1
|
Kelam N, Ogorevc M, Gotovac I, Kuzmić Prusac I, Vukojević K, Saraga-Babić M, Mardešić S. Analysis of Kallikrein 6, Acetyl-α-Tubulin, and Aquaporin 1 and 2 Expression Patterns During Normal Human Nephrogenesis and in Congenital Anomalies of the Kidney and Urinary Tract (CAKUT). Genes (Basel) 2025; 16:499. [PMID: 40428321 PMCID: PMC12111363 DOI: 10.3390/genes16050499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: The human kallikrein-related peptidase 6 (KLK6), a serine protease with trypsin-like properties, belongs to the 15-member kallikrein (KLK) gene family and is predominantly recognized for its role in oncogenesis, neurodegenerative disorders, and skin conditions. Aquaporins (AQPs) are integral membrane proteins that facilitate water transport across cell membranes. AQP1 is constitutively active in the kidneys and plays a crucial role in reabsorbing filtered water, while AQP2 is regulated by vasopressin and is essential for maintaining body fluid homeostasis. The primary objective of the present study is to investigate the spatio-temporal expression patterns of KLK6, AQP1, and AQP2 throughout normal human nephrogenesis and congenital kidney and urinary tract (CAKUT) abnormalities: duplex kidneys, horseshoe kidneys, and dysplastic kidneys. Methods: An immunofluorescence analysis of KLK6, AQP1, and AQP2 was performed on 37 paraffin-embedded fetal kidney samples. The area percentage of KLK6 in the kidney cortex was calculated in normal developing samples during developmental phases 2, 3, and 4 and compared with CAKUT samples. Results: KLK6 exhibits distinct spatiotemporal expression patterns during human kidney development, with consistent localization in proximal tubules. Its subcellular positioning shifts from the basolateral cytoplasm in early phases to the apical cytoplasm in later stages, which may be strategically positioned to act on its substrate in either the peritubular space or the tubular fluid. KLK6 expression followed a quadratic trajectory, peaking at Ph4. This marked increase in the final developmental phase aligns with its strong expression in mature kidneys, suggesting a potential role in proximal tubule differentiation and functional maturation through facilitating extracellular matrix remodeling and activating proteinase-activated receptors, modulating the signaling pathways that are essential for tubular development. In duplex kidneys, structural abnormalities such as ureteral obstruction and hydronephrosis may upregulate KLK6 as part of a reparative response, while its downregulation could impair epithelial remodeling and cytoskeletal integrity, exacerbating dysplastic phenotypes. Conclusions: These findings highlight the potential of KLK6 involvement in normal kidney development and the pathology of CAKUT.
Collapse
Affiliation(s)
- Nela Kelam
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia; (N.K.); (K.V.); (S.M.)
| | - Marin Ogorevc
- Department of Pathology, University Hospital Split, Spinciceva 1, 21000 Split, Croatia; (M.O.); (I.K.P.)
| | - Ivona Gotovac
- Clinical Department of Diagnostic and Interventional Radiology, University Hospital Centre Split, Spinciceva 1, 21000 Split, Croatia;
| | - Ivana Kuzmić Prusac
- Department of Pathology, University Hospital Split, Spinciceva 1, 21000 Split, Croatia; (M.O.); (I.K.P.)
| | - Katarina Vukojević
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia; (N.K.); (K.V.); (S.M.)
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Mediterranean Institute for Life Sciences, University of Split, Meštrovićevo Šetalište 45, 21000 Split, Croatia
| | - Mirna Saraga-Babić
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia; (N.K.); (K.V.); (S.M.)
| | - Snježana Mardešić
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia; (N.K.); (K.V.); (S.M.)
| |
Collapse
|
2
|
Yousif G, Murugesan S, Djekidel MN, Terranegra A, Gentilcore G, Grivel JC, Al Khodor S. Distinctive blood and salivary proteomics signatures in Qatari individuals at high risk for cardiovascular disease. Sci Rep 2025; 15:4056. [PMID: 39901062 PMCID: PMC11790934 DOI: 10.1038/s41598-025-87596-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/20/2025] [Indexed: 02/05/2025] Open
Abstract
Cardiovascular disease (CVD) remains a leading cause of global morbidity and mortality. Timely diagnosis is important in reducing both short and long-term health complications. Saliva has emerged as a potential source for biomarker discovery, offering a non-invasive tool for early detection of individuals at elevated risk for CVD, yet large-scale extensive proteomic analysis using saliva for a comprehensive biomarker discovery remains limited. In an effort to develop a diagnostic tool using saliva samples, our study aims to assess the salivary and plasma proteomes in subjects with high risk of developing CVD using a large-scale proteomic approach. Leveraging on the SOMAscan platform, we analyzed 1,317 proteins in saliva and plasma collected from subjects at a high risk of CVD (HR-CVD) and compared the profiles to subjects with low risk of CVD (LR-CVD). Our analysis revealed significant differences in the plasma and salivary proteins between the two groups. Pathway enrichment analysis of the differentially detected proteins revealed that the immune system activation and extracellular matrix remodeling are the most enriched pathways in the CVD-HR group. Comparing proteomic signatures between plasma and saliva, we found approximately 42 and 17 differentially expressed proteins associated with CVD-HR uniquely expressed in plasma and saliva respectively. Additionally, we identified eight common CVD-risk biomarkers shared between both plasma and saliva, demonstrating promising diagnostic tools for identifying individuals at high risk of developing CVD. In conclusion, saliva proteomics holds a significant promise to identify subjects with a high risk to develop CVD. Further studies are needed to validate our findings.
Collapse
Affiliation(s)
- Ghada Yousif
- Research Department, Sidra Medicine, Doha, Qatar
| | | | | | | | | | | | | |
Collapse
|
3
|
Song Y, Bai G, Li X, Zhou L, Si Y, Liu X, Deng Y, Shi Y. Bioinformatics analysis of human kallikrein 5 ( KLK5) expression in metaplastic triple-negative breast cancer. CANCER INNOVATION 2023; 2:376-390. [PMID: 38090381 PMCID: PMC10686124 DOI: 10.1002/cai2.96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/25/2023] [Accepted: 09/03/2023] [Indexed: 10/15/2024]
Abstract
Background Metaplastic breast carcinoma (MBC) is a rare breast cancer subtype; most cases are triple-negative breast cancers (TNBCs) and are poorly responsive to conventional systemic therapy. Few potential diagnostic and prognostic markers for distinguishing between metaplastic TNBC and nonmetaplastic TNBC have been discovered. We performed bioinformatic analysis to explore the underlying mechanism by which metaplastic TNBC differs from nonmetaplastic TNBC and provides potential pathogenic genes of metaplastic TNBC. Methods Differentially expressed genes (DEGs) in metaplastic tumors and nonmetaplastic tumors from TNBC patients were screened using GSE165407. The GSE76275 data set and The Cancer Genome Atlas (TCGA) database were used to screen DEGs in TNBC and non-TNBC. Metascape and DAVID were used for the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis and Gene Ontology (GO) analysis of DEGs. Online databases, including UALCAN, GEPIA, HPA, Breast Cancer Gene-Expression Miner, and quantitative PCR and western blot, were used to examine KLK5 messenger RNA and protein expression in breast cancer. Analysis of KLK5‑associated genes was performed with TCGA data, and the LinkedOmics database was used to detect the genes co-expressed with KLK5. STRING (Search Tool for the Retrieval of Interacting Genes) and Cytoscape were used to screen for hub genes. Kaplan‑Meier plotter was used for survival analysis. Results KLK5 was identified among the DEGs in nonmetaplastic TNBC and metaplastic TNBC. The KLK5 gene was overexpressed in nonmetaplastic TNBC but downregulated in metaplastic TNBC. KEGG and GO analyses revealed that epithelial-to-mesenchymal transition was a pathogenic mechanism in metaplastic TNBC and an important pathway by which KLK5 and its associated genes DSG1 and DSG3 influence metaplastic TNBC progression. Prognosis analysis showed that only low expression of KLK5 in metaplastic TNBC had clinical significance. Conclusion Our research indicated that KLK5 may be a pivotal molecule with a key role in the mechanism of tumorigenesis in metaplastic TNBC.
Collapse
Affiliation(s)
- Yue Song
- Department of Phase I Clinical TrialTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Guiying Bai
- Department of Phase I Clinical TrialTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Xiaoqing Li
- Department of Phase I Clinical TrialTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Liyan Zhou
- Department of Phase I Clinical TrialTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Yiran Si
- Department of Phase I Clinical TrialTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Xiaohui Liu
- Department of Phase I Clinical TrialTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Yilin Deng
- Department of Phase I Clinical TrialTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Yehui Shi
- Department of Phase I Clinical TrialTianjin Medical University Cancer Institute and HospitalTianjinChina
- Medical Oncology Department of Breast CancerTianjin Medical University Cancer Institute and HospitalTianjinChina
- National Clinical Research Center for CancerTianjinChina
| |
Collapse
|
4
|
Ho HY, Lin CC, Lo YS, Chuang YC, Abomughaid MM, Hsieh MJ. Picrasidine J, a Dimeric β-Carboline-Type Alkaloid from Picrasma quassioides, Inhibits Metastasis of Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2023; 24:13230. [PMID: 37686036 PMCID: PMC10487624 DOI: 10.3390/ijms241713230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) are associated with recurrence, distant metastasis, and poor overall survival. This highlights the need for identifying potential therapeutics with minimal side-effects. The present study was designed to investigate the anticancer effects of picrasidine J, a dimeric β-carboline-type alkaloid isolated from the southern Asian plant Picrasma quassioides. The results showed that picrasidine J significantly inhibits HNSCC cell motility, migration, and invasion. Specifically, picrasidine J inhibited the EMT process by upregulating E-cadherin and ZO-1 and downregulating beta-catenin and Snail. Moreover, picrasidine J reduced the expression of the serine protease KLK-10. At the signaling level, the compound reduced the phosphorylation of ERK. All these factors collectively facilitated the inhibition of HNSCC metastasis with picrasidine J. Taken together, the study identifies picrasidine J as a potential anticancer compound of plant origin that might be used clinically to prevent the distant metastasis and progression of HNSCC.
Collapse
Affiliation(s)
- Hsin-Yu Ho
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan; (H.-Y.H.); (C.-C.L.); (Y.-S.L.); (Y.-C.C.)
| | - Chia-Chieh Lin
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan; (H.-Y.H.); (C.-C.L.); (Y.-S.L.); (Y.-C.C.)
| | - Yu-Sheng Lo
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan; (H.-Y.H.); (C.-C.L.); (Y.-S.L.); (Y.-C.C.)
| | - Yi-Ching Chuang
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan; (H.-Y.H.); (C.-C.L.); (Y.-S.L.); (Y.-C.C.)
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Ming-Ju Hsieh
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan; (H.-Y.H.); (C.-C.L.); (Y.-S.L.); (Y.-C.C.)
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
5
|
Zhou S, Liu S, Tian G, Zhao L, Wang H, Li Y, Shen Y, Han L. KLK5 is associated with the radioresistance, aggression, and progression of cervical cancer. Gynecol Oncol 2022; 166:138-147. [PMID: 35595569 DOI: 10.1016/j.ygyno.2022.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The role of kallikrein-related peptidase 5 (KLK5) has been studied in several diseases, including skin and ovarian cancers. However, its role in cervical cancer remains unclear, particularly in regulating the radiation resistance and growth of cervical cancer cells. Radiation resistance of cervical cancer is associated with local recurrence, distant metastasis, and reduced survival. METHODS We first analyzed radiotherapy-naive samples and relevant clinical data from patients with cervical cancer who received radiotherapy without surgery or other antitumor treatment from 2014 to 2016. Subsequently, biopsied tissues, in vitro cells, and transplanted tumors in nude mice were investigated. RESULTS Gene sequencing and clinical data analysis showed that KLK5 overexpression was associated with a poor prognosis post-radiotherapy. In in vitro cell and tumor transplantation experiments, KLK5 overexpression significantly increased radiation resistance. However, downregulating KLK5 expression increased radiosensitivity. CONCLUSION Our results confirm that KLK5 is vital to the radioresistance of cervical cancer, and provide a new target and marker for the treatment of radioresistance in cervical cancer.
Collapse
Affiliation(s)
- Shunqing Zhou
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China.
| | - Shuyan Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China.
| | - Geng Tian
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China.
| | - Lin Zhao
- Department of Obstetrics and Gynecology, The People's Hospital of LIAONING PROVINCE, Shenyang 110000, China
| | - Haichen Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Ying Li
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China
| | - Yannan Shen
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Liying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China.
| |
Collapse
|
6
|
Srinivasan S, Kryza T, Batra J, Clements J. Remodelling of the tumour microenvironment by the kallikrein-related peptidases. Nat Rev Cancer 2022; 22:223-238. [PMID: 35102281 DOI: 10.1038/s41568-021-00436-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 02/07/2023]
Abstract
Kallikrein-related peptidases (KLKs) are critical regulators of the tumour microenvironment. KLKs are proteolytic enzymes regulating multiple functions of bioactive molecules including hormones and growth factors, membrane receptors and the extracellular matrix architecture involved in cancer progression and metastasis. Perturbations of the proteolytic cascade generated by these peptidases, and their downstream signalling actions, underlie tumour emergence or blockade of tumour growth. Recent studies have also revealed their role in tumour immune suppression and resistance to cancer therapy. Here, we present an overview of the complex biology of the KLK family and its context-dependent nature in cancer, and discuss the different therapeutic strategies available to potentially target these proteases.
Collapse
Affiliation(s)
- Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Thomas Kryza
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Mater Research Institute, The University of Queensland, Woolloongabba, Brisbane, Queensland, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Centre for Genomics and Personalised Medicine, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia.
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
7
|
Lavergne M, Guillon-Munos A, Lenga Ma Bonda W, Attucci S, Kryza T, Barascu A, Moreau T, Petit-Courty A, Sizaret D, Courty Y, Iochmann S, Reverdiau P. Tissue factor pathway inhibitor 2 is a potent kallikrein-related protease 12 inhibitor. Biol Chem 2021; 402:1257-1268. [PMID: 33977679 DOI: 10.1515/hsz-2020-0389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/30/2021] [Indexed: 11/15/2022]
Abstract
The protease activities are tightly regulated by inhibitors and dysregulation contribute to pathological processes such as cancer and inflammatory disorders. Tissue factor pathway inhibitor 2 (TFPI-2) is a serine proteases inhibitor, that mainly inhibits plasmin. This protease activated matrix metalloproteases (MMPs) and degraded extracellular matrix. Other serine proteases are implicated in these mechanisms like kallikreins (KLKs). In this study, we identified for the first time that TFPI-2 is a potent inhibitor of KLK5 and 12. Computer modeling showed that the first Kunitz domain of TFPI-2 could interact with residues of KLK12 near the catalytic triad. Furthermore, like plasmin, KLK12 was able to activate proMMP-1 and -3, with no effect on proMMP-9. Thus, the inhibition of KLK12 by TFPI-2 greatly reduced the cascade activation of these MMPs and the cleavage of cysteine-rich 61, a matrix signaling protein. Moreover, when TFPI-2 bound to extracellular matrix, its classical localisation, the KLK12 inhibition was retained. Finally, TFPI-2 was downregulated in human non-small-cell lung tumour tissue as compared with non-affected lung tissue. These data suggest that TFPI-2 is a potent inhibitor of KLK12 and could regulate matrix remodeling and cancer progression mediated by KLK12.
Collapse
Affiliation(s)
- Marion Lavergne
- Université de Tours, F-37032 Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
| | - Audrey Guillon-Munos
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
- Groupe IMT, Bio3 Institute, 15 rue du Plat D'Etain, F-37020 Tours Cedex 1, France
| | - Woodys Lenga Ma Bonda
- Université de Tours, F-37032 Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
| | - Sylvie Attucci
- Université de Tours, F-37032 Tours, France
- INSERM, Imagerie et Cerveau (iBrain), UMR 1253, F-37000 Tours, France
| | - Thomas Kryza
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
- Mater Research Institute, The University of Queensland, Woollongabba Brisbane, QLD, Australia
| | - Aurélia Barascu
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
- UMR 8226-CNRS/UPMC, Institut de Biologie Physico-chimique, Sorbonne Université, 13 rue Pierre et Marie Curie, F-75005 Paris, France
| | - Thierry Moreau
- Université de Tours, F-37032 Tours, France
- INRA, UMR INRA 0083 - Biologie des Oiseaux et Aviculture (BOA), F-37380 Nouzilly, France
| | - Agnès Petit-Courty
- Université de Tours, F-37032 Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
| | - Damien Sizaret
- Département d'Anatomie et Cytologie Pathologiques, Hôpital Trousseau, CHRU de Tours, F-37044 Tours, France
| | - Yves Courty
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
| | - Sophie Iochmann
- Université de Tours, F-37032 Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
- Institut Universitaire de Technologie, F-37082 Tours, France
| | - Pascale Reverdiau
- Université de Tours, F-37032 Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
- Institut Universitaire de Technologie, F-37082 Tours, France
| |
Collapse
|
8
|
Abuduhadeer X, Xu X, Aihesan K, Yilihamu M, Zhao Y, Zhang W. Clinical significance of kallikrein 5 as a novel prognostic biomarker in gastric adenocarcinoma. J Clin Lab Anal 2021; 35:e23958. [PMID: 34510543 PMCID: PMC8529136 DOI: 10.1002/jcla.23958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 06/19/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Backgrounds Gastric cancer is one of the most common cancers with unsatisfied prognosis. It is challenging to predict gastric cancer prognosis due to its highly heterogeneous nature. Kallikrein 5 (KLK5) belongs to the family of kallikreins, which plays a crucial role in serine proteolysis and exerts diverse physiological functions. The role of KLK5 in human gastric adenocarcinoma (GAC) has not been elucidated. In the present study, we aimed to examine the expression level of KLK5 and dissect whether the KLK5 expression was associated with GAC prognosis. Patients and methods Clinicopathological analyses were performed in a retrospective GAC patient cohort (n = 138). The expression of KLK5 was tested by quantitative RT‐PCR and immunohistochemistry staining. The prognostic role of KLK5 in GAC was assessed by univariate and multivariate analyses. The effects of KLK5 on cell proliferation, migration, and invasion were examined through cellular experiments. Results The data showed that KLK5 expression was elevated in GAC tissues compared with normal stomach tissues. Protein expression of KLK5 was positively correlated with tumor invasion depth and lymph node metastasis. Patients with higher KLK5 expression had poorer overall survival. KLK5 was identified to be an independent risk factor according to multivariate analysis. Using human GAC cell lines, we found that KLK5 can promote tumor cell migration and invasion. Conclusions Our study demonstrated that higher expression of KLK5 was significantly correlated with a poorer prognosis of GAC patients, implying the potential of KLK5 as a novel prognostic biomarker in GAC.
Collapse
Affiliation(s)
- Xiaokaiti Abuduhadeer
- Day Operation Treatment Ward, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xincai Xu
- Department of Gastrointestinal (Tumor) Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Kamali Aihesan
- Day Operation Treatment Ward, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Maimaiti Yilihamu
- Day Operation Treatment Ward, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yanjun Zhao
- Day Operation Treatment Ward, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wenbin Zhang
- Department of Gastrointestinal (Tumor) Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
9
|
Chang JS, Kim N, Kim JY, Do SI, Cho Y, Kim HS, Kim YB. Kallikrein 5 overexpression is associated with poor prognosis in uterine cervical cancer. J Gynecol Oncol 2021; 31:e78. [PMID: 33078588 PMCID: PMC7593226 DOI: 10.3802/jgo.2020.31.e78] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/01/2020] [Accepted: 06/07/2020] [Indexed: 01/16/2023] Open
Abstract
Objective Kallikrein 5 (KLK5), which is frequently observed in normal cervico-vaginal fluid, is known to be related to prognosis in several solid tumors. We investigated the prognostic significance of KLK5 in uterine cervical cancer using tumor tissue microarray and immunohistochemistry staining. Methods We analyzed samples of 165 patients with uterine cervical cancer who received definitive radiation therapy between 2004 and 2012. We divided patients into two groups stratified by their KLK5 activity by immunohistochemistry staining: negative/weak (0–1+) (n=120 patients) and moderate/strong (2–3+) group (n=45 patients). Patient and tumor characteristics, patterns of failure, and survival outcomes were compared. Univariable and multivariable analyses were performed to identify prognostic factors. Results Patients with KLK5 2–3+ were younger (median: 52 vs. 60 years) and had frequent paraaortic lymph node involvement (40.0% vs. 18.3%) than those with KLK5 0–1+. With a median follow-up of 60.8 (interquartile range, 47.5–77.9) months, patients with KLK5 2–3+ had inferior 5-year locoregional recurrence-free survival and distant metastasis-free survival of 61.7% (vs. 77.5% in KLK5 0–1+ group) and 59.4% (vs. 72.8% in the KLK5 0–1+ group), respectively (all p<0.05). KLK5 2–3+ expression retained its significance after adjusting for other well-known prognostic factors of tumor size and stage in multivariable analysis. Conclusions KLK5 overexpression is associated with the aggressiveness of cervical cancer and may underlie the diminished response to conventional treatments. Therefore, KLK5 could be a reliable prognostic factor in cervical cancer.
Collapse
Affiliation(s)
- Jee Suk Chang
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Nalee Kim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Ye Kim
- Department of Pathology, Ilsan Paik Hospital, Inje University, Goyang, Korea
| | - Sung Im Do
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeona Cho
- Department of Radiation Oncology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Soo Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Yong Bae Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
10
|
Pampalakis G, Zingkou E, Panagiotidis C, Sotiropoulou G. Kallikreins emerge as new regulators of viral infections. Cell Mol Life Sci 2021; 78:6735-6744. [PMID: 34459952 PMCID: PMC8404027 DOI: 10.1007/s00018-021-03922-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/23/2021] [Accepted: 08/12/2021] [Indexed: 01/13/2023]
Abstract
Kallikrein-related peptidases (KLKs) or kallikreins have been linked to diverse (patho) physiological processes, such as the epidermal desquamation and inflammation, seminal clot liquefaction, neurodegeneration, and cancer. Recent mounting evidence suggests that KLKs also represent important regulators of viral infections. It is well-established that certain enveloped viruses, including influenza and coronaviruses, require proteolytic processing of their hemagglutinin or spike proteins, respectively, to infect host cells. Similarly, the capsid protein of the non-enveloped papillomavirus L1 should be proteolytically cleaved for viral uncoating. Consequently, extracellular or membrane-bound proteases of the host cells are instrumental for viral infections and represent potential targets for drug development. Here, we summarize how extracellular proteolysis mediated by the kallikreins is implicated in the process of influenza (and potentially coronavirus and papillomavirus) entry into host cells. Besides direct proteolytic activation of viruses, KLK5 and 12 promote viral entry indirectly through proteolytic cascade events, like the activation of thrombolytic enzymes that also can process hemagglutinin, while additional functions of KLKs in infection cannot be excluded. In the light of recent evidence, KLKs represent potential host targets for the development of new antivirals. Humanized animal models to validate their key functions in viral infections will be valuable.
Collapse
Affiliation(s)
- Georgios Pampalakis
- Department of Pharmacognosy-Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece.
| | - Eleni Zingkou
- Department of Pharmacy, School of Health Sciences, University of Patras, 265 04, Rion-Patras, Greece
| | - Christos Panagiotidis
- Department of Pharmacognosy-Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece
| | - Georgia Sotiropoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, 265 04, Rion-Patras, Greece
| |
Collapse
|
11
|
Sotiropoulou G, Zingkou E, Pampalakis G. Redirecting drug repositioning to discover innovative cosmeceuticals. Exp Dermatol 2021; 30:628-644. [PMID: 33544970 DOI: 10.1111/exd.14299] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/23/2021] [Accepted: 02/01/2021] [Indexed: 12/18/2022]
Abstract
Skin appearance is essential for self-esteem and quality of life; consequently, skin care products represent a huge market. In particular, cosmeceuticals constitute a hybrid category of skin care formulations, at the interphase of cosmetics and pharmaceuticals, rationally designed to target (patho) physiological mechanisms aiming to enhance skin health and appearance. Cosmeceuticals are marketed as anti-ageing, anti-wrinkle, hair regrowth, skin whitening and wound healing agents with special emphasis on scar-free healing. An overview on recent cutting-edge advances concerning the discovery and development of enhanced performance cosmeceuticals by drug repositioning approaches is presented here. In this context, we propose "target repositioning," a new term, to highlight that druggable protein targets implicated in multiple diseases (hubs in the diseasome) can be exploited to accelerate the discovery of molecularly targeted cosmeceuticals that can promote skin health as an added benefit, which is a novel concept not described before. In this direction, emphasis is placed on the role of mouse models, for often untreatable skin diseases, as well as recent breakthroughs on monogenic rare skin syndromes, in promoting compound repositioning to innovative cosmeceuticals.
Collapse
Affiliation(s)
- Georgia Sotiropoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Greece
| | - Eleni Zingkou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Greece
| | - Georgios Pampalakis
- Department of Pharmacognosy-Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
12
|
Tian J, Wang V, Wang N, Khadang B, Boudreault J, Bakdounes K, Ali S, Lebrun JJ. Identification of MFGE8 and KLK5/7 as mediators of breast tumorigenesis and resistance to COX-2 inhibition. Breast Cancer Res 2021; 23:23. [PMID: 33588911 PMCID: PMC7885389 DOI: 10.1186/s13058-021-01401-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 01/31/2021] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Cyclooxygenase 2 (COX-2) promotes stemness in triple negative breast cancer (TNBC), highlighting COX-2 as a promising therapeutic target in these tumors. However, to date, clinical trials using COX-2 inhibitors in breast cancer only showed variable patient responses with no clear significant clinical benefits, suggesting underlying molecular mechanisms contributing to resistance to COX-2 inhibitors. METHODS By combining in silico analysis of human breast cancer RNA-seq data with interrogation of public patient databases and their associated transcriptomic, genomic, and clinical profiles, we identified COX-2 associated genes whose expression correlate with aggressive TNBC features and resistance to COX-2 inhibitors. We then assessed their individual contributions to TNBC metastasis and resistance to COX-2 inhibitors, using CRISPR gene knockout approaches in both in vitro and in vivo preclinical models of TNBC. RESULTS We identified multiple COX-2 associated genes (TPM4, RGS2, LAMC2, SERPINB5, KLK7, MFGE8, KLK5, ID4, RBP1, SLC2A1) that regulate tumor lung colonization in TNBC. Furthermore, we found that silencing MFGE8 and KLK5/7 gene expression in TNBC cells markedly restored sensitivity to COX-2 selective inhibitor both in vitro and in vivo. CONCLUSIONS Together, our study supports the establishment and use of novel COX-2 inhibitor-based combination therapies as future strategies for TNBC treatment.
Collapse
Affiliation(s)
- Jun Tian
- Department of Medicine, McGill University Health Center, Cancer Research Program, 1001 Decarie Blvd, Bloc E, Suite E02.6224, Montreal, QC, H4A 3J1, Canada
| | - Vivian Wang
- Department of Medicine, McGill University Health Center, Cancer Research Program, 1001 Decarie Blvd, Bloc E, Suite E02.6224, Montreal, QC, H4A 3J1, Canada
| | - Ni Wang
- Department of Medicine, McGill University Health Center, Cancer Research Program, 1001 Decarie Blvd, Bloc E, Suite E02.6224, Montreal, QC, H4A 3J1, Canada
| | - Baharak Khadang
- Department of Medicine, McGill University Health Center, Cancer Research Program, 1001 Decarie Blvd, Bloc E, Suite E02.6224, Montreal, QC, H4A 3J1, Canada
| | - Julien Boudreault
- Department of Medicine, McGill University Health Center, Cancer Research Program, 1001 Decarie Blvd, Bloc E, Suite E02.6224, Montreal, QC, H4A 3J1, Canada
| | - Khldoun Bakdounes
- Department of Medicine, McGill University Health Center, Cancer Research Program, 1001 Decarie Blvd, Bloc E, Suite E02.6224, Montreal, QC, H4A 3J1, Canada
| | - Suhad Ali
- Department of Medicine, McGill University Health Center, Cancer Research Program, 1001 Decarie Blvd, Bloc E, Suite E02.6224, Montreal, QC, H4A 3J1, Canada
| | - Jean-Jacques Lebrun
- Department of Medicine, McGill University Health Center, Cancer Research Program, 1001 Decarie Blvd, Bloc E, Suite E02.6224, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
13
|
Peraman R, Sure SK, Dusthackeer VNA, Chilamakuru NB, Yiragamreddy PR, Pokuri C, Kutagulla VK, Chinni S. Insights on recent approaches in drug discovery strategies and untapped drug targets against drug resistance. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021; 7:56. [PMID: 33686369 PMCID: PMC7928709 DOI: 10.1186/s43094-021-00196-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 02/03/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Despite the various strategies undertaken in the clinical practice, the mortality rate due to antibiotic-resistant microbes has been markedly increasing worldwide. In addition to multidrug-resistant (MDR) microbes, the "ESKAPE" bacteria are also emerging. Of course, the infection caused by ESKAPE cannot be treated even with lethal doses of antibiotics. Now, the drug resistance is also more prevalent in antiviral, anticancer, antimalarial and antifungal chemotherapies. MAIN BODY To date, in the literature, the quantum of research reported on the discovery strategies for new antibiotics is remarkable but the milestone is still far away. Considering the need of the updated strategies and drug discovery approaches in the area of drug resistance among researchers, in this communication, we consolidated the insights pertaining to new drug development against drug-resistant microbes. It includes drug discovery void, gene paradox, transposon mutagenesis, vitamin biosynthesis inhibition, use of non-conventional media, host model, target through quorum sensing, genomic-chemical network, synthetic viability to targets, chemical versus biological space, combinational approach, photosensitization, antimicrobial peptides and transcriptome profiling. Furthermore, we optimally briefed about antievolution drugs, nanotheranostics and antimicrobial adjuvants and then followed by twelve selected new feasible drug targets for new drug design against drug resistance. Finally, we have also tabulated the chemical structures of potent molecules against antimicrobial resistance. CONCLUSION It is highly recommended to execute the anti-drug resistance research as integrated approach where both molecular and genetic research needs to be as integrative objective of drug discovery. This is time to accelerate new drug discovery research with advanced genetic approaches instead of conventional blind screening.
Collapse
Affiliation(s)
- Ramalingam Peraman
- RERDS-CPR, Raghavendra Institute of Pharmaceutical Education and Research (RIPER)-Autonomous, Anantapur, Andhra Pradesh India
| | - Sathish Kumar Sure
- RERDS-CPR, Raghavendra Institute of Pharmaceutical Education and Research (RIPER)-Autonomous, Anantapur, Andhra Pradesh India
| | - V. N. Azger Dusthackeer
- grid.417330.20000 0004 1767 6138ICMR-National Institute of Research in Tuberculosis, Chennai, Tamilnadu India
| | - Naresh Babu Chilamakuru
- RERDS-CPR, Raghavendra Institute of Pharmaceutical Education and Research (RIPER)-Autonomous, Anantapur, Andhra Pradesh India
| | - Padmanabha Reddy Yiragamreddy
- RERDS-CPR, Raghavendra Institute of Pharmaceutical Education and Research (RIPER)-Autonomous, Anantapur, Andhra Pradesh India
| | - Chiranjeevi Pokuri
- RERDS-CPR, Raghavendra Institute of Pharmaceutical Education and Research (RIPER)-Autonomous, Anantapur, Andhra Pradesh India
| | - Vinay Kumar Kutagulla
- RERDS-CPR, Raghavendra Institute of Pharmaceutical Education and Research (RIPER)-Autonomous, Anantapur, Andhra Pradesh India
| | - Santhivardhan Chinni
- RERDS-CPR, Raghavendra Institute of Pharmaceutical Education and Research (RIPER)-Autonomous, Anantapur, Andhra Pradesh India
| |
Collapse
|
14
|
Petrova E, Hovnanian A. Advances in understanding of Netherton syndrome and therapeutic implications. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1857724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Evgeniya Petrova
- Laboratory of genetic skin diseases, Université de Paris, Imagine Institute, INSERM UMR1163, Paris, France
| | - Alain Hovnanian
- Laboratory of genetic skin diseases, Université de Paris, Imagine Institute, INSERM UMR1163, Paris, France
- Departement of Genetics, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
15
|
Maffioli E, Jiang Z, Nonnis S, Negri A, Romeo V, Lietz CB, Hook V, Ristagno G, Baselli G, Kistler EB, Aletti F, O’Donoghue AJ, Tedeschi G. High-Resolution Mass Spectrometry-Based Approaches for the Detection and Quantification of Peptidase Activity in Plasma. Molecules 2020; 25:molecules25184071. [PMID: 32899982 PMCID: PMC7571063 DOI: 10.3390/molecules25184071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/28/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022] Open
Abstract
Proteomic technologies have identified 234 peptidases in plasma but little quantitative information about the proteolytic activity has been uncovered. In this study, the substrate profile of plasma proteases was evaluated using two nano-LC-ESI-MS/MS methods. Multiplex substrate profiling by mass spectrometry (MSP-MS) quantifies plasma protease activity in vitro using a global and unbiased library of synthetic peptide reporter substrates, and shotgun peptidomics quantifies protein degradation products that have been generated in vivo by proteases. The two approaches gave complementary results since they both highlight key peptidase activities in plasma including amino- and carboxypeptidases with different substrate specificity profiles. These assays provide a significant advantage over traditional approaches, such as fluorogenic peptide reporter substrates, because they can detect active plasma proteases in a global and unbiased manner, in comparison to detecting select proteases using specific reporter substrates. We discovered that plasma proteins are cleaved by endoproteases and these peptide products are subsequently degraded by amino- and carboxypeptidases. The exopeptidases are more active and stable in plasma and therefore were found to be the most active proteases in the in vitro assay. The protocols presented here set the groundwork for studies to evaluate changes in plasma proteolytic activity in shock.
Collapse
Affiliation(s)
- Elisa Maffioli
- Department of Veterinary Medicine, University of Milano, 20133 Milano, Italy; (E.M.); (S.N.); (A.N.); (V.R.)
- Centre for Nanostructured Materials and Interfaces (CIMAINA), University of Milano, 20133 Milano, Italy
| | - Zhenze Jiang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (Z.J.); (C.B.L.); (V.H.)
| | - Simona Nonnis
- Department of Veterinary Medicine, University of Milano, 20133 Milano, Italy; (E.M.); (S.N.); (A.N.); (V.R.)
- Centre for Nanostructured Materials and Interfaces (CIMAINA), University of Milano, 20133 Milano, Italy
| | - Armando Negri
- Department of Veterinary Medicine, University of Milano, 20133 Milano, Italy; (E.M.); (S.N.); (A.N.); (V.R.)
- Centre for Nanostructured Materials and Interfaces (CIMAINA), University of Milano, 20133 Milano, Italy
| | - Valentina Romeo
- Department of Veterinary Medicine, University of Milano, 20133 Milano, Italy; (E.M.); (S.N.); (A.N.); (V.R.)
| | - Christopher B. Lietz
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (Z.J.); (C.B.L.); (V.H.)
| | - Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (Z.J.); (C.B.L.); (V.H.)
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Giuseppe Ristagno
- Department of Pathophysiology and Transplantation, University of Milan, 20133 Milan, Italy;
| | - Giuseppe Baselli
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, 20133 Milan, Italy;
| | - Erik B. Kistler
- Department of Anesthesiology & Critical Care, University of California San Diego, La Jolla, CA 92093, USA;
- Department of Anesthesiology & Critical Care, VA San Diego HealthCare System, San Diego, CA 92161, USA
| | - Federico Aletti
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA;
| | - Anthony J. O’Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (Z.J.); (C.B.L.); (V.H.)
- Correspondence: (A.J.O.); (G.T.); Tel.: +1-8585345360 (A.J.O.); +39-02-50318127 (G.T.)
| | - Gabriella Tedeschi
- Department of Veterinary Medicine, University of Milano, 20133 Milano, Italy; (E.M.); (S.N.); (A.N.); (V.R.)
- Centre for Nanostructured Materials and Interfaces (CIMAINA), University of Milano, 20133 Milano, Italy
- Correspondence: (A.J.O.); (G.T.); Tel.: +1-8585345360 (A.J.O.); +39-02-50318127 (G.T.)
| |
Collapse
|
16
|
Anamthathmakula P, Winuthayanon W. Mechanism of semen liquefaction and its potential for a novel non-hormonal contraception†. Biol Reprod 2020; 103:411-426. [PMID: 32529252 PMCID: PMC7523691 DOI: 10.1093/biolre/ioaa075] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 12/21/2022] Open
Abstract
Semen liquefaction is a proteolytic process where a gel-like ejaculated semen becomes watery due to the enzymatic activity of prostate-derived serine proteases in the female reproductive tract. The liquefaction process is crucial for the sperm to gain their motility and successful transport to the fertilization site in Fallopian tubes (or oviducts in animals). Hyperviscous semen or failure in liquefaction is one of the causes of male infertility. Therefore, the biochemical inhibition of serine proteases in the female reproductive tract after ejaculation is a prime target for novel contraceptive development. Herein, we will discuss protein components in the ejaculates responsible for semen liquefaction and any developments of contraceptive methods in the past that involve the liquefaction process.
Collapse
Affiliation(s)
- Prashanth Anamthathmakula
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Wipawee Winuthayanon
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
17
|
Chen X, Leahy D, Van Haeften J, Hartfield P, Prentis PJ, van der Burg CA, Surm JM, Pavasovic A, Madio B, Hamilton BR, King GF, Undheim EAB, Brattsand M, Harris JM. A Versatile and Robust Serine Protease Inhibitor Scaffold from Actinia tenebrosa. Mar Drugs 2019; 17:E701. [PMID: 31842369 PMCID: PMC6950308 DOI: 10.3390/md17120701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022] Open
Abstract
Serine proteases play pivotal roles in normal physiology and a spectrum of patho-physiological processes. Accordingly, there is considerable interest in the discovery and design of potent serine protease inhibitors for therapeutic applications. This led to concerted efforts to discover versatile and robust molecular scaffolds for inhibitor design. This investigation is a bioprospecting study that aims to isolate and identify protease inhibitors from the cnidarian Actinia tenebrosa. The study isolated two Kunitz-type protease inhibitors with very similar sequences but quite divergent inhibitory potencies when assayed against bovine trypsin, chymostrypsin, and a selection of human sequence-related peptidases. Homology modeling and molecular dynamics simulations of these inhibitors in complex with their targets were carried out and, collectively, these methodologies enabled the definition of a versatile scaffold for inhibitor design. Thermal denaturation studies showed that the inhibitors were remarkably robust. To gain a fine-grained map of the residues responsible for this stability, we conducted in silico alanine scanning and quantified individual residue contributions to the inhibitor's stability. Sequences of these inhibitors were then used to search for Kunitz homologs in an A. tenebrosa transcriptome library, resulting in the discovery of a further 14 related sequences. Consensus analysis of these variants identified a rich molecular diversity of Kunitz domains and expanded the palette of potential residue substitutions for rational inhibitor design using this domain.
Collapse
Affiliation(s)
- Xingchen Chen
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Darren Leahy
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Jessica Van Haeften
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Perry Hartfield
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Peter J. Prentis
- School of Earth, Environmental and Biological Sciences, Science and Engineering Faculty, Queensland University of Technology, Brisbane, QLD 4000, Australia;
- Institute for Future Environments, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Chloé A. van der Burg
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Joachim M. Surm
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Ana Pavasovic
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Bruno Madio
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD 4072, Australia; (B.M.); (G.F.K.)
| | - Brett R. Hamilton
- Centre for Advanced Imaging, University of Queensland, St Lucia, QLD 4072, Australia; (B.R.H.); (E.A.B.U.)
| | - Glenn F. King
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD 4072, Australia; (B.M.); (G.F.K.)
| | - Eivind A. B. Undheim
- Centre for Advanced Imaging, University of Queensland, St Lucia, QLD 4072, Australia; (B.R.H.); (E.A.B.U.)
- Centre for Biodiversity Dynamics, Department of Biology, Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, PO Box 1066 Blindern, 0316 Oslo, Norway
| | - Maria Brattsand
- Department of Medical Biosciences, Umeå University, 901 87 Umeå, Sweden;
| | - Jonathan M. Harris
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia; (X.C.); (D.L.); (J.V.H.); (C.A.v.d.B.); (A.P.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| |
Collapse
|
18
|
Nauroy P, Nyström A. Kallikreins: Essential epidermal messengers for regulation of the skin microenvironment during homeostasis, repair and disease. Matrix Biol Plus 2019; 6-7:100019. [PMID: 33543017 PMCID: PMC7852331 DOI: 10.1016/j.mbplus.2019.100019] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
As the outermost layer of the skin, the epidermis is playing a major role in organism homeostasis providing the first barrier against external aggressions. Although considered as an extracellular matrix (ECM)-poor subtissue, the epidermal microenvironment is a key regulator of skin homeostasis and functionality. Among the proteins essential for upholding the epidermal microenvironment are the members of the kallikrein (KLK) family composed of 15 secreted serine proteases. Most of the members of these epithelial-specific proteins are present in skin and regulate skin desquamation and inflammation. However, although epidermal products, the consequences of KLK activities are not confined to the epidermis but widespread in the skin. In this review starting with the location and proteolytic activation cascade of KLKs, we present KLKs involvement in skin homeostasis, regeneration and pathology. KLKs have a large variety of substrates including ECM proteins, and evidence suggests that they are involved in the different steps of skin wound healing as discussed here. KLKs are also used as prognosis/diagnosis markers for many cancer types and we are focusing later on KLKs in cutaneous cancers, although their pathogenicity remains to be fully elucidated. Dysregulation of the KLK cascade is directly responsible for skin diseases with heavy inflammatory aspects, highlighting their involvement in skin immune homeostasis. Future studies will be needed to support the therapeutic potential of adjusting KLK activities for treatment of inflammatory skin diseases and wound healing pathologies. Regulation of the microenvironment even in an extracellular matrix-poor tissue can heavily impact organ function. Extracellular activities of kallikreins maintain skin homeostasis by regulating desquamation and inflammation. The activation of skin epidermal-specific kallikrein family of proteases is regulated by an intricate proteolytic cascade. Kallikreins are emerging as players during skin wound healing. Dysregulated kallikrein expression and activity occur in cancers and inflammatory skin diseases.
Collapse
Key Words
- AD, atopic dermatitis
- CDSN, corneodesmosin
- DSC1, desmocollin 1
- DSG1, desmoglein 1
- Diseases
- ECM, extracellular matrix
- EMT, epithelial-to-mesenchymal transition
- Epidermal microenvironment
- Epidermis
- Inflammation
- KLKs, kallikreins
- Kallikrein
- LEKTI, lympho-epithelial Kazal-type inhibitor
- NS, Netherton syndrome
- PAR1/2, protease activated-receptor 1/2
- SCC, squamous cell carcinoma
- Wound healing
- tPA, tissue plasminogen activator
- uPA, urokinase plasminogen activator
Collapse
Affiliation(s)
- Pauline Nauroy
- Department of Dermatology, Medical Center - University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Medical Center - University of Freiburg, Hauptstrasse 7, 79104 Freiburg, Germany
| |
Collapse
|
19
|
Papachristopoulou G, Malachias A, Devetzi M, Kamouza E, Scorilas A, Xynopoulos D, Talieri M. Uncovering the clinical impact of kallikrein-related peptidase 5 (KLK5) mRNA expression in the colorectal adenoma-carcinoma sequence. Clin Chem Lab Med 2019; 57:1251-1260. [PMID: 30759066 DOI: 10.1515/cclm-2018-1010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 01/08/2019] [Indexed: 12/11/2022]
Abstract
Background Kallikrein-related peptidases (KLKs) are a subgroup of serine proteases located on chromosome 19q13.3. Most KLKs have been extensively studied as potential biomarkers for several carcinomas and other diseases. KLK5 was originally identified from a keratinocyte library, and its enzyme was purified from the stratum corneum of human skin. KLK5 was shown to be differentially expressed in a variety of endocrine tumors, although it is not as yet examined widely in colorectal cancer (CRC). Methods In this study, we quantitatively assessed the mRNA expression status of KLK5 in 197 colorectal tissues from 133 patients (70 cancerous and their paired normal colonic mucosa for 64 of them, as well as 63 colorectal adenomas) by quantitative real-time PCR (qPCR) using TaqMan probes. Statistical analysis evaluated the results. Results It was shown that KLK5 expression is reduced following the histologically non-cancerous-adenoma sequence (p<0.001), whereas it is increased during the sequence adenoma-carcinoma (p<0.001). Furthermore, KLK5 positive expression is associated with positive nodal status (p=0.022), advanced tumor stage (p=0.038) and high histological grade (p=0.033). Cox univariate analysis revealed that KLK5 positive expression is associated with disease-free survival (DFS) (p=0.028) and overall survival (OS) of patients (p=0.048). Kaplan-Meyer survival models showed that patients with positive KLK5 expression have lower DFS (p=0.009) and OS (p=0.019). Receiver operating characteristic (ROC) analysis demonstrated for first time that KLK5 expression had significant discriminatory values between cancer and adenoma tissues (area under the curve [AUC] 0.77; 95% confidence interval [CI]=0.69-0.85, p=0.03). Conclusions KLK5 mRNA expression may be useful for the differentiation of CRC from colorectal adenoma and represents a potential unfavorable prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Georgia Papachristopoulou
- Department of Pathology, "Saint Savvas" Cancer Hospital of Athens, Athens, Greece.,Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Apostolos Malachias
- Department of Gastroenterology, "Saint Savvas" Cancer Hospital of Athens, Athens, Greece
| | - Marina Devetzi
- Department of Cellular Physiology, G. Papanicolaou Research Center of Oncology, "Saint Savvas" Cancer Hospital of Athens, Athens, Greece
| | - Evdoxia Kamouza
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Xynopoulos
- Department of Gastroenterology, "Saint Savvas" Cancer Hospital of Athens, Athens, Greece
| | - Maroulio Talieri
- Department of Cellular Physiology, G. Papanicolaou Research Center of Oncology, "Saint Savvas" Cancer Hospital of Athens, Athens, Greece
| |
Collapse
|
20
|
Gong W, Liu Y, Seidl C, Diamandis EP, Kiechle M, Drecoll E, Kotzsch M, Magdolen V, Dorn J. Quantitative assessment and clinical relevance of kallikrein-related peptidase 5 mRNA expression in advanced high-grade serous ovarian cancer. BMC Cancer 2019; 19:696. [PMID: 31307411 PMCID: PMC6631576 DOI: 10.1186/s12885-019-5901-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 07/02/2019] [Indexed: 11/10/2022] Open
Abstract
Background In ovarian cancer, dysregulation of mRNA expression of several components of the family of the kallikrein-related peptidases (KLKs) is observed. In this study, we have analyzed the KLK5 mRNA expression pattern in tumor tissue of patients suffering from high-grade serous ovarian cancer stage FIGO III/IV. Moreover, we have correlated the KLK5 mRNA levels with clinical outcome. Methods We assessed the mRNA expression levels of KLK5 in tumor tissue of 138 patients using quantitative PCR (qPCR). The mRNA levels were correlated with KLK5 antigen tumor tissue levels measured by ELISA (available for 41 of the 138 patients), established clinical features as well as patients’ outcome, using Chi-square-tests, Mann-Whitney U-tests and Spearman rank calculations as well as Cox regression models, Kaplan-Meier survival analysis and the log-rank test. Results A highly significant correlation between the mRNA expression levels and protein levels of KLK5 in tumor tissues was observed (rs = 0.683, p < 0.001). In univariate Cox regression analysis, elevated KLK5 mRNA expression was remarkably associated with reduced progression-free survival (PFS; p = 0.047), but not with overall survival (OS). Association of KLK5 mRNA expression with PFS was validated in silico using The Cancer Genome Atlas. For this, Affymetrix-based mRNA data (n = 377) were analyzed applying the Kaplan-Meier Plotter tool (p = 0.027). In multivariable Cox analysis, KLK5 mRNA values revealed a trend towards statistical significance for PFS (p = 0.095), whereas residual tumor mass (0 mm vs. > 0 mm), but not ascites fluid volume (≤500 ml vs. > 500 ml), remained an independent indicator for both OS and PFS (p < 0.001, p = 0.005, respectively). Conclusions These results obtained with a homogenous patient group with all patients suffering from advanced high-grade serous ovarian cancer support previous results suggesting elevated KLK5 mRNA levels as an unfavorable marker in ovarian cancer.
Collapse
Affiliation(s)
- Weiwei Gong
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
| | - Yueyang Liu
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
| | - Christof Seidl
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
| | - Eleftherios P Diamandis
- Division of Clinical Biochemistry, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Marion Kiechle
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
| | - Enken Drecoll
- Department of Institute of Pathology, Technical University of Munich, Munich, Germany
| | | | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany.
| | - Julia Dorn
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
| |
Collapse
|
21
|
Wei Q, Tian H, Zhang F, Sai W, Ge Y, Gao X, Yao W. Establishment of an HPLC-based method to identify key proteases of proteins in vitro. Anal Biochem 2019; 573:1-7. [PMID: 30849379 DOI: 10.1016/j.ab.2019.02.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 02/03/2023]
Abstract
Given that the biological functions of proteins may decrease or even be lost due to degradation by proteases, it is of great significance to identify potential proteases that degrade protein drugs during systemic circulation. In this work, we describe a method based on high-performance liquid chromatography (HPLC) to identify key proteases that degrade therapeutic proteins in blood, including endopeptidases and exopeptidases. Here, the degradation of proteins was detected by competition with standard substrates of proteases and is shown as the relative residue rate. Four protein drugs were subjected to this method, and the results suggested that growth hormone was degraded by aminopeptidase N and kallikrein-related peptidase 5, pertuzumab was hardly degraded by the proteases, factor VII was degraded by carboxypeptidase B, neprilysin, dipeptidyl peptidase-4 and peptidyl dipeptidase A, and fibrinogen was degraded by carboxypeptidase B and kallikrein-related peptidase 5, findings consistent with the literature. The results were confirmed by microscale thermophoresis; additionally, activity detection in vitro substantiated that the degradation of factor VII decreased its activity. We demonstrate that this method can be used to identify key proteases of proteins with high accuracy, precision and durability.
Collapse
Affiliation(s)
- Qingqing Wei
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Hong Tian
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Fan Zhang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Wenbo Sai
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yang Ge
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Wenbing Yao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
22
|
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: 2.0] [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
|
23
|
Pampalakis G, Zingkou E, Sotiropoulou G. KLK5, a novel potential suppressor of vaginal carcinogenesis. Biol Chem 2018; 399:1107-1111. [PMID: 29561728 DOI: 10.1515/hsz-2017-0302] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/11/2018] [Indexed: 01/15/2023]
Abstract
Vaginal cancer is rare and largely unexplored. We found here that kallikrein-related peptidase 5 (KLK5) is coordinately expressed along with other KLKs in all stratified epithelia, including vagina, pointing to potential role(s) in differentiation. Further, we propose that KLK5 could be implicated in vaginal cancer development based on the fact that Klk5-/- mice are prone to develop vaginal tumors when exposed to 7,12-dimethylbenz[a]anthracene. Nf-κb activation is markedly enhanced in Klk5-/-, leading to increased resistance to apoptosis of mutated vaginal cells. This explains the higher tumor numbers observed in Klk5-/- compared to wildtype. Thus, KLK5 may represent a putative suppressor of vaginal cancer.
Collapse
Affiliation(s)
- Georgios Pampalakis
- Department of Pharmacy, School of Health Sciences, University of Patras, GR-265 04 Rion-Patras, Greece
| | - Eleni Zingkou
- Department of Pharmacy, School of Health Sciences, University of Patras, GR-265 04 Rion-Patras, Greece
| | - Georgia Sotiropoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, GR-265 04 Rion-Patras, Greece
| |
Collapse
|
24
|
De Vita E, Schüler P, Lovell S, Lohbeck J, Kullmann S, Rabinovich E, Sananes A, Heßling B, Hamon V, Papo N, Hess J, Tate EW, Gunkel N, Miller AK. Depsipeptides Featuring a Neutral P1 Are Potent Inhibitors of Kallikrein-Related Peptidase 6 with On-Target Cellular Activity. J Med Chem 2018; 61:8859-8874. [DOI: 10.1021/acs.jmedchem.8b01106] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Elena De Vita
- Cancer Drug Development Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- Biosciences Faculty, University of Heidelberg, Heidelberg 69120, Germany
| | - Peter Schüler
- Cancer Drug Development Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Scott Lovell
- Department of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, U.K
| | - Jasmin Lohbeck
- Cancer Drug Development Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Sven Kullmann
- Cancer Drug Development Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Eitan Rabinovich
- Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Amiram Sananes
- Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Bernd Heßling
- Center for Molecular Biology, University of Heidelberg, Heidelberg 69120, Germany
| | - Veronique Hamon
- European Screening Centre, Biocity Scotland, University of Dundee, Newhouse ML1 5UH, U.K
| | - Niv Papo
- Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg 69120, Germany
- Research Group Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Edward W. Tate
- Department of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, U.K
| | - Nikolas Gunkel
- Cancer Drug Development Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- German Cancer Consortium (DKTK), Heidelberg 69120, Germany
| | - Aubry K. Miller
- Cancer Drug Development Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- German Cancer Consortium (DKTK), Heidelberg 69120, Germany
| |
Collapse
|
25
|
Filippou PS, Ren AH, Bala S, Papaioannou MD, Brinc D, Prassas I, Karakosta T, Diamandis EP. Biochemical characterization of human tissue kallikrein 15 and examination of its potential role in cancer. Clin Biochem 2018; 58:108-115. [PMID: 29928903 DOI: 10.1016/j.clinbiochem.2018.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/25/2018] [Accepted: 06/12/2018] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Human tissue kallikrein 15 (KLK15) is the last cloned member of the KLK-related gene family. Despite being implicated in multiple cancers, its pathophysiological role remains unknown. We aimed to biochemically characterize KLK15 and preliminarily study its role in cancer. DESIGN & METHODS Recombinant KLK15 protein was produced, purified to homogeneity and quantified by mass spectrometry (parallel reaction monitoring analysis). We profiled the enzymatic activity of KLK15 using fluorogenic peptide substrates, and performed kinetic analysis to discover the cleavage sites. As KLK15 has mainly been associated with prostate cancer, we used a degradomic approach and subsequent KEGG pathway analysis to identify a number of putative protein substrates in the KLK15-treated prostate cancer cell line PC3. RESULTS We discovered trypsin-like activity in KLK15, finding that it cleaves preferentially after arginine (R). The enzymatic activity of KLK15 was regulated by different factors such as pH, cations and serine protease inhibitors. Notably, we revealed that KLK15 most likely interacts with the extracellular matrix (ECM) receptor group. CONCLUSION To our knowledge, this is the first study that experimentally verifies the trypsin-like activity of KLK15. We show here for the first time that KLK15 may be able to cleave many ECM components, similar to several members of the KLK family. Thus the protease could potentially be linked to tumorigenesis by promoting metastasis via this mechanism.
Collapse
Affiliation(s)
- Panagiota S Filippou
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Annie H Ren
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Mount Sinai Hospital, Lunenfeld-Tanenbaum Research Institute, Toronto, Canada
| | - Sudarshan Bala
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | | | - Davor Brinc
- Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Ioannis Prassas
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Theano Karakosta
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Clinical Biochemistry, University Health Network, Toronto, Canada; Mount Sinai Hospital, Lunenfeld-Tanenbaum Research Institute, Toronto, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada.
| |
Collapse
|
26
|
Limón D, Fábrega MJ, Calpena AC, Badia J, Baldomà L, Pérez-García L. Multifunctional Serine Protease Inhibitor-Coated Water-Soluble Gold Nanoparticles as a Novel Targeted Approach for the Treatment of Inflammatory Skin Diseases. Bioconjug Chem 2018; 29:1060-1072. [PMID: 29406699 DOI: 10.1021/acs.bioconjchem.7b00717] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The overexpression and increased activity of the serine protease Kallikrein 5 (KLK5) is characteristic of inflammatory skin diseases such as Rosacea. The use of inhibitors of this enzyme-such as 4-(2-aminoethyl)benzenesulfonyl fluoride hydrochloride (AEBSF·HCl) or the anti-human recombinant Kallikrein 5 (anti-KLK5) antibody-in the treatment of the disease has been limited due to their low bioavailability, for which their immobilization in drug delivery agents can contribute to making serine protease inhibitors clinically useful. In this work, we synthesized gold nanoparticles (GNP) coated with a mixture of hydroxyl- and carboxyl-terminated thiolates (GNP.OH/COOH), whose carboxyl groups were used to further functionalize the nanoparticles with the serine protease inhibitor AEBSF·HCl either electrostatically or covalently (GNP.COOH AEBSF and GNP.AEBSF, respectively), or with the anti-KLK5 antibody (GNP.antiKLK5). The synthesized and functionalized GNP were highly water-soluble, and they were extensively characterized using UV-vis absorption spectroscopy, Transmission Electron Microscopy (TEM), Dynamic Light Scattering (DLS), and Thermogravimetric Analysis (TGA). GNP.OH/COOH and their subsequent functionalizations effectively inhibited KLK5 in vitro. Internalization of fluorophore-coated GNP.OH/COOH in human keratinocytes (HaCaT cells) was proven using confocal fluorescence microscopy. Cell viability assays revealed that the cytotoxicity of free AEBSF is importantly decreased when it is incorporated in the nanoparticles, either ionically (GNP.COOH AEBSF) or, most importantly, covalently (GNP.AEBSF). The functionalized nanoparticles GNP.AEBSF and GNP.antiKLK5 inhibited intracellular KLK5 activity in HaCaT cells and diminished secretion of IL-8 under inflammatory conditions triggered by TLR-2 ligands. This study points to the great potential of these GNP as a new intracellular delivery strategy for both small drugs and antibodies in the treatment of skin diseases such as Rosacea.
Collapse
|
27
|
Darling MR, Tsai S, Jackson-Boeters L, Daley TD, Diamandis EP. Human Kallikrein 3 (prostate-specific antigen) and Human Kallikrein 5 Expression in Salivary Gland Tumors. Int J Biol Markers 2018; 21:201-5. [PMID: 17177156 DOI: 10.1177/172460080602100401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The human kallikrein 5 protein (hK5) is expressed in many normal tissues, most notably in skin, breast, salivary gland and esophagus. It has also been shown to be a potential biomarker for breast, ovarian and testicular cancer. Human kallikrein 3 (hK3; prostate-specific antigen) is the most useful marker for adenocarcinoma of the prostate gland. The aim of this study was to determine whether hK3 and hK5 are expressed in salivary gland tissues and salivary gland tumors (both benign and malignant), in order to compare normal with tumor tissues. Pleomorphic adenomas, adenoid cystic carcinomas, polymorphous low-grade adenocarcinomas, acinic cell carcinomas, mucoepidermoid carcinomas and adenocarcinomas not otherwise specified of both minor and major salivary glands were examined. The results of this study indicate that most salivary gland tumors do not show high levels of expression of hK5. Staining was most prominent in keratinizing epithelia in pleomorphic adenomas. hK3 is not expressed in salivary gland tumors.
Collapse
Affiliation(s)
- M R Darling
- Division of Oral Pathology, Department of Pathology, University of Western Ontario, London, Ontario N6A 5C1, Canada.
| | | | | | | | | |
Collapse
|
28
|
Dufresne J, Florentinus-Mefailoski A, Ajambo J, Ferwa A, Bowden P, Marshall J. Random and independent sampling of endogenous tryptic peptides from normal human EDTA plasma by liquid chromatography micro electrospray ionization and tandem mass spectrometry. Clin Proteomics 2017; 14:41. [PMID: 29234243 PMCID: PMC5721679 DOI: 10.1186/s12014-017-9176-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/26/2017] [Indexed: 12/12/2022] Open
Abstract
Background Normal human EDTA plasma samples were collected on ice, processed ice cold, and stored in a freezer at – 80 °C prior to experiments. Plasma test samples from the – 80 °C freezer were thawed on ice or intentionally warmed to room temperature. Methods Protein content was measured by CBBR binding and the release of alcohol soluble amines by the Cd ninhydrin assay. Plasma peptides released over time were collected over C18 for random and independent sampling by liquid chromatography micro electrospray ionization and tandem mass spectrometry (LC–ESI–MS/MS) and correlated with X!TANDEM. Results Fully tryptic peptides by X!TANDEM returned a similar set of proteins, but was more computationally efficient, than “no enzyme” correlations. Plasma samples maintained on ice, or ice with a cocktail of protease inhibitors, showed lower background amounts of plasma peptides compared to samples incubated at room temperature. Regression analysis indicated that warming plasma to room temperature, versus ice cold, resulted in a ~ twofold increase in the frequency of peptide identification over hours–days of incubation at room temperature. The type I error rate of the protein identification from the X!TANDEM algorithm combined was estimated to be low compared to a null model of computer generated random MS/MS spectra. Conclusion The peptides of human plasma were identified and quantified with low error rates by random and independent sampling that revealed 1000s of peptides from hundreds of human plasma proteins from endogenous tryptic peptides. Electronic supplementary material The online version of this article (10.1186/s12014-017-9176-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jaimie Dufresne
- Ryerson University, 350 Victoria Street, Toronto, ON M5B 2K3 Canada
| | | | - Juliet Ajambo
- Ryerson University, 350 Victoria Street, Toronto, ON M5B 2K3 Canada
| | - Ammara Ferwa
- Ryerson University, 350 Victoria Street, Toronto, ON M5B 2K3 Canada
| | - Peter Bowden
- Ryerson University, 350 Victoria Street, Toronto, ON M5B 2K3 Canada
| | - John Marshall
- Ryerson University, 350 Victoria Street, Toronto, ON M5B 2K3 Canada.,Integrated BioBank of Luxembourg, 6 r. Nicolas-Ernest Barblé, Dudelange, 1210 Luxembourg
| |
Collapse
|
29
|
The proteins cleaved by endogenous tryptic proteases in normal EDTA plasma by C18 collection of peptides for liquid chromatography micro electrospray ionization and tandem mass spectrometry. Clin Proteomics 2017; 14:39. [PMID: 29213220 PMCID: PMC5712186 DOI: 10.1186/s12014-017-9174-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 11/21/2017] [Indexed: 02/08/2023] Open
Abstract
The tryptic peptides from ice cold versus room temperature plasma were identified by C18 liquid chromatography and micro electrospray ionization tandem mass spectrometry (LC–ESI–MS/MS). Samples collected on ice showed low levels of endogenous tryptic peptides compared to the same samples incubated at room temperature. Plasma on ice contained peptides from albumin, complement, and apolipoproteins and others that were observed by the X!TANDEM and SEQUEST algorithms. In contrast to ice cold samples, after incubation at room temperature, greater numbers of tryptic peptides from well characterized plasma proteins, and from cellular proteins were observed. A total of 583,927 precursor ions and MS/MS spectra were correlated to 94,669 best fit peptides that reduced to 22,287 correlations to the best accession within a gene symbol and to 7174 correlations to at least 510 gene symbols with ≥ 5 independent MS/MS correlations (peptide counts) that showed FDR q-values ranging from E−9 (i.e. FDR = 0.000000001) to E−227. A set of 528 gene symbols identified by X!TANDEM and SEQUEST including C4B showed ≥ fivefold variation between ice cold versus room temperature incubation. STRING analysis of the protein gene symbols observed from endogenous peptides in normal plasma revealed an extensive protein-interaction network of cellular factors associated with cell signalling and regulation, the formation of membrane bound organelles, cellular exosomes and exocytosis network proteins. Taken together the results indicated that a pool of cellular proteins, or protein complexes, in plasma are apparently not stable and degrade soon after incubation at room temperature.
Collapse
|
30
|
Solís-Calero C, Carvalho HF. KLK14 interactions with HAI-1 and HAI-2 serine protease inhibitors: A molecular dynamics and relative free-energy calculations study. Cell Biol Int 2017; 41:1246-1264. [PMID: 28817220 DOI: 10.1002/cbin.10839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 08/12/2017] [Indexed: 01/13/2023]
Abstract
Kallikrein 14 (KLK14) is a serine protease linked to several pathologies including prostate cancer and positively correlates with Gleason score. Though KLK14 functioning in cancer is poorly understood, it has been implicated in HGF/Met signaling, given that KLK14 proteolytically inhibits HGF activator-inhibitor 1 (HAI-1), which strongly inhibits pro-HGF activators, thereby contributing to tumor progression. In this work, KLK14 binding to either hepatocyte growth factor activator inhibitor type-1 (HAI-1) or type-2 (HAI-2) was essayed using homology modeling, molecular dynamic simulations and free-energy calculations through MM/PBSA and MM/GBSA. KLK14 was successfully modeled. Calculated free energies suggested higher binding affinity for the KLK14/HAI-1 interaction than for KLK14/HAI-2. This difference in binding affinity is largely explained by the higher stability of the hydrogen-bond networks in KLK14/HAI-1 along the simulation trajectory. A key arginine residue in both HAI-1 and HAI-2 is responsible for their interaction with the S1 pocket in KLK14. Additionally, MM/GBSA free-energy decomposition postulates that KLK14 Asp174 and Trp196 are hotspots for binding HAI-1 and HAI-2.
Collapse
Affiliation(s)
- Christian Solís-Calero
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, State University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
31
|
Biochemical and functional characterization of the human tissue kallikrein 9. Biochem J 2017; 474:2417-2433. [DOI: 10.1042/bcj20170174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 12/23/2022]
Abstract
Human tissue kallikrein 9 (KLK9) is a member of the kallikrein-related family of proteases. Despite its known expression profile, much less is known about the functional roles of this protease and its implications in normal physiology and disease. We present here the first data on the biochemical characterization of KLK9, investigate parameters that affect its enzymatic activity (such as inhibitors) and provide preliminary insights into its putative substrates. We show that mature KLK9 is a glycosylated chymotrypsin-like enzyme with strong preference for tyrosine over phenylalanine at the P1 cleavage position. The enzyme activity is enhanced by Mg2+ and Ca2+, but is reversibly attenuated by Zn2+. KLK9 is inhibited in vitro by many naturally occurring or synthetic protease inhibitors. Using a combination of degradomic and substrate specificity assays, we identified candidate KLK9 substrates in two different epithelial cell lines [the non-tumorigenic human keratinocyte cells (HaCaT) and the tumorigenic tongue squamous carcinoma cells (SCC9)]. Two potential KLK9 substrates [KLK10 and midkine (MDK)] were subjected to further validation. Taken together, our data delineate some functional and biochemical properties of KLK9 for future elucidation of the role of this enzyme in health and disease.
Collapse
|
32
|
Li S, Garcia M, Gewiss RL, Winuthayanon W. Crucial role of estrogen for the mammalian female in regulating semen coagulation and liquefaction in vivo. PLoS Genet 2017; 13:e1006743. [PMID: 28414719 PMCID: PMC5411094 DOI: 10.1371/journal.pgen.1006743] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 05/01/2017] [Accepted: 04/05/2017] [Indexed: 11/18/2022] Open
Abstract
Semen liquefaction changes semen from a gel-like to watery consistency and is required for sperm to gain mobility and swim to the fertilization site in the Fallopian tubes. Kallikrein-related peptidases 3 (KLK3) and other kallikrein-related peptidases from male prostate glands are responsible for semen liquefaction by cleaving gel-forming proteins (semenogelin and collagen). In a physiological context, the liquefaction process occurs within the female reproductive tract. How seminal proteins interact with the female reproductive environment is still largely unexplored. We previously reported that conditional genetic ablation of Esr1 (estrogen receptor α) in the epithelial cells of the female reproductive tract (Wnt7aCre/+;Esr1f/f) causes female infertility, partly due to a drastic reduction in the number of motile sperm entering the oviduct. In this study, we found that post-ejaculated semen from fertile wild-type males was solidified and the sperm were entrapped in Wnt7aCre/+;Esr1f/f uteri, compared to the watery semen (liquefied) found in Esr1f/f controls. In addition, semenogelin and collagen were not degraded in Wnt7aCre/+;Esr1f/f uteri. Amongst multiple gene families aberrantly expressed in the absence of epithelial ESR1, we have identified that a lack of Klks in the uterus is a potential cause for the liquefaction defect. Pharmacological inhibition of KLKs in the uterus replicated the phenotype observed in Wnt7aCre/+;Esr1f/f uteri, suggesting that loss of uterine and seminal KLK function causes this liquefaction defect. In human cervical cell culture, expression of several KLKs and their inhibitors (SPINKs) was regulated by estrogen in an ESR1-dependent manner. Our study demonstrates that estrogen/ESR1 signaling in the female reproductive tract plays an indispensable role in normal semen liquefaction, providing fundamental evidence that exposure of post-ejaculated semen to the suboptimal microenvironment in the female reproductive tract leads to faulty liquefaction and subsequently causes a fertility defect. Semen liquefaction has been considered to be solely modulated by prostate-derived kallikrein-related peptidases (KLKs), especially KLK3 (or prostate specific antigen). However, our research demonstrated that female mice lacking estrogen receptor alpha (ERα) in the uterine epithelial cells had a drastic decrease in Klk transcripts and semen from fertile males fails to liquefy within the uteri of these females. Therefore, our results provide a novel aspect that, due to an interplay between semen and female reproductive tract secretions, the physiology of semen liquefaction is more complicated than previously assumed. This information will advance research on semen liquefaction in the female reproductive tract, an area that has never been explored, and could lead to the development of diagnostic tools for unexplained infertility cases and non-invasive contraception technologies.
Collapse
Affiliation(s)
- Shuai Li
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Marleny Garcia
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Rachel L. Gewiss
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Wipawee Winuthayanon
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
- * E-mail:
| |
Collapse
|
33
|
Chen W, Kinsler VA, Macmillan D, Di WL. Tissue Kallikrein Inhibitors Based on the Sunflower Trypsin Inhibitor Scaffold - A Potential Therapeutic Intervention for Skin Diseases. PLoS One 2016; 11:e0166268. [PMID: 27824929 PMCID: PMC5100903 DOI: 10.1371/journal.pone.0166268] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/25/2016] [Indexed: 01/27/2023] Open
Abstract
Tissue kallikreins (KLKs), in particular KLK5, 7 and 14 are the major serine proteases in the skin responsible for skin shedding and activation of inflammatory cell signaling. In the normal skin, their activities are controlled by an endogenous protein protease inhibitor encoded by the SPINK5 gene. Loss-of-function mutations in SPINK5 leads to enhanced skin kallikrein activities and cause the skin disease Netherton Syndrome (NS). We have been developing inhibitors based on the Sunflower Trypsin Inhibitor 1 (SFTI-1) scaffold, a 14 amino acids head-to-tail bicyclic peptide with a disulfide bond. To optimize a previously reported SFTI-1 analogue (I10H), we made five analogues with additional substitutions, two of which showed improved inhibition. We then combined those substitutions and discovered a variant (Analogue 6) that displayed dual inhibition of KLK5 (tryptic) and KLK7 (chymotryptic). Analogue 6 attained a tenfold increase in KLK5 inhibition potency with an Isothermal Titration Calorimetry (ITC) Kd of 20nM. Furthermore, it selectively inhibits KLK5 and KLK14 over seven other serine proteases. Its biological function was ascertained by full suppression of KLK5-induced Protease-Activated Receptor 2 (PAR-2) dependent intracellular calcium mobilization and postponement of Interleukin-8 (IL-8) secretion in cell model. Moreover, Analogue 6 permeates through the cornified layer of in vitro organotypic skin equivalent culture and inhibits protease activities therein, providing a potential drug lead for the treatment of NS.
Collapse
Affiliation(s)
- Wenjie Chen
- Infection, Immunity and Inflammation Programme, Immunobiology Section, UCL GOS Institute of Child Health, London, United Kingdom
| | - Veronica A. Kinsler
- Genetics and Genomic Medicine Programme, UCL GOS Institute of Child Health, London, United Kingdom
| | - Derek Macmillan
- Department of Chemistry, University College London, London, United Kingdom
| | - Wei-Li Di
- Infection, Immunity and Inflammation Programme, Immunobiology Section, UCL GOS Institute of Child Health, London, United Kingdom
- * E-mail:
| |
Collapse
|
34
|
de Veer SJ, Furio L, Swedberg JE, Munro CA, Brattsand M, Clements JA, Hovnanian A, Harris JM. Selective Substrates and Inhibitors for Kallikrein-Related Peptidase 7 (KLK7) Shed Light on KLK Proteolytic Activity in the Stratum Corneum. J Invest Dermatol 2016; 137:430-439. [PMID: 27697464 DOI: 10.1016/j.jid.2016.09.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/05/2016] [Accepted: 09/16/2016] [Indexed: 01/01/2023]
Abstract
Proteases have pivotal roles in the skin's outermost layer, the epidermis. In the stratum corneum, serine proteases from the kallikrein-related peptidase (KLK) family have been implicated in several key homeostatic processes, including desquamation. However, the precise contribution of specific KLKs to each process remains unclear. To address this, we used a chemical biology approach and designed selective substrates and inhibitors for KLK7, the most abundant KLK protease in the stratum corneum. The resulting KLK7 inhibitor is the most potent inhibitor of this protease reported to date (Ki = 140 pM), and displays at least 1,000-fold selectivity over several proteases that are related by function (KLK5 and KLK14) or specificity (chymotrypsin). We then used substrates and inhibitors for KLK5, KLK7, and KLK14 to explore the activity of each protease in the stratum corneum using casein zymography and an ex vivo desquamation assay. These experiments provide the most detailed assessment of each KLK's contribution to corneocyte shedding in the plantar stratum corneum, revealing that inhibition of KLK7 alone is sufficient to block shedding, whereas KLK5 is also a major contributor. Collectively, these findings unveil chemical tools for studying KLK activity and demonstrate their potential for characterizing KLK biological functions in epidermal homeostasis.
Collapse
Affiliation(s)
- Simon J de Veer
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia; Laboratory of Genetic Skin Diseases, INSERM UMR 1163 and Imagine Institute of Genetic Diseases, Paris, France; Université Paris V Descartes-Sorbonne Paris Cité, Paris, France
| | - Laetitia Furio
- Laboratory of Genetic Skin Diseases, INSERM UMR 1163 and Imagine Institute of Genetic Diseases, Paris, France; Université Paris V Descartes-Sorbonne Paris Cité, Paris, France
| | - Joakim E Swedberg
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Christopher A Munro
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Maria Brattsand
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Judith A Clements
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia; Australian Prostate Cancer Research Centre, Translational Research Institute, Brisbane, Queensland, Australia
| | - Alain Hovnanian
- Laboratory of Genetic Skin Diseases, INSERM UMR 1163 and Imagine Institute of Genetic Diseases, Paris, France; Université Paris V Descartes-Sorbonne Paris Cité, Paris, France; Department of Genetics, Necker Hospital for Sick Children, Paris, France
| | - Jonathan M Harris
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.
| |
Collapse
|
35
|
Abstract
Cervical-vaginal fluid (CVF) is a complex biological fluid that hydrates the mucosa of the lower female reproductive system. In-depth proteomic and biochemical studies on CVF have revealed that it contains large amounts of endogenous proteases and protease inhibitors, including an abundance of several members of the tissue kallikrein-related peptidase (KLK) family. Despite their ubiquitous presence in human tissues and fluids, KLK expression levels vary considerably, with maximum expression observed in reproduction-related tissues and fluids. The roles of KLKs in the lower female reproductive system are not fully understood. The activation of KLKs in CVF is dependent on pH and various modes of KLK regulation in the vagina exist. KLKs have been postulated to have roles in physiological functions related to antimicrobial processes, vaginal and cervical epithelial desquamation, sperm transport, and the processing of fetal membranes as observed in preterm premature rupture of membranes. Increased understanding of the functional roles of KLKs in the lower female reproductive system could lead to new diagnostic and therapeutic modalities for conditions such as vaginal infections and vaginal atrophy.
Collapse
|
36
|
Wu Y, Chen Y, Li Q, Gong Y, Liu X, Bi L, Hu C. Upregulation of kallikrein-related peptidase 5 is associated with the malignant behavior of colorectal cancer. Mol Med Rep 2016; 14:2164-70. [DOI: 10.3892/mmr.2016.5516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 02/22/2016] [Indexed: 11/06/2022] Open
|
37
|
Filippou PS, Karagiannis GS, Musrap N, Diamandis EP. Kallikrein-related peptidases (KLKs) and the hallmarks of cancer. Crit Rev Clin Lab Sci 2016; 53:277-91. [PMID: 26886390 DOI: 10.3109/10408363.2016.1154643] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The kallikrein-related peptidases (KLKs) represent the largest family of serine proteases within the human genome and are expressed in various tissues. Although they regulate several important physiological functions, KLKs have also been implicated in numerous pathophysiological processes, including cancer. Growing evidence describing the deregulation of KLK expression and secretion, as well as activation in various malignancies, has uncovered their potential as mediators of cancer progression, biomarkers of disease and as candidate therapeutic targets. The diversity of signalling pathways and proteolytic cascades involving KLKs and their downstream targets appears to affect cancer biology through multiple mechanisms, including those related to the hallmarks of cancer. The aim of this review is to provide an update on the importance of KLK-driven molecular pathways in relation to cancer cell traits associated with the hallmarks of cancer and to highlight their potential in personalized therapeutics.
Collapse
Affiliation(s)
- Panagiota S Filippou
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada
| | - George S Karagiannis
- b Department of Anatomy & Structural Biology , Albert Einstein College of Medicine, Yeshiva University Bronx , New York , NY , USA
| | - Natasha Musrap
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada
| | - Eleftherios P Diamandis
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada .,c Department of Clinical Biochemistry , University Health Network , Toronto , ON , Canada , and.,d Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
38
|
The kallikrein-related peptidase family: Dysregulation and functions during cancer progression. Biochimie 2015; 122:283-99. [PMID: 26343558 DOI: 10.1016/j.biochi.2015.09.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/01/2015] [Indexed: 01/07/2023]
Abstract
Cancer is the second leading cause of death with 14 million new cases and 8.2 million cancer-related deaths worldwide in 2012. Despite the progress made in cancer therapies, neoplastic diseases are still a major therapeutic challenge notably because of intra- and inter-malignant tumour heterogeneity and adaptation/escape of malignant cells to/from treatment. New targeted therapies need to be developed to improve our medical arsenal and counter-act cancer progression. Human kallikrein-related peptidases (KLKs) are secreted serine peptidases which are aberrantly expressed in many cancers and have great potential in developing targeted therapies. The potential of KLKs as cancer biomarkers is well established since the demonstration of the association between KLK3/PSA (prostate specific antigen) levels and prostate cancer progression. In addition, a constantly increasing number of in vitro and in vivo studies demonstrate the functional involvement of KLKs in cancer-related processes. These peptidases are now considered key players in the regulation of cancer cell growth, migration, invasion, chemo-resistance, and importantly, in mediating interactions between cancer cells and other cell populations found in the tumour microenvironment to facilitate cancer progression. These functional roles of KLKs in a cancer context further highlight their potential in designing new anti-cancer approaches. In this review, we comprehensively review the biochemical features of KLKs, their functional roles in carcinogenesis, followed by the latest developments and the successful utility of KLK-based therapeutics in counteracting cancer progression.
Collapse
|
39
|
Lizama AJ, Andrade Y, Colivoro P, Sarmiento J, Matus CE, Gonzalez CB, Bhoola KD, Ehrenfeld P, Figueroa CD. Expression and bioregulation of the kallikrein-related peptidases family in the human neutrophil. Innate Immun 2015; 21:575-586. [DOI: 10.1177/1753425914566083] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
The family of kallikrein-related peptidases (KLKs) has been identified in a variety of immunolabeled human tissue sections, but no previous study has experimentally confirmed their presence in the human neutrophil. We have investigated the expression and bioregulation of particular KLKs in the human neutrophil and, in addition, examined whether stimulation by a kinin B1receptor (B1R) agonist or fMet-Leu-Phe (fMLP) induces their secretion. Western blot analysis of neutrophil homogenates indicated that the MM of the KLKs ranged from 27 to 50 kDa. RT-PCR showed that blood neutrophils expressed only KLK1, KLK4, KLK10, KLK13, KLK14 and KLK15 mRNAs, whereas the non-differentiated HL-60 cells expressed most of them, with exception of KLK3 and KLK7. Nevertheless, mRNAs for KLK2, KLK5, KLK6 and KLK9 that were previously undetectable appeared after challenging with a mixture of cytokines. Both kinin B1R agonist and fMLP induced secretion of KLK1, KLK6, KLK10, KLK13 and KLK14 into the culture medium in similar amounts, whereas the B1R agonist caused the release of lower amounts of KLK2, KLK4 and KLK5. When secreted, the differing proteolytic activity of KLKs provides the human neutrophil with a multifunctional enzymatic capacity supporting a new dimension for its role in human disorders of diverse etiology.
Collapse
Affiliation(s)
- Alejandro J Lizama
- Laboratorio de Patologia Celular, Instituto de Anatomia, Histologia y Patologia, Universidad Austral de Chile, Valdivia, Chile
| | - Yessica Andrade
- Laboratorio de Patologia Celular, Instituto de Anatomia, Histologia y Patologia, Universidad Austral de Chile, Valdivia, Chile
| | - Patricio Colivoro
- Laboratorio de Patologia Celular, Instituto de Anatomia, Histologia y Patologia, Universidad Austral de Chile, Valdivia, Chile
| | - Jose Sarmiento
- Instituto de Fisiologia, Universidad Austral de Chile, Valdivia, Chile
| | - Carola E Matus
- Laboratorio de Patologia Celular, Instituto de Anatomia, Histologia y Patologia, Universidad Austral de Chile, Valdivia, Chile
| | - Carlos B Gonzalez
- Instituto de Fisiologia, Universidad Austral de Chile, Valdivia, Chile
| | - Kanti D Bhoola
- Laboratorio de Patologia Celular, Instituto de Anatomia, Histologia y Patologia, Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Laboratorio de Patologia Celular, Instituto de Anatomia, Histologia y Patologia, Universidad Austral de Chile, Valdivia, Chile
| | - Carlos D Figueroa
- Laboratorio de Patologia Celular, Instituto de Anatomia, Histologia y Patologia, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
40
|
Jimenez L, Sharma VP, Condeelis J, Harris T, Ow TJ, Prystowsky MB, Childs G, Segall JE. MicroRNA-375 Suppresses Extracellular Matrix Degradation and Invadopodial Activity in Head and Neck Squamous Cell Carcinoma. Arch Pathol Lab Med 2015; 139:1349-61. [PMID: 26172508 DOI: 10.5858/arpa.2014-0471-oa] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
CONTEXT Head and neck squamous cell carcinoma (HNSCC) is a highly invasive cancer with an association with locoregional recurrence and lymph node metastasis. We have previously reported that low microRNA-375 (miR-375) expression levels correlate with poor patient survival, increased locoregional recurrence, and distant metastasis. Increasing miR-375 expression in HNSCC cell lines to levels found in normal cells results in suppressed invasive properties. HNSCC invasion is mediated in part by invadopodia-associated degradation of the extracellular matrix. OBJECTIVE To determine whether elevated miR-375 expression in HNSCC cell lines also affects invadopodia formation and activity. DESIGN For evaluation of the matrix degradation properties of the HNSCC lines, an invadopodial matrix degradation assay was used. The total protein levels of invadopodia-associated proteins were measured by Western blot analyses. Immunoprecipitation experiments were conducted to evaluate the tyrosine phosphorylation state of cortactin. Human protease arrays were used for the detection of the secreted proteases. Quantitative real time-polymerase chain reaction measurements were used to evaluate the messenger RNA (mRNA) expression of the commonly regulated proteases. RESULTS Increased miR-375 expression in HNSCC cells suppresses extracellular matrix degradation and reduces the number of mature invadopodia. Higher miR-375 expression does not reduce cellular levels of selected invadopodia-associated proteins, nor is tyrosine phosphorylation of cortactin altered. However, HNSCC cells with higher miR-375 expression had significant reductions in the mRNA expression levels and secreted levels of specific proteases. CONCLUSIONS MicroRNA-375 regulates invadopodia maturation and function potentially by suppressing the expression and secretion of proteases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jeffrey E Segall
- From the Departments of Pathology (Ms Jimenez and Drs Harris, Ow, Prystowsky, Childs, and Segall) and Anatomy & Structural Biology (Ms Jimenez and Drs Sharma, Condeelis, and Segall), Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
41
|
Sidiropoulos KG, White NMA, Bui A, Ding Q, Boulos P, Pampalakis G, Khella H, Samuel JN, Sotiropoulou G, Yousef GM. Kallikrein-related peptidase 5 induces miRNA-mediated anti-oncogenic pathways in breast cancer. Oncoscience 2014; 1:709-24. [PMID: 25593998 PMCID: PMC4278268 DOI: 10.18632/oncoscience.91] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 10/23/2014] [Indexed: 12/15/2022] Open
Abstract
Kallikrein-related peptidase 5 (KLK5) displays aberrant expression in cancer. Recently, we showed KLK5 reconstitution in breast cancer cell lines suppresses malignancy. Present study aims to investigate the functional KLK5 mediated miRNA network on breast cancer progression, molecular subtype and survival. 28 miRNAs were up-regulated and 62 miRNAs were down-regulated upon KLK5 expression. Extracellular matrix (ECM) molecules and cell-adhesion pathways were the most significant KLK5-induced miRNA-mediated regulatory targets. Validation from The Cancer Genome Atlas (TCGA) database indicated KLK5 was specifically down-regulated in luminal B and basal-like breast cancer subtypes. There was a correlation between KLK5, miRNAs and their downstream ECM gene targets. Long-term patient survival correlated with dysregulation of KLK5 and interacting ECM target genes. It suggests biological differences between breast cancer molecular subtypes, patient survival, and their propensity for invasion and metastasis can be explained in part by altered miRNA networks induced by KLK5 dysregulation. We provide the first evidence that KLK5 can affect miRNA networks, which regulate MMPs and other novel ECM targets and a new compelling hypothesis of interplay between serine proteases and miRNAs. We developed a combined KLK5-(ITGB1+COL12A1) predictive score for recurrence-free survival that could be exploited in clinical applications.
Collapse
Affiliation(s)
- Konstantinos G Sidiropoulos
- The Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute and Department of Laboratory Medicine, St. Michael's Hospital, Toronto, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Nicole M A White
- The Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute and Department of Laboratory Medicine, St. Michael's Hospital, Toronto, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Anna Bui
- The Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute and Department of Laboratory Medicine, St. Michael's Hospital, Toronto, Canada
| | - Qiang Ding
- The Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute and Department of Laboratory Medicine, St. Michael's Hospital, Toronto, Canada
| | - Peter Boulos
- The Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute and Department of Laboratory Medicine, St. Michael's Hospital, Toronto, Canada
| | - Georgios Pampalakis
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Greece
| | - Heba Khella
- The Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute and Department of Laboratory Medicine, St. Michael's Hospital, Toronto, Canada
| | - Joseph N Samuel
- The Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute and Department of Laboratory Medicine, St. Michael's Hospital, Toronto, Canada
| | - Georgia Sotiropoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Greece
| | - George M Yousef
- The Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute and Department of Laboratory Medicine, St. Michael's Hospital, Toronto, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
42
|
Skala W, Utzschneider DT, Magdolen V, Debela M, Guo S, Craik CS, Brandstetter H, Goettig P. Structure-function analyses of human kallikrein-related peptidase 2 establish the 99-loop as master regulator of activity. J Biol Chem 2014; 289:34267-83. [PMID: 25326387 PMCID: PMC4256358 DOI: 10.1074/jbc.m114.598201] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Human kallikrein-related peptidase 2 (KLK2) is a tryptic serine protease predominantly expressed in prostatic tissue and secreted into prostatic fluid, a major component of seminal fluid. Most likely it activates and complements chymotryptic KLK3 (prostate-specific antigen) in cleaving seminal clotting proteins, resulting in sperm liquefaction. KLK2 belongs to the “classical” KLKs 1–3, which share an extended 99- or kallikrein loop near their non-primed substrate binding site. Here, we report the 1.9 Å crystal structures of two KLK2-small molecule inhibitor complexes. In both structures discontinuous electron density for the 99-loop indicates that this loop is largely disordered. We provide evidence that the 99-loop is responsible for two biochemical peculiarities of KLK2, i.e. reversible inhibition by micromolar Zn2+ concentrations and permanent inactivation by autocatalytic cleavage. Indeed, several 99-loop mutants of KLK2 displayed an altered susceptibility to Zn2+, which located the Zn2+ binding site at the 99-loop/active site interface. In addition, we identified an autolysis site between residues 95e and 95f in the 99-loop, whose elimination prevented the mature enzyme from limited autolysis and irreversible inactivation. An exhaustive comparison of KLK2 with related structures revealed that in the KLK family the 99-, 148-, and 220-loop exist in open and closed conformations, allowing or preventing substrate access, which extends the concept of conformational selection in trypsin-related proteases. Taken together, our novel biochemical and structural data on KLK2 identify its 99-loop as a key player in activity regulation.
Collapse
Affiliation(s)
- Wolfgang Skala
- From the Division of Structural Biology, Department of Molecular Biology, University of Salzburg, A-5020 Salzburg, Austria
| | - Daniel T Utzschneider
- Klinische Forschergruppe der Frauenklinik, Klinikum rechts der Isar der TU München, D-81675 Munich, Germany
| | - Viktor Magdolen
- Klinische Forschergruppe der Frauenklinik, Klinikum rechts der Isar der TU München, D-81675 Munich, Germany
| | - Mekdes Debela
- Max-Planck-Institut for Biochemistry, Proteinase Research Group, D-82152 Martinsried, Germany, and
| | - Shihui Guo
- From the Division of Structural Biology, Department of Molecular Biology, University of Salzburg, A-5020 Salzburg, Austria
| | - Charles S Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143
| | - Hans Brandstetter
- From the Division of Structural Biology, Department of Molecular Biology, University of Salzburg, A-5020 Salzburg, Austria
| | - Peter Goettig
- From the Division of Structural Biology, Department of Molecular Biology, University of Salzburg, A-5020 Salzburg, Austria,
| |
Collapse
|
43
|
Shariff L, Zhu Y, Cowper B, Di WL, Macmillan D. Sunflower trypsin inhibitor (SFTI-1) analogues of synthetic and biological origin via N→S acyl transfer: potential inhibitors of human Kallikrein-5 (KLK5). Tetrahedron 2014. [DOI: 10.1016/j.tet.2014.06.059] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
44
|
Pampalakis G, Obasuyi O, Papadodima O, Chatziioannou A, Zoumpourlis V, Sotiropoulou G. The KLK5 protease suppresses breast cancer by repressing the mevalonate pathway. Oncotarget 2014; 5:2390-403. [PMID: 24158494 PMCID: PMC4058013 DOI: 10.18632/oncotarget.1235] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 09/01/2013] [Indexed: 12/21/2022] Open
Abstract
Kallikrein-related peptidase 5 (KLK5) displays aberrant expression in cancer. However, any functional association is missing. Here, we show that reconstitution of KLK5 expression in non-expressing MDA-MB-231 breast cancer cells suppresses malignancy in vitro and in vivo dose-dependently. Reactivation of KLK5 suppressed key EMT genes. Unexpectedly, we identified altered expression of genes encoding enzymes of the mevalonate pathway typical of those observed upon cholesterol starvation. Consistently, we found that SREBF1, the master regulator of the mevalonate pathway was induced. KLK5 re-expression leads to reduced cellular cholesterol and fatty acid synthesis and enhanced uptake of LDL-cholesterol. Suppression of the mevalonate pathway in KLK5 transfectants was further shown by reduced synthesis of isoprenoids. Indeed, we found diminished levels of active RhoA, a signaling oncoprotein that requires prenylation for activation. We propose that reduced RhoA activation plays a dominant role in suppression of malignancy by KLK5, since geranylgeranyl pyrophosphate restored active RhoA in KLK5-reverted cells resulting in increased malignancy. For the first time, we suggest that a protease may suppress breast cancer by modulating the mevalonate pathway.
Collapse
Affiliation(s)
| | - Osahon Obasuyi
- Department of Pharmacy, University of Patras, Rion-Patras 26500
| | | | | | | | | |
Collapse
|
45
|
Fuhrman-Luck RA, Silva ML, Dong Y, Irving-Rodgers H, Stoll T, Hastie ML, Loessner D, Gorman JJ, Clements JA. Proteomic and other analyses to determine the functional consequences of deregulated kallikrein-related peptidase (KLK) expression in prostate and ovarian cancer. Proteomics Clin Appl 2014; 8:403-15. [PMID: 24535680 DOI: 10.1002/prca.201300098] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/23/2013] [Accepted: 11/30/2013] [Indexed: 02/06/2023]
Abstract
Rapidly developing proteomic tools are improving detection of deregulated kallikrein-related peptidase (KLK) expression, at the protein level, in prostate and ovarian cancer, as well as facilitating the determination of functional consequences downstream. MS-driven proteomics uniquely allows for the detection, identification, and quantification of thousands of proteins in a complex protein pool, and this has served to identify certain KLKs as biomarkers for these diseases. In this review, we describe applications of this technology in KLK biomarker discovery and elucidate MS-based techniques that have been used for unbiased, global screening of KLK substrates within complex protein pools. Although MS-based KLK degradomic studies are limited to date, they helped to discover an array of novel KLK substrates. Substrates identified by MS-based degradomics are reported with improved confidence over those determined by incubating a purified or recombinant substrate and protease of interest, in vitro. We propose that these novel proteomic approaches represent the way forward for KLK research, in order to correlate proteolysis of biological substrates with tissue-related consequences, toward clinical targeting of KLK expression and function for cancer diagnosis, prognosis, and therapies.
Collapse
Affiliation(s)
- Ruth Anna Fuhrman-Luck
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dong Y, Loessner D, Irving-Rodgers H, Obermair A, Nicklin JL, Clements JA. Metastasis of ovarian cancer is mediated by kallikrein related peptidases. Clin Exp Metastasis 2014; 31:135-47. [PMID: 24043563 PMCID: PMC3892111 DOI: 10.1007/s10585-013-9615-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/26/2013] [Indexed: 12/16/2022]
Abstract
Ovarian cancer, in particular epithelial ovarian cancer (EOC), is commonly diagnosed when the tumor has metastasized into the abdominal cavity with an accumulation of ascites fluid. Combining histopathology and genetic variations, EOC can be sub-grouped into Type-I and Type-II tumors, of which the latter are more aggressive and metastatic. Metastasis and chemoresistance are the key events associated with the tumor microenvironment that lead to a poor patient outcome. Kallikrein-related peptidases (KLKs) are aberrantly expressed in EOC, in particular, in the more metastatic Type-II tumors. KLKs are a family of 15 serine proteases that are expressed in diverse human tissues and involved in various patho-physiological processes. As extracellular enzymes, KLKs function in the hydrolysis of growth factors, proteases, cell membrane bound receptors, adhesion proteins, and cytokines initiating intracellular signaling pathways and their downstream events. High KLK levels are differentially associated with the prognosis of ovarian cancer patients, suggesting that they not only have application as biomarkers but also function in disease progression, and therefore are potential therapeutic targets. Recent studies have demonstrated the function of these proteases in promoting and/or suppressing the invasive behavior of ovarian cancer cells in metastasis in vitro and in vivo. Both conventional cell culture methods and three-dimensional platforms have been applied to mimic the ovarian cancer microenvironment of patients, such as the solid stromal matrix and ascites fluid. Here we summarize published studies to provide an overview of our understanding of the role of KLKs in EOC, and to lay the foundation for future research directions.
Collapse
Affiliation(s)
- Ying Dong
- Cancer Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Brisbane, QLD, 4059, Australia,
| | | | | | | | | | | |
Collapse
|
47
|
Shahinian H, Loessner D, Biniossek ML, Kizhakkedathu JN, Clements JA, Magdolen V, Schilling O. Secretome and degradome profiling shows that Kallikrein-related peptidases 4, 5, 6, and 7 induce TGFβ-1 signaling in ovarian cancer cells. Mol Oncol 2013; 8:68-82. [PMID: 24120346 PMCID: PMC5528505 DOI: 10.1016/j.molonc.2013.09.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/04/2013] [Accepted: 09/18/2013] [Indexed: 11/16/2022] Open
Abstract
Kallikrein‐related peptidases, in particular KLK4, 5, 6 and 7 (4–7), often have elevated expression levels in ovarian cancer. In OV‐MZ‐6 ovarian cancer cells, combined expression of KLK4–7 reduces cell adhesion and increases cell invasion and resistance to paclitaxel. The present work investigates how KLK4–7 shape the secreted proteome (“secretome”) and proteolytic profile (“degradome”) of ovarian cancer cells. The secretome comparison consistently identified >900 proteins in three replicate analyses. Expression of KLK4–7 predominantly affected the abundance of proteins involved in cell–cell communication. Among others, this includes increased levels of transforming growth factor β‐1 (TGFβ‐1). KLK4–7 co‐transfected OV‐MZ‐6 cells share prominent features of elevated TGFβ‐1 signaling, including increased abundance of neural cell adhesion molecule L1 (L1CAM). Augmented levels of TGFβ‐1 and L1CAM upon expression of KLK4–7 were corroborated in vivo by an ovarian cancer xenograft model. The degradomic analysis showed that KLK4–7 expression mostly affected cleavage sites C‐terminal to arginine, corresponding to the preference of kallikreins 4, 5 and 6. Putative kallikrein substrates include chemokines, such as growth differentiation factor 15 (GDF 15) and macrophage migration inhibitory factor (MIF). Proteolytic maturation of TGFβ‐1 was also elevated. KLK4–7 have a pronounced, yet non‐degrading impact on the secreted proteome, with a strong association between these proteases and TGFβ‐1 signaling in tumor biology. Expression of KLK4, 5, 6, and 7 yields subtle secretome and degradome alterations. Pro‐ and anti‐tumorigenic proteins are affected. TGFβ‐1 signaling is elevated. Degradomic analysis supports cell‐contextual KLK activity. Potential substrates of KLK4–7 have been identified.
Collapse
Affiliation(s)
- Hasmik Shahinian
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | - Daniela Loessner
- Cancer Program, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | - Martin L Biniossek
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Judith A Clements
- Cancer Program, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, D-81675, Germany
| | - Oliver Schilling
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, D-79104 Freiburg, Germany.
| |
Collapse
|
48
|
Loessner D, Kobel S, Clements JA, Lutolf MP, Hutmacher DW. Hydrogel Microwell Arrays Allow the Assessment of Protease-Associated Enhancement of Cancer Cell Aggregation and Survival. MICROARRAYS 2013; 2:208-27. [PMID: 27605189 PMCID: PMC5003461 DOI: 10.3390/microarrays2030208] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 07/31/2013] [Accepted: 08/13/2013] [Indexed: 01/03/2023]
Abstract
Current routine cell culture techniques are only poorly suited to capture the physiological complexity of tumor microenvironments, wherein tumor cell function is affected by intricate three-dimensional (3D), integrin-dependent cell-cell and cell-extracellular matrix (ECM) interactions. 3D cell cultures allow the investigation of cancer-associated proteases like kallikreins as they degrade ECM proteins and alter integrin signaling, promoting malignant cell behaviors. Here, we employed a hydrogel microwell array platform to probe using a high-throughput mode how ovarian cancer cell aggregates of defined size form and survive in response to the expression of kallikreins and treatment with paclitaxel, by performing microscopic, quantitative image, gene and protein analyses dependent on the varying microwell and aggregate sizes. Paclitaxel treatment increased aggregate formation and survival of kallikrein-expressing cancer cells and levels of integrins and integrin-related factors. Cancer cell aggregate formation was improved with increasing aggregate size, thereby reducing cell death and enhancing integrin expression upon paclitaxel treatment. Therefore, hydrogel microwell arrays are a powerful tool to screen the viability of cancer cell aggregates upon modulation of protease expression, integrin engagement and anti-cancer treatment providing a micro-scaled yet high-throughput technique to assess malignant progression and drug-resistance.
Collapse
Affiliation(s)
- Daniela Loessner
- Faculty of Health, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia.
| | - Stefan Kobel
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Building AI 3138, Station 15, CH-1015 Lausanne, Switzerland.
| | - Judith A Clements
- Faculty of Health, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia.
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Building AI 3138, Station 15, CH-1015 Lausanne, Switzerland.
| | - Dietmar W Hutmacher
- Faculty of Science and Engineering, IHBI, QUT, 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia.
| |
Collapse
|
49
|
Burda JE, Radulovic M, Yoon H, Scarisbrick IA. Critical role for PAR1 in kallikrein 6-mediated oligodendrogliopathy. Glia 2013; 61:1456-70. [PMID: 23832758 DOI: 10.1002/glia.22534] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 04/28/2013] [Accepted: 05/01/2013] [Indexed: 11/10/2022]
Abstract
Kallikrein 6 (KLK6) is a secreted serine protease preferentially expressed by oligodendroglia in CNS white matter. Elevated levels of KLK6 occur in actively demyelinating multiple sclerosis (MS) lesions and in cases of spinal cord injury (SCI), stroke, and glioblastoma. Taken with recent evidence establishing KLK6 as a CNS-endogenous activator of protease-activated receptors (PARs), we hypothesized that KLK6 activates a subset of PARs to regulate oligodendrocyte physiology and potentially pathophysiology. Here, primary oligodendrocyte cultures derived from wild type or PAR1-deficient mice and the murine oligodendrocyte cell line, Oli-neu, were used to demonstrate that Klk6 (rodent form) mediates loss of oligodendrocyte processes and impedes morphological differentiation of oligodendrocyte progenitor cells (OPCs) in a PAR1-dependent fashion. Comparable gliopathy was also elicited by the canonical PAR1 agonist, thrombin, as well as PAR1-activating peptides (PAR1-APs). Klk6 also exacerbated ATP-mediated oligodendrogliopathy in vitro, pointing to a potential role in augmenting excitotoxicity. In addition, Klk6 suppressed the expression of proteolipid protein (PLP) RNA in cultured oligodendrocytes by a mechanism involving PAR1-mediated Erk1/2 signaling. Microinjection of PAR1 agonists, including Klk6 or PAR1-APs, into the dorsal column white matter of PAR1(+/+) but not PAR1(-/-) mice promoted vacuolating myelopathy and a loss of immunoreactivity for myelin basic protein (MBP) and CC-1(+) oligodendrocytes. These results demonstrate a functional role for Klk6-PAR1 signaling in oligodendroglial pathophysiology and suggest that antagonists of PAR1 or its protease agonists may represent new modalities to moderate demyelination and to promote myelin regeneration in cases of CNS white matter injury or disease.
Collapse
Affiliation(s)
- Joshua E Burda
- Neurobiology of Disease Program, Mayo Medical and Graduate School, Rochester, Minnesota, USA
| | | | | | | |
Collapse
|
50
|
Loessner D, Rizzi SC, Stok KS, Fuehrmann T, Hollier B, Magdolen V, Hutmacher DW, Clements JA. A bioengineered 3D ovarian cancer model for the assessment of peptidase-mediated enhancement of spheroid growth and intraperitoneal spread. Biomaterials 2013; 34:7389-400. [PMID: 23827191 DOI: 10.1016/j.biomaterials.2013.06.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 06/07/2013] [Indexed: 12/19/2022]
Abstract
Cancer-associated proteases promote peritoneal dissemination and chemoresistance in malignant progression. In this study, kallikrein-related peptidases 4, 5, 6, and 7 (KLK4-7)-cotransfected OV-MZ-6 ovarian cancer cells were embedded in a bioengineered three-dimensional (3D) microenvironment that contains RGD motifs for integrin engagement to analyze their spheroid growth and survival after chemotreatment. KLK4-7-cotransfected cells formed larger spheroids and proliferated more than controls in 3D, particularly within RGD-functionalized matrices, which was reduced upon integrin inhibition. In contrast, KLK4-7-expressing cell monolayers proliferated less than controls, emphasizing the relevance of the 3D microenvironment and integrin engagement. In a spheroid-based animal model, KLK4-7-overexpression induced tumor growth after 4 weeks and intraperitoneal spread after 8 weeks. Upon paclitaxel administration, KLK4-7-expressing tumors declined in size by 91% (controls: 87%) and showed 90% less metastatic outgrowth (controls: 33%, P < 0.001). KLK4-7-expressing spheroids showed 53% survival upon paclitaxel treatment (controls: 51%), accompanied by enhanced chemoresistance-related factors, and their survival was further reduced by combination treatment of paclitaxel with KLK4/5/7 (22%, P = 0.007) or MAPK (6%, P = 0.006) inhibition. The concomitant presence of KLK4-7 in ovarian cancer cells together with integrin activation drives spheroid formation and proliferation. Combinatorial approaches of paclitaxel and KLK/MAPK inhibition may be more efficient for late-stage disease than chemotherapeutics alone as these inhibitory regimens reduced cancer spheroid growth to a greater extent than paclitaxel alone.
Collapse
Affiliation(s)
- Daniela Loessner
- Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland 4059, Australia.
| | | | | | | | | | | | | | | |
Collapse
|