1
|
Shi J, Qi Y, Sun Y, Huang Y. Kallikrein-Related Peptidase 4 Promotes Proliferation, Migration, Invasion, and Pro-Angiogenesis of Endometrial Stromal Cells via Regulation of Brain-Derived Neurotrophic Factor Production in Endometriosis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:121-134. [PMID: 37918799 DOI: 10.1016/j.ajpath.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 11/04/2023]
Abstract
Endometriosis is a common benign gynecologic condition. Endometriosis lesions are associated with endometrial cell proliferation, migration, invasion, and neovascularization, while the specific molecular mechanisms are still elusive. Transcriptome sequencing has been used for the identification of diagnostic markers in endometriosis. Here, transcriptome profiling revealed that kallikrein-related peptidase 4 (KLK4) expression was up-regulated in ectopic endometrium (EC) tissues of patients with endometriosis. KLK4 mediates the degradation of extracellular matrix proteins, and its proteolytic activity activates many tumorigenic and metastatic pathways via tumor invasion and migration. Nevertheless, whether KLK4 serves as an important regulatory factor in endometriosis remains unclear. This study confirmed that KLK4 was highly expressed in ectopic endometrial stromal cells (EC-ESCs). KLK4 overexpression promoted proliferation and suppressed apoptosis of EC-ESCs, induced cell migration and invasion, and enhanced angiogenesis in vivo. Mechanistically, KLK4 overexpression mediated the protein cleavage of pro-brain-derived neurotrophic factor in EC-ESCs. Finally, brain-derived neurotrophic factor was a vital downstream substrate of KLK4 maintained the proliferation, metastasis, and pro-angiogenesis abilities and inhibited apoptosis of ESCs through a rescue study. Together, these findings demonstrate the promotive role of KLK4 in endometriosis development. In addition, the study provides a new insight that KLK4 might be a potential therapeutic target and prognostic marker for patients with endometriosis.
Collapse
Affiliation(s)
- Jingwen Shi
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yue Qi
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yuanchen Sun
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Ying Huang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
2
|
Shukla N, Kour B, Sharma D, Vijayvargiya M, Sadasukhi TC, Medicherla KM, Malik B, Bissa B, Vuree S, Lohiya NK, Suravajhala P. Towards Understanding the Key Signature Pathways Associated from Differentially Expressed Gene Analysis in an Indian Prostate Cancer Cohort. Diseases 2023; 11:diseases11020072. [PMID: 37218885 DOI: 10.3390/diseases11020072] [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: 02/20/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent cancers among men in India. Although studies on PCa have dealt with genetics, genomics, and the environmental influence in the causality of PCa, not many studies employing the Next Generation Sequencing (NGS) approaches of PCa have been carried out. In our previous study, we identified some causal genes and mutations specific to Indian PCa using Whole Exome Sequencing (WES). In the recent past, with the help of different cancer consortiums such as The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC), along with differentially expressed genes (DEGs), many cancer-associated novel non-coding RNAs have been identified as biomarkers. In this work, we attempt to identify differentially expressed genes (DEGs) including long non-coding RNAs (lncRNAs) associated with signature pathways from an Indian PCa cohort using the RNA-sequencing (RNA-seq) approach. From a cohort of 60, we screened six patients who underwent prostatectomy; we performed whole transcriptome shotgun sequencing (WTSS)/RNA-sequencing to decipher the DEGs. We further normalized the read counts using fragments per kilobase of transcript per million mapped reads (FPKM) and analyzed the DEGs using a cohort of downstream regulatory tools, viz., GeneMANIA, Stringdb, Cytoscape-Cytohubba, and cbioportal, to map the inherent signatures associated with PCa. By comparing the RNA-seq data obtained from the pairs of normal and PCa tissue samples using our benchmarked in-house cuffdiff pipeline, we observed some important genes specific to PCa, such as STEAP2, APP, PMEPA1, PABPC1, NFE2L2, and HN1L, and some other important genes known to be involved in different cancer pathways, such as COL6A1, DOK5, STX6, BCAS1, BACE1, BACE2, LMOD1, SNX9, CTNND1, etc. We also identified a few novel lncRNAs such as LINC01440, SOX2OT, ENSG00000232855, ENSG00000287903, and ENST00000647843.1 that need to be characterized further. In comparison with publicly available datasets, we have identified characteristic DEGs and novel lncRNAs implicated in signature PCa pathways in an Indian PCa cohort which perhaps have not been reported. This has set a precedent for us to validate candidates further experimentally, and we firmly believe this will pave a way toward the discovery of biomarkers and the development of novel therapies.
Collapse
Affiliation(s)
- Nidhi Shukla
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research (BISR), Statue Circle, Jaipur 302001, India
- Department of Chemistry, School of Basic Sciences, Manipal University Jaipur, Jaipur 303007, India
| | - Bhumandeep Kour
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144001, India
| | - Devendra Sharma
- Department of Urology, Rukmani Birla Hospital, Jaipur 302018, India
| | - Maneesh Vijayvargiya
- Department of Pathology, Mahatma Gandhi University of Medical Sciences and Technology, Jaipur 302022, India
| | - T C Sadasukhi
- Department of Urology, Mahatma Gandhi University of Medical Sciences and Technology, Jaipur 302022, India
| | - Krishna Mohan Medicherla
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research (BISR), Statue Circle, Jaipur 302001, India
- Department of Bioengineering, Birla Institute of Technology, Mesra Jaipur Campus, 27-Malaviya Industrial Area, Jaipur 302017, India
| | - Babita Malik
- Department of Chemistry, School of Basic Sciences, Manipal University Jaipur, Jaipur 303007, India
| | - Bhawana Bissa
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India
| | - Sugunakar Vuree
- Bioclues.org, Hyderabad 500072, India
- MNR Foundation for Research & Innovation, MNR University, Sangareddy 502294, India
| | - Nirmal Kumar Lohiya
- Department of Zoology, Center for Advanced Studies, University of Rajasthan, Jaipur 302004, India
| | - Prashanth Suravajhala
- Bioclues.org, Hyderabad 500072, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, India
| |
Collapse
|
3
|
Charytonowicz D, Brody R, Sebra R. Interpretable and context-free deconvolution of multi-scale whole transcriptomic data with UniCell deconvolve. Nat Commun 2023; 14:1350. [PMID: 36906603 PMCID: PMC10008582 DOI: 10.1038/s41467-023-36961-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 02/27/2023] [Indexed: 03/13/2023] Open
Abstract
We introduce UniCell: Deconvolve Base (UCDBase), a pre-trained, interpretable, deep learning model to deconvolve cell type fractions and predict cell identity across Spatial, bulk-RNA-Seq, and scRNA-Seq datasets without contextualized reference data. UCD is trained on 10 million pseudo-mixtures from a fully-integrated scRNA-Seq training database comprising over 28 million annotated single cells spanning 840 unique cell types from 898 studies. We show that our UCDBase and transfer-learning models achieve comparable or superior performance on in-silico mixture deconvolution to existing, reference-based, state-of-the-art methods. Feature attribute analysis uncovers gene signatures associated with cell-type specific inflammatory-fibrotic responses in ischemic kidney injury, discerns cancer subtypes, and accurately deconvolves tumor microenvironments. UCD identifies pathologic changes in cell fractions among bulk-RNA-Seq data for several disease states. Applied to lung cancer scRNA-Seq data, UCD annotates and distinguishes normal from cancerous cells. Overall, UCD enhances transcriptomic data analysis, aiding in assessment of cellular and spatial context.
Collapse
Affiliation(s)
- Daniel Charytonowicz
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Brody
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Genomics Institute, New York, NY, USA.
- Black Family Stem Cell Institute, New York, NY, USA.
| |
Collapse
|
4
|
An B, Cai Y, Zhu J, Liu Y. Long Noncoding RNA LINC01503 Silencing Suppresses KLK4 Expression to Impede Pancreatic Cancer Development as miR-1321 Sponge. BIOMED RESEARCH INTERNATIONAL 2023; 2023:5403344. [PMID: 36785666 PMCID: PMC9922183 DOI: 10.1155/2023/5403344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 02/05/2023]
Abstract
Background Long intergenic nonprotein coding RNA 1503 (LINC01503) was reportedly oncogenic in several malignancies, whereas whether it contributed to pancreatic cancer tumorigenesis and progression requires to be verified. Methods The expression pattern of LINC01503 was monitored via qRT-PCR assay in normal cells and cancerous pancreatic cancer cells. The introduction of silencing LINC01503 was to verify the relation between LINC01503 expression and cell growth. Then, the targeting relationship of LINC01503 to miR-1321 was confirmed by bioinformatics predication and luciferase reporter assay. In addition, luciferase reporter assays evaluated the binding of miR-1321 to the 3'-untranslated region of KLK4. Overexpressing KLK4 and inhibiting LINC01503 was introduced in tumor cells to investigate the corresponding impacts on pancreatic cancer cell proliferation and migration. Results LINC01503 and KLK4 were highly abundant in pancreatic cancer cells. Mechanistically, miR-1321 bound to LINC01503 and KLK4. Downregulating LINC01503 promoted the availability of miR-1321 in pancreatic cancer cells and thus repressed KLK4 expression. KLK4 overexpression abolished the impediment of LINC01503 depletion on cell proliferation and migration. Conclusion Oncogenic function of LINC01503 was dependent on KLK4 upregulation by sponging miR-1321. Revealing the tumor-promoting property of LINC01503 in pancreatic cancer may confer new biomarkers for this malignancy.
Collapse
Affiliation(s)
- Baiping An
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu City 610032, China
| | - Yi Cai
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu City 610032, China
| | - Jie Zhu
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu City 610032, China
| | - Yuan Liu
- Department of Interventional Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu City 610032, China
| |
Collapse
|
5
|
Calls A, Torres‐Espin A, Tormo M, Martínez‐Escardó L, Bonet N, Casals F, Navarro X, Yuste VJ, Udina E, Bruna J. A transient inflammatory response contributes to oxaliplatin neurotoxicity in mice. Ann Clin Transl Neurol 2022; 9:1985-1998. [PMID: 36369764 PMCID: PMC9735376 DOI: 10.1002/acn3.51691] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/14/2022] [Accepted: 10/16/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Peripheral neuropathy is a relevant dose-limiting adverse event that can affect up to 90% of oncologic patients with colorectal cancer receiving oxaliplatin treatment. The severity of neurotoxicity often leads to dose reduction or even premature cessation of chemotherapy. Unfortunately, the limited knowledge about the molecular mechanisms related to oxaliplatin neurotoxicity leads to a lack of effective treatments to prevent the development of this clinical condition. In this context, the present work aimed to determine the exact molecular mechanisms involved in the development of oxaliplatin neurotoxicity in a murine model to try to find new therapeutical targets. METHODS By single-cell RNA sequencing (scRNA-seq), we studied the transcriptomic profile of sensory neurons and satellite glial cells (SGC) of the Dorsal Root Ganglia (DRG) from a well-characterized mouse model of oxaliplatin neurotoxicity. RESULTS Analysis of scRNA-seq data pointed to modulation of inflammatory processes in response to oxaliplatin treatment. In this line, we observed increased levels of NF-kB p65 protein, pro-inflammatory cytokines, and immune cell infiltration in DRGs and peripheral nerves of oxaliplatin-treated mice, which was accompanied by mechanical allodynia and decrease in sensory nerve amplitudes. INTERPRETATION Our data show that, in addition to the well-described DNA damage, oxaliplatin neurotoxicity is related to an exacerbated pro-inflammatory response in DRG and peripheral nerves, and open new insights in the development of anti-inflammatory strategies as a treatment for preventing peripheral neuropathy induced by oxaliplatin.
Collapse
Affiliation(s)
- Aina Calls
- Department of Cell Biology, Physiology, and Immunology, Institute of NeuroscienceUniversitat Autònoma de BarcelonaBellaterraSpain,Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Abel Torres‐Espin
- Department of Neurological Surgery, Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Marc Tormo
- Genomics Core Facility, Departament de Ciències Experimentals i de la SalutUniversitat Pompeu Fabra, Parc de Recerca Biomèdica de BarcelonaBarcelonaSpain,Scientific IT Core Facility, Departament de Ciències Experimentals i de la SalutUniversitat Pompeu Fabra, Parc de Recerca Biomèdica de BarcelonaBarcelonaSpain
| | - Laura Martínez‐Escardó
- Department of Biochemistry, Institute of NeuroscienceUniversitat Autònoma de BarcelonaBellaterraSpain
| | - Núria Bonet
- Genomics Core Facility, Departament de Ciències Experimentals i de la SalutUniversitat Pompeu Fabra, Parc de Recerca Biomèdica de BarcelonaBarcelonaSpain
| | - Ferran Casals
- Genomics Core Facility, Departament de Ciències Experimentals i de la SalutUniversitat Pompeu Fabra, Parc de Recerca Biomèdica de BarcelonaBarcelonaSpain,Departament de Genètica, Microbiologia i Estadística, Facultat de BiologiaUniversitat de BarcelonaBarcelonaSpain
| | - Xavier Navarro
- Department of Cell Biology, Physiology, and Immunology, Institute of NeuroscienceUniversitat Autònoma de BarcelonaBellaterraSpain,Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Víctor J. Yuste
- Department of Biochemistry, Institute of NeuroscienceUniversitat Autònoma de BarcelonaBellaterraSpain
| | - Esther Udina
- Department of Cell Biology, Physiology, and Immunology, Institute of NeuroscienceUniversitat Autònoma de BarcelonaBellaterraSpain,Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Jordi Bruna
- Department of Cell Biology, Physiology, and Immunology, Institute of NeuroscienceUniversitat Autònoma de BarcelonaBellaterraSpain,Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain,Unit of Neuro‐Oncology, Hospital Universitari de BellvitgeBellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de LlobregatBarcelonaSpain
| |
Collapse
|
6
|
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
|
7
|
KLK4T2 Is a Hormonally Regulated Transcript from the KLK4 Locus. Int J Mol Sci 2021; 22:ijms222313023. [PMID: 34884832 PMCID: PMC8657796 DOI: 10.3390/ijms222313023] [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: 10/20/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
The human kallikrein-related peptidase 4 (KLK4) and the transcribed pseudogene KLKP1 are reported to be highly expressed in the prostate. When trying to clone transcripts of KLKP1, we partly failed. Instead, we identified an androgen-regulated transcript, KLK4T2, which appeared to be a splice variant of KLK4 that also contained exons of KLKP1. Expression analysis of KLK4, KLK4T2, and KLKP1 transcripts in prostate cancer cell lines showed high levels of KLKP1 transcripts in the nucleus and in unfractionated cell extract, whereas it was almost completely absent in the cytoplasmatic fraction. This was in contrast to KLK4 and KLK4T2, which displayed high to moderate levels in the cytoplasm. In patient cohorts we found significantly higher expression of both KLK4T2 and KLK4 in benign prostatic hyperplasia compared to both primary prostate cancer and bone metastasis. Analysis of tissue panels demonstrated the highest expression of KLK4T2 in the prostate, but in contrast to the classical KLK4, relatively high levels were also found in placenta. So far, the function of KLK4T2 is still to be explored, but the structure of the translation product indicated that it generates a 17.4 kDa intracellular protein with possible regulatory function.
Collapse
|
8
|
Pihlstrøm N, Jin Y, Nenseth Z, Kuzu OF, Saatcioglu F. STAMP2 Expression Mediated by Cytokines Attenuates Their Growth-Limiting Effects in Prostate Cancer Cells. Cancers (Basel) 2021; 13:cancers13071579. [PMID: 33808059 PMCID: PMC8036285 DOI: 10.3390/cancers13071579] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 01/22/2023] Open
Abstract
Simple Summary Prostate cancer (PCa) is the most common non-skin cancer and one of the leading causes of cancer death in men. Despite significant developments in therapy options with improved survival, no curative treatment is currently available. We have previously identified six transmembrane protein of prostate 2 (STAMP2) as an important factor for PCa growth and survival. We now show that STAMP2 expression is regulated by inflammatory signaling, which has recently been implicated in PCa. Two proinflammatory cytokines, interleukin 6 and interleukin 1 beta, synergize with each other to induce STAMP2 expression. Interestingly, STAMP2 knockdown increased the sensitivity of PCa cells to cytokine treatment. Thus, STAMP2 that acts as a survival factor in PCa, is both independently and synergistically regulated by inflammatory signaling that may affect disease progression. Abstract Inflammatory events and dysregulated cytokine expression are implicated in prostate cancer (PCa), but the underlying molecular mechanisms are poorly understood at present. We have previously identified six transmembrane protein of the prostate 2 (STAMP2, also known as STEAP4) as an androgen-regulated gene, as well as a key regulator of PCa growth and survival. STAMP2 is also regulated by, and participates in, inflammatory signaling in other tissues and pathologies. Here, we show that the proinflammatory cytokines interleukin 6 (IL-6) and Interleukin 1 beta (IL-1β) significantly increase and strongly synergize in promoting STAMP2 expression in PCa cells. The two cytokines increase androgen-induced STAMP2 expression, but not expression of other known androgen target genes, suggesting a unique interplay of androgens and cytokines in regulating STAMP2 expression. Interestingly, STAMP2 knockdown significantly increased the ability of IL-6 and IL-1β to inhibit PCa cell growth in vitro. These results suggest that STAMP2 may represent a unique node through which inflammatory events mediate their effects on PCa growth and survival.
Collapse
Affiliation(s)
- Nicklas Pihlstrøm
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
| | - Yang Jin
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
| | - Zeynep Nenseth
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
| | - Omer F. Kuzu
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
- Correspondence: (O.F.K.); (F.S.); Tel.: +47-22-854-569 (F.S.); Fax: +47-22-857-207 (F.S.)
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0188 Oslo, Norway
- Correspondence: (O.F.K.); (F.S.); Tel.: +47-22-854-569 (F.S.); Fax: +47-22-857-207 (F.S.)
| |
Collapse
|
9
|
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: 2] [Impact Index Per Article: 0.7] [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
|
10
|
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: 0.5] [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
|
11
|
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: 6.5] [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
|
12
|
Abstract
Alternative splicing of precursor mRNA is a key mediator of gene expression regulation leading to greater diversity of the proteome in complex organisms. Systematic sequencing of the human genome and transcriptome has led to our understanding of how alternative splicing of critical genes leads to multiple pathological conditions such as cancer. For many years, proteases were known only for their roles as proteolytic enzymes, acting to regulate/process proteins associated with diverse cellular functions. However, the differential expression and altered function of various protease isoforms, such as (i) anti-apoptotic activities, (ii) mediating intercellular adhesion, and (iii) modifying the extracellular matrix, are evidence of their specific contribution towards shaping the tumor microenvironment. Revealing the alternative splicing of protease genes and characterization of their protein products/isoforms with distinct and opposing functions creates a platform to understand how protease isoforms contribute to specific cancer hallmarks. Here, in this review, we address cancer-specific isoforms produced by the alternative splicing of proteases and their distinctive roles in the tumor microenvironment.
Collapse
Affiliation(s)
- Chamikara Liyanage
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Achala Fernando
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), 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.
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Translational Research Institute, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
13
|
Pseudogene Associated Recurrent Gene Fusion in Prostate Cancer. Neoplasia 2019; 21:989-1002. [PMID: 31446281 PMCID: PMC6713813 DOI: 10.1016/j.neo.2019.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 02/08/2023] Open
Abstract
We present the functional characterization of a pseudogene associated recurrent gene fusion in prostate cancer. The fusion gene KLK4-KLKP1 is formed by the fusion of the protein coding gene KLK4 with the noncoding pseudogene KLKP1. Screening of a cohort of 659 patients (380 Caucasian American; 250 African American, and 29 patients from other races) revealed that the KLK4-KLKP1 is expressed in about 32% of prostate cancer patients. Correlative analysis with other ETS gene fusions and SPINK1 revealed a concomitant expression pattern of KLK4-KLKP1 with ERG and a mutually exclusive expression pattern with SPINK1, ETV1, ETV4, and ETV5. Development of an antibody specific to KLK4-KLKP1 fusion protein confirmed the expression of the full-length KLK4-KLKP1 protein in prostate tissues. The in vitro and in vivo functional assays to study the oncogenic properties of KLK4-KLKP1 confirmed its role in cell proliferation, cell invasion, intravasation, and tumor formation. Presence of strong ERG and AR binding sites located at the fusion junction in KLK4-KLKP1 suggests that the fusion gene is regulated by ERG and AR. Correlative analysis of clinical data showed an association of KLK4-KLKP1 with lower preoperative PSA values and in young men (<50 years) with prostate cancer. Screening of patient urine samples showed that KLK4-KLKP1 can be detected noninvasively in urine. Taken together, we present KLK4-KLKP1 as a class of pseudogene associated fusion transcript in cancer with potential applications as a biomarker for routine screening of prostate cancer.
Collapse
|
14
|
Cui Z, Sun S, Liu Q, Zhou X, Gao S, Peng P, Li Q. MicroRNA-378-3p/5p suppresses the migration and invasiveness of oral squamous carcinoma cells by inhibiting KLK4 expression. Biochem Cell Biol 2019; 98:154-163. [PMID: 31265790 DOI: 10.1139/bcb-2019-0017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Distant metastasis frequently occurs in oral squamous cell carcinoma (OSCC) and contributes to the adverse prognosis for patients with OSCC. However, the potential mechanisms behind the metastasis have not yet been clarified. This study investigated the role of miR-378 in the migration and invasiveness of OSCC in vitro and in vivo. According to our results, the migration and invasiveness of OSCC cells were increased in cells overexpressing miR-378, and reduced in cells where miR-378-3p/5p was silenced. In addition, overexpression of miR-378 suppressed the expressions and activities of matrix metalloproteinase 9 (MMP-9) and MMP-2. Epithelial-mesenchymal transition (EMT) was restrained by overexpression of miR-378, as evidenced by an increase in E-cadherin expression and decrease in N-cadherin and uPA expression. However, knockdown of miR-378-3p/5p produced the opposite results. Moreover, kallikrein-related peptidase 4 (KLK4) was confirmed to be a target gene of miR-378. Overexpression of KLK4 reversed the induced decrease in migration and invasiveness of cells overexpressing miR-378 by upregulating the levels of MMP-9, MMP-2, and N-cadherin, and downregulating the level of E-cadhrin. Finally, the number of metastasis nodules in the lung tissues of nude mice was reduced by overexpression of miR-378, whereas the number of metastases increased with knockdown of miR-378. Taken together, our results suggest that the miR-378-KLK4 axis is involved in the mechanisms behind the migration and invasiveness of OSCC cells. Targeting the miR-378-KLK4 axis may be an effective measure for treating OSCC.
Collapse
Affiliation(s)
- Zhi Cui
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Jilin University, Changchun 130021, P.R. China
| | - Shiqun Sun
- Department of Prosthodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, P.R. China
| | - Qilin Liu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Jilin University, Changchun 130021, P.R. China
| | - Xuechun Zhou
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, P.R. China
| | - Siyu Gao
- Department of Pedodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, P.R. China
| | - Peixuan Peng
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, P.R. China
| | - Qianpeng Li
- VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun 130021, P.R. China
| |
Collapse
|
15
|
Riley BT, Ilyichova O, de Veer SJ, Swedberg JE, Wilson E, Hoke DE, Harris JM, Buckle AM. KLK4 Inhibition by Cyclic and Acyclic Peptides: Structural and Dynamical Insights into Standard-Mechanism Protease Inhibitors. Biochemistry 2019; 58:2524-2533. [PMID: 31058493 DOI: 10.1021/acs.biochem.9b00191] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sunflower trypsin inhibitor (SFTI-1) is a 14 amino acid serine protease inhibitor. The dual antiparallel β-sheet arrangement of SFTI-1 is stabilized by an N-terminal-C-terminal backbone cyclization and a further disulfide bridge to form a final bicyclic structure. This constrained structure is further rigidified by an extensive network of internal hydrogen bonds. Thus, the structure of SFTI-1 in solution resembles the protease-bound structure, reducing the entropic penalty upon protease binding. When cleaved at the scissile bond, it is thought that the rigidifying features of SFTI-1 maintain its structure, allowing the scissile bond to be reformed. The lack of structural plasticity for SFTI-1 is proposed to favor initial protease binding and continued occupancy in the protease active site, resulting in an equilibrium between the cleaved and uncleaved inhibitor in the presence of a protease. We have determined, at 1.15 Å resolution, the X-ray crystal structures of complexes between human kallikrein-related peptidase 4 (KLK4) and SFTI-FCQR(Asn14) and between KLK4 and an acyclic form of the same inhibitor, SFTI-FCQR(Asn14)[1,14], with the latter displaying a cleaved scissile bond. Structural analysis and MD simulations together reveal the roles of the altered contact sequence, intramolecular hydrogen bonding network, and backbone cyclization in altering the state of SFTI's scissile bond ligation at the protease active site. Taken together, the data presented reveal insights into the role of dynamics in the standard-mechanism inhibition and suggest that modifications on the non-contact strand may be a useful, underexplored approach for generating further potent or selective SFTI-based inhibitors against members of the serine protease family.
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
| | - Simon J de Veer
- Institute for Molecular Bioscience , The University of Queensland , Brisbane , Queensland 4072 , Australia
| | - Joakim E Swedberg
- Institute for Molecular Bioscience , The University of Queensland , Brisbane , Queensland 4072 , Australia
| | - Emily Wilson
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute , Monash University , Clayton , Victoria 3800 , Australia
| | - David E Hoke
- 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
| | - Ashley M Buckle
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute , Monash University , Clayton , Victoria 3800 , Australia
| |
Collapse
|
16
|
Figueroa CD, Molina L, Bhoola KD, Ehrenfeld P. Overview of tissue kallikrein and kallikrein-related peptidases in breast cancer. Biol Chem 2019; 399:937-957. [PMID: 29885274 DOI: 10.1515/hsz-2018-0111] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/17/2018] [Indexed: 12/11/2022]
Abstract
The kallikrein family comprises tissue kallikrein and 14 kallikrein-related peptidases (KLKs) recognized as a subgroup of secreted trypsin- or chymotrypsin-like serine proteases. KLKs are expressed in many cellular types where they regulate important physiological activities such as semen liquefaction, immune response, neural development, blood pressure, skin desquamation and tooth enamel formation. Tissue kallikrein, the oldest member and kinin-releasing enzyme, and KLK3/PSA, a tumor biomarker for prostate cancer are the most prominent components of the family. Additionally, other KLKs have shown an abnormal expression in neoplasia, particularly in breast cancer. Thus, increased levels of some KLKs may increase extracellular matrix degradation, invasion and metastasis; other KLKs modulate cell growth, survival and angiogenesis. On the contrary, KLKs can also inhibit angiogenesis and produce tumor suppression. However, there is a lack of knowledge on how KLKs are regulated in tumor microenvironment by molecules present at the site, namely cytokines, inflammatory mediators and growth factors. Little is known about the signaling pathways that control expression/secretion of KLKs in breast cancer, and further how activation of PAR receptors may contribute to functional activity in neoplasia. A better understanding of these molecular events will allow us to consider KLKs as relevant therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Carlos D Figueroa
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Luis Molina
- Department of Science, Universidad San Sebastián, sede De la Patagonia, Puerto Montt, Chile
| | - Kanti D Bhoola
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Centro de Investigaciones del Sistema Nervioso (CISNe), Valdivia, Chile, e-mail:
| |
Collapse
|
17
|
Di Meo A, Wang C, Cheng Y, Diamandis EP, Yousef GM. The miRNA-kallikrein interaction: a mosaic of epigenetic regulation in cancer. Biol Chem 2019; 399:973-982. [PMID: 29604203 DOI: 10.1515/hsz-2018-0112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/26/2018] [Indexed: 12/28/2022]
Abstract
The kallikrein-related peptidases (KLKs) constitute a family of 15 highly conserved serine proteases with trypsin- and chymotrypsin-like activities. Dysregulated expression and/or aberrant activation of KLKs has been linked to various pathophysiological processes, including cancer. Many KLKs have been identified as potential cancer biomarkers. microRNAs (miRNAs) are a class of small non-coding RNAs that regulate gene expression by pairing to the 3' untranslated region (UTR) of complimentary mRNA targets. miRNAs are dysregulated in many cancers, including prostate, kidney and ovarian cancers. Several studies have shown that miRNAs are involved in the post-transcriptional regulation of KLKs. However, recent evidence suggests that miRNAs can also act as downstream effectors of KLKs. In this review, we provide an update on the epigenetic regulation of KLKs by miRNAs. We also present recent experimental evidence that supports the regulatory role of KLKs on miRNA networks. The potential diagnostic and therapeutic applications of miRNA-kallikrein interactions are also discussed.
Collapse
Affiliation(s)
- Ashley Di Meo
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Laboratory Medicine, and the Keenan Research Centre for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada
| | - Cong Wang
- Department of Laboratory Medicine, and the Keenan Research Centre for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, M5T 3L9, Canada
| | - George M Yousef
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Laboratory Medicine, and the Keenan Research Centre for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada
| |
Collapse
|
18
|
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: 3.0] [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
|
19
|
Mitchell AC, Kannan D, Hunter SA, Parra Sperberg RA, Chang CH, Cochran JR. Engineering a potent inhibitor of matriptase from the natural hepatocyte growth factor activator inhibitor type-1 (HAI-1) protein. J Biol Chem 2018; 293:4969-4980. [PMID: 29386351 DOI: 10.1074/jbc.m117.815142] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 01/17/2018] [Indexed: 01/17/2023] Open
Abstract
Dysregulated matriptase activity has been established as a key contributor to cancer progression through its activation of growth factors, including the hepatocyte growth factor (HGF). Despite its critical role and prevalence in many human cancers, limitations to developing an effective matriptase inhibitor include weak binding affinity, poor selectivity, and short circulating half-life. We applied rational and combinatorial approaches to engineer a potent inhibitor based on the hepatocyte growth factor activator inhibitor type-1 (HAI-1), a natural matriptase inhibitor. The first Kunitz domain (KD1) of HAI-1 has been well established as a minimal matriptase-binding and inhibition domain, whereas the second Kunitz domain (KD2) is inactive and involved in negative regulation. Here, we replaced the inactive KD2 domain of HAI-1 with an engineered chimeric variant of KD2/KD1 domains and fused the resulting construct to an antibody Fc domain to increase valency and circulating serum half-life. The final protein variant contains four stoichiometric binding sites that we showed were needed to effectively inhibit matriptase with a Ki of 70 ± 5 pm, an increase of 120-fold compared with the natural HAI-1 inhibitor, to our knowledge making it one of the most potent matriptase inhibitors identified to date. Furthermore, the engineered inhibitor demonstrates a protease selectivity profile similar to that of wildtype KD1 but distinct from that of HAI-1. It also inhibits activation of the natural pro-HGF substrate and matriptase expressed on cancer cells with at least an order of magnitude greater efficacy than KD1.
Collapse
Affiliation(s)
| | | | - Sean A Hunter
- Cancer Biology Program, Stanford University, Stanford, California 94305
| | | | | | - Jennifer R Cochran
- From the Departments of Bioengineering and .,Cancer Biology Program, Stanford University, Stanford, California 94305.,Chemical Engineering and
| |
Collapse
|
20
|
Mitchell AC, Alford SC, Hunter SA, Kannan D, Sperberg RAP, Chang CH, Cochran JR. Development of a Protease Biosensor Based on a Dimerization-Dependent Red Fluorescent Protein. ACS Chem Biol 2018; 13:66-72. [PMID: 29125730 PMCID: PMC6453536 DOI: 10.1021/acschembio.7b00715] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dysregulated activity of the protease matriptase is a key contributor to aggressive tumor growth, cancer metastasis, and osteoarthritis. Methods for the detection and quantification of matriptase activity and inhibition would be useful tools. To address this need, we developed a matriptase-sensitive protein biosensor based on a dimerization-dependent red fluorescent protein (ddRFP) reporter system. In this platform, two adjoining protein domains, connected by a protease-labile linker, produce fluorescence when assembled and are nonfluorescent when the linker is cleaved by matriptase. A panel of ddRFP-based matriptase biosensor designs was created that contained different linker lengths between the protein domains. These constructs were characterized for linker-specific cleavage, matriptase activity, and matriptase selectivity; a biosensor containing a RSKLRVGGH linker (termed B4) was expressed at high yields and displayed both high catalytic efficiency and matriptase specificity. This biosensor detects matriptase inhibition by soluble and yeast cell surface expressed inhibitor domains with up to a 5-fold dynamic range and also detects matriptase activity expressed by human cancer cell lines. In addition to matriptase, we highlight a strategy that can be used to create effective biosensors for quantifying activity and inhibition of other proteases of interest.
Collapse
Affiliation(s)
- Aaron C. Mitchell
- Department of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Spencer C. Alford
- Department of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Sean A. Hunter
- Cancer Biology Program, Stanford University, Stanford, California 94305, United States
| | - Deepti Kannan
- Cancer Biology Program, Stanford University, Stanford, California 94305, United States
| | | | - Cheryl H. Chang
- Department of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Jennifer R. Cochran
- Department of Bioengineering, Stanford University, Stanford, California 94305, United States
- Cancer Biology Program, Stanford University, Stanford, California 94305, United States
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
21
|
Dorn J, Bayani J, Yousef GM, Yang F, Magdolen V, Kiechle M, Diamandis EP, Schmitt M. Clinical utility of kallikrein-related peptidases (KLK) in urogenital malignancies. Thromb Haemost 2017; 110:408-22. [DOI: 10.1160/th13-03-0206] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 05/25/2013] [Indexed: 12/19/2022]
Abstract
SummaryKallikrein-related peptidases (KLK), which represent a major tissue-associated proteolytic system, stand for a rich source of biomarkers that may allow molecular classification, early diagnosis and prognosis of human malignancies as well as prediction of response or failure to cancer-directed drugs. International research points to an important role of certain KLKs in female and male urogenital tract malignancies, in addition to cancers of the lung, brain, skin, head and neck, and the gastrointestinal tract. Regarding the female/male urogenital tract, remarkably, all of the KLKs are expressed in the normal prostate, testis, and kidney whereas the uterus, the ovary, and the urinary bladder are expressing a limited number of KLKs only. Most of the information regarding KLK expression in tumour-affected organs is available for ovarian cancer; all of the 12 KLKs tested so far were found to be elevated in the malignant state, depicting them as valuable biomarkers to distinguish between the normal and the cancerous phenotype. In contrast, for kidney cancer, a series of KLKs was found to be downregulated, while other KLKs were not expressed. Evidently, depending on the type of cancer or cancer stage, individual KLKs may show characteristics of a Janus-faced behaviour, by either expanding or inhibiting cancer progression and metastasis.
Collapse
|
22
|
Cui Z, Cui Y, Luo G, Yang S, Ling X, Lou Y, Sun X. Kallikrein-related peptidase 4 contributes to the tumor metastasis of oral squamous cell carcinoma. Biosci Biotechnol Biochem 2017; 81:1768-1777. [PMID: 28743213 DOI: 10.1080/09168451.2017.1356216] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is a disfiguring malignancy and significantly impacts the quality of patient's life. Kallikrein-related peptidase 4 (KLK4), which is closely related to cancers, is highly expressed in OSCC. To explore the biological function of KLK4 in OSCC, a KLK4-specific shRNA was used to silence its endogenous expression, and then the migration and invasion of OSCC cells were explored. Results of our study showed that silencing KLK4 inhibited the migration and invasion of OSCC cells. The protein levels of epithelial mesenchymal transition-associated markers and proteases were also altered by KLK4 silencing. Further study showed that the phosphatidylinositol 3-kinase (PI3 K)/protein kinase B (AKT) signaling pathway was involved in the function of KLK4. Treatment with a PI3 K/AKT activator reversed the migration-inhibitory effect of KLK4 shRNA. Our study suggests that KLK4 may contribute to the metastasis of OSCC through the PI3 K/AKT signaling pathway.
Collapse
Affiliation(s)
- Zhi Cui
- a Department of Oral and Maxillofacial Surgery, School of Stomatology , Jilin University , Changchun , People's Republic of China
| | - Ye Cui
- b Department of Orthodontics, School of Stomatology , Jilin University , Changchun , People's Republic of China
| | - Gan Luo
- b Department of Orthodontics, School of Stomatology , Jilin University , Changchun , People's Republic of China
| | - Shuting Yang
- c Department of Prosthodontics, School of Stomatology , Jilin University , Changchun , People's Republic of China
| | - Xinlian Ling
- b Department of Orthodontics, School of Stomatology , Jilin University , Changchun , People's Republic of China
| | - Yixin Lou
- b Department of Orthodontics, School of Stomatology , Jilin University , Changchun , People's Republic of China
| | - Xinhua Sun
- b Department of Orthodontics, School of Stomatology , Jilin University , Changchun , People's Republic of China
| |
Collapse
|
23
|
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: 1.3] [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
|
24
|
Liu B, Cui X, Zheng S, Dong K, Dong R. Aberrant KLK4 gene promoter hypomethylation in pediatric hepatoblastomas. Oncol Lett 2017; 13:1360-1364. [PMID: 28454262 DOI: 10.3892/ol.2017.5558] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/24/2016] [Indexed: 12/20/2022] Open
Abstract
DNA methylation has a crucial role in cancer biology and has been recognized as an activator of oncogenes and inactivator of tumor suppressor genes, both of which are mechanisms for tumorigenesis. Kallikrein-related peptidase 4 (KLK4), has been suggested to be an oncogene in various types of cancer. The aim of the present study was to assess the DNA methylation patterns of the KLK4 gene in cancerous samples harvested from patients with hepatoblastoma (HB). KLK4 mRNA expression levels were detected using reverse transcription-quantitative polymerase chain reaction and assessed its DNA methylation patterns using high-throughput mass spectrometry on a matrix-assisted laser desorption/ionization time-of-flight mass array. A total of 10 HB and 10 normal liver tissue samples were obtained from patients with HB. The results of the present study showed that a significantly higher level of KLK4 mRNA expression levels were detected in HB tissues, as compared with the matched controls. Furthermore, the KLK4 gene promoter region was distinctively less methylated in the HB samples compared with the controls and negatively correlated with KLK4 mRNA expression levels. These findings indicate that aberrant methylation of KLK4 may contribute to its upregulated mRNA expression in HB.
Collapse
Affiliation(s)
- Baihui Liu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defects and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, P.R. China
| | - Ximao Cui
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defects and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, P.R. China
| | - Shan Zheng
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defects and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, P.R. China
| | - Kuiran Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defects and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, P.R. China
| | - Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai Key Laboratory of Birth Defects and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai 201102, P.R. China
| |
Collapse
|
25
|
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: 3.1] [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
|
26
|
Fuhrman-Luck RA, Stansfield SH, Stephens CR, Loessner D, Clements JA. Prostate Cancer-Associated Kallikrein-Related Peptidase 4 Activates Matrix Metalloproteinase-1 and Thrombospondin-1. J Proteome Res 2016; 15:2466-78. [PMID: 27378148 DOI: 10.1021/acs.jproteome.5b01148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Prostate cancer metastasis to bone is terminal; thus, novel therapies are required to prevent end-stage disease. Kallikrein-related peptidase 4 (KLK4) is a serine protease that is overproduced in localized prostate cancer and is abundant in prostate cancer bone metastases. In vitro, KLK4 induces tumor-promoting phenotypes; however, the underlying proteolytic mechanism is undefined. The protein topography and migration analysis platform (PROTOMAP) was used for high-depth identification of KLK4 substrates secreted by prostate cancer bone metastasis-derived PC-3 cells to delineate the mechanism of KLK4 action in advanced prostate cancer. Thirty-six putative novel substrates were determined from the PROTOMAP analysis. In addition, KLK4 cleaved the established substrate, urokinase-type plasminogen activator, thus validating the approach. KLK4 activated matrix metalloproteinase-1 (MMP1), a protease that promotes prostate tumor growth and metastasis. MMP1 was produced in the tumor compartment of prostate cancer bone metastases, highlighting its accessibility to KLK4 at this site. KLK4 further liberated an N-terminal product, with purported angiogenic activity, from thrombospondin-1 (TSP1) and cleaved TSP1 in an osteoblast-derived matrix. This is the most comprehensive analysis of the proteolytic action of KLK4 in an advanced prostate cancer model to date, highlighting KLK4 as a potential multifunctional regulator of prostate cancer progression.
Collapse
Affiliation(s)
- Ruth A Fuhrman-Luck
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute , 37 Kent Street, Woolloongabba, Queensland 4102, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology , 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia
| | - Scott H Stansfield
- Institute of Health and Biomedical Innovation, Queensland University of Technology , 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia
| | - Carson R Stephens
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute , 37 Kent Street, Woolloongabba, Queensland 4102, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology , 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia
| | - Daniela Loessner
- Institute of Health and Biomedical Innovation, Queensland University of Technology , 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia
| | - Judith A Clements
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute , 37 Kent Street, Woolloongabba, Queensland 4102, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology , 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia
| |
Collapse
|
27
|
Karakosta TD, Soosaipillai A, Diamandis EP, Batruch I, Drabovich AP. Quantification of Human Kallikrein-Related Peptidases in Biological Fluids by Multiplatform Targeted Mass Spectrometry Assays. Mol Cell Proteomics 2016; 15:2863-76. [PMID: 27371727 DOI: 10.1074/mcp.m115.057695] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Indexed: 12/30/2022] Open
Abstract
Human kallikrein-related peptidases (KLKs) are a group of 15 secreted serine proteases encoded by the largest contiguous cluster of protease genes in the human genome. KLKs are involved in coordination of numerous physiological functions including regulation of blood pressure, neuronal plasticity, skin desquamation, and semen liquefaction, and thus represent promising diagnostic and therapeutic targets. Until now, quantification of KLKs in biological and clinical samples was accomplished by enzyme-linked immunosorbent assays (ELISA). Here, we developed multiplex targeted mass spectrometry assays for the simultaneous quantification of all 15 KLKs. Proteotypic peptides for each KLK were carefully selected based on experimental data and multiplexed in single assays. Performance of assays was evaluated using three different mass spectrometry platforms including triple quadrupole, quadrupole-ion trap, and quadrupole-orbitrap instruments. Heavy isotope-labeled synthetic peptides with a quantifying tag were used for absolute quantification of KLKs in sweat, cervico-vaginal fluid, seminal plasma, and blood serum, with limits of detection ranging from 5 to 500 ng/ml. Analytical performance of assays was evaluated by measuring endogenous KLKs in relevant biological fluids, and results were compared with selected ELISAs. The multiplex targeted proteomic assays were demonstrated to be accurate, reproducible, sensitive, and specific alternatives to antibody-based assays. Finally, KLK4, a highly prostate-specific protein and a speculated biomarker of prostate cancer, was unambiguously detected and quantified by immunoenrichment-SRM assay in seminal plasma and blood serum samples from individuals with confirmed prostate cancer and negative biopsy. Mass spectrometry revealed exclusively the presence of a secreted isoform and thus unequivocally resolved earlier disputes about KLK4 identity in seminal plasma. Measurements of KLK4 in either 41 seminal plasma or 58 blood serum samples revealed no statistically significant differences between patients with confirmed prostate cancer and negative biopsy. The presented multiplex targeted proteomic assays are an alternative analytical tool to study the biological and pathological roles of human KLKs.
Collapse
Affiliation(s)
- Theano D Karakosta
- From the ‡Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; §Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada
| | - Antoninus Soosaipillai
- ¶Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Eleftherios P Diamandis
- From the ‡Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; ‖Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ihor Batruch
- ‖Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Andrei P Drabovich
- From the ‡Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; ¶Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada;
| |
Collapse
|
28
|
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: 3.5] [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
|
29
|
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: 79] [Impact Index Per Article: 9.9] [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
|
30
|
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: 5.3] [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
|
31
|
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: 6.8] [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
|
32
|
Functional characterization of BC039389-GATM and KLK4-KRSP1 chimeric read-through transcripts which are up-regulated in renal cell cancer. BMC Genomics 2015; 16:247. [PMID: 25888189 PMCID: PMC4422297 DOI: 10.1186/s12864-015-1446-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2015] [Indexed: 11/10/2022] Open
Abstract
Background Chimeric read-through RNAs are transcripts originating from two directly adjacent genes (<10 kb) on the same DNA strand. Although they are found in next-generation whole transcriptome sequencing (RNA-Seq) data on a regular basis, investigating them further has usually been refrained from. Therefore, their expression patterns or functions in general, and in oncogenesis in particular, are poorly understood. Results We used paired-end RNA-Seq and a specifically designed computational data analysis pipeline (FusionSeq) to nominate read-through events in a small discovery set of renal cell carcinomas (RCC) and confirmed them in a larger validation cohort. 324 read-through events were called overall; 22/27 (81%) selected nominees passed validation with conventional PCR and were sequenced at the junction region. We frequently identified various isoforms of a given read-through event. 2/22 read-throughs were up-regulated: BC039389-GATM was higher expressed in RCC compared to benign adjacent kidney; KLK4-KRSP1 was expressed in 46/169 (27%) RCCs, but rarely in normal tissue. KLK4-KRSP1 expression was associated with worse clinical outcome in the patient cohort. In cell lines, both read-throughs influenced molecular mechanisms (i.e. target gene expression or migration/invasion) in a way that counteracted the effect of the respective parent transcript GATM or KLK4. Conclusions Our data suggests that the up-regulation of read-through RNA chimeras in tumors is not random but causes regulatory effects on cellular mechanisms and may impact patient survival. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1446-z) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
Lisle JE, Mertens-Walker I, Stephens CR, Stansfield SH, Clements JA, Herington AC, Stephenson SA. Murine, but not human, ephrin-B2 can be efficiently cleaved by the serine protease kallikrein-4: implications for xenograft models of human prostate cancer. Exp Cell Res 2015; 333:136-46. [PMID: 25724897 DOI: 10.1016/j.yexcr.2015.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/24/2014] [Accepted: 02/14/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND Ephrin-B2 is the sole physiologically-relevant ligand of the receptor tyrosine kinase EphB4, which is over-expressed in many epithelial cancers, including 66% of prostate cancers, and contributes to cancer cell survival, invasion and migration. Crucially, however, the cancer-promoting EphB4 signalling pathways are independent of interaction with its ligand ephrin-B2, as activation of ligand-dependent signalling causes tumour suppression. Ephrin-B2, however, is often found on the surface of endothelial cells of the tumour vasculature, where it can regulate angiogenesis to support tumour growth. Proteolytic cleavage of endothelial cell ephrin-B2 has previously been suggested as one mechanism whereby the interaction between tumour cell-expressed EphB4 and endothelial cell ephrin-B2 is regulated to support both cancer promotion and angiogenesis. METHODS An in silico approach was used to search accessible surfaces of 3D protein models for cleavage sites for the key prostate cancer serine protease, KLK4, and this identified murine ephrin-B2 as a potential KLK4 substrate. Mouse ephrin-B2 was then confirmed as a KLK4 substrate by in vitro incubation of recombinant mouse ephrin-B2 with active recombinant human KLK4. Cleavage products were visualised by SDS-PAGE, silver staining and Western blot and confirmed by N-terminal sequencing. RESULTS At low molar ratios, KLK4 cleaved murine ephrin-B2 but other prostate-specific KLK family members (KLK2 and KLK3/PSA) were less efficient, suggesting cleavage was KLK4-selective. The primary KLK4 cleavage site in murine ephrin-B2 was verified and shown to correspond to one of the in silico predicted sites between extracellular domain residues arginine 178 and asparagine 179. Surprisingly, the highly homologous human ephrin-B2 was poorly cleaved by KLK4 at these low molar ratios, likely due to the 3 amino acid differences at this primary cleavage site. CONCLUSION These data suggest that in in vivo mouse xenograft models, endogenous mouse ephrin-B2, but not human tumour ephrin-B2, may be a downstream target of cancer cell secreted human KLK4. This is a critical consideration when interpreting data from murine explants of human EphB4+/KLK4+ cancer cells, such as prostate cancer cells, where differential effects may be seen in mouse models as opposed to human clinical situations.
Collapse
Affiliation(s)
- J E Lisle
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - I Mertens-Walker
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - C R Stephens
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - S H Stansfield
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - J A Clements
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - A C Herington
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - S-A Stephenson
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia.
| |
Collapse
|
34
|
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: 0.9] [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
|
35
|
Kularathna PK, Pagel CN, Mackie EJ. Tumour progression and cancer-induced pain: a role for protease-activated receptor-2? Int J Biochem Cell Biol 2014; 57:149-56. [PMID: 25448411 DOI: 10.1016/j.biocel.2014.10.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 10/23/2014] [Indexed: 02/08/2023]
Abstract
The role of proteases in modifying the microenvironment of tumour cells has long been recognised. With the discovery of the protease-activated receptor family of G protein-coupled receptors a mechanism for cells to sense and respond directly to proteases in their microenvironment was revealed. Many early studies described the roles of protease-activated receptors in the cellular events that occur during blood coagulation and inflammation. More recently, studies have begun to focus on the roles of protease-activated receptors in the establishment, progression and metastasis of a variety of tumours. This review will focus on the expression of protease-activated receptor-2 and its activators by normal and neoplastic tissues, and describe current evidence that activation of protease-activated receptor-2 is an important event at multiple stages of tumour progression and in pain associated with cancer.
Collapse
Affiliation(s)
- Pamuditha K Kularathna
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Charles N Pagel
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Eleanor J Mackie
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
36
|
Mavridis K, Avgeris M, Scorilas A. Targeting kallikrein-related peptidases in prostate cancer. Expert Opin Ther Targets 2014; 18:365-83. [DOI: 10.1517/14728222.2014.880693] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
37
|
Molecular circuit involving KLK4 integrates androgen and mTOR signaling in prostate cancer. Proc Natl Acad Sci U S A 2013; 110:E2572-81. [PMID: 23798432 DOI: 10.1073/pnas.1304318110] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The androgen receptor (AR) and the phosphoinositide 3-kinase (PI3K)/protein kinase B/mammalian target of rapamycin (mTOR) signaling are two of the major proliferative pathways in a number of tissues and are the main therapeutic targets in various disorders, including prostate cancer (PCa). Previous work has shown that there is reciprocal feedback regulation of PI3K and AR signaling in PCa, suggesting that cotargeting both pathways may enhance therapeutic efficacy. Here we show that proteins encoded by two androgen-regulated genes, kallikrein related peptidase 4 (KLK4) and promyelocytic leukemia zinc finger (PLZF), integrate optimal functioning of AR and mTOR signaling in PCa cells. KLK4 interacts with PLZF and decreases its stability. PLZF in turn interacts with AR and inhibits its function as a transcription factor. PLZF also activates expression of regulated in development and DNA damage responses 1, an inhibitor of mTORC1. Thus, a unique molecular switch is generated that regulates both AR and PI3K signaling. Consistently, KLK4 knockdown results in a significant decline in PCa cell proliferation in vitro and in vivo, decreases anchorage-independent growth, induces apoptosis, and dramatically sensitizes PCa cells to apoptosis-inducing agents. Furthermore, in vivo nanoliposomal KLK4 siRNA delivery in mice bearing PCa tumors results in profound remission. These results demonstrate that the activities of AR and mTOR pathways are maintained by KLK4, which may thus be a viable target for therapy.
Collapse
|
38
|
Qu S, Olafsrud SM, Meza-Zepeda LA, Saatcioglu F. Rapid gene expression changes in peripheral blood lymphocytes upon practice of a comprehensive yoga program. PLoS One 2013; 8:e61910. [PMID: 23613970 PMCID: PMC3629142 DOI: 10.1371/journal.pone.0061910] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 03/18/2013] [Indexed: 11/18/2022] Open
Abstract
One of the most common integrative medicine (IM) modalities is yoga and related practices. Previous work has shown that yoga may improve wellness in healthy people and have benefits for patients. However, the mechanisms of how yoga may positively affect the mind-body system are largely unknown. Here we have assessed possible rapid changes in global gene expression profiles in the peripheral blood mononuclear cells (PBMCs) in healthy people that practiced either a comprehensive yoga program or a control regimen. The experimental sessions included gentle yoga postures, breathing exercises, and meditation (Sudarshan Kriya and Related Practices – SK&P) compared with a control regimen of a nature walk and listening to relaxing music. We show that the SK&P program has a rapid and significantly greater effect on gene expression in PBMCs compared with the control regimen. These data suggest that yoga and related practices result in rapid gene expression alterations which may be the basis for their longer term cell biological and higher level health effects.
Collapse
Affiliation(s)
- Su Qu
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Solveig Mjelstad Olafsrud
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Genomics Core Facility, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Leonardo A. Meza-Zepeda
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Genomics Core Facility, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway
- * E-mail:
| |
Collapse
|
39
|
Lose F, Srinivasan S, O’Mara T, Marquart L, Chambers S, Gardiner RA, Aitken JF, Spurdle AB, Batra J, Clements JA. Genetic association of the KLK4 locus with risk of prostate cancer. PLoS One 2012; 7:e44520. [PMID: 22970239 PMCID: PMC3435290 DOI: 10.1371/journal.pone.0044520] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Accepted: 08/08/2012] [Indexed: 11/25/2022] Open
Abstract
The Kallikrein-related peptidase, KLK4, has been shown to be significantly overexpressed in prostate tumours in numerous studies and is suggested to be a potential biomarker for prostate cancer. KLK4 may also play a role in prostate cancer progression through its involvement in epithelial-mesenchymal transition, a more aggressive phenotype, and metastases to bone. It is well known that genetic variation has the potential to affect gene expression and/or various protein characteristics and hence we sought to investigate the possible role of single nucleotide polymorphisms (SNPs) in the KLK4 gene in prostate cancer. Assessment of 61 SNPs in the KLK4 locus (± 10 kb) in approximately 1300 prostate cancer cases and 1300 male controls for associations with prostate cancer risk and/or prostate tumour aggressiveness (Gleason score <7 versus ≥ 7) revealed 7 SNPs to be associated with a decreased risk of prostate cancer at the P(trend)<0.05 significance level. Three of these SNPs, rs268923, rs56112930 and the HapMap tagSNP rs7248321, are located several kb upstream of KLK4; rs1654551 encodes a non-synonymous serine to alanine substitution at position 22 of the long isoform of the KLK4 protein, and the remaining 3 risk-associated SNPs, rs1701927, rs1090649 and rs806019, are located downstream of KLK4 and are in high linkage disequilibrium with each other (r(2) ≥ 0.98). Our findings provide suggestive evidence of a role for genetic variation in the KLK4 locus in prostate cancer predisposition.
Collapse
Affiliation(s)
- Felicity Lose
- Molecular Cancer Epidemiology Group, Genetics and Population Health Division, Queensland Institute of Medical Research, 300 Herston Rd, Herston, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Srilakshmi Srinivasan
- Australian Prostate Cancer Research Centre – Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Tracy O’Mara
- Molecular Cancer Epidemiology Group, Genetics and Population Health Division, Queensland Institute of Medical Research, 300 Herston Rd, Herston, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre – Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Louise Marquart
- Statistics Unit, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Suzanne Chambers
- Griffith Health Institute, Griffith University, Brisbane, Queensland, Australia
- Viertel Centre for Cancer Research, Cancer Council Queensland, Brisbane, Queensland, Australia
- University of Queensland Centre for Clinical Research, Royal Brisbane Hospital, Brisbane, Queensland, Australia
| | - Robert A. Gardiner
- University of Queensland Centre for Clinical Research, Royal Brisbane Hospital, Brisbane, Queensland, Australia
| | - Joanne F. Aitken
- Griffith Health Institute, Griffith University, Brisbane, Queensland, Australia
- Viertel Centre for Cancer Research, Cancer Council Queensland, Brisbane, Queensland, Australia
| | - Amanda B. Spurdle
- Molecular Cancer Epidemiology Group, Genetics and Population Health Division, Queensland Institute of Medical Research, 300 Herston Rd, Herston, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Jyotsna Batra
- Molecular Cancer Epidemiology Group, Genetics and Population Health Division, Queensland Institute of Medical Research, 300 Herston Rd, Herston, Brisbane, Queensland, Australia
| | - Judith A. Clements
- Australian Prostate Cancer Research Centre – Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
40
|
White NMA, Youssef YM, Fendler A, Stephan C, Jung K, Yousef GM. The miRNA-kallikrein axis of interaction: a new dimension in the pathogenesis of prostate cancer. Biol Chem 2012; 393:379-89. [PMID: 22505520 DOI: 10.1515/hsz-2011-0246] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 01/11/2012] [Indexed: 11/15/2022]
Abstract
Kallikrein-related peptidases (KLKs) are a family of serine proteases that were shown to be useful cancer biomarkers. KLKs have been shown to be dysregulated in prostate cancer (PCa). microRNAs (miRNAs) are short RNA nucleotides that negatively regulate gene expression and have been reportedly dysregulated in PCa. We compiled a comprehensive list of 55 miRNAs that are differentially expressed in PCa from previous microarray analysis and published literature. Target prediction analyses showed that 29 of these miRNAs are predicted to target 10 KLKs. Eight of these miRNAs were predicted to target more than one KLK. Quantitative real-time (qRT)-PCR demonstrated that there was an inverse correlation pattern in the expression (normal vs. cancer) between dysregulated miRNAs and their target KLKs. In addition, we experientially validated the miRNA-KLK interaction by transfecting miR-331-3p and miR-143 into a PCa cell line. Decreased expression of targets KLK4 and KLK10, respectively, and decreased cellular growth were observed. In addition to KLKs, dysregulated miRNAs were predicted to target other genes involved in the pathogenesis of PCa. These data show that miRNAs can contribute to KLK regulation in PCa. The miRNA-KLK axis of interaction projects a new element in the pathogenesis of PCa that may have therapeutic implications.
Collapse
Affiliation(s)
- Nicole M A White
- Department of Laboratory Medicine and the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
41
|
Sanchez WY, de Veer SJ, Swedberg JE, Hong EJ, Reid JC, Walsh TP, Hooper JD, Hammond GL, Clements JA, Harris JM. Selective cleavage of human sex hormone-binding globulin by kallikrein-related peptidases and effects on androgen action in LNCaP prostate cancer cells. Endocrinology 2012; 153:3179-89. [PMID: 22547569 DOI: 10.1210/en.2012-1011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Stimulation of the androgen receptor via bioavailable androgens, including testosterone and testosterone metabolites, is a key driver of prostate development and the early stages of prostate cancer. Androgens are hydrophobic and as such require carrier proteins, including sex hormone-binding globulin (SHBG), to enable efficient distribution from sites of biosynthesis to target tissues. The similarly hydrophobic corticosteroids also require a carrier protein whose affinity for steroid is modulated by proteolysis. However, proteolytic mechanisms regulating the SHBG/androgen complex have not been reported. Here, we show that the cancer-associated serine proteases, kallikrein-related peptidase (KLK)4 and KLK14, bind strongly to SHBG in glutathione S-transferase interaction analyses. Further, we demonstrate that active KLK4 and KLK14 cleave human SHBG at unique sites and in an androgen-dependent manner. KLK4 separated androgen-free SHBG into its two laminin G-like (LG) domains that were subsequently proteolytically stable even after prolonged digestion, whereas a catalytically equivalent amount of KLK14 reduced SHBG to small peptide fragments over the same period. Conversely, proteolysis of 5α-dihydrotestosterone (DHT)-bound SHBG was similar for both KLKs and left the steroid binding LG4 domain intact. Characterization of this proteolysis fragment by [(3)H]-labeled DHT binding assays revealed that it retained identical affinity for androgen compared with full-length SHBG (dissociation constant = 1.92 nM). Consistent with this, both full-length SHBG and SHBG-LG4 significantly increased DHT-mediated transcriptional activity of the androgen receptor compared with DHT delivered without carrier protein. Collectively, these data provide the first evidence that SHBG is a target for proteolysis and demonstrate that a stable fragment derived from proteolysis of steroid-bound SHBG retains binding function in vitro.
Collapse
Affiliation(s)
- Washington Y Sanchez
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
The effects of short-term genistein intervention on prostate biomarker expression in patients with localised prostate cancer before radical prostatectomy. Br J Nutr 2012; 108:2138-47. [PMID: 22397815 DOI: 10.1017/s0007114512000384] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nutritionally relevant levels of genistein, the predominant isoflavone in soyabean associated with lower risk of prostate cancer (PCa), may modulate the expression of prostate tissue biomarkers associated with cancer prediction and progression. A phase 2 placebo-controlled, randomised, double-blind clinical trial was conducted in forty-seven Norwegian patients before prostatectomy. Intervention was 30 mg genistein or placebo capsules daily for 3-6 weeks. Luminal cells from malignant and benign glands were isolated with laser capture microdissection and the mRNA levels of androgen-related biomarkers (androgen receptor, NK3 homeobox 1, kallikrein-related peptide 4 (KLK4)) and cell cycle-related genes (p21 Waf1/Cip1 , p27 Kip1 , p53) were analysed with real-time semiquantitative PCR. Immunohistochemistry of androgen-, cell cycle-, proliferative- (Ki67 nuclear antigen), apoptotic- (B-cell CLL/lymphoma 2 (BCL-2) and BCL-2-associated X protein) and neuroendocrine differentiation-related biomarkers (neuron-specific enolase and cytoplasmic chromogranin A) was performed using tissue microarrays containing normal, Gleason grade 3 and grade 4 prostate tissues. There were no significant effects by genistein intervention on proliferation-, cell cycle-, apoptosis- or neuroendocrine biomarkers. Genistein intervention, however, significantly reduced the mRNA level of KLK4 in tumour cells (P = 0·033) and there was a non-significant reduction in androgen and cell cycle-related biomarkers, except for p27Kip1, whose expression in the nuclear compartment was increased. Genistein intervention modulated the expression of several biomarkers which may be related to PCa prediction and progression. The present study supports genistein as a chemopreventive agent in PCa. Further investigation is warranted in larger and longer-duration studies.
Collapse
|
43
|
Wilkinson R, Woods K, D'Rozario R, Prue R, Vari F, Hardy MY, Dong Y, Clements JA, Hart DNJ, Radford KJ. Human kallikrein 4 signal peptide induces cytotoxic T cell responses in healthy donors and prostate cancer patients. Cancer Immunol Immunother 2012; 61:169-179. [PMID: 21874303 PMCID: PMC11028920 DOI: 10.1007/s00262-011-1095-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 07/30/2011] [Indexed: 12/16/2022]
Abstract
Immunotherapy is a promising new treatment for patients with advanced prostate and ovarian cancer, but its application is limited by the lack of suitable target antigens that are recognized by CD8+ cytotoxic T lymphocytes (CTL). Human kallikrein 4 (KLK4) is a member of the kallikrein family of serine proteases that is significantly overexpressed in malignant versus healthy prostate and ovarian tissue, making it an attractive target for immunotherapy. We identified a naturally processed, HLA-A*0201-restricted peptide epitope within the signal sequence region of KLK4 that induced CTL responses in vitro in most healthy donors and prostate cancer patients tested. These CTL lysed HLA-A*0201+ KLK4 + cell lines and KLK4 mRNA-transfected monocyte-derived dendritic cells. CTL specific for the HLA-A*0201-restricted KLK4 peptide were more readily expanded to a higher frequency in vitro compared to the known HLA-A*0201-restricted epitopes from prostate cancer antigens; prostate-specific antigen (PSA), prostate-specific membrane antigen (PSMA) and prostatic acid phosphatase (PAP). These data demonstrate that KLK4 is an immunogenic molecule capable of inducing CTL responses and identify it as an attractive target for prostate and ovarian cancer immunotherapy.
Collapse
Affiliation(s)
- Ray Wilkinson
- Dendritic Cell Program, Mater Medical Research Institute, Level 3 Aubigny Place, South Brisbane, QLD, 4101, Australia
- Renal Research Laboratory, Queensland Health/Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | - Katherine Woods
- Dendritic Cell Program, Mater Medical Research Institute, Level 3 Aubigny Place, South Brisbane, QLD, 4101, Australia
| | - Rachael D'Rozario
- Dendritic Cell Program, Mater Medical Research Institute, Level 3 Aubigny Place, South Brisbane, QLD, 4101, Australia
| | - Rebecca Prue
- Dendritic Cell Program, Mater Medical Research Institute, Level 3 Aubigny Place, South Brisbane, QLD, 4101, Australia
| | - Frank Vari
- Dendritic Cell Program, Mater Medical Research Institute, Level 3 Aubigny Place, South Brisbane, QLD, 4101, Australia
- Clinical Immunohematology, Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | - Melinda Y Hardy
- Dendritic Cell Program, Mater Medical Research Institute, Level 3 Aubigny Place, South Brisbane, QLD, 4101, Australia
| | - Ying Dong
- Institute of Health and Biomedical Innovation and Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Brisbane, QLD, Australia
| | - Judith A Clements
- Institute of Health and Biomedical Innovation and Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Brisbane, QLD, Australia
| | - Derek N J Hart
- Dendritic Cell Program, Mater Medical Research Institute, Level 3 Aubigny Place, South Brisbane, QLD, 4101, Australia
- Dendritic Cell Biology and Therapeutics, ANZAC Research Institute, Concord Hospital, Hospital Road, Sydney, NSW, 2139, Australia
| | - Kristen J Radford
- Dendritic Cell Program, Mater Medical Research Institute, Level 3 Aubigny Place, South Brisbane, QLD, 4101, Australia.
| |
Collapse
|
44
|
Avgeris M, Stravodimos K, Scorilas A. Kallikrein-related peptidase 4 gene (KLK4) in prostate tumors: quantitative expression analysis and evaluation of its clinical significance. Prostate 2011; 71:1780-9. [PMID: 21520157 DOI: 10.1002/pros.21395] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Accepted: 03/16/2011] [Indexed: 01/02/2023]
Abstract
BACKGROUND Recently accumulating evidences underline the central role of the kallikrein-related peptidases family (KLKs) in prostate cancer (PCa) development and progression. The KLK4 is a prostate highly expressed gene under the transcriptional control of androgens, encoding for the KLK4 extracellular serine protease. The aim of this study is to investigate the expression status of KLK4 in PCa patients in order to reveal its utility in PCa establishment and clinical management. METHODS Prostatic tissue specimens were obtained from 60 PCa and 59 benign prostate hyperplasia (BPH) randomly chosen patients. Using a developed quantitative real-time RT-PCR method, KLK4 expression levels were determined in the specimens of the two patients' cohorts. Advance biostatistical analysis was completed to explore the clinical value of KLK4 expression in PCa and BPH patients. RESULTS PCa patients presented a statistically significant (P = 0.002) elevation, more than threefold, of the KLK4 transcripts compared to BPH ones. The KLK4 expression levels were also positive correlated with PCa patients' stage (P = 0.031) and preoperative prostate-specific antigen (PSA) serum concentrations (P < 0.001). ROC curve and logistic regression analysis revealed the significant (P = 0.002) and the independent (P = 0.044) clinical value of the KLK4 expression for the discrimination of PCa from BPH patients. CONCLUSIONS The KLK4 expression analysis reveals its up-regulation in PCa cells, which is significantly associated with the advanced stages of the disease and the patients' preoperative PSA serum levels. KLK4 quantification serves as an independent biomarker for the discrimination between the malignant and the benign nature of prostate tumors.
Collapse
Affiliation(s)
- Margaritis Avgeris
- Department of Biochemistry and Molecular Biology, University of Athens, Panepistimiopolis, Athens, Greece
| | | | | |
Collapse
|
45
|
Skårn M, Namløs HM, Noordhuis P, Wang MY, Meza-Zepeda LA, Myklebost O. Adipocyte differentiation of human bone marrow-derived stromal cells is modulated by microRNA-155, microRNA-221, and microRNA-222. Stem Cells Dev 2011; 21:873-83. [PMID: 21756067 DOI: 10.1089/scd.2010.0503] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Human mesenchymal stromal cells (hMSCs) are capable of limited self-renewal and multilineage differentiation in vitro. Several studies have demonstrated that microRNAs (miRNAs, miRs), post-transcriptional modifiers of mRNA stability and protein translation, play crucial roles in the regulation of these complex processes. To gain knowledge regarding the role of miRNAs in human adipocyte differentiation, we examined the miRNA expression profile of the immortalized human bone marrow-derived stromal cell line hMSC-Tert20. Such a model system has the advantage of a reproducible and consistent phenotype while maintaining important properties of the primary donor cells, including the potential to differentiate to adipocytes, osteoblasts, and chondrocytes. We identified 12 miRNAs that were differentially expressed during adipogenesis, of which several have been previously shown to play important roles in adipocyte biology. Among these, the expression of miRNA-155, miRNA-221, and miRNA-222 decreased during the adipogenic program of both immortalized and primary hMSCs, suggesting that they act as negative regulators of differentiation. Interestingly, ectopic expression of the miRNAs significantly inhibited adipogenesis and repressed induction of the master regulators PPARγ and CCAAT/enhancer-binding protein alpha. Our study provides the first experimental evidence that miRNA-155, miRNA-221, and miRNA-222 have an important function in human adipocyte differentiation, and that their downregulation is necessary to relieve the repression of genes crucial for this process.
Collapse
Affiliation(s)
- Magne Skårn
- Department of Tumor Biology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | | | | | | | | | | |
Collapse
|
46
|
Lawrence MG, Lai J, Clements JA. Kallikreins on steroids: structure, function, and hormonal regulation of prostate-specific antigen and the extended kallikrein locus. Endocr Rev 2010; 31:407-46. [PMID: 20103546 DOI: 10.1210/er.2009-0034] [Citation(s) in RCA: 169] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The 15 members of the kallikrein-related serine peptidase (KLK) family have diverse tissue-specific expression profiles and putative proteolytic functions. The kallikrein family is also emerging as a rich source of disease biomarkers with KLK3, commonly known as prostate-specific antigen, being the current serum biomarker for prostate cancer. The kallikrein locus is also notable because it is extraordinarily responsive to steroids and other hormones. Indeed, at least 14 functional hormone response elements have been identified in the kallikrein locus. A more comprehensive understanding of the transcriptional regulation of kallikreins may help the field make more informed hypotheses about the physiological functions of kallikreins and their effectiveness as biomarkers. In this review, we describe the organization of the kallikrein locus and the structure of kallikrein genes and proteins. We also focus on the transcriptional regulation of kallikreins by androgens, progestins, glucocorticoids, mineralocorticoids, estrogens, and other hormones in animal models and human prostate, breast, and reproductive tract tissues. The interaction of the androgen receptor with androgen response elements in the promoter and enhancer of KLK2 and KLK3 is also summarized in detail. There is evidence that all kallikreins are regulated by multiple nuclear receptors. Yet, apart from KLK2 and KLK3, it is not clear whether all kallikreins are direct transcriptional targets. Therefore, we argue that gaining more detailed information about the mechanisms that regulate kallikrein expression should be a priority of future studies and that the kallikrein locus will continue to be an important model in the era of genome-wide analyses.
Collapse
Affiliation(s)
- Mitchell G Lawrence
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | | | | |
Collapse
|
47
|
Seiz L, Kotzsch M, Grebenchtchikov NI, Geurts-Moespot AJ, Fuessel S, Goettig P, Gkazepis A, Wirth MP, Schmitt M, Lossnitzer A, Sweep FCGJ, Magdolen V. Polyclonal antibodies against kallikrein-related peptidase 4 (KLK4): immunohistochemical assessment of KLK4 expression in healthy tissues and prostate cancer. Biol Chem 2010; 391:391-401. [PMID: 20180634 DOI: 10.1515/bc.2010.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
KLK4 is a member of the human kallikrein-related peptidase family of (chymo)trypsin-like serine proteases. The aim of the present study was to generate polyclonal antibodies (pAb) directed against KLK4 for the analysis of KLK4 by immunohistochemistry in human tissues. Recombinantly expressed human mature KLK4 was used for immunization of chickens. pAb 617A is an affinity-purified monospecific pAb fraction reacting with a linear epitope within a flexible surface-exposed loop of KLK4. pAb 617C is the KLK-directed pAb fraction completely depleted from pAb 617A. In healthy adult tissues, KLK4 was immunodetected by both antibody fractions in kidney, liver, and prostate, but not in other organs such as colon and lung. To evaluate protein expression of KLK4 in prostate cancer, samples of tumor tissue plus corresponding tumor-free areas of 44 prostate cancer patients, represented on a tissue microarray, were investigated. Distinct KLK4 immunostaining was observed with both antibodies in cancerous glandular epithelial cells, but not in surrounding stromal cells. KLK4 expression was lower in stage pT3+4 than in pT1+2 tumors, which was highly significant when employing pAb 617A. Thus, our results indicate that KLK4, which is expressed in the healthy prostate, is upregulated in early-stage but not late-stage prostate cancer.
Collapse
Affiliation(s)
- Lina Seiz
- Department of Obstetrics and Gynecology, Technical University of Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wang L, Jin Y, Arnoldussen YJ, Jonson I, Qu S, Mælandsmo GM, Kristian A, Risberg B, Wæhre H, Danielsen HE, Saatcioglu F. STAMP1 Is Both a Proliferative and an Antiapoptotic Factor in Prostate Cancer. Cancer Res 2010; 70:5818-28. [DOI: 10.1158/0008-5472.can-09-4697] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
Lai J, Lehman ML, Dinger ME, Hendy SC, Mercer TR, Seim I, Lawrence MG, Mattick JS, Clements JA, Nelson CC. A variant of the KLK4 gene is expressed as a cis sense-antisense chimeric transcript in prostate cancer cells. RNA (NEW YORK, N.Y.) 2010; 16:1156-1166. [PMID: 20406994 PMCID: PMC2874168 DOI: 10.1261/rna.2019810] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 02/18/2010] [Indexed: 05/29/2023]
Abstract
In humans, more than 30,000 chimeric transcripts originating from 23,686 genes have been identified. The mechanisms and association of chimeric transcripts arising from chromosomal rearrangements with cancer are well established, but much remains unknown regarding the biogenesis and importance of other chimeric transcripts that arise from nongenomic alterations. Recently, a SLC45A3-ELK4 chimera has been shown to be androgen-regulated, and is overexpressed in metastatic or high-grade prostate tumors relative to local prostate cancers. Here, we characterize the expression of a KLK4 cis sense-antisense chimeric transcript, and show other examples in prostate cancer. Using non-protein-coding microarray analyses, we initially identified an androgen-regulated antisense transcript within the 3' untranslated region of the KLK4 gene in LNCaP cells. The KLK4 cis-NAT was validated by strand-specific linker-mediated RT-PCR and Northern blotting. Characterization of the KLK4 cis-NAT by 5' and 3' rapid amplification of cDNA ends (RACE) revealed that this transcript forms multiple fusions with the KLK4 sense transcript. Lack of KLK4 antisense promoter activity using reporter assays suggests that these transcripts are unlikely to arise from a trans-splicing mechanism. 5' RACE and analyses of deep sequencing data from LNCaP cells treated +/-androgens revealed six high-confidence sense-antisense chimeras of which three were supported by the cDNA databases. In this study, we have shown complex gene expression at the KLK4 locus that might be a hallmark of cis sense-antisense chimeric transcription.
Collapse
Affiliation(s)
- John Lai
- Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology and Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Mavridis K, Scorilas A. Prognostic value and biological role of the kallikrein-related peptidases in human malignancies. Future Oncol 2010; 6:269-85. [PMID: 20146586 DOI: 10.2217/fon.09.149] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cancer is a substantial health problem for the populations of the Western world. The discovery of new molecular biomarkers for diagnosis, prognosis and monitoring patients' response to therapy can aid in combating this complicated disease. The human kallikrein-related peptidases (human tissue kallikreins [KLKs]) are encoded by a continuous multigene family, located on chromosomal region 19q13.3-4. KLK3 (prostate-specific antigen) is the most efficient cancer biomarker ever employed. KLK genes are expressed abnormally in various malignancies, where they affect cancer-cell growth and metastasis. Their deregulated expression pattern, often associated with various clinicopathological characteristics of cancer patients, can be exploited, solely or within multiparametric panels, as a prognostic biomarker. Recent data illustrate that discernible molecular modulations of KLKs, occurring as a result of cancer cells' treatment with antitumor agents, may serve as new potential biomarkers, possibly predicting patients' treatment response. It is believed that KLKs might be employed in future clinical practice as novel and effective tumor markers.
Collapse
Affiliation(s)
- Konstantinos Mavridis
- Department of Biochemistry & Molecular Biology, University of Athens, Athens, Greece.
| | | |
Collapse
|