAD) structure, including the merging of TADs, whereas TAD boundaries are not altered where persistent sites are maintained. We propose that the subset of essential CTCF sites are involved in cell-type constitutive, higher order chromatin architecture. The architectural protein CTCF is a mediator of chromatin conformation, but how CTCF binding to DNA is regulated remains poorly understood. Here the authors find that there is a shared subset of CTCF-bound sites resistant to protein depletion in different cell lines, which are enriched at domain boundaries and chromatin loops constitutive to all cell types.
Collapse
|
21
|
Testa U, Castelli G, Pelosi E. Cellular and Molecular Mechanisms Underlying Prostate Cancer Development: Therapeutic Implications. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E82. [PMID: 31366128 PMCID: PMC6789661 DOI: 10.3390/medicines6030082] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/19/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
Abstract
Prostate cancer is the most frequent nonskin cancer and second most common cause of cancer-related deaths in man. Prostate cancer is a clinically heterogeneous disease with many patients exhibiting an aggressive disease with progression, metastasis, and other patients showing an indolent disease with low tendency to progression. Three stages of development of human prostate tumors have been identified: intraepithelial neoplasia, adenocarcinoma androgen-dependent, and adenocarcinoma androgen-independent or castration-resistant. Advances in molecular technologies have provided a very rapid progress in our understanding of the genomic events responsible for the initial development and progression of prostate cancer. These studies have shown that prostate cancer genome displays a relatively low mutation rate compared with other cancers and few chromosomal loss or gains. The ensemble of these molecular studies has led to suggest the existence of two main molecular groups of prostate cancers: one characterized by the presence of ERG rearrangements (~50% of prostate cancers harbor recurrent gene fusions involving ETS transcription factors, fusing the 5' untranslated region of the androgen-regulated gene TMPRSS2 to nearly the coding sequence of the ETS family transcription factor ERG) and features of chemoplexy (complex gene rearrangements developing from a coordinated and simultaneous molecular event), and a second one characterized by the absence of ERG rearrangements and by the frequent mutations in the E3 ubiquitin ligase adapter SPOP and/or deletion of CDH1, a chromatin remodeling factor, and interchromosomal rearrangements and SPOP mutations are early events during prostate cancer development. During disease progression, genomic and epigenomic abnormalities accrued and converged on prostate cancer pathways, leading to a highly heterogeneous transcriptomic landscape, characterized by a hyperactive androgen receptor signaling axis.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
22
|
Pällmann N, Livgård M, Tesikova M, Zeynep Nenseth H, Akkus E, Sikkeland J, Jin Y, Koc D, Kuzu OF, Pradhan M, Danielsen HE, Kahraman N, Mokhlis HM, Ozpolat B, Banerjee PP, Uren A, Fazli L, Rennie PS, Jin Y, Saatcioglu F. Regulation of the unfolded protein response through ATF4 and FAM129A in prostate cancer. Oncogene 2019; 38:6301-6318. [DOI: 10.1038/s41388-019-0879-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/31/2019] [Accepted: 04/10/2019] [Indexed: 12/26/2022]
|
23
|
Current Transport Systems and Clinical Applications for Small Interfering RNA (siRNA) Drugs. Mol Diagn Ther 2019; 22:551-569. [PMID: 29926308 DOI: 10.1007/s40291-018-0338-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Small interfering RNAs (siRNAs) are an attractive new agent with potential as a therapeutic tool because of its ability to inhibit specific genes for many conditions, including viral infections and cancers. However, despite this potential, many challenges remain, including off-target effects, difficulties with delivery, immune responses, and toxicity. Traditional genetic vectors do not guarantee that siRNAs will silence genes in vivo. Rational design strategies, such as chemical modification, viral vectors, and non-viral vectors, including cationic liposomes, polymers, nanocarriers, and bioconjugated siRNAs, provide important opportunities to overcome these challenges. We summarize the results of research into vector delivery of siRNAs as a therapeutic agent from their design to clinical trials in ophthalmic diseases, cancers, respiratory diseases, and liver virus infections. Finally, we discuss the current state of siRNA delivery methods and the need for greater understanding of the requirements.
Collapse
|
24
|
Wang J, Li J, Xu W, Xia Q, Gu Y, Song W, Zhang X, Yang Y, Wang W, Li H, Zou K. Androgen promotes differentiation of PLZF + spermatogonia pool via indirect regulatory pattern. Cell Commun Signal 2019; 17:57. [PMID: 31142324 PMCID: PMC6542041 DOI: 10.1186/s12964-019-0369-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/14/2019] [Indexed: 12/24/2022] Open
Abstract
Background Androgen plays a pivotal role in spermatogenesis, accompanying a question how androgen acts on germ cells in testis since germ cells lack of androgen receptors (AR). Promyelocytic leukemia zinc-finger (PLZF) is essential for maintenance of undifferentiated spermatogonia population which is terminologically called spermatogonia progenitor cells (SPCs). Aims We aim to figure out the molecular connections between androgen and fates of PLZF+ SPCs population. Method Immunohistochemistry was conducted to confirm that postnatal testicular germ cells lacked endogenous AR. Subsequently, total cells were isolated from 5 dpp (day post partum) mouse testes, and dihydrotestosterone (DHT) and/or bicalutamide treatment manifested that Plzf was indirectly regulated by androgen. Then, Sertoli cells were purified to screen downstream targets of AR using ChIP-seq, and gene silence and overexpression were used to attest these interactions in Sertoli cells or SPCs-Sertoli cells co-culture system. Finally, these connections were further verified in vivo using androgen pharmacological deprivation mouse model. Results Gata2 is identified as a target of AR, and β1-integrin is a target of Wilms’ tumor 1 (WT1) in Sertoli cells. Androgen signal negatively regulate β1-integrin on Sertoli cells via Gata2 and WT1, and β1-integrin on Sertoli cells interacts with E-cadherin on SPCs to regulate SPCs fates. Conclusion Androgen promotes differentiation of PLZF+ spermatogonia pool via indirect regulatory pattern. Electronic supplementary material The online version of this article (10.1186/s12964-019-0369-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jingjing Wang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Weigang NO.1, Xuanwu District, Nanjing, 210095, China
| | - Jinmei Li
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Weigang NO.1, Xuanwu District, Nanjing, 210095, China
| | - Wei Xu
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Xia
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Weigang NO.1, Xuanwu District, Nanjing, 210095, China
| | - Yunzhao Gu
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Weixiang Song
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Weigang NO.1, Xuanwu District, Nanjing, 210095, China
| | - Xiaoyu Zhang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Weigang NO.1, Xuanwu District, Nanjing, 210095, China
| | - Yang Yang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Weigang NO.1, Xuanwu District, Nanjing, 210095, China
| | - Wei Wang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Weigang NO.1, Xuanwu District, Nanjing, 210095, China.,National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Hua Li
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Weigang NO.1, Xuanwu District, Nanjing, 210095, China.
| |
Collapse
|
25
|
Matin F, Jeet V, Srinivasan S, Cristino AS, Panchadsaram J, Clements JA, Batra J. MicroRNA-3162-5p-Mediated Crosstalk between Kallikrein Family Members Including Prostate-Specific Antigen in Prostate Cancer. Clin Chem 2019; 65:771-780. [PMID: 31018918 DOI: 10.1373/clinchem.2018.295824] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 02/05/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND MicroRNAs mediate biological processes through preferential binding to the 3' untranslated region (3' UTR) of target genes. Studies have shown their association with prostate cancer (PCa) risk through single-nucleotide polymorphisms (SNPs), known as miRSNPs. In a European cohort, 22 PCa risk-associated miRSNPs have been identified. The most significant miRSNP in the 3' UTR of Kallikrein-related peptidase 3 (KLK3) created a binding site for miR-3162-5p. Here we investigated the miR-3162-5p-KLK interaction and the clinical implication of miR-3162-5p in PCa. METHODS We tested the role of miR-3162-5p in PCa etiology using IncuCyte live-cell imaging and anchorage-independent growth assays. The effect of miR-3162-5p on KLK and androgen receptor (AR) expression was measured by RT-quantitative (q)PCR and target pulldown assays. KLK3 proteolytic activity was determined by DELFIA® immunoassay. Mass spectrometry identified pathways affected by miR-3162-5p. miR-3162-5p expression was measured in clinical samples using RT-qPCR. RESULTS miR-3162-5p affected proliferation, migration, and colony formation of LNCaP cells by regulating the expression of KLK2-4 and AR by direct targeting. KLK3 protein expression was regulated by miR-3162-5p consistent with lower KLK3 proteolytic activity observed in LNCaP-conditioned media. KLK/AR pulldown and mass spectrometry analysis showed a potential role of miR-3162-5p in metabolic pathways via KLK/AR and additional targets. Increased miR-3162-5p expression was observed in prostate tumor tissues with higher Gleason grade. CONCLUSIONS Our study provides an insight into possible involvement of miR-3162-5p in PCa etiology by targeting KLKs and AR. It highlights clinical utility of miR-3162-5p and its interactive axis as a new class of biomarkers and therapeutic targets for PCa.
Collapse
Affiliation(s)
- Farhana Matin
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Brisbane, Australia
| | - Varinder Jeet
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Brisbane, Australia
| | - Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Brisbane, Australia
| | - Alexandre S Cristino
- University of Queensland Diamantina Institute (UQDI), Faculty of Medicine, Translational Research Institute, University of Queensland, Brisbane, Australia
| | - Janaththani Panchadsaram
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Brisbane, Australia
| | | | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Brisbane, Australia;
| | | |
Collapse
|
26
|
Stopsack KH, Gerke T, Tyekucheva S, Mazzu YZ, Lee GSM, Chakraborty G, Abida W, Mucci LA, Kantoff PW. Low Expression of the Androgen-Induced Tumor Suppressor Gene PLZF and Lethal Prostate Cancer. Cancer Epidemiol Biomarkers Prev 2019; 28:707-714. [PMID: 30602500 PMCID: PMC6532645 DOI: 10.1158/1055-9965.epi-18-1014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/19/2018] [Accepted: 12/26/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND 4%-9% of prostate cancers harbor homozygous deletions of the androgen-induced tumor suppressor gene, promyelocytic leukemia zinc finger (PLZF, ZBTB16). PLZF loss induces an in vitro phenotype of castration resistance and enzalutamide resistance. The association of low expression of PLZF and clinical outcomes is unclear. METHODS We assessed PLZF mRNA expression in patients diagnosed with primary prostate cancer during prospective follow-up of the Health Professionals Follow-up Study (HPFS; n = 254) and the Physicians' Health Study (PHS; n = 150), as well as in The Cancer Genome Atlas (n = 333). We measured PTEN status (using copy numbers and IHC) and transcriptional activation of the MAPK pathway. Patients from HPFS and PHS were followed for metastases and prostate cancer-specific mortality (median, 15.3 years; 113 lethal events). RESULTS PLZF mRNA expression was lower in tumors with PLZF deletions. There was a strong, positive association between intratumoral androgen receptor (AR) signaling and PLZF expression. PLZF expression was also lower in tumors with PTEN loss. Low PLZF expression was associated with higher MAPK signaling. Patients in the lowest quartile of PLZF expression compared with those in the highest quartile were more likely to develop lethal prostate cancer, independent of clinicopathologic features, Gleason score, and AR signaling (odds ratio, 3.17; 95% confidence interval, 1.32-7.60). CONCLUSIONS Low expression of the tumor suppressor gene PLZF is associated with a worse prognosis in primary prostate cancer. IMPACT Suppression of PLZF as a consequence of androgen deprivation may be undesirable. PLZF should be tested as a predictive marker for resistance to androgen deprivation therapy.
Collapse
Affiliation(s)
- Konrad H Stopsack
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Travis Gerke
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida
| | - Svitlana Tyekucheva
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ying Z Mazzu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gwo-Shu Mary Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Goutam Chakraborty
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Wassim Abida
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
27
|
Swedberg JE, Ghani HA, Harris JM, de Veer SJ, Craik DJ. Potent, Selective, and Cell-Penetrating Inhibitors of Kallikrein-Related Peptidase 4 Based on the Cyclic Peptide MCoTI-II. ACS Med Chem Lett 2018; 9:1258-1262. [PMID: 30613336 DOI: 10.1021/acsmedchemlett.8b00422] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022] Open
Abstract
Kallikrein-related peptidase 4 (KLK4) is a serine protease that has putative intracellular and extracellular functions in prostate cancer progression. Here we show that MCoTI-II, a 34-amino acid cyclic peptide found in the seeds of red gac (Momordica cochinchinensis), is an inhibitor of KLK4. By grafting a preferred KLK4 cleavage sequence into MCoTI-II, we produced a highly potent KLK4 inhibitor (K i = 0.1 nM) that displayed 100,000-fold selectivity over related KLKs and the ability to penetrate cells. Additionally, by substituting positively charged noncontact residues in this compound, we produced a potent and selective KLK4 inhibitor that does not penetrate cells. The inhibitors were shown to be nontoxic to human cells and stable in human serum. These KLK4 inhibitors provide useful chemical tools to further define the role(s) of both intracellular and extracellular KLK4 in prostate cancer cell lines and disease models.
Collapse
Affiliation(s)
- Joakim E. Swedberg
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Hafiza Abdul Ghani
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jonathan M. Harris
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Simon J. de Veer
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - David J. Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
28
|
Zhu S, Shi J, Zhang S, Li Z. KLK6 Promotes Growth, Migration, and Invasion of Gastric Cancer Cells. J Gastric Cancer 2018; 18:356-367. [PMID: 30607299 PMCID: PMC6310766 DOI: 10.5230/jgc.2018.18.e35] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/02/2018] [Accepted: 11/04/2018] [Indexed: 11/20/2022] Open
Abstract
Purpose Kallikrein (KLK) proteases are hormone-like signaling molecules with critical functions in different cancers. This study investigated the expression of KLK6 in gastric cancer and its potential role in the growth, migration, and invasion of gastric cancer cells. Materials and Methods In this study, we compared protein levels of KLK6, vascular endothelial growth factor (VEGF), and matrix metallopeptidase (MMP) 9 in normal gastric epithelial and gastric cancer cell lines by western blot. Fluorescence-activated cell sorting was employed to sort 2 clones of SGC-7901 cells with distinct KLK6 expression, namely, KLK6-high (KLK6high) and KLK6-low (KLK6low), which were then expanded. Lastly, immunohistochemical analysis was performed to investigate KLK6 expression in gastric cancer patients. Results The expression levels of KLK6, VEGF, and MMP 9, were significantly higher in the gastric cancer cell lines SGC-7901, BGC-823, MKN-28, and MGC-803 than in the normal gastric epithelial cell line GES-1. Compared to KLK6low cells, KLK6high cells showed enhanced viability, colony-forming ability, migration, and invasion potential in vitro. Importantly, immunohistochemical analysis of a human gastric cancer tissue cohort revealed that the staining for KLK6, VEGF, and MMP9 was markedly stronger in the cancerous tissues than in the adjacent normal tissues. KLK6 expression also correlated with that of VEGF and MMP9 expression, as well as several key clinicopathological parameters. Conclusions Together, these results suggest an important role for KLK6 in human gastric cancer progression.
Collapse
Affiliation(s)
- Shengxing Zhu
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of The Second General Surgery, People's Hospital of Zhengzhou, Zhengzhou, China
| | - Jihua Shi
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanfeng Zhang
- School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Zhen Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
29
|
Du JP, Li L, Zheng J, Zhang D, Liu W, Zheng WH, Li XS, Yao RC, Wang F, Liu S, Tan X. Kallikrein-related peptidase 7 is a potential target for the treatment of pancreatic cancer. Oncotarget 2018; 9:12894-12906. [PMID: 29560118 PMCID: PMC5849182 DOI: 10.18632/oncotarget.24132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/01/2017] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer is one of the deadliest cancers with very poor prognosis, and the five-year survival rate of the patients is less than 5% after diagnosis. Kallikrein-related peptidases (KLKs) belong to a serine protease family with 15 members that play important roles in cellular physiological behavior and diseases. The high expression level of KLK7 in pancreatic cancer tissues is considered to be a marker for the poor prognosis of this disease. In this work, we set out to investigate whether KLK7 could be a target for the treatment of pancreatic cancer. Short hairpin RNAs (shRNAs) were designed and constructed in lentivirus to knock down KLK7 in pancreatic cancer cell line PANC-1, and the real time cellular analysis (RTCA) was used to evaluate cell proliferation, migration and invasion abilities. Small molecules inhibiting KLK7 were discovered by computer-aided drug screening and used to inhibit PANC-1 cells. Our results confirmed that KLK7 is significantly up-regulated in pancreatic cancer tissue, and knocking down or inhibiting KLK7 efficiently inhibited the proliferation, migration and invasion of pancreatic cancer cells. This study suggested that KLK7 could be a potential chemotherapy target for treatment of pancreatic cancer, which would provide us a novel strategy for the treatment of this disease.
Collapse
Affiliation(s)
- Jian Ping Du
- Institute of Hepatopancreatobilary Surgery, China Three Gorges University, Yichang 443003, P.R. China
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, P.R. China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, Medical School of China Three Gorges University, Yichang 443002, P.R. China
- Department of Vascular surgery, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, 437100, P.R. China
| | - Lin Li
- Institute of Hepatopancreatobilary Surgery, China Three Gorges University, Yichang 443003, P.R. China
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, P.R. China
| | - Jun Zheng
- Institute of Hepatopancreatobilary Surgery, China Three Gorges University, Yichang 443003, P.R. China
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, P.R. China
| | - Ding Zhang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, P.R. China
| | - Wei Liu
- Institute of Hepatopancreatobilary Surgery, China Three Gorges University, Yichang 443003, P.R. China
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, P.R. China
| | - Wei Hong Zheng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, Medical School of China Three Gorges University, Yichang 443002, P.R. China
| | - Xiao Song Li
- Institute of Hepatopancreatobilary Surgery, China Three Gorges University, Yichang 443003, P.R. China
| | - Ru Cheng Yao
- Institute of Hepatopancreatobilary Surgery, China Three Gorges University, Yichang 443003, P.R. China
| | - Fangyu Wang
- College of Life Science and Environment, Hengyang Normal University, Hengyang, 421008, P.R. China
| | - Sen Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, Medical School of China Three Gorges University, Yichang 443002, P.R. China
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan 430068, P.R. China
| | - Xiao Tan
- Institute of Hepatopancreatobilary Surgery, China Three Gorges University, Yichang 443003, P.R. China
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443003, P.R. China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, Medical School of China Three Gorges University, Yichang 443002, P.R. China
| |
Collapse
|
30
|
Mirkheshti N, Park S, Jiang S, Cropper J, Werner SL, Song CS, Chatterjee B. Dual targeting of androgen receptor and mTORC1 by salinomycin in prostate cancer. Oncotarget 2018; 7:62240-62254. [PMID: 27557496 PMCID: PMC5308723 DOI: 10.18632/oncotarget.11404] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/09/2016] [Indexed: 12/25/2022] Open
Abstract
Androgen receptor (AR) and PI3K/AKT/mTORC1 are major survival signals that drive prostate cancer to a lethal disease. Reciprocal activation of these oncogenic pathways from negative cross talks contributes to low/limited success of pathway-selective inhibitors in curbing prostate cancer progression. We report that the antibiotic salinomycin, a cancer stem cell blocker, is a dual-acting AR and mTORC1 inhibitor, inhibiting PTEN-deficient castration-sensitive and castration-resistant prostate cancer in culture and xenograft tumors. AR expression, its transcriptional activity, and androgen biosynthesis regulating enzymes CYP17A1, HSD3β1 were reduced by sub-micro molar salinomycin. Estrogen receptor-α expression was unchanged. Loss of phosphorylated AR at serine-81, which is an index for nuclear AR activity, preceded total AR reduction. Rapamycin enhanced the AR protein level without altering phosphoAR-Ser81 and CYP17A1. Inactivation of mTORC1, evident from reduced phosphorylation of mTOR and downstream effectors, as well as AMPK activation led to robust autophagy induction. Apoptosis increased modestly, albeit significantly, by sub-micro molar salinomycin. Enhanced stimulatory TSC2 phosphorylation at Ser-1387 by AMPK, and reduced inhibitory TSC2 phosphorylation at Ser-939/Thr-1462 catalyzed by AKT augmented TSC2/TSC1 activity, which led to mTORC1 inhibition. AMPK-mediated raptor phosphorylation further reduced mTOR's kinase function and mTORC1 activity. Our novel finding on dual inhibition of AR and mTORC1 suggests that salinomycin is potentially active as monotherapy against advanced prostate cancer.
Collapse
Affiliation(s)
- Nooshin Mirkheshti
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
| | - Sulgi Park
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
| | - Shoulei Jiang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
| | - Jodie Cropper
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
| | - Sherry L Werner
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
| | - Chung S Song
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA
| | - Bandana Chatterjee
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78245, USA.,South Texas Veterans Health Care System, San Antonio, Texas 78229, USA
| |
Collapse
|
31
|
Shen H, Zhan M, Zhang Y, Huang S, Xu S, Huang X, He M, Yao Y, Man M, Wang J. PLZF inhibits proliferation and metastasis of gallbladder cancer by regulating IFIT2. Cell Death Dis 2018; 9:71. [PMID: 29358655 PMCID: PMC5833736 DOI: 10.1038/s41419-017-0107-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 02/06/2023]
Abstract
Gallbladder cancer (GBC) is a malignant cancer with very poor prognosis. Although promyelocytic leukemia zinc-finger protein (PLZF) was reported to be deregulated in numerous cancers and also relevant to clinical prognosis, its role in GBC progression has been little known. In this study, we found PLZF expression was decreased in GBC, correlating to advanced TNM stage, distant metastasis, and shorter overall survival. Moreover, ectopic PLZF expression in GBC cells (NOZ and GBC-SD) significantly reduced the cell proliferation, migration, and invasion. Consistently, overexpression of PLZF in xenograft mice model could suppress tumor growth and liver metastasis. Mechanical investigations verified PLZF could regulate the expression of cell cycle arrest-associated gene p21 and epithelial-mesenchymal transition (EMT)-related genes (E-cadherin and N-cadherin) in GBC cell lines. Importantly, PLZF remarkably increased the mRNA transcription of interferon-induced protein with tetratricopeptide repeat 2 (IFIT2) by increasing STAT1 protein level, a known factor involved in tumor progression. Furthermore, ablation of IFIT2 in PLZF overexpression cells abrogated the tumor-suppressive function of PLZF, at least partially, leading to impaired tumor growth and EMT program. These studies indicated PLZF inhibited the proliferation and metastasis via regulation of IFIT2. In conclusion, our study demonstrated PLZF could be a promising tumor biomarker for GBC, and also be a potential therapeutic target.
Collapse
Affiliation(s)
- Hui Shen
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Ming Zhan
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Yonglong Zhang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Shuai Huang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Sunwang Xu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Xince Huang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Min He
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Yanhua Yao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Institutes of Medical Sciences, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Mohan Man
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Institutes of Medical Sciences, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Jian Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China.
| |
Collapse
|
32
|
Zhu SP, Wang JY, Wang XG, Zhao JP. Long intergenic non-protein coding RNA 00858 functions as a competing endogenous RNA for miR-422a to facilitate the cell growth in non-small cell lung cancer. Aging (Albany NY) 2017; 9:475-486. [PMID: 28177876 PMCID: PMC5361675 DOI: 10.18632/aging.101171] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 01/31/2017] [Indexed: 01/17/2023]
Abstract
The expression of long non-coding RNAs (lncRNAs) is dysregulated in non-small cell lung cancer (NSCLC). However, the functions and contributions of lncRNAs remain largely unknown. Here, we identified a critical role of long intergenic non-protein coding RNA 00858 (LINC00858) in NSCLC. Ectopic expression of LINC00858 in NSCLC cells promoted cell proliferation and induced cell migration and invasion. Moreover, LINC00858 functioned as a competitive endogenous RNA (ceRNA), effectively becoming sponge for miR-422a and thereby modulating the expression of kallikrein-related peptidase 4 (KLK4). In NSCLC patients, high expression of LINC00858 closely correlated with tumor progression. Thus, targeting the ceRNA network involving LINC00858 may be used as a treatment strategy against NSCLC.
Collapse
Affiliation(s)
- Shao-Ping Zhu
- Department of Cardiothoracic Surgery, ZhongNan Hospital of Wuhan University, 430071 Wuhan, P. R. China
| | - Jun-Yu Wang
- Department of Oncology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, 430015 Wuhan, P. R. China
| | - Xian-Guo Wang
- Department of Cardiothoracic Surgery, ZhongNan Hospital of Wuhan University, 430071 Wuhan, P. R. China
| | - Jin-Ping Zhao
- Department of Cardiothoracic Surgery, ZhongNan Hospital of Wuhan University, 430071 Wuhan, P. R. China
| |
Collapse
|
33
|
Luo W, Tan P, Rodriguez M, He L, Tan K, Zeng L, Siwko S, Liu M. Leucine-rich repeat-containing G protein-coupled receptor 4 (Lgr4) is necessary for prostate cancer metastasis via epithelial-mesenchymal transition. J Biol Chem 2017; 292:15525-15537. [PMID: 28768769 DOI: 10.1074/jbc.m116.771931] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 08/01/2017] [Indexed: 01/01/2023] Open
Abstract
Prostate cancer is a highly penetrant disease among men in industrialized societies, but the factors regulating the transition from indolent to aggressive and metastatic cancer remain poorly understood. We found that men with prostate cancers expressing high levels of the G protein-coupled receptor LGR4 had a significantly shorter recurrence-free survival compared with patients with cancers having low LGR4 expression. LGR4 expression was elevated in human prostate cancer cell lines with metastatic potential. We therefore generated a novel transgenic adenocarcinoma of the mouse prostate (TRAMP) mouse model to investigate the role of Lgr4 in prostate cancer development and metastasis in vivo TRAMP Lgr4-/- mice exhibited an initial delay in prostate intraepithelial neoplasia formation, but the frequency of tumor formation was equivalent between TRAMP and TRAMP Lgr4-/- mice by 12 weeks. The loss of Lgr4 significantly improved TRAMP mouse survival and dramatically reduced the occurrence of lung metastases. LGR4 knockdown impaired the migration, invasion, and colony formation of DU145 cells and reversed epithelial-mesenchymal transition (EMT), as demonstrated by up-regulation of E-cadherin and decreased expression of the EMT transcription factors ZEB, Twist, and Snail. Overexpression of LGR4 in LNCaP cells had the opposite effects. Orthotopic injection of DU145 cells stably expressing shRNA targeting LGR4 resulted in decreased xenograft tumor size, reduced tumor EMT marker expression, and impaired metastasis, in accord with our findings in TRAMP Lgr4-/- mice. In conclusion, we propose that Lgr4 is a key protein necessary for prostate cancer EMT and metastasis.
Collapse
Affiliation(s)
- Weijia Luo
- From the Center for Translational Cancer Research, Institute of Bioscience and Technology, Department of Molecular and Cellular Medicine, Texas A&M University System Health Science Center, Houston, Texas 77030 and
| | - Peng Tan
- From the Center for Translational Cancer Research, Institute of Bioscience and Technology, Department of Molecular and Cellular Medicine, Texas A&M University System Health Science Center, Houston, Texas 77030 and.,the Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Melissa Rodriguez
- From the Center for Translational Cancer Research, Institute of Bioscience and Technology, Department of Molecular and Cellular Medicine, Texas A&M University System Health Science Center, Houston, Texas 77030 and
| | - Lian He
- From the Center for Translational Cancer Research, Institute of Bioscience and Technology, Department of Molecular and Cellular Medicine, Texas A&M University System Health Science Center, Houston, Texas 77030 and
| | - Kunrong Tan
- From the Center for Translational Cancer Research, Institute of Bioscience and Technology, Department of Molecular and Cellular Medicine, Texas A&M University System Health Science Center, Houston, Texas 77030 and
| | - Li Zeng
- the Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Stefan Siwko
- From the Center for Translational Cancer Research, Institute of Bioscience and Technology, Department of Molecular and Cellular Medicine, Texas A&M University System Health Science Center, Houston, Texas 77030 and
| | - Mingyao Liu
- From the Center for Translational Cancer Research, Institute of Bioscience and Technology, Department of Molecular and Cellular Medicine, Texas A&M University System Health Science Center, Houston, Texas 77030 and .,the Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
34
|
Jin Y, Nenseth HZ, Saatcioglu F. Role of PLZF as a tumor suppressor in prostate cancer. Oncotarget 2017; 8:71317-71324. [PMID: 29050363 PMCID: PMC5642638 DOI: 10.18632/oncotarget.19813] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/12/2017] [Indexed: 01/07/2023] Open
Abstract
The promyelocytic leukemia zinc finger (PLZF), also known as ZBTB16 (Zinc Finger And BTB Domain Containing 16), is a transcription factor involved in the regulation of diverse biological processes, including cell proliferation, differentiation, organ development, stem cell maintenance and innate immune cell development. A number of recent studies have now implicated PLZF in cancer progression as a tumor suppressor. However, in certain cancer types, PLZF may function as an oncoprotein. Here, we summarize our current knowledge on the role of PLZF in various cancer types, in particular prostate cancer, including its deregulation, genomic alterations and potential functions in prostate cancer progression.
Collapse
Affiliation(s)
- Yang Jin
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | | | - Fahri Saatcioglu
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
35
|
Cui Z, Cui Y, Yang S, Luo G, Wang Y, Lou Y, Sun X. KLK4 silencing inhibits the growth of oral squamous cell carcinoma through Wnt/β-catenin signaling pathway. Cell Biol Int 2017; 41:392-404. [PMID: 28150891 DOI: 10.1002/cbin.10736] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 01/28/2017] [Indexed: 12/17/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is a malignancy that largely impacts the quality of people's daily life. Kallikrein-related peptidase 4 (KLK4) is highly expressed in OSCC; however, its roles in OSCC cells are unclear. In the present study, the effect of KLK4 silencing on the growth of OSCC cells was investigated. Our study showed that the proliferation and colony formation of OSCC cells was inhibited by KLK4 silencing and their cell cycle was arrested. Additionally, apoptosis of OSCC cells was enhanced by KLK4 silencing, with increased protein levels of cleaved PARP, cleaved caspase-3, Bax and decreased levels of Bcl-2. KLK4 silencing inhibited the Wnt/β-catenin signaling pathway, as evidence by decreased protein levels of Wnt1, β-catenin, reduced GSK-3β phosphorylation as well as decreased levels of cyclinD1 and c-myc proteins. We further showed that Wnt/β-catenin activator reversed the effects of KLK4 silencing on the proliferation and apoptosis of OSCC cells. We concluded that KLK4 silencing inhibited the growth of OSCC cells through Wnt/β-catenin signaling pathway, suggesting that KLK4 may become a promising therapeutic target for the treatment of OSCC.
Collapse
Affiliation(s)
- Zhi Cui
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Ye Cui
- Department of Orthodontics, School of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Shuting Yang
- Department of Prosthodontics, School of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Gan Luo
- Department of Orthodontics, School of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Yang Wang
- Department of Orthodontics, School of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Yixin Lou
- Department of Orthodontics, School of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Xinhua Sun
- Department of Orthodontics, School of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| |
Collapse
|
36
|
Messina M. Soy and Health Update: Evaluation of the Clinical and Epidemiologic Literature. Nutrients 2016; 8:E754. [PMID: 27886135 PMCID: PMC5188409 DOI: 10.3390/nu8120754] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/17/2016] [Accepted: 11/18/2016] [Indexed: 01/10/2023] Open
Abstract
Soyfoods have long been recognized as sources of high-quality protein and healthful fat, but over the past 25 years these foods have been rigorously investigated for their role in chronic disease prevention and treatment. There is evidence, for example, that they reduce risk of coronary heart disease and breast and prostate cancer. In addition, soy alleviates hot flashes and may favorably affect renal function, alleviate depressive symptoms and improve skin health. Much of the focus on soyfoods is because they are uniquely-rich sources of isoflavones. Isoflavones are classified as both phytoestrogens and selective estrogen receptor modulators. Despite the many proposed benefits, the presence of isoflavones has led to concerns that soy may exert untoward effects in some individuals. However, these concerns are based primarily on animal studies, whereas the human research supports the safety and benefits of soyfoods. In support of safety is the recent conclusion of the European Food Safety Authority that isoflavones do not adversely affect the breast, thyroid or uterus of postmenopausal women. This review covers each of the major research areas involving soy focusing primarily on the clinical and epidemiologic research. Background information on Asian soy intake, isoflavones, and nutrient content is also provided.
Collapse
Affiliation(s)
- Mark Messina
- Nutrition Matters, Inc., 26 Spadina Parkway, Pittsfield, MA 01201, USA.
| |
Collapse
|
37
|
Riley BT, Ilyichova O, Costa MGS, Porebski BT, de Veer SJ, Swedberg JE, Kass I, Harris JM, Hoke DE, Buckle AM. Direct and indirect mechanisms of KLK4 inhibition revealed by structure and dynamics. Sci Rep 2016; 6:35385. [PMID: 27767076 PMCID: PMC5073354 DOI: 10.1038/srep35385] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 09/28/2016] [Indexed: 11/09/2022] Open
Abstract
The kallikrein-related peptidase (KLK) family of proteases is involved in many aspects of human health and disease. One member of this family, KLK4, has been implicated in cancer development and metastasis. Understanding mechanisms of inactivation are critical to developing selective KLK4 inhibitors. We have determined the X-ray crystal structures of KLK4 in complex with both sunflower trypsin inhibitor-1 (SFTI-1) and a rationally designed SFTI-1 derivative to atomic (~1 Å) resolution, as well as with bound nickel. These structures offer a structural rationalization for the potency and selectivity of these inhibitors, and together with MD simulation and computational analysis, reveal a dynamic pathway between the metal binding exosite and the active site, providing key details of a previously proposed allosteric mode of inhibition. Collectively, this work provides insight into both direct and indirect mechanisms of inhibition for KLK4 that have broad implications for the enzymology of the serine protease superfamily, and may potentially be exploited for the design of therapeutic inhibitors.
Collapse
Affiliation(s)
- Blake T Riley
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Olga Ilyichova
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Mauricio G S Costa
- Programa de Computação Científica, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Benjamin T Porebski
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Simon J de Veer
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4059, Australia
| | - Joakim E Swedberg
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Itamar Kass
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Jonathan M Harris
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4059, Australia
| | - David E Hoke
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Ashley M Buckle
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
38
|
Lee SJ, Kim MJ, Kwon IC, Roberts TM. Delivery strategies and potential targets for siRNA in major cancer types. Adv Drug Deliv Rev 2016; 104:2-15. [PMID: 27259398 DOI: 10.1016/j.addr.2016.05.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 02/24/2016] [Accepted: 05/15/2016] [Indexed: 02/08/2023]
Abstract
Small interfering RNA (siRNA) has gained attention as a potential therapeutic reagent due to its ability to inhibit specific genes in many genetic diseases. For many years, studies of siRNA have progressively advanced toward novel treatment strategies against cancer. Cancer is caused by various mutations in hundreds of genes including both proto-oncogenes and tumor suppressor genes. In order to develop siRNAs as therapeutic agents for cancer treatment, delivery strategies for siRNA must be carefully designed and potential gene targets carefully selected for optimal anti-cancer effects. In this review, various modifications and delivery strategies for siRNA delivery are discussed. In addition, we present current thinking on target gene selection in major tumor types.
Collapse
|
39
|
Filippou PS, Karagiannis GS, Musrap N, Diamandis EP. Kallikrein-related peptidases (KLKs) and the hallmarks of cancer. Crit Rev Clin Lab Sci 2016; 53:277-91. [PMID: 26886390 DOI: 10.3109/10408363.2016.1154643] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The kallikrein-related peptidases (KLKs) represent the largest family of serine proteases within the human genome and are expressed in various tissues. Although they regulate several important physiological functions, KLKs have also been implicated in numerous pathophysiological processes, including cancer. Growing evidence describing the deregulation of KLK expression and secretion, as well as activation in various malignancies, has uncovered their potential as mediators of cancer progression, biomarkers of disease and as candidate therapeutic targets. The diversity of signalling pathways and proteolytic cascades involving KLKs and their downstream targets appears to affect cancer biology through multiple mechanisms, including those related to the hallmarks of cancer. The aim of this review is to provide an update on the importance of KLK-driven molecular pathways in relation to cancer cell traits associated with the hallmarks of cancer and to highlight their potential in personalized therapeutics.
Collapse
Affiliation(s)
- Panagiota S Filippou
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada
| | - George S Karagiannis
- b Department of Anatomy & Structural Biology , Albert Einstein College of Medicine, Yeshiva University Bronx , New York , NY , USA
| | - Natasha Musrap
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada
| | - Eleftherios P Diamandis
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada .,c Department of Clinical Biochemistry , University Health Network , Toronto , ON , Canada , and.,d Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
40
|
Avgeris M, Scorilas A. Kallikrein-related peptidases (KLKs) as emerging therapeutic targets: focus on prostate cancer and skin pathologies. Expert Opin Ther Targets 2016; 20:801-18. [PMID: 26941073 DOI: 10.1517/14728222.2016.1147560] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Tissue kallikrein and the kallikrein-related peptidases (KLKs) constitute a family of 15 homologous secreted serine proteases with trypsin- or chymotrypsin-like activities, which participate in a broad spectrum of physiological procedures. Deregulated expression and/or activation of the majority of the family members have been reported in several human diseases, thereby making KLKs ideal targets for therapeutic intervention. AREAS COVERED In the present review, we summarize the role of KLKs in normal human physiology and pathology, focusing on prostate cancer and skin diseases. Furthermore, we discuss the recent advances in the development of KLK-based therapies. A great number of diverse engineered KLKs inhibitors with improved potency, selectivity and immunogenicity have been synthesized by redesigning examples that are endogenous and naturally occurring. Moreover, encouraging results have been documented using KLKs-based vaccines and immunotherapies, as well as KLKs-mediated activation of pro-drugs. Finally, KLKs-targeting aptamers and KLKs-based imaging tools represent novel approaches towards the exploitation of KLKs' therapeutic value. EXPERT OPINION The central/critical roles of KLK family in several human pathologies highlight KLKs as attractive molecular targets for developing novel therapeutics.
Collapse
Affiliation(s)
- Margaritis Avgeris
- a Department of Biochemistry and Molecular Biology, Faculty of Biology , University of Athens , Athens , Greece
| | - Andreas Scorilas
- a Department of Biochemistry and Molecular Biology, Faculty of Biology , University of Athens , Athens , Greece
| |
Collapse
|
41
|
Sheng X, Arnoldussen YJ, Storm M, Tesikova M, Nenseth HZ, Zhao S, Fazli L, Rennie P, Risberg B, Wæhre H, Danielsen H, Mills IG, Jin Y, Hotamisligil G, Saatcioglu F. Divergent androgen regulation of unfolded protein response pathways drives prostate cancer. EMBO Mol Med 2016; 7:788-801. [PMID: 25864123 PMCID: PMC4459818 DOI: 10.15252/emmm.201404509] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The unfolded protein response (UPR) is a homeostatic mechanism to maintain endoplasmic reticulum (ER) function. The UPR is activated by various physiological conditions as well as in disease states, such as cancer. As androgens regulate secretion and development of the normal prostate and drive prostate cancer (PCa) growth, they may affect UPR pathways. Here, we show that the canonical UPR pathways are directly and divergently regulated by androgens in PCa cells, through the androgen receptor (AR), which is critical for PCa survival. AR bound to gene regulatory sites and activated the IRE1α branch, but simultaneously inhibited PERK signaling. Inhibition of the IRE1α arm profoundly reduced PCa cell growth in vitro as well as tumor formation in preclinical models of PCa in vivo. Consistently, AR and UPR gene expression were correlated in human PCa, and spliced XBP-1 expression was significantly upregulated in cancer compared with normal prostate. These data establish a genetic switch orchestrated by AR that divergently regulates the UPR pathways and suggest that targeting IRE1α signaling may have therapeutic utility in PCa.
Collapse
Affiliation(s)
- Xia Sheng
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | | | | | | | - Sen Zhao
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Ladan Fazli
- The Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Paul Rennie
- The Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Bjørn Risberg
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway Division of Pathology, Oslo University Hospital, Oslo, Norway
| | - Håkon Wæhre
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway Division of Surgery, Oslo University Hospital, Oslo, Norway Center for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Håvard Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway Center for Cancer Biomedicine, University of Oslo, Oslo, Norway Department of Informatics, University of Oslo, Oslo, Norway
| | - Ian G Mills
- The Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway Department of Urology, Oslo University Hospital, Oslo, Norway Department of Cancer Prevention, Institute of Cancer Research, Radium Hospital, Oslo, Norway
| | - Yang Jin
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Gökhan Hotamisligil
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Harvard University, Boston, MA, USA
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
42
|
Involvement of Kallikrein-Related Peptidases in Normal and Pathologic Processes. DISEASE MARKERS 2015; 2015:946572. [PMID: 26783378 PMCID: PMC4689925 DOI: 10.1155/2015/946572] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/16/2015] [Accepted: 10/29/2015] [Indexed: 12/31/2022]
Abstract
Human kallikrein-related peptidases (KLKs) are a subgroup of serine proteases that participate in proteolytic pathways and control protein levels in normal physiology as well as in several pathological conditions. Their complex network of stimulatory and inhibitory interactions may induce inflammatory and immune responses and contribute to the neoplastic phenotype through the regulation of several cellular processes, such as proliferation, survival, migration, and invasion. This family of proteases, which includes one of the most useful cancer biomarkers, kallikrein-related peptidase 3 or PSA, also has a protective effect against cancer promoting apoptosis or counteracting angiogenesis and cell proliferation. Therefore, they represent attractive therapeutic targets and may have important applications in clinical oncology. Despite being intensively studied, many gaps in our knowledge on several molecular aspects of KLK functions still exist. This review aims to summarize recent data on their involvement in different processes related to health and disease, in particular those directly or indirectly linked to the neoplastic process.
Collapse
|
43
|
Jin Y, Wang L, Qu S, Sheng X, Kristian A, Mælandsmo GM, Pällmann N, Yuca E, Tekedereli I, Gorgulu K, Alpay N, Sood A, Lopez-Berestein G, Fazli L, Rennie P, Risberg B, Wæhre H, Danielsen HE, Ozpolat B, Saatcioglu F. STAMP2 increases oxidative stress and is critical for prostate cancer. EMBO Mol Med 2015; 7:315-31. [PMID: 25680860 PMCID: PMC4364948 DOI: 10.15252/emmm.201404181] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The six transmembrane protein of prostate 2 (STAMP2) is an androgen-regulated gene whose mRNA expression is increased in prostate cancer (PCa). Here, we show that STAMP2 protein expression is increased in human PCa compared with benign prostate that is also correlated with tumor grade and treatment response. We also show that STAMP2 significantly increased reactive oxygen species (ROS) in PCa cells through its iron reductase activity which also depleted NADPH levels. Knockdown of STAMP2 expression in PCa cells inhibited proliferation, colony formation, and anchorage-independent growth, and significantly increased apoptosis. Furthermore, STAMP2 effects were, at least in part, mediated by activating transcription factor 4 (ATF4), whose expression is regulated by ROS. Consistent with in vitro findings, silencing STAMP2 significantly inhibited PCa xenograft growth in mice. Finally, therapeutic silencing of STAMP2 by systemically administered nanoliposomal siRNA profoundly inhibited tumor growth in two established preclinical PCa models in mice. These data suggest that STAMP2 is required for PCa progression and thus may serve as a novel therapeutic target.
Collapse
Affiliation(s)
- Yang Jin
- Department of Biosciences, University of Oslo, Oslo, Norway Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Ling Wang
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Su Qu
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Xia Sheng
- Department of Biosciences, University of Oslo, Oslo, Norway Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | | | | | - Nora Pällmann
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Erkan Yuca
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | - Ibrahim Tekedereli
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | - Kivanc Gorgulu
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | - Neslihan Alpay
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | - Anil Sood
- Gynecological Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Ladan Fazli
- The Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Paul Rennie
- The Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Bjørn Risberg
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway Division of Pathology, Oslo University Hospital, Oslo, Norway Division of Surgery, Oslo University Hospital, Oslo, Norway
| | - Håkon Wæhre
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway Division of Pathology, Oslo University Hospital, Oslo, Norway Division of Surgery, Oslo University Hospital, Oslo, Norway Center for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Håvard E Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway Center for Cancer Biomedicine, University of Oslo, Oslo, Norway Department of Informatics, University of Oslo, Oslo, Norway
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
44
|
The kallikrein-related peptidase family: Dysregulation and functions during cancer progression. Biochimie 2015; 122:283-99. [PMID: 26343558 DOI: 10.1016/j.biochi.2015.09.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/01/2015] [Indexed: 01/07/2023]
Abstract
Cancer is the second leading cause of death with 14 million new cases and 8.2 million cancer-related deaths worldwide in 2012. Despite the progress made in cancer therapies, neoplastic diseases are still a major therapeutic challenge notably because of intra- and inter-malignant tumour heterogeneity and adaptation/escape of malignant cells to/from treatment. New targeted therapies need to be developed to improve our medical arsenal and counter-act cancer progression. Human kallikrein-related peptidases (KLKs) are secreted serine peptidases which are aberrantly expressed in many cancers and have great potential in developing targeted therapies. The potential of KLKs as cancer biomarkers is well established since the demonstration of the association between KLK3/PSA (prostate specific antigen) levels and prostate cancer progression. In addition, a constantly increasing number of in vitro and in vivo studies demonstrate the functional involvement of KLKs in cancer-related processes. These peptidases are now considered key players in the regulation of cancer cell growth, migration, invasion, chemo-resistance, and importantly, in mediating interactions between cancer cells and other cell populations found in the tumour microenvironment to facilitate cancer progression. These functional roles of KLKs in a cancer context further highlight their potential in designing new anti-cancer approaches. In this review, we comprehensively review the biochemical features of KLKs, their functional roles in carcinogenesis, followed by the latest developments and the successful utility of KLK-based therapeutics in counteracting cancer progression.
Collapse
|
45
|
mTOR and its tight regulation for iNKT cell development and effector function. Mol Immunol 2015; 68:536-45. [PMID: 26253278 DOI: 10.1016/j.molimm.2015.07.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/09/2015] [Accepted: 07/19/2015] [Indexed: 12/26/2022]
Abstract
Invariant NKT (iNKT) cells, which express the invariant Vα14Jα18 TCR that recognizes lipid antigens, have the ability to rapidly respond to agonist stimulation, producing a variety of cytokines that can shape both innate and adaptive immunity. iNKT cells have been implicated in host defense against microbial infection, in anti-tumor immunity, and a multitude of diseases such as allergies, asthma, graft versus host disease, and obesity. Emerging evidence has demonstrated crucial role for mammalian target of rapamycin (mTOR) in immune cells, including iNKT. In this review we will discuss current understanding of how mTOR and its tight regulation control iNKT cell development, effector lineage differentiation, and function.
Collapse
|
46
|
Cereda V, Formica V, Menghi A, Pellicori S, Roselli M. Kallikrein-related peptidases targeted therapies in prostate cancer: perspectives and challenges. Expert Opin Investig Drugs 2015; 24:929-47. [PMID: 25858813 DOI: 10.1517/13543784.2015.1035708] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Despite the emergence of several new effective treatments for metastatic castration-resistant prostate cancer patients, disease progression inevitably occurs, leading scientific community to carefully look for novel therapeutic targets of prostate cancer. Kallikrein (KLK)-related peptidases have been demonstrated to facilitate prostate tumorigenesis and disease progression through the development of an oncogenic microenvironment for prostate cells. AREAS COVERED This review first summarizes the large amount of preclinical data showing the involvement of KLKs in prostate cancer pathobiology. In the second part, the authors assess the current status and future directions for KLK-targeted therapy and briefly describe the advances and challenges implicated in the design of effective manufactured drugs. The authors then focus on the preclinical data and on Phase I/II studies of the most promising KLK-targeted agents in prostate cancer. The drugs discussed here are divided on the basis of their mechanism of action: KLK-engineered inhibitors; KLK-activated pro-drugs; KLK-targeted microRNAs and small interfering RNAs(-/)small hairpin RNAs; KLK vaccines and antibodies. EXPERT OPINION Targeting KLK expression and/or activity could be a promising direction in prostate cancer treatment. Future human clinical trials will help us to evaluate the real benefits, toxicities and the consequent optimal use of KLK-targeted drugs, as mono-therapy or in combination regimens.
Collapse
Affiliation(s)
- Vittore Cereda
- 1 University of Rome Tor Vergata, Tor Vergata University Clinical Center, Department of Systems Medicine, Medical Oncology , Viale Oxford 81, 00133 Rome , Italy +39 0620908190 ; +39 0620903504 ;
| | | | | | | | | |
Collapse
|
47
|
Papagerakis P, Pannone G, Zheng LI, Athanassiou-Papaefthymiou M, Yamakoshi Y, McGuff HS, Shkeir O, Ghirtis K, Papagerakis S. Clinical significance of kallikrein-related peptidase-4 in oral cancer. Anticancer Res 2015; 35:1861-1866. [PMID: 25862839 PMCID: PMC4577232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Kallikrein-related-peptidase-4 (KLK4), a serine protease originally discovered in developing tooth with broad target sequence specificity, serves vital functions in dental enamel formation. KLK4 is involved in degradation of extracellular matrix proteins and it is thought that this proteolytic activity could also promote tumor invasion and metastasis. Recent studies have associated KLK4 expression with tumor progression and clinical outcome, particularly in prostate and ovarian cancer. Very little is known in regard KLK4 involvement in oral squamous cell carcinomas (OSCCs). Our objective was to investigate KLK4 expression in OSCC pathogenesis and disease progression. KLK4 expression was evaluated by immunohistochemistry, western blots and zymograms in OSCC lines. Invasion assays using high versus low/undetectable KLK4-expressing OSCC cell lines were performed jointly with KLK4 siRNA inhibition. A large collection of OSCC specimens was evaluated for KLK4 expression and correlation with patients' characteristics and outcomes were determined. Our data indicate that KLK4 is differentially expressed in oral carcinomas. OSCC cell lines with high invasive and metastatic potential have the highest levels of KLK4 expression. KLK4 mRNA and protein expression correlated with enzyme activity detected by zymograms. Inhibition of KLK4 expression results in diminished invasive potential in OSCC cell lines. Consistently, KLK4 expression is stronger in primary tumors that later either recurred or developed metastases, suggesting that its preferential expression in OSCC might contribute to individual tumor biology. Therefore, this study provides supportive evidence in favor of a prognostic value for KLK4 in OSCC and suggests that KLK4 could serve as a potential therapeutic target in patients with oral cancer.
Collapse
Affiliation(s)
- Petros Papagerakis
- Department of Pediatric Dentistry and Orthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, U.S.A. Department of Computational Medicine and Bioinformatics, Medical School, University of Michigan, Ann Arbor, MI, U.S.A
| | - Giuseppe Pannone
- Department of Clinical and Experimental Medicine, Section of Anatomic Pathology, University of Foggia, Foggia, Italy
| | - L I Zheng
- Department of Pediatric Dentistry and Orthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, U.S.A. Department of Otolaryngology-Head & Neck Surgery, Medical School, University of Michigan, Ann Arbor, MI, U.S.A
| | - Maria Athanassiou-Papaefthymiou
- Department of Pediatric Dentistry and Orthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, U.S.A. Department of Otolaryngology-Head & Neck Surgery, Medical School, University of Michigan, Ann Arbor, MI, U.S.A
| | - Yashuo Yamakoshi
- Department of Biomaterials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, U.S.A
| | - Howard Stan McGuff
- Department of Pathology, School of Medicine, University of Texas Health Science Center, San Antonio, TX, U.S.A
| | - Omar Shkeir
- Department of Otolaryngology-Head & Neck Surgery, Medical School, University of Michigan, Ann Arbor, MI, U.S.A
| | - Konstantinos Ghirtis
- Department of Pediatric Dentistry and Orthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, U.S.A. Department of Otolaryngology-Head & Neck Surgery, Medical School, University of Michigan, Ann Arbor, MI, U.S.A
| | - Silvana Papagerakis
- Department of Otolaryngology-Head & Neck Surgery, Medical School, University of Michigan, Ann Arbor, MI, U.S.A.
| |
Collapse
|
48
|
Prassas I, Eissa A, Poda G, Diamandis EP. Unleashing the therapeutic potential of human kallikrein-related serine proteases. Nat Rev Drug Discov 2015; 14:183-202. [DOI: 10.1038/nrd4534] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
49
|
van Soom J, Cuzzucoli Crucitti G, Gladysz R, van der Veken P, Di Santo R, Stuyver I, Buck V, Lambeir AM, Magdolen V, Joossens J, Augustyns K. The first potent diphenyl phosphonate KLK4 inhibitors with unexpected binding kinetics. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00288e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We report the first highly potent and selective small-molecule KLK4 inhibitors, showing surprising reversible binding kinetics.
Collapse
|
50
|
Fuhrman-Luck RA, Loessner D, Clements JA. Kallikrein-Related Peptidases in Prostate Cancer: From Molecular Function to Clinical Application. EJIFCC 2014; 25:269-81. [PMID: 27683474 PMCID: PMC4975200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Prostate cancer is a leading contributor to male cancer-related deaths worldwide. Kallikrein-related peptidases (KLKs) are serine proteases that exhibit deregulated expression in prostate cancer, with KLK3, or prostate specific antigen (PSA), being the widely-employed clinical biomarker for prostate cancer. Other KLKs, such as KLK2, show promise as prostate cancer biomarkers and, additionally, their altered expression has been utilised for the design of KLK-targeted therapies. There is also a large body of in vitro and in vivo evidence supporting their role in cancer-related processes. Here, we review the literature on studies to date investigating the potential of other KLKs, in addition to PSA, as biomarkers and in therapeutic options, as well as their current known functional roles in cancer progression. Increased knowledge of these KLK-mediated functions, including degradation of the extracellular matrix, local invasion, cancer cell proliferation, interactions with fibroblasts, angiogenesis, migration, bone metastasis and tumour growth in vivo, may help define new roles as prognostic biomarkers and novel therapeutic targets for this cancer.
Collapse
Affiliation(s)
| | | | - Judith A. Clements
- Cancer Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|