1
|
Eiumtrakul W, Wittayakornrerk S, Wongkittichote P, Nujaroen S, Pakhathirathien P, Wattanasirichaigoon D. KLK11-Related Disorder of Cornification Presenting as Inflammatory Skin Disease: A Familial Case Report and Literature Review. Am J Med Genet A 2025:e64018. [PMID: 39902525 DOI: 10.1002/ajmg.a.64018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/05/2025]
Abstract
Mendelian disorders of cornification (MeDoC) are a group of heterogeneous inherited skin disorders characterized by hyperkeratosis or scaling. Clinical assessment and skin biopsy are primarily used to differentiate MeDoC from overlapping diseases, including inflammatory skin diseases, and to identify the specific type of MeDoC; however, the results can sometimes be inconclusive. We present two individuals from a family who were initially misdiagnosed with familial pityriasis rubra pilaris, however clinical exome sequencing revealed a likely pathogenic variant in KLK11, a gene recently identified to be associated with MeDoC. This study emphasizes the importance of genetic testing in dermatologic diagnosis.
Collapse
Affiliation(s)
- Wongsathorn Eiumtrakul
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sanchawan Wittayakornrerk
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Parith Wongkittichote
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Saisuda Nujaroen
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pattima Pakhathirathien
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
2
|
Daneva GN, Tsiakanikas P, Adamopoulos PG, Scorilas A. Kallikrein-related peptidases: mechanistic understanding for potential therapeutic targeting in cancer. Expert Opin Ther Targets 2024; 28:875-894. [PMID: 39431595 DOI: 10.1080/14728222.2024.2415014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Human kallikrein-related peptidases (KLKs) represent a subgroup of 15 serine endopeptidases involved in various physiological processes and pathologies, including cancer. AREAS COVERED This review aims to provide a comprehensive overview of the KLK family, highlighting their genomic structure, expression profiles and substrate specificity. We explore the role of KLKs in tumorigenesis, emphasizing their potential as biomarkers and therapeutic targets in cancer treatment. The dysregulated activity of KLKs has been linked to various malignancies, making them promising candidates for cancer diagnostics and therapy. EXPERT OPINION : Recent advancements in understanding the mechanistic pathways of KLK-related tumorigenesis offer new prospects for developing targeted cancer treatments. Expert opinion suggests that while significant progress has been made, further research is necessary to fully exploit KLKs' potential in clinical applications.
Collapse
Affiliation(s)
- Glykeria N Daneva
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Tsiakanikas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis G Adamopoulos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
3
|
Martins D, Pinoteau MA, Leger R. Development of a back-titration assay to quantitate functional lympho-epithelial Kazal-type inhibitors (LEKTI) in skin samples. Anal Biochem 2024; 690:115524. [PMID: 38556114 DOI: 10.1016/j.ab.2024.115524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
The lympho-epithelial Kazal-type inhibitors (LEKTI) are key to control skin turnover, and their absence causes Netherton syndrome. For clinical sample testing of LEKTI-based therapies, a robust analytical method to measure LEKTI-like activity in skin is required. This work reports on the development of a back-titration method to determine incremental LEKTI-like activity in skin samples. The method meets the analytical requirements for study sample testing, and reliable quantification can be achieved with negligible skin matrix interference. This assay does not provide analyte identity, but it can be used to measure treatment-driven increments of LEKTI-like activity within the skin epidermis.
Collapse
Affiliation(s)
- Dorival Martins
- Azitra Inc, 21 Business Park Drive, Branford, CT, 06405, USA.
| | | | - Roger Leger
- Azitra Inc, 21 Business Park Drive, Branford, CT, 06405, USA
| |
Collapse
|
4
|
Radisky ES. Extracellular proteolysis in cancer: Proteases, substrates, and mechanisms in tumor progression and metastasis. J Biol Chem 2024; 300:107347. [PMID: 38718867 PMCID: PMC11170211 DOI: 10.1016/j.jbc.2024.107347] [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: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/02/2024] Open
Abstract
A vast ensemble of extracellular proteins influences the development and progression of cancer, shaped and reshaped by a complex network of extracellular proteases. These proteases, belonging to the distinct classes of metalloproteases, serine proteases, cysteine proteases, and aspartic proteases, play a critical role in cancer. They often become dysregulated in cancer, with increases in pathological protease activity frequently driven by the loss of normal latency controls, diminished regulation by endogenous protease inhibitors, and changes in localization. Dysregulated proteases accelerate tumor progression and metastasis by degrading protein barriers within the extracellular matrix (ECM), stimulating tumor growth, reactivating dormant tumor cells, facilitating tumor cell escape from immune surveillance, and shifting stromal cells toward cancer-promoting behaviors through the precise proteolysis of specific substrates to alter their functions. These crucial substrates include ECM proteins and proteoglycans, soluble proteins secreted by tumor and stromal cells, and extracellular domains of cell surface proteins, including membrane receptors and adhesion proteins. The complexity of the extracellular protease web presents a significant challenge to untangle. Nevertheless, technological strides in proteomics, chemical biology, and the development of new probes and reagents are enabling progress and advancing our understanding of the pivotal importance of extracellular proteolysis in cancer.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA.
| |
Collapse
|
5
|
Souza-Silva IM, Steckelings UM, Assersen KB. The role of vasoactive peptides in skin homeostasis-focus on adiponectin and the kallikrein-kinin system. Am J Physiol Cell Physiol 2023; 324:C741-C756. [PMID: 36745527 DOI: 10.1152/ajpcell.00269.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vasoactive peptides often serve a multitude of functions aside from their direct effects on vasodynamics. This article will review the existing literature on two vasoactive peptides and their involvement in skin homeostasis: adiponectin and-as the main representative of the kallikrein-kinin system-bradykinin. Adiponectin is the most abundantly expressed adipokine in the human organism, where it is mainly localized in fat depots including subcutaneous adipose tissue, from where adiponectin can exert paracrine effects. The involvement of adiponectin in skin homeostasis is supported by a number of studies reporting the effects of adiponectin in isolated human keratinocytes, sebocytes, fibroblasts, melanocytes, and immune cells. Regarding skin pathology, the potential involvement of adiponectin in psoriasis, atopic dermatitis, scleroderma, keloid, and melanogenesis is discussed in this article. The kallikrein-kinin system is composed of a variety of enzymes and peptides, most of which have been identified to be expressed in the skin. This also includes the expression of bradykinin receptors on most skin cells. Bradykinin is one of the very few hormones that is targeted by treatment in routine clinical use in dermatology-in this case for the treatment of hereditary angioedema. The potential involvement of bradykinin in wound healing, psoriasis, and melanoma is further discussed in this article. This review concludes with a call for additional preclinical and clinical studies to further explore the therapeutic potential of adiponectin supplementation (for psoriasis, atopic dermatitis, wound healing, scleroderma, and keloid) or pharmacological interference with the kallikrein-kinin system (for wound healing, psoriasis, and melanoma).
Collapse
Affiliation(s)
- Igor M Souza-Silva
- Department of Cardiovascular & Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - U Muscha Steckelings
- Department of Cardiovascular & Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Kasper Bostlund Assersen
- Department of Cardiovascular & Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Dermatology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
6
|
Chan B, Cheng IC, Rozita J, Gorshteyn I, Huang Y, Shaffer I, Chang C, Li W, Lytton J, Den Besten P, Zhang Y. Sodium/(calcium + potassium) exchanger NCKX4 optimizes KLK4 activity in the enamel matrix microenvironment to regulate ECM modeling. Front Physiol 2023; 14:1116091. [PMID: 36814474 PMCID: PMC9939835 DOI: 10.3389/fphys.2023.1116091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/16/2023] [Indexed: 02/09/2023] Open
Abstract
Enamel development is a process in which extracellular matrix models from a soft proteinaceous matrix to the most mineralized tissue in vertebrates. Patients with mutant NCKX4, a gene encoding a K+-dependent Na+/Ca2+-exchanger, develop a hypomineralized and hypomature enamel. How NCKX4 regulates enamel protein removal to achieve an almost protein-free enamel is unknown. We characterized the upregulation pattern of Nckx4 in the progressively differentiating enamel-forming ameloblasts by qPCR, and as well as confirmed NCKX4 protein to primarily localize at the apical surface of wild-type ruffle-ended maturation ameloblasts by immunostaining of the continuously growing mouse incisors, posing the entire developmental trajectory of enamel. In contrast to the normal mature enamel, where ECM proteins are hydrolyzed and removed, we found significant protein retention in the maturation stage of Nckx4 -/- mouse enamel. The Nckx4 -/- enamel held less Ca2+ and K+ but more Na+ than the Nckx4 +/+ enamel did, as measured by EDX. The alternating acidic and neutral pH zones at the surface of mineralizing Nckx4 +/+ enamel were replaced by a largely neutral pH matrix in the Nckx4 -/- enamel. In situ zymography revealed a reduced kallikrein-related peptidase 4 (KLK4) activity in the Nckx4 -/- enamel. We showed that KLK4 took on 90% of proteinase activity in the maturation stage of normal enamel, and that recombinant KLK4 as well as native mouse enamel KLK4 both performed less effectively in a buffer with increased [Na+] and pH, conditions found in the Nckx4 -/- developing enamel. This study, for the first time to our knowledge, provides evidence demonstrating the impaired in situ KLK4 activity in Nckx4 -/- enamel and suggests a novel function of NCKX4 in facilitating KLK4-mediated hydrolysis and removal of ECM proteins, warranting the completion of enamel matrix modeling.
Collapse
Affiliation(s)
- Barry Chan
- Department of Orofacial Sciences, University of California, San Francisco, CA, San Francisco, United States
| | - Ieong Cheng Cheng
- Department of Orofacial Sciences, University of California, San Francisco, CA, San Francisco, United States
| | - Jalali Rozita
- Department of Orofacial Sciences, University of California, San Francisco, CA, San Francisco, United States
| | - Ida Gorshteyn
- Department of Orofacial Sciences, University of California, San Francisco, CA, San Francisco, United States
| | - Yulei Huang
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun-Yat-sen University, Guangzhou, China
| | - Ida Shaffer
- Department of Orofacial Sciences, University of California, San Francisco, CA, San Francisco, United States
| | - Chih Chang
- Department of Orofacial Sciences, University of California, San Francisco, CA, San Francisco, United States
| | - Wu Li
- Department of Orofacial Sciences, University of California, San Francisco, CA, San Francisco, United States
| | - Jonathan Lytton
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Pamela Den Besten
- Department of Orofacial Sciences, University of California, San Francisco, CA, San Francisco, United States
| | - Yan Zhang
- Department of Orofacial Sciences, University of California, San Francisco, CA, San Francisco, United States
| |
Collapse
|
7
|
Gong Z, Dai S, Jiang X, Lee M, Zhu X, Wang H, Lin Z. Variants in KLK11, affecting signal peptide cleavage of kallikrein-related peptidase 11, cause an autosomal-dominant cornification disorder. Br J Dermatol 2023; 188:100-111. [PMID: 36689511 DOI: 10.1093/bjd/ljac029] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/21/2022] [Accepted: 09/24/2022] [Indexed: 01/22/2023]
Abstract
BACKGROUND Mendelian disorders of cornification (MeDOC) are a group of heterogeneous genodermatoses with different genetic bases. The pathogenesis of a substantial group of MeDOC remains to be elucidated. OBJECTIVES To identify a new causative gene and the pathogenesis of a previously undescribed autosomal-dominant cornification disorder. METHODS Whole-exome sequencing was performed in three families with the novel cornification disorder to identify the disease-causing variants. As the variants were located around the signal peptide (SP) cleavage site of a kallikrein-related peptidase, SP cleavage, subcellular localization and extracellular secretion of the variants were evaluated in eukaryotic overexpression systems by Western blotting or immunocytochemistry. Then the trypsin-like and chymotrypsin-like proteolytic activity of the peptidase and degradation of its catalytic substrate were assayed using the patients' stratum corneum (SC) samples. The morphology of the lamellar bodies and corneodesmosomes (CDs) in the patients' SC was ultrastructurally examined. A mouse model harbouring the equivalent variant was constructed and evaluated histologically. RESULTS We identified two heterozygous variants affecting Gly50 in kallikrein-related peptidase (KLK)11 in a familial case and two sporadic cases with the new disorder, which is characterized by early-onset ichthyosiform erythroderma or erythrokeratoderma. KLK11 belongs to the family of kallikrein-related peptidases participating in skin desquamation by decomposing CDs, a process essential for shedding of the SC. In vitro experiments demonstrated that the variants perturbed the SP cleavage of KLK11, leading to subcellular mislocalization and impaired extracellular secretion of the KLK11 Gly50Glu variant. Both trypsin-like and chymotrypsin-like proteolytic activities were significantly decreased in the patients' SC samples. Reduced proteolysis of desmoglein 1 and delayed degeneration of CDs were detected in patients' SC, indicating delayed skin desquamation. Consistently, the patients showed a thickened, dense SC, indicating abnormal skin desquamation. Mice harbouring the homozygous c.131G>A (p.Gly44Glu) Klk11 variant, which is equivalent to KLK11 c.149G>A (p.Gly50Glu) in humans, exhibited hyperkeratosis and abnormal desquamation, partially recapitulating the phenotype. CONCLUSIONS We provide evidence that variants at Gly50 affecting the SP cleavage of KLK11 cause a new autosomal-dominant cornification disorder with abnormal desquamation. Our findings highlight the essential role of KLKs in maintaining homeostasis of skin keratinization and desquamation.
Collapse
Affiliation(s)
- Zhuoqing Gong
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Shangzhi Dai
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Xingyuan Jiang
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Mingyang Lee
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Xuejun Zhu
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Huijun Wang
- Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Zhimiao Lin
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
- Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen 361026, China
| |
Collapse
|
8
|
Liu Y, Gong W, Preis S, Dorn J, Kiechle M, Reuning U, Magdolen V, Dreyer TF. A Pair of Prognostic Biomarkers in Triple-Negative Breast Cancer: KLK10 and KLK11 mRNA Expression. Life (Basel) 2022; 12:life12101517. [PMID: 36294951 PMCID: PMC9605449 DOI: 10.3390/life12101517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/27/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with poor patient prognosis and limited therapeutic options. A lack of prognostic biomarkers and therapeutic targets fuels the need for new approaches to tackle this severe disease. Extracellular matrix degradation, release, and modulation of the activity of growth factors/cytokines/chemokines, and the initiation of signaling pathways by extracellular proteolytic networks, have been identified as major processes in the carcinogenesis of breast cancer. Members of the kallikrein-related peptidase (KLK) family contribute to these tumor-relevant processes, and are associated with breast cancer progression and metastasis. In this study, the clinical relevance of mRNA expression of two members of this family, KLK10 and KLK11, has been evaluated in TNBC. For this, their expression levels were quantified in tumor tissue of a large, well-characterized patient cohort (n = 123) via qPCR. Although, in general, the overall expression of both factors are lower in tumor tissue of breast cancer patients (encompassing all subtypes) compared to normal tissue of healthy donors, in the TNBC subtype, expression is even increased. In our cohort, a significant, positive correlation between the expression levels of both KLKs was detected, indicating a coordinate expression mode of these proteases. Elevated KLK10 and KLK11 mRNA levels were associated with poor patient prognosis. Moreover, both factors were found to be independent of other established clinical factors such as age, lymph node status, or residual tumor mass, as determined by multivariable Cox regression analysis. Thus, both proteases, KLK10 and KLK11, may represent unfavorable prognostic factors for TNBC patients and, furthermore, appear as promising potential targets for therapy in TNBC.
Collapse
Affiliation(s)
- Yueyang Liu
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
- Department of Gynecology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 519041, China
| | - Weiwei Gong
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
- Department of Hematology/Oncology, Guangzhou Women and Children’s Medical Center, Guangzhou 519041, China
| | - Sarah Preis
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
| | - Julia Dorn
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
| | - Marion Kiechle
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
| | - Tobias F. Dreyer
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-4140-7408
| |
Collapse
|
9
|
Wikström T, Abrahamsson S, Bengtsson‐Palme J, Ek J, Kuusela P, Rekabdar E, Lindgren P, Wennerholm U, Jacobsson B, Valentin L, Hagberg H. Microbial and human transcriptome in vaginal fluid at midgestation: Association with spontaneous preterm delivery. Clin Transl Med 2022; 12:e1023. [PMID: 36103557 PMCID: PMC9473488 DOI: 10.1002/ctm2.1023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/03/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Intrauterine infection and inflammation caused by microbial transfer from the vagina are believed to be important factors causing spontaneous preterm delivery (PTD). Multiple studies have examined the relationship between the cervicovaginal microbiome and spontaneous PTD with divergent results. Most studies have applied a DNA-based assessment, providing information on the microbial composition but not transcriptional activity. A transcriptomic approach was applied to investigate differences in the active vaginal microbiome and human transcriptome at midgestation between women delivering spontaneously preterm versus those delivering at term. METHODS Vaginal swabs were collected in women with a singleton pregnancy at 18 + 0 to 20 + 6 gestational weeks. For each case of spontaneous PTD (delivery <37 + 0 weeks) two term controls were randomized (39 + 0 to 40 + 6 weeks). Vaginal specimens were subject to sequencing of both human and microbial RNA. Microbial reads were taxonomically classified using Kraken2 and RefSeq as a reference. Statistical analyses were performed using DESeq2. GSEA and HUMAnN3 were used for pathway analyses. RESULTS We found 17 human genes to be differentially expressed (false discovery rate, FDR < 0.05) in the preterm group (n = 48) compared to the term group (n = 96). Gene expression of kallikrein-2 (KLK2), KLK3 and four isoforms of metallothioneins 1 (MT1s) was higher in the preterm group (FDR < 0.05). We found 11 individual bacterial species to be differentially expressed (FDR < 0.05), most with a low occurrence. No statistically significant differences in bacterial load, diversity or microbial community state types were found between the groups. CONCLUSIONS In our mainly white population, primarily bacterial species of low occurrence were differentially expressed at midgestation in women who delivered preterm versus at term. However, the expression of specific human transcripts including KLK2, KLK3 and several isoforms of MT1s was higher in preterm cases. This is of interest, because these genes may be involved in critical inflammatory pathways associated with spontaneous PTD.
Collapse
Affiliation(s)
- Tove Wikström
- Centre of Perinatal Medicine and HealthDepartment of Obstetrics and GynecologyInstitute of Clinical SciencesSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of ObstetricsRegion Västra GötalandSahlgrenska University HospitalGothenburgSweden
| | - Sanna Abrahamsson
- Bioinformatics Core FacilitySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Johan Bengtsson‐Palme
- Department of Infectious DiseasesInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Centre for Antibiotic Resistance Research (CARe) at University of GothenburgGothenburgSweden
- Division of Systems and Synthetic BiologyDepartment ofBiology and Biological EngineeringChalmers University of TechnologyGothenburgSweden
| | - Joakim Ek
- Institute of Neuroscience and PhysiologyDepartment of Physiology Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | | | - Elham Rekabdar
- Bioinformatics Core FacilitySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Peter Lindgren
- Department of Clinical ScienceIntervention and TechnologyKarolinska InstitutetStockholmSweden
- Centre for Fetal MedicineKarolinska University HospitalStockholmSweden
| | - Ulla‐Britt Wennerholm
- Centre of Perinatal Medicine and HealthDepartment of Obstetrics and GynecologyInstitute of Clinical SciencesSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of ObstetricsRegion Västra GötalandSahlgrenska University HospitalGothenburgSweden
| | - Bo Jacobsson
- Centre of Perinatal Medicine and HealthDepartment of Obstetrics and GynecologyInstitute of Clinical SciencesSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of ObstetricsRegion Västra GötalandSahlgrenska University HospitalGothenburgSweden
| | - Lil Valentin
- Department of Obstetrics and GynecologySkåne University HospitalMalmöSweden
- Department of Clinical Sciences MalmöLund UniversityLundSweden
| | - Henrik Hagberg
- Centre of Perinatal Medicine and HealthDepartment of Obstetrics and GynecologyInstitute of Clinical SciencesSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of ObstetricsRegion Västra GötalandSahlgrenska University HospitalGothenburgSweden
| |
Collapse
|
10
|
Srinivasan S, Kryza T, Batra J, Clements J. Remodelling of the tumour microenvironment by the kallikrein-related peptidases. Nat Rev Cancer 2022; 22:223-238. [PMID: 35102281 DOI: 10.1038/s41568-021-00436-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 02/07/2023]
Abstract
Kallikrein-related peptidases (KLKs) are critical regulators of the tumour microenvironment. KLKs are proteolytic enzymes regulating multiple functions of bioactive molecules including hormones and growth factors, membrane receptors and the extracellular matrix architecture involved in cancer progression and metastasis. Perturbations of the proteolytic cascade generated by these peptidases, and their downstream signalling actions, underlie tumour emergence or blockade of tumour growth. Recent studies have also revealed their role in tumour immune suppression and resistance to cancer therapy. Here, we present an overview of the complex biology of the KLK family and its context-dependent nature in cancer, and discuss the different therapeutic strategies available to potentially target these proteases.
Collapse
Affiliation(s)
- Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Thomas Kryza
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Mater Research Institute, The University of Queensland, Woolloongabba, Brisbane, Queensland, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Centre for Genomics and Personalised Medicine, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia.
- Australian Prostate Cancer Research Centre-Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
11
|
Candido JB, Maiques O, Boxberg M, Kast V, Peerani E, Tomás-Bort E, Weichert W, Sananes A, Papo N, Magdolen V, Sanz-Moreno V, Loessner D. Kallikrein-Related Peptidase 6 Is Associated with the Tumour Microenvironment of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13163969. [PMID: 34439122 PMCID: PMC8392253 DOI: 10.3390/cancers13163969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Kallikrein-related peptidases have tumour-biological roles and are dysregulated in many cancers. Only a few studies have reported their upregulation in pancreatic cancer and linked them to poor prognosis. By interrogating publicly available and our own datasets, we studied their expression in patient-derived tissues and pancreatic cancer cells. We found several kallikrein-related peptidases that were upregulated, in particular kallikrein-related peptidase 6 at the forefront of the tumour area. We then tested the effect of a kallikrein-related peptidase 6 inhibitor on cancer cell functions. Because the majority of patients present with inoperable disease, a targeted therapeutic intervention may have a positive impact on the survival of this patient population. Abstract As cancer-associated factors, kallikrein-related peptidases (KLKs) are components of the tumour microenvironment, which represents a rich substrate repertoire, and considered attractive targets for the development of novel treatments. Standard-of-care therapy of pancreatic cancer shows unsatisfactory results, indicating the need for alternative therapeutic approaches. We aimed to investigate the expression of KLKs in pancreatic cancer and to inhibit the function of KLK6 in pancreatic cancer cells. KLK6, KLK7, KLK8, KLK10 and KLK11 were coexpressed and upregulated in tissues from pancreatic cancer patients compared to normal pancreas. Their high expression levels correlated with each other and were linked to shorter survival compared to low KLK levels. We then validated KLK6 mRNA and protein expression in patient-derived tissues and pancreatic cancer cells. Coexpression of KLK6 with KRT19, αSMA or CD68 was independent of tumour stage, while KLK6 was coexpressed with KRT19 and CD68 in the invasive tumour area. High KLK6 levels in tumour and CD68+ cells were linked to shorter survival. KLK6 inhibition reduced KLK6 mRNA expression, cell metabolic activity and KLK6 secretion and increased the secretion of other serine and aspartic lysosomal proteases. The association of high KLK levels and poor prognosis suggests that inhibiting KLKs may be a therapeutic strategy for precision medicine.
Collapse
Affiliation(s)
- Juliana B. Candido
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.B.C.); (O.M.); (E.P.); (E.T.-B.); (V.S.-M.)
| | - Oscar Maiques
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.B.C.); (O.M.); (E.P.); (E.T.-B.); (V.S.-M.)
| | - Melanie Boxberg
- Institute of Pathology, Technical University of Munich, 81657 Munich, Germany; (M.B.); (W.W.)
| | - Verena Kast
- Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden e.V., Hohe Straβe 6, 01069 Dresden, Germany;
| | - Eleonora Peerani
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.B.C.); (O.M.); (E.P.); (E.T.-B.); (V.S.-M.)
| | - Elena Tomás-Bort
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.B.C.); (O.M.); (E.P.); (E.T.-B.); (V.S.-M.)
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, 81657 Munich, Germany; (M.B.); (W.W.)
| | - Amiram Sananes
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (A.S.); (N.P.)
| | - Niv Papo
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (A.S.); (N.P.)
| | - Viktor Magdolen
- Department of Obstetrics and Gynaecology, Technical University of Munich, 81675 Munich, Germany;
| | - Victoria Sanz-Moreno
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.B.C.); (O.M.); (E.P.); (E.T.-B.); (V.S.-M.)
| | - Daniela Loessner
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (J.B.C.); (O.M.); (E.P.); (E.T.-B.); (V.S.-M.)
- Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden e.V., Hohe Straβe 6, 01069 Dresden, Germany;
- Department of Chemical Engineering and Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia
- Correspondence:
| |
Collapse
|
12
|
Lovell S, Zhang L, Kryza T, Neodo A, Bock N, De Vita E, Williams ED, Engelsberger E, Xu C, Bakker AT, Maneiro M, Tanaka RJ, Bevan CL, Clements JA, Tate EW. A Suite of Activity-Based Probes To Dissect the KLK Activome in Drug-Resistant Prostate Cancer. J Am Chem Soc 2021; 143:8911-8924. [PMID: 34085829 PMCID: PMC9282638 DOI: 10.1021/jacs.1c03950] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
Kallikrein-related
peptidases (KLKs) are a family of secreted serine
proteases, which form a network (the KLK activome) with an important
role in proteolysis and signaling. In prostate cancer (PCa), increased
KLK activity promotes tumor growth and metastasis through multiple
biochemical pathways, and specific quantification and tracking of
changes in the KLK activome could contribute to validation of KLKs
as potential drug targets. Herein we report a technology platform
based on novel activity-based probes (ABPs) and inhibitors enabling
simultaneous orthogonal analysis of KLK2, KLK3, and KLK14 activity
in hormone-responsive PCa cell lines and tumor homogenates. Importantly,
we identifed a significant decoupling of KLK activity and abundance
and suggest that KLK proteolysis should be considered as an additional
parameter, along with the PSA blood test, for accurate PCa diagnosis
and monitoring. Using selective inhibitors and multiplexed fluorescent
activity-based protein profiling (ABPP), we dissect the KLK activome
in PCa cells and show that increased KLK14 activity leads to a migratory
phenotype. Furthermore, using biotinylated ABPs, we show that active
KLK molecules are secreted into the bone microenvironment by PCa cells
following stimulation by osteoblasts suggesting KLK-mediated signaling
mechanisms could contribute to PCa metastasis to bone. Together our
findings show that ABPP is a powerful approach to dissect dysregulation
of the KLK activome as a promising and previously underappreciated
therapeutic target in advanced PCa.
Collapse
Affiliation(s)
- Scott Lovell
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Leran Zhang
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Thomas Kryza
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Anna Neodo
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Nathalie Bock
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Elena De Vita
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Elisabeth Engelsberger
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Congyi Xu
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Alexander T Bakker
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Maria Maneiro
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Reiko J Tanaka
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K
| | - Charlotte L Bevan
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| | - Judith A Clements
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Edward W Tate
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.,The Francis Crick Institute, London NW1 1AT, U.K
| |
Collapse
|
13
|
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
|
14
|
Gong W, Liu Y, Diamandis EP, Kiechle M, Bronger H, Dorn J, Dreyer T, Magdolen V. Prognostic value of kallikrein-related peptidase 7 (KLK7) mRNA expression in advanced high-grade serous ovarian cancer. J Ovarian Res 2020; 13:125. [PMID: 33087135 PMCID: PMC7579813 DOI: 10.1186/s13048-020-00725-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND High-grade serous ovarian cancer (HGSOC) is the most common and lethal subtype of ovarian cancer. A growing body of evidence suggests tumor-supporting roles of several members of the kallikrein-related peptidase (KLK) family, including KLK5 and KLK7, in this cancer subtype. In normal physiology, KLK5 and KLK7 are the major proteases involved in skin desquamation. Moreover, in several cancer types KLK5 and KLK7 co-expression has been observed. Recently, we have shown that elevated KLK5 mRNA levels are associated with an unfavorable prognosis in HGSOC. Therefore, the aim of this study was to investigate the clinical significance of KLK7 mRNA expression and to explore its relation to KLK5 levels in HGSOC. METHODS mRNA expression levels of KLK7 were quantified by qPCR in a well-characterized patient cohort afflicted with advanced high-grade serous ovarian cancer (FIGO III/IV, n = 139). Previously determined KLK5 mRNA as well as KLK5 and KLK7 antigen concentrations were used to evaluate the relationship between the expression patterns of both factors on the mRNA as well as protein level in tumor tissue of HGSOC patients. RESULTS There were strong, significant positive correlations between KLK5 and KLK7 both at the mRNA and the protein level, suggesting coordinate expression of these proteases in HGSOC. In univariate analyses, elevated KLK7 levels as well as the combination of KLK5 + KLK7 (high and/or high versus low/low) were significantly associated with worse progression-free survival (PFS). High mRNA expression levels of KLK7 and the combination of KLK5 and KLK7 showed a trend towards significance for overall survival (OS). In multivariate analyses, KLK7 mRNA expression represented an unfavorable, statistically significant independent predictor for PFS and OS. CONCLUSIONS The findings imply that both increased KLK5 and KLK7 mRNA expression levels represent unfavorable prognostic biomarkers in advanced high-grade serous ovarian cancer, whereby multivariate analyses indicate that KLK7 mRNA exhibits a stronger predictive value as compared to KLK5 mRNA and the combination of KLK5 and KLK7.
Collapse
Affiliation(s)
- Weiwei Gong
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany.,Department of Hematology-Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, People's Republic of China
| | - Yueyang Liu
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany.,Department of Gynecology, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Marion Kiechle
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany
| | - Holger Bronger
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany
| | - Julia Dorn
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany
| | - Tobias Dreyer
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany.
| |
Collapse
|
15
|
Di Paolo CT, Diamandis EP, Prassas I. The role of kallikreins in inflammatory skin disorders and their potential as therapeutic targets. Crit Rev Clin Lab Sci 2020; 58:1-16. [PMID: 32568598 DOI: 10.1080/10408363.2020.1775171] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The skin is a vital organ of the human body, serving numerous protective and functional roles that are essential for survival. Residing in the epidermis are various epidermal proteases responsible for the establishment and regulation of barrier function. The human tissue kallikrein-related peptidase family conserves homeostasis of the skin barrier through their roles in desquamation, antimicrobial defense, innate immune response, and barrier maintenance. The activity of kallikreins is tightly regulated and dysregulation of kallikrein activity is seen to contribute to the formation of several inflammatory skin disorders. This review highlights the roles of kallikreins in skin homeostasis and pathologies. Due to their part in these skin disorders, inhibitors of the skin kallikreins have become attractive therapeutics. Over the past few years, both natural and synthetic inhibitors of several kallikreins have been identified and are undergoing further development as treatments to restore compromised barrier function. This review summarizes the kallikrein inhibitors under development for this purpose. These inhibitors remain promising therapeutics in cases of severe skin inflammation not well managed by current therapies.
Collapse
Affiliation(s)
- Caitlin T Di Paolo
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada.,Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Ioannis Prassas
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| |
Collapse
|
16
|
Kallikrein-Related Peptidase 14 Activates Zymogens of Membrane Type Matrix Metalloproteinases (MT-MMPs)-A CleavEx Based Analysis. Int J Mol Sci 2020; 21:ijms21124383. [PMID: 32575583 PMCID: PMC7352328 DOI: 10.3390/ijms21124383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/17/2020] [Indexed: 01/02/2023] Open
Abstract
Kallikrein-related peptidases (KLKs) and matrix metalloproteinases (MMPs) are secretory proteinases known to proteolytically process components of the extracellular matrix, modulating the pericellular environment in physiology and in pathologies. The interconnection between these families remains elusive. To assess the cross-activation of these families, we developed a peptide, fusion protein-based exposition system (Cleavage of exposed amino acid sequences, CleavEx) aiming at investigating the potential of KLK14 to recognize and hydrolyze proMMP sequences. Initial assessment identified ten MMP activation domain sequences which were validated by Edman degradation. The analysis revealed that membrane-type MMPs (MT-MMPs) are targeted by KLK14 for activation. Correspondingly, proMMP14-17 were investigated in vitro and found to be effectively processed by KLK14. Again, the expected neo-N-termini of the activated MT-MMPs was confirmed by Edman degradation. The effectiveness of proMMP activation was analyzed by gelatin zymography, confirming the release of fully active, mature MT-MMPs upon KLK14 treatment. Lastly, MMP14 was shown to be processed on the cell surface by KLK14 using murine fibroblasts overexpressing human MMP14. Herein, we propose KLK14-mediated selective activation of cell-membrane located MT-MMPs as an additional layer of their regulation. As both, KLKs and MT-MMPs, are implicated in cancer, their cross-activation may constitute an important factor in tumor progression and metastasis.
Collapse
|
17
|
Boon L, Ugarte-Berzal E, Vandooren J, Opdenakker G. Protease propeptide structures, mechanisms of activation, and functions. Crit Rev Biochem Mol Biol 2020; 55:111-165. [PMID: 32290726 DOI: 10.1080/10409238.2020.1742090] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Proteases are a diverse group of hydrolytic enzymes, ranging from single-domain catalytic molecules to sophisticated multi-functional macromolecules. Human proteases are divided into five mechanistic classes: aspartate, cysteine, metallo, serine and threonine proteases, based on the catalytic mechanism of hydrolysis. As a protective mechanism against uncontrolled proteolysis, proteases are often produced and secreted as inactive precursors, called zymogens, containing inhibitory N-terminal propeptides. Protease propeptide structures vary considerably in length, ranging from dipeptides and propeptides of about 10 amino acids to complex multifunctional prodomains with hundreds of residues. Interestingly, sequence analysis of the different protease domains has demonstrated that propeptide sequences present higher heterogeneity compared with their catalytic domains. Therefore, we suggest that protease inhibition targeting propeptides might be more specific and have less off-target effects than classical inhibitors. The roles of propeptides, besides keeping protease latency, include correct folding of proteases, compartmentalization, liganding, and functional modulation. Changes in the propeptide sequence, thus, have a tremendous impact on the cognate enzymes. Small modifications of the propeptide sequences modulate the activity of the enzymes, which may be useful as a therapeutic strategy. This review provides an overview of known human proteases, with a focus on the role of their propeptides. We review propeptide functions, activation mechanisms, and possible therapeutic applications.
Collapse
Affiliation(s)
- Lise Boon
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| | - Estefania Ugarte-Berzal
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| | - Jennifer Vandooren
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| | - Ghislain Opdenakker
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, KU Leuven, Leuven, Belgium
| |
Collapse
|
18
|
Kawaguchi M, Yamamoto K, Kataoka H, Izumi A, Yamashita F, Kiwaki T, Nishida T, Camerer E, Fukushima T. Protease-activated receptor-2 accelerates intestinal tumor formation through activation of nuclear factor-κB signaling and tumor angiogenesis in Apc Min/+ mice. Cancer Sci 2020; 111:1193-1202. [PMID: 31997435 PMCID: PMC7156842 DOI: 10.1111/cas.14335] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/19/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocyte growth factor activator inhibitor‐1 (HAI‐1), encoded by the SPINT1 gene, is a membrane‐bound protease inhibitor expressed on the surface of epithelial cells. Hepatocyte growth factor activator inhibitor‐1 regulates type II transmembrane serine proteases that activate protease‐activated receptor‐2 (PAR‐2). We previously reported that deletion of Spint1 in ApcMin/+ mice resulted in accelerated formation of intestinal tumors, possibly through enhanced nuclear factor‐κB signaling. In this study, we examined the role of PAR‐2 in accelerating tumor formation in the ApcMin/+ model in the presence or absence of Spint1. We observed that knockout of the F2rl1 gene, encoding PAR‐2, not only eliminated the enhanced formation of intestinal tumors caused by Spint1 deletion, but also reduced tumor formation in the presence of Spint1. Exacerbation of anemia and weight loss associated with HAI‐1 deficiency was also normalized by compound deficiency of PAR‐2. Mechanistically, signaling triggered by deregulated protease activities increased nuclear translocation of RelA/p65, vascular endothelial growth factor expression, and vascular density in ApcMin/+‐induced intestinal tumors. These results suggest that serine proteases promote intestinal carcinogenesis through activation of PAR‐2, and that HAI‐1 plays a critical tumor suppressor role as an inhibitor of matriptase, kallikreins, and other PAR‐2 activating proteases.
Collapse
Affiliation(s)
- Makiko Kawaguchi
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Koji Yamamoto
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Hiroaki Kataoka
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Aya Izumi
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Fumiki Yamashita
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Takumi Kiwaki
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Takahiro Nishida
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Eric Camerer
- Inserm U970, Paris Cardiovascular Research Center, Université de Paris, Paris, France
| | | |
Collapse
|
19
|
Moradi A, Srinivasan S, Clements J, Batra J. Beyond the biomarker role: prostate-specific antigen (PSA) in the prostate cancer microenvironment. Cancer Metastasis Rev 2020; 38:333-346. [PMID: 31659564 DOI: 10.1007/s10555-019-09815-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prostate-specific antigen (PSA) blood test is the accepted biomarker of tumor recurrence. PSA levels in serum correlate with disease progression, though its diagnostic accuracy is questionable. As a result, significant progress has been made in developing modified PSA tests such as PSA velocity, PSA density, 4Kscore, PSA glycoprofiling, Prostate Health Index, and the STHLM3 test. PSA, a serine protease, is secreted from the epithelial cells of the prostate. PSA has been suggested as a molecular target for prostate cancer therapy due to the fact that it is not only active in prostate tissue but also has a pivotal role on prostate cancer signaling pathways including proliferation, invasion, metastasis, angiogenesis, apoptosis, immune response, and tumor microenvironment regulation. Here, we summarize the current standing of PSA in prostate cancer progression as well as its utility in prostate cancer therapeutic approaches with an emphasis on the role of PSA in the tumor microenvironment.
Collapse
Affiliation(s)
- Afshin Moradi
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Srilakshmi Srinivasan
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia. .,Translational Research Institute, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
20
|
Gong W, Liu Y, Preis S, Geng X, Petit-Courty A, Kiechle M, Muckenhuber A, Dreyer T, Dorn J, Courty Y, Magdolen V. Prognostic value of kallikrein-related peptidase 12 (KLK12) mRNA expression in triple-negative breast cancer patients. Mol Med 2020; 26:19. [PMID: 32028882 PMCID: PMC7006133 DOI: 10.1186/s10020-020-0145-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/28/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The serine protease KLK12 belongs to the human fifteen-member family of kallikrein-related peptidases. Differential expression accompanied by either increased or decreased enzymatic activity has been linked to several diseases including cancer. Triple-negative breast cancer (TNBC) represents a very aggressive subgroup of breast cancer with high tumor recurrence rates and poor patient prognosis. Here, we quantified the KLK12 mRNA expression levels in tumor tissue of TNBC patients and analyzed their prognostic value. METHODS In the present study, KLK12 mRNA expression in tumor tissue of TNBC patients (n = 116) was determined by quantitative real-time PCR assay. The association of KLK12 mRNA levels with clinical parameters, and patients' outcome was analyzed using Chi-square tests, Cox regression models and Kaplan-Meier survival analysis. RESULTS Positive, but low KLK12 mRNA levels were detected in about half of the cases (54 out of 116; 47%), the other samples were negative for KLK12 mRNA expression. No significant association was observed between KLK12 mRNA levels and clinicopathological variables (age, lymph node status, tumor size, and histological grade). In univariate Cox analyses, positive KLK12 mRNA expression was significantly associated with shortened disease-free survival (DFS; hazard ratio [HR] = 2.12, 95% CI = 1.19-3.78, p = 0.010) as well as overall survival (OS; HR = 1.91, 95% CI = 1.04-3.50, p = 0.037). In multivariable Cox analysis, including all clinical parameters plus KLK12 mRNA, the latter - together with age - remained an independent unfavorable predictive marker for DFS (HR = 2.33, 95% CI = 1.28-4.24, p = 0.006) and showed a trend towards significance in case of OS (HR = 1.80, 95% CI = 0.96-3.38, p = 0.066). CONCLUSIONS Positive KLK12 expression is remarkably associated with shortened DFS and OS, suggesting that KLK12 plays a tumor-supporting role in TNBC.
Collapse
Affiliation(s)
- Weiwei Gong
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, 81576, Munich, Germany
| | - Yueyang Liu
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, 81576, Munich, Germany.,Department of Gynecology, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Sarah Preis
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, 81576, Munich, Germany
| | - Xiaocong Geng
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, 81576, Munich, Germany
| | - Agnes Petit-Courty
- INSERM, U1100 - Centre d'Etude des Pathologies Respiratoires, Tours, France
| | - Marion Kiechle
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, 81576, Munich, Germany
| | | | - Tobias Dreyer
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, 81576, Munich, Germany
| | - Julia Dorn
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, 81576, Munich, Germany
| | - Yves Courty
- INSERM, U1100 - Centre d'Etude des Pathologies Respiratoires, Tours, France
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, Technical University of Munich, Ismaninger Str. 22, 81576, Munich, Germany.
| |
Collapse
|
21
|
De Vita E, Smits N, van den Hurk H, Beck EM, Hewitt J, Baillie G, Russell E, Pannifer A, Hamon V, Morrison A, McElroy SP, Jones P, Ignatenko NA, Gunkel N, Miller AK. Synthesis and Structure-Activity Relationships of N-(4-Benzamidino)-Oxazolidinones: Potent and Selective Inhibitors of Kallikrein-Related Peptidase 6. ChemMedChem 2020; 15:79-95. [PMID: 31675166 PMCID: PMC7004151 DOI: 10.1002/cmdc.201900536] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/23/2019] [Indexed: 12/16/2022]
Abstract
Kallikrein-related peptidase 6 (KLK6) is a secreted serine protease that belongs to the family of tissue kallikreins. Aberrant expression of KLK6 has been found in different cancers and neurodegenerative diseases, and KLK6 is currently studied as a potential target in these pathologies. We report a novel series of KLK6 inhibitors discovered in a high-throughput screen within the European Lead Factory program. Structure-guided design based on docking studies enabled rapid progression of a hit cluster to inhibitors with improved potency, selectivity and pharmacokinetic properties. In particular, inhibitors 32 ((5R)-3-(4-carbamimidoylphenyl)-N-((S)-1-(naphthalen-1-yl)propyl)-2-oxooxazolidine-5-carboxamide) and 34 ((5R)-3-(6-carbamimidoylpyridin-3-yl)-N-((1S)-1-(naphthalen-1-yl)propyl)-2-oxooxazolidine-5-carboxamide) have single-digit nanomolar potency against KLK6, with over 25-fold and 100-fold selectivities against the closely related enzyme trypsin, respectively. The most potent compound, 32, effectively reduces KLK6-dependent invasion of HCT116 cells. The high potency in combination with good solubility and low clearance of 32 make it a good chemical probe for KLK6 target validation in vitro and potentially in vivo.
Collapse
Affiliation(s)
- Elena De Vita
- Cancer Drug Development GroupGerman Cancer Research Center (DKFZ)Im Neuenheimer Feld 28069120HeidelbergGermany
- Faculty of BiosciencesUniversity of Heidelberg69120HeidelbergGermany
| | - Niels Smits
- Pivot Park Screening CentreKloosterstraat 95349 ABOss (TheNetherlands
| | | | - Elizabeth M. Beck
- European Screening Centre Newhouse (ESC) Biocity ScotlandBo'ness RoadML15UHNewhouseScotland
| | - Joanne Hewitt
- European Screening Centre Newhouse (ESC) Biocity ScotlandBo'ness RoadML15UHNewhouseScotland
| | - Gemma Baillie
- European Screening Centre Newhouse (ESC) Biocity ScotlandBo'ness RoadML15UHNewhouseScotland
| | - Emily Russell
- European Screening Centre Newhouse (ESC) Biocity ScotlandBo'ness RoadML15UHNewhouseScotland
| | - Andrew Pannifer
- European Screening Centre Newhouse (ESC) Biocity ScotlandBo'ness RoadML15UHNewhouseScotland
| | - Véronique Hamon
- European Screening Centre Newhouse (ESC) Biocity ScotlandBo'ness RoadML15UHNewhouseScotland
| | - Angus Morrison
- European Screening Centre Newhouse (ESC) Biocity ScotlandBo'ness RoadML15UHNewhouseScotland
| | - Stuart P. McElroy
- European Screening Centre Newhouse (ESC) Biocity ScotlandBo'ness RoadML15UHNewhouseScotland
| | - Philip Jones
- European Screening Centre Newhouse (ESC) Biocity ScotlandBo'ness RoadML15UHNewhouseScotland
| | - Natalia A. Ignatenko
- University of Arizona Cancer CenterUniversity of ArizonaTucsonAZ 85721USA
- Department of Cellular and Molecular MedicineUniversity of ArizonaTucsonAZ 85721USA
| | - Nikolas Gunkel
- Cancer Drug Development GroupGerman Cancer Research Center (DKFZ)Im Neuenheimer Feld 28069120HeidelbergGermany
- German Cancer Consortium (DKTK)69120HeidelbergGermany
| | - Aubry K. Miller
- Cancer Drug Development GroupGerman Cancer Research Center (DKFZ)Im Neuenheimer Feld 28069120HeidelbergGermany
- German Cancer Consortium (DKTK)69120HeidelbergGermany
| |
Collapse
|
22
|
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
|
23
|
Kryza T, Bock N, Lovell S, Rockstroh A, Lehman ML, Lesner A, Panchadsaram J, Silva LM, Srinivasan S, Snell CE, Williams ED, Fazli L, Gleave M, Batra J, Nelson C, Tate EW, Harris J, Hooper JD, Clements JA. The molecular function of kallikrein-related peptidase 14 demonstrates a key modulatory role in advanced prostate cancer. Mol Oncol 2019; 14:105-128. [PMID: 31630475 PMCID: PMC6944120 DOI: 10.1002/1878-0261.12587] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/06/2019] [Accepted: 10/18/2019] [Indexed: 12/20/2022] Open
Abstract
Kallikrein-related peptidase 14 (KLK14) is one of the several secreted KLK serine proteases involved in prostate cancer (PCa) pathogenesis. While relatively understudied, recent reports have identified KLK14 as overexpressed during PCa development. However, the modulation of KLK14 expression during PCa progression and the molecular and biological functions of this protease in the prostate tumor microenvironment remain unknown. To determine the modulation of KLK14 expression during PCa progression, we analyzed the expression levels of KLK14 in patient samples using publicly available databases and immunohistochemistry. In order to delineate the molecular mechanisms involving KLK14 in PCa progression, we integrated proteomic, transcriptomic, and in vitro assays with the goal to identify substrates, related-signaling pathways, and functional roles of this protease. We showed that KLK14 expression is elevated in advanced PCa, and particularly in metastasis. Additionally, KLK14 levels were found to be decreased in PCa tissues from patients responsive to neoadjuvant therapy compared to untreated patients. Furthermore, we also identified that KLK14 expression reoccurred in patients who developed castrate-resistant PCa. The combination of proteomic and transcriptomic analysis as well as functional assays revealed several new KLK14 substrates (agrin, desmoglein 2, vitronectin, laminins) and KLK14-regulated genes (Interleukin 32, midkine, SRY-Box 9), particularly an involvement of the mitogen-activated protein kinase 1 and interleukin 1 receptor pathways, and an involvement of KLK14 in the regulation of cellular migration, supporting its involvement in aggressive features of PCa progression. In conclusion, our work showed that KLK14 expression is associated with the development of aggressive PCa suggesting that targeting this protease could offer a novel route to limit the progression of prostate tumors. Additional work is necessary to determine the benefits and implications of targeting/cotargeting KLK14 in PCa as well as to determine the potential use of KLK14 expression as a predictor of PCa aggressiveness or response to treatment.
Collapse
Affiliation(s)
- Thomas Kryza
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation, Queensland University of Technology, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia.,Mater Research Institute - The University of Queensland, Brisbane, Australia
| | - Nathalie Bock
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation, Queensland University of Technology, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia
| | - Scott Lovell
- Department of Chemistry, Imperial College London, UK
| | - Anja Rockstroh
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation, Queensland University of Technology, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia
| | - Melanie L Lehman
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation, Queensland University of Technology, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia.,Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Canada
| | - Adam Lesner
- Faculty of Chemistry, University of Gdansk, Poland
| | - Janaththani Panchadsaram
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation, Queensland University of Technology, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia
| | - Lakmali Munasinghage Silva
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia
| | - Srilakshmi Srinivasan
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation, Queensland University of Technology, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia
| | - Cameron E Snell
- Mater Research Institute - The University of Queensland, Brisbane, Australia.,Mater Health Services, South Brisbane, Australia
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation, Queensland University of Technology, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia
| | - Ladan Fazli
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Canada
| | - Jyotsna Batra
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation, Queensland University of Technology, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia
| | - Colleen Nelson
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation, Queensland University of Technology, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia
| | - Edward W Tate
- Department of Chemistry, Imperial College London, UK
| | - Jonathan Harris
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia
| | - John D Hooper
- Mater Research Institute - The University of Queensland, Brisbane, Australia.,Mater Health Services, South Brisbane, Australia
| | - Judith A Clements
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation, Queensland University of Technology, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Woolloongabba, Australia.,Translational Research Institute, Woolloongabba, Australia
| |
Collapse
|
24
|
Goettig P, Brandstetter H, Magdolen V. Surface loops of trypsin-like serine proteases as determinants of function. Biochimie 2019; 166:52-76. [PMID: 31505212 PMCID: PMC7615277 DOI: 10.1016/j.biochi.2019.09.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023]
Abstract
Trypsin and chymotrypsin-like serine proteases from family S1 (clan PA) constitute the largest protease group in humans and more generally in vertebrates. The prototypes chymotrypsin, trypsin and elastase represent simple digestive proteases in the gut, where they cleave nearly any protein. Multidomain trypsin-like proteases are key players in the tightly controlled blood coagulation and complement systems, as well as related proteases that are secreted from diverse immune cells. Some serine proteases are expressed in nearly all tissues and fluids of the human body, such as the human kallikreins and kallikrein-related peptidases with specialization for often unique substrates and accurate timing of activity. HtrA and membrane-anchored serine proteases fulfill important physiological tasks with emerging roles in cancer. The high diversity of all family members, which share the tandem β-barrel architecture of the chymotrypsin-fold in the catalytic domain, is conferred by the large differences of eight surface loops, surrounding the active site. The length of these loops alters with insertions and deletions, resulting in remarkably different three-dimensional arrangements. In addition, metal binding sites for Na+, Ca2+ and Zn2+ serve as regulatory elements, as do N-glycosylation sites. Depending on the individual tasks of the protease, the surface loops determine substrate specificity, control the turnover and allow regulation of activation, activity and degradation by other proteins, which are often serine proteases themselves. Most intriguingly, in some serine proteases, the surface loops interact as allosteric network, partially tuned by protein co-factors. Knowledge of these subtle and complicated molecular motions may allow nowadays for new and specific pharmaceutical or medical approaches.
Collapse
Affiliation(s)
- Peter Goettig
- Division of Structural Biology, Department of Biosciences, University of Salzburg, Billrothstrasse 11, 5020, Salzburg, Austria.
| | - Hans Brandstetter
- Division of Structural Biology, Department of Biosciences, University of Salzburg, Billrothstrasse 11, 5020, Salzburg, Austria
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, School of Medicine, Technical University of Munich, Ismaninger Strasse 22, 81675, München, Germany
| |
Collapse
|
25
|
Nishimiya D, Kawaguchi Y, Kodama S, Nasu H, Yano H, Yamaguchi A, Tamura M, Hashimoto R. A protein scaffold, engineered SPINK2, for generation of inhibitors with high affinity and specificity against target proteases. Sci Rep 2019; 9:11436. [PMID: 31391482 PMCID: PMC6686015 DOI: 10.1038/s41598-019-47615-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 07/19/2019] [Indexed: 12/21/2022] Open
Abstract
Proteases are one of attractive therapeutic targets to play key roles in pharmacological action. There are many protease inhibitors in nature, and most of them structurally have cystine knot motifs. Their structures are favorable for recognition of active pockets of proteases, leading to the potent inhibition. However, they also have drawbacks, such as broad cross-reactivity, on the therapeutic application. To create therapeutic proteins derived from a disulfide-rich scaffold, we selected human serine protease inhibitor Kazal type 2 (SPINK2) through a scaffold screening, as a protein scaffold with requirements for therapeutic proteins. We then constructed a diverse library of the engineered SPINK2 by introducing random mutations into its flexible loop region with the designed method. By phage panning against four serine proteases, we isolated potent inhibitors against each target with picomolar KD and sub-nanomolar Ki values. Also, they exhibited the desired specificities against target proteases without inhibiting non-target proteases. The crystal structure of kallikrein related peptidase 4 (KLK4)-engineered SPINK2 complex revealed the interface with extensive conformational complementarity. Our study demonstrates that engineered SPINK2 can serve as a scaffold to generate therapeutic molecules against target proteins with groove structures.
Collapse
Affiliation(s)
- Daisuke Nishimiya
- DAIICHI SANKYO CO., LTD., Biologics Division, Modality Research Laboratories, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan.
| | - Yoshirou Kawaguchi
- DAIICHI SANKYO CO., LTD., Biologics Division, Modality Research Laboratories, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Shiho Kodama
- DAIICHI SANKYO CO., LTD., Biologics Division, Modality Research Laboratories, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Hatsumi Nasu
- DAIICHI SANKYO CO., LTD., Biologics Division, Modality Research Laboratories, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Hidenori Yano
- DAIICHI SANKYO CO., LTD., Biologics Division, Modality Research Laboratories, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Aya Yamaguchi
- DAIICHI SANKYO CO., LTD., Biologics Division, Modality Research Laboratories, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Masakazu Tamura
- DAIICHI SANKYO CO., LTD., Biologics Division, Modality Research Laboratories, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Ryuji Hashimoto
- DAIICHI SANKYO CO., LTD., Biologics Division, Modality Research Laboratories, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| |
Collapse
|
26
|
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
|
27
|
Horn M, Zbodakova O, Kasparek P, Srp J, Haneckova R, Hradilek M, Mares M, Sedlacek R. Profiling system for skin kallikrein proteolysis applied in gene-deficient mouse models. Biol Chem 2019; 399:1085-1089. [PMID: 29885275 DOI: 10.1515/hsz-2018-0116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/14/2018] [Indexed: 12/22/2022]
Abstract
Kallikrein-related proteases (KLKs) play a critical role in epidermis physiology and have been implicated in skin pathologies such as Netherton syndrome. The contribution of individual KLKs to skin proteolysis is poorly understood. Monitoring of their activities in skin proteome is hampered by overlapping substrate specificities, and there is a need for novel assays. Here, we present a platform of selective and sensitive fluorogenic substrates and inhibitors for profiling KLK5, KLK7 and KLK14. These chemical tools were evaluated using recombinant KLKs and tissue from a unique set of mice deficient in eight combinations of KLKs and their natural regulator LEKTI.
Collapse
Affiliation(s)
- Martin Horn
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague, Czech Republic
| | - Olga Zbodakova
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Petr Kasparek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, Prumyslova 595, 252 50 Vestec, Czech Republic.,Czech Centre for Phenogenomics, Institute of Molecular Genetics of the ASCR, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Jaroslav Srp
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague, Czech Republic.,Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 128 00 Prague, Czech Republic
| | - Radka Haneckova
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Martin Hradilek
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague, Czech Republic
| | - Michael Mares
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague, Czech Republic
| | - Radislav Sedlacek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, Prumyslova 595, 252 50 Vestec, Czech Republic.,Czech Centre for Phenogenomics, Institute of Molecular Genetics of the ASCR, Prumyslova 595, 252 50 Vestec, Czech Republic
| |
Collapse
|
28
|
Schilling O, Biniossek ML, Mayer B, Elsässer B, Brandstetter H, Goettig P, Stenman UH, Koistinen H. Specificity profiling of human trypsin-isoenzymes. Biol Chem 2019; 399:997-1007. [PMID: 29883318 DOI: 10.1515/hsz-2018-0107] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/05/2018] [Indexed: 01/18/2023]
Abstract
In humans, three different trypsin-isoenzymes have been described. Of these, trypsin-3 appears to be functionally different from the others. In order to systematically study the specificity of the trypsin-isoenzymes, we utilized proteome-derived peptide libraries and quantitative proteomics. We found similar specificity profiles dominated by the well-characterized preference for cleavage after lysine and arginine. Especially, trypsin-1 slightly favored lysine over arginine in this position, while trypsin-3 did not discriminate between them. In the P1' position, which is the residue C-terminal to the cleavage site, we noticed a subtle enrichment of alanine and glycine for all three trypsins and for trypsin-3 there were additional minor P1' and P2' preferences for threonine and aspartic acid, respectively. These findings were confirmed by FRET peptide substrates showing different susceptibility to cleavage by different trypsins. The preference of trypsin-3 for aspartic acid in P2' is explained by salt bridge formation with the unique Arg193. This salt bridge enables and stabilizes a canonical oxyanion conformation by the amides of Ser195 and Arg193, thus manifesting a selective substrate-assisted catalysis. As trypsin-3 has been proposed to be a therapeutic target and marker for cancers, our results may aid the development of specific inhibitors for cancer therapy and diagnostic probes.
Collapse
Affiliation(s)
- Oliver Schilling
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, D-79104 Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Martin L Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany
| | - Bettina Mayer
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, D-79104 Freiburg, Germany
| | - Brigitta Elsässer
- Department of Biosciences, University of Salzburg, Billrothstr. 11, A-5020 Salzburg, Austria
| | - Hans Brandstetter
- Department of Biosciences, University of Salzburg, Billrothstr. 11, A-5020 Salzburg, Austria
| | - Peter Goettig
- Department of Biosciences, University of Salzburg, Billrothstr. 11, A-5020 Salzburg, Austria
| | - Ulf-Håkan Stenman
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Central Hospital, Haartmaninkatu 8, FI-00290 Helsinki, Finland
| | - Hannu Koistinen
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Central Hospital, Haartmaninkatu 8, FI-00290 Helsinki, Finland
| |
Collapse
|
29
|
Adamopoulos PG, Kontos CK, Scorilas A. Novel splice variants of the human kallikrein-related peptidases 11 (KLK11) and 12 (KLK12), unraveled by next-generation sequencing technology. Biol Chem 2019; 399:1065-1071. [PMID: 29874189 DOI: 10.1515/hsz-2017-0294] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 04/15/2018] [Indexed: 01/18/2023]
Abstract
Tissue kallikrein, kallikrein-related peptidases (KLKs), and plasma kallikrein form the largest group of serine proteases in the human genome, sharing many structural and functional characteristics. In this study, we describe the molecular cloning of four novel splice variants of the human KLK11 and KLK12 genes, discovered by combining 3' rapid amplification of cDNA ends (3' RACE), next-generation sequencing (NGS) technology, advanced bioinformatic analysis and Sanger sequencing. Expression analysis of these new transcripts in cell lines originating from 17 cancerous and two normal tissues revealed the expression pattern of each transcript. These novel KLK11 and KLK12 splice variants represent new potential cancer biomarkers.
Collapse
Affiliation(s)
- Panagiotis G Adamopoulos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Panepistimiopolis, GR-15701 Athens, Greece
| | - Christos K Kontos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Panepistimiopolis, GR-15701 Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Panepistimiopolis, GR-15701 Athens, Greece
| |
Collapse
|
30
|
Alexander-Curtis M, Pauls R, Chao J, Volpi JJ, Bath PM, Verdoorn TA. Human tissue kallikrein in the treatment of acute ischemic stroke. Ther Adv Neurol Disord 2019; 12:1756286418821918. [PMID: 30719079 PMCID: PMC6348491 DOI: 10.1177/1756286418821918] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/24/2018] [Indexed: 01/08/2023] Open
Abstract
Acute ischemic stroke (AIS) remains a major cause of death and disability throughout the world. The most severe form of stroke results from large vessel occlusion of the major branches of the Circle of Willis. The treatment strategies currently available in western countries for large vessel occlusion involve rapid restoration of blood flow through removal of the offending blood clot using mechanical or pharmacological means (e.g. tissue plasma activator; tPA). This review assesses prospects for a novel pharmacological approach to enhance the availability of the natural enzyme tissue kallikrein (KLK1), an important regulator of local blood flow. KLK1 is responsible for the generation of kinins (bradykinin and kallidin), which promote local vasodilation and long-term vascularization. Moreover, KLK1 has been used clinically as a direct treatment for multiple diseases associated with impaired local blood flow including AIS. A form of human KLK1 isolated from human urine is approved in the People's Republic of China for subacute treatment of AIS. Here we review the rationale for using KLK1 as an additional pharmacological treatment for AIS by providing the biochemical mechanism as well as the human clinical data that support this approach.
Collapse
Affiliation(s)
| | - Rick Pauls
- DiaMedica Therapeutics, Minneapolis, MN, USA
| | - Julie Chao
- Medical University of South Carolina, Department of Biochemistry and Molecular Biology, Charleston, SC, USA
| | - John J Volpi
- Houston Methodist, Stanley H. Appel Department of Neurology, Houston, TX, USA
| | - Philip M Bath
- Stroke Trials Unit, University of Nottingham, City Hospital Campus, Nottingham, UK
| | | |
Collapse
|
31
|
Muytjens CMJ, Yu Y, Diamandis EP. Functional proteomic profiling reveals KLK13 and TMPRSS11D as active proteases in the lower female reproductive tract. F1000Res 2019; 7:1666. [PMID: 30647911 PMCID: PMC6329257 DOI: 10.12688/f1000research.16255.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/13/2018] [Indexed: 11/20/2022] Open
Abstract
Background: Cervical-vaginal fluid (CVF) hydrates the mucosa of the lower female reproductive tract and is known to contain numerous proteases. The low pH of CVF (4.5 or below in healthy women of reproductive age) is a uniquely human attribute and poses a challenge for the proteolytic functioning of the proteases identified in this complex biological fluid. Despite the abundance of certain proteases in CVF, the proteolytic activity and function of proteases in CVF is not well characterized. Methods: In the present study, we employed fluorogenic substrate screening to investigate the influence of pH and inhibitory compounds on the proteolytic activity in CVF. Activity-based probe (ABP) proteomics has evolved as a powerful tool to investigate active proteases within complex proteomes and a trypsin-specific ABP was used to identify active proteases in CVF. Results: Serine proteases are among the most abundant proteins in the CVF proteome. Labeling human CVF samples with the trypsin-specific ABP revealed serine proteases transmembrane protein serine 11D and kallikrein-related peptidase 13 as active proteases in CVF. Furthermore, we demonstrated that the proteolytic activity in CVF is highly pH-dependent with an almost absolute inhibition of trypsin-like proteolytic activity at physiological pH levels. Conclusions: These findings provide a framework to understand proteolytic activity in CVF. Furthermore, the present results provide clues for a novel regulatory mechanism in which fluctuations in CVF pH have the potential to control the catalytic activity in the lower female reproductive tract.
Collapse
Affiliation(s)
- Carla M J Muytjens
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Toronto, Canada
| | - Yijing Yu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Toronto, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Toronto, Canada.,Department of Clinical Biochemistry, University Health Network, Canada, Toronto, Canada
| |
Collapse
|
32
|
Meunier M, Scandolera A, Chapuis E, Lambert C, Jarrin C, Robe P, Chajra H, Auriol D, Reynaud R. From stem cells protection to skin microbiota balance: Orobanche rapum extract, a new natural strategy. J Cosmet Dermatol 2018; 18:1140-1154. [PMID: 30485658 PMCID: PMC7379634 DOI: 10.1111/jocd.12804] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/17/2018] [Indexed: 12/25/2022]
Abstract
Background Healthy skin is a delicate balance between skin renewal and microbiota homeostasis, and its imbalance promotes premature aging and dermatological disorders. Skin stem cells are key actors in this process but their sensitivity to aging and external stressors such as UV reduces the skin renewal power. The skin microbiota has been recently described as active in the healthy skin, and its imbalance could trigger some disorders. Aims We hypothesized that reactivation of stem cells and maintenance of microbiota could be a disruptive strategy for younger and healthier skin. We thus developed a new plant extract that restores the entire skin renewal process by sequential activation from stem cells stimulation to microbiota protection. Methods We studied stem cells comportment in the presence of Orobanche rapum extract by survivin immunocytochemistry and caspases 3 and 9 dosages. We also analyzed epidermal differentiation markers by immunohistochemistry and lipids organization by GC/MS At the clinical level, we investigated the impact of O. rapum extract on microbiota and on skin aspect. Results We demonstrated an active protection of skin stem cells through the maintenance of their clone‐forming capacity and resistance to UV through the overexpression of survivin coupled to caspases inhibition. Furthermore, we showed the restoration of epidermal differentiation markers and ceramide biosynthesis favorable to orthorhombic organization. Clinical studies, including microbiota analysis, showed an active skin surface renewal coupled with microbiota protection. Conclusion We evidenced that our active ingredient is able to stimulate skin rejuvenation while protecting the cutaneous microbiota, creating healthier skin and thereby beauty.
Collapse
Affiliation(s)
- Marie Meunier
- Research and Development, Givaudan France SAS, Pomacle, France
| | | | - Emilie Chapuis
- Research and Development, Givaudan France SAS, Pomacle, France
| | - Carole Lambert
- Research and Development, Givaudan France SAS, Toulouse, France
| | - Cyrille Jarrin
- Research and Development, Givaudan France SAS, Toulouse, France
| | - Patrick Robe
- Research and Development, Givaudan France SAS, Toulouse, France
| | - Hanane Chajra
- Research and Development, Givaudan France SAS, Toulouse, France
| | - Daniel Auriol
- Research and Development, Givaudan France SAS, Toulouse, France
| | - Romain Reynaud
- Research and Development, Givaudan France SAS, Pomacle, France
| |
Collapse
|
33
|
Kahler U, Fuchs JE, Goettig P, Liedl KR. An unexpected switch in peptide binding mode: from simulation to substrate specificity. J Biomol Struct Dyn 2018; 36:4072-4084. [PMID: 29210603 PMCID: PMC6334781 DOI: 10.1080/07391102.2017.1407674] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022]
Abstract
A ten microsecond molecular dynamics simulation of a kallikrein-related peptidase 7 peptide complex revealed an unexpected change in binding mode. After more than two microseconds unrestrained sampling we observe a spontaneous transition of the binding pose including a 180° rotation around the P1 residue. Subsequently, the substrate peptide occupies the prime side region rather than the cognate non-prime side in a stable conformation. We characterize the unexpected binding mode in terms of contacts, solvent-accessible surface area, molecular interactions and energetic properties. We compare the new pose to inhibitor-bound structures of kallikreins with occupied prime side and find that a similar orientation is adopted. Finally, we apply in silico mutagenesis based on the alternative peptide binding position to explore the prime side specificity of kallikrein-related peptidase 7 and compare it to available experimental data. Our study provides the first microsecond time scale simulation data on a kallikrein protease and shows previously unexplored prime side interactions. Therefore, we expect our study to advance the rational design of inhibitors targeting kallikrein-related peptidase 7, an emerging drug target involved in several skin diseases as well as cancer.
Collapse
Affiliation(s)
- Ursula Kahler
- Faculty of Chemistry and Pharmacy, Institute of General, Inorganic and Theoretical Chemistry, University Innsbruck, Innrain 82, InnsbruckA-6020, Austria
| | - Julian E. Fuchs
- Faculty of Chemistry and Pharmacy, Institute of General, Inorganic and Theoretical Chemistry, University Innsbruck, Innrain 82, InnsbruckA-6020, Austria
| | - Peter Goettig
- Division of Structural Biology, Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, SalzburgA-5020, Austria
| | - Klaus R. Liedl
- Faculty of Chemistry and Pharmacy, Institute of General, Inorganic and Theoretical Chemistry, University Innsbruck, Innrain 82, InnsbruckA-6020, Austria
| |
Collapse
|
34
|
Guo S, Briza P, Magdolen V, Brandstetter H, Goettig P. Activation and activity of glycosylated KLKs 3, 4 and 11. Biol Chem 2018; 399:1009-1022. [DOI: 10.1515/hsz-2018-0148] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/21/2018] [Indexed: 01/10/2023]
Abstract
Abstract
Human kallikrein-related peptidases 3, 4, 11, and KLK2, the activator of KLK3/PSA, belong to the prostatic group of the KLKs, whose major physiological function is semen liquefaction during the fertilization process. Notably, these KLKs are upregulated in prostate cancer and are used as clinical biomarkers or have been proposed as therapeutic targets. However, this potential awaits a detailed characterization of these proteases. In order to study glycosylated prostatic KLKs resembling the natural proteases, we used Leishmania (LEXSY) and HEK293 cells for secretory expression. Both systems allowed the subsequent purification of soluble pro-KLK zymogens with correct propeptides and of the mature forms. Periodic acid-Schiff reaction, enzymatic deglycosylation assays, and mass spectrometry confirmed the glycosylation of these KLKs. Activation of glycosylated pro-KLKs 4 and 11 turned out to be most efficient by glycosylated KLK2 and KLK4, respectively. By comparing the glycosylated prostatic KLKs with their non-glycosylated counterparts from Escherichia coli, it was observed that the N-glycans stabilize the KLK proteases and change their activation profiles and their enzymatic activity to some extent. The functional role of glycosylation in prostate-specific KLKs could pave the way to a deeper understanding of their biology and to medical applications.
Collapse
|
35
|
Loessner D, Goettig P, Preis S, Felber J, Bronger H, Clements JA, Dorn J, Magdolen V. Kallikrein-related peptidases represent attractive therapeutic targets for ovarian cancer. Expert Opin Ther Targets 2018; 22:745-763. [PMID: 30114962 DOI: 10.1080/14728222.2018.1512587] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Aberrant levels of kallikrein-related peptidases (KLK1-15) have been linked to cancer cell proliferation, invasion and metastasis. In ovarian cancer, the KLK proteolytic network has a crucial role in the tissue and tumor microenvironment. Publically available ovarian cancer genome and expression data from multiple patient cohorts show an upregulation of most KLKs. Areas covered: Here, we review the expression levels of all 15 members of this family in normal and ovarian cancer tissues, categorizing them into highly and moderately or weakly expressed KLKs, and their association with patient prognosis and survival. We summarize their tumor-biological functions determined in cell-based assays and xenograft models, further highlighting their suitability as cancer biomarkers and attractive candidates for drug development. Finally, we discuss some different pharmaceutical approaches, including peptide-based and small molecule inhibitors, cyclic peptides, depsipeptides, engineered natural inhibitors, antibodies, RNA/DNA-based aptamers, prodrugs, miRNA and siRNA. Expert opinion: In light of the results from clinical and tumor-biological studies, together with the available pharmaceutical tools, we suggest KLK4, KLK5, KLK6 and possibly KLK7 as preferred targets for inhibition in ovarian cancer.
Collapse
Affiliation(s)
- Daniela Loessner
- a Barts Cancer Institute , Queen Mary University of London , London , UK.,b Institute of Health and Biomedical Innovation , Queensland University of Technology (QUT) , Brisbane , Australia
| | - Peter Goettig
- c Department of Biosciences , University of Salzburg , Salzburg , Austria
| | - Sarah Preis
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Johanna Felber
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Holger Bronger
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Judith A Clements
- b Institute of Health and Biomedical Innovation , Queensland University of Technology (QUT) , Brisbane , Australia.,e Australian Prostate Cancer Research Centre - Queensland , Queensland University of Technology (QUT), Translational Research Institute , Brisbane , Australia
| | - Julia Dorn
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| | - Viktor Magdolen
- d Department of Obstetrics and Gynecology , Technical University of Munich , Munich , Germany
| |
Collapse
|
36
|
Structural determinants of specificity and regulation of activity in the allosteric loop network of human KLK8/neuropsin. Sci Rep 2018; 8:10705. [PMID: 30013126 PMCID: PMC6048020 DOI: 10.1038/s41598-018-29058-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/27/2018] [Indexed: 11/12/2022] Open
Abstract
Human KLK8/neuropsin, a kallikrein-related serine peptidase, is mostly expressed in skin and the hippocampus regions of the brain, where it regulates memory formation by synaptic remodeling. Substrate profiles of recombinant KLK8 were analyzed with positional scanning using fluorogenic tetrapeptides and the proteomic PICS approach, which revealed the prime side specificity. Enzyme kinetics with optimized substrates showed stimulation by Ca2+ and inhibition by Zn2+, which are physiological regulators. Crystal structures of KLK8 with a ligand-free active site and with the inhibitor leupeptin explain the subsite specificity and display Ca2+ bound to the 75-loop. The variants D70K and H99A confirmed the antagonistic role of the cation binding sites. Molecular docking and dynamics calculations provided insights in substrate binding and the dual regulation of activity by Ca2+ and Zn2+, which are important in neuron and skin physiology. Both cations participate in the allosteric surface loop network present in related serine proteases. A comparison of the positional scanning data with substrates from brain suggests an adaptive recognition by KLK8, based on the tertiary structures of its targets. These combined findings provide a comprehensive picture of the molecular mechanisms underlying the enzyme activity of KLK8.
Collapse
|
37
|
Frombach J, Lohan SB, Lemm D, Gruner P, Hasler J, Ahlberg S, Blume-Peytavi U, Unbehauen M, Haag R, Meinke MC, Vogt A. Protease-mediated Inflammation: An In Vitro Human Keratinocyte-based Screening Tool for Anti-inflammatory Drug Nanocarrier Systems. ACTA ACUST UNITED AC 2018. [DOI: 10.1515/zpch-2017-1048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Abstract
Background:
Refined encapsulation approaches in dermatotherapy gain increased interest. There is need of reproducible in vitro systems representing disease features to screen drug delivery systems for preclinical assessment. Inflammatory human skin diseases are commonly accompanied by abnormal epidermal differentiation and barrier impairment. Serine proteases (SPs) and their inhibitors play a critical role in such dysfunctional differentiation. SPs also initiate cellular pathways via activation of protease-activated receptors, which contribute to inflammation. Thus, function and activity of SPs should be considered for the design of new therapies of such disorders.
Objectives:
Herein, we established a novel simplified cell culture model, based on SP-mediated inflammation suitable to assess nanocarriers loaded with anti-inflammatory drugs.
Methods:
SP-mediated inflammation and the regulatory effect of free or encapsulated dexamethasone were determined by measuring interleukin-6 and interleukin-8 in culture medium of HaCaT (human adult low calcium temperature)-keratinocytes. Additionally, radical formation was analyzed by electron paramagnetic resonance spectroscopy. Cellular uptake of core-multishell nanocarriers was investigated by fluorescence microscopy. Cytotoxicity of all additives was determined by a viability assay.
Results:
SP-Stimulation of keratinocytes resulted in increased radical production and release of inflammatory cytokines without affecting cell viability. Induced inflammation was successfully downregulated by addition of free or encapsulated dexamethasone.
Conclusion:
SP-addition can be used as inflammatory stimulus in cell culture to mimic effects of aberrant enzymatic activities found in skin of atopic dermatitis patients. The set-up is appropriate as a preliminary test to examine the effectiveness of new molecules or delivery-systems to counteract serine protease-mediated inflammatory processes prior to skin studies.
Collapse
Affiliation(s)
- Janna Frombach
- Clinical Research Center for Hair and Skin Science , Department of Dermatology, Venerology and Allergy , Charité – Universitätsmedizin Berlin , Berlin , Germany
| | - Silke B. Lohan
- Center of Experimental and Applied Cutaneous Physiology , Department of Dermatology, Venerology and Allergy , Charité – Universitätsmedizin Berlin , Berlin , Germany
| | - Davina Lemm
- Clinical Research Center for Hair and Skin Science , Department of Dermatology, Venerology and Allergy , Charité – Universitätsmedizin Berlin , Berlin , Germany
| | - Paul Gruner
- Clinical Research Center for Hair and Skin Science , Department of Dermatology, Venerology and Allergy , Charité – Universitätsmedizin Berlin , Berlin , Germany
| | - Julia Hasler
- Center of Experimental and Applied Cutaneous Physiology , Department of Dermatology, Venerology and Allergy , Charité – Universitätsmedizin Berlin , Berlin , Germany
| | - Sebastian Ahlberg
- Center of Experimental and Applied Cutaneous Physiology , Department of Dermatology, Venerology and Allergy , Charité – Universitätsmedizin Berlin , Berlin , Germany
| | - Ulrike Blume-Peytavi
- Clinical Research Center for Hair and Skin Science , Department of Dermatology, Venerology and Allergy , Charité – Universitätsmedizin Berlin , Berlin , Germany
| | - Michael Unbehauen
- Freie Universität Berlin , Institut für Chemie und Biochemie , Berlin , Germany
| | - Rainer Haag
- Freie Universität Berlin , Institut für Chemie und Biochemie , Berlin , Germany
| | - Martina C. Meinke
- Center of Experimental and Applied Cutaneous Physiology , Department of Dermatology, Venerology and Allergy , Charité – Universitätsmedizin Berlin , Berlin , Germany
| | - Annika Vogt
- Clinical Research Center for Hair and Skin Science , Department of Dermatology, Venerology and Allergy , Charité – Universitätsmedizin Berlin, Charitéplatz 1 , Berlin , Germany , Phone: +4930450518207, 10117
| |
Collapse
|
38
|
Kryza T, Parent C, Pardessus J, Petit A, Burlaud-Gaillard J, Reverdiau P, Iochmann S, Labas V, Courty Y, Heuzé-Vourc'h N. Human kallikrein-related peptidase 12 stimulates endothelial cell migration by remodeling the fibronectin matrix. Sci Rep 2018; 8:6331. [PMID: 29679011 PMCID: PMC5910384 DOI: 10.1038/s41598-018-24576-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/05/2018] [Indexed: 12/30/2022] Open
Abstract
Kallikrein-related peptidase 12 (KLK12) is a kallikrein family peptidase involved in angiogenesis - a complex biological process in which the sprouting, migration and stabilization of endothelial cells requires extracellular matrix remodeling. To characterize the molecular mechanisms associated with KLK12's proangiogenic activity, we evaluated its ability to hydrolyze various matrix proteins. Our results show that KLK12 efficiently cleaved the human extracellular matrix proteins fibronectin and tenascin, both of which are involved in the regulation of endothelial cell adhesion and migration. For fibronectin, the major proteolytic product generated by KLK12 was a 29 kDa fragment containing the amino-terminal domain and the first five type I fibronectin-domains, which are essential for regulating fibronectin assembly. We also demonstrated that KLK12-mediated fibronectin proteolysis antagonizes fibronectin polymerization and fibronectin fibril formation by endothelial cells, leading to an increase in cell migration. Furthermore, a polyclonal antibody raised against KLK12's proteolytic cleavage site on fibronectin prevented the KLK12-dependent inhibition of fibronectin polymerization and the KLK12-mediated pro-migratory effect on endothelial cells. Taken as a whole, our results indicate that KLK12's proangiogenic effect is mediated through several molecular mechanisms.
Collapse
Affiliation(s)
- T Kryza
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France.,Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Australia
| | - C Parent
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - J Pardessus
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - A Petit
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - J Burlaud-Gaillard
- Université François Rabelais de Tours, F-37032, Tours, France.,Plateforme IBiSA de Microscopie Electronique, Université François Rabelais de Tours, F-37032, Tours, France
| | - P Reverdiau
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - S Iochmann
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - V Labas
- PRC, INRA, CNRS, Université François Rabelais de Tours, IFCE, F-37380, Nouzilly, France.,PAIB, CIRE, INRA, CHRU de Tours, Université François Rabelais de Tours, F-37380, Nouzilly, France
| | - Y Courty
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - N Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France. .,Université François Rabelais de Tours, F-37032, Tours, France.
| |
Collapse
|
39
|
Fischer J, Meyer-Hoffert U. Regulation of kallikrein-related peptidases in the skin – from physiology to diseases to therapeutic options. Thromb Haemost 2017; 110:442-9. [DOI: 10.1160/th12-11-0836] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 01/25/2013] [Indexed: 12/21/2022]
Abstract
SummaryKallikrein-related peptidases (KLKs) constitute a family of 15 highly conserved serine proteases, which show a tissue-specific expression profile. This made them valuable tumour expression markers. It became evident that KLKs are involved in many physiological processes like semen liquefaction and skin desquamation. More recently, we have learnt that they are involved in many pathophysiological conditions and diseases making them promising target of therapeutic intervention. Therefore, regulation of KLKs raised the interest of numerous reports. Herein, we summarise the current knowledge on KLKs regulation with an emphasis on skin-relevant KLKs regulation processes. Regulation of KLKs takes place on the level of transcription, on protease activation and on protease inactivation. A variety of protease inhibitors has been described to interact with KLKs including the irreversible serine protease inhibitors (SERPINs) and the reversible serine protease inhibitors of Kazal-type (SPINKs). In an attempt to integrate current knowledge, we propose that KLK regulation has credentials as targets for therapeutic intervention.
Collapse
|
40
|
Clinical relevance of kallikrein-related peptidase 9, 10, 11, and 15 mRNA expression in advanced high-grade serous ovarian cancer. PLoS One 2017; 12:e0186847. [PMID: 29095848 PMCID: PMC5667830 DOI: 10.1371/journal.pone.0186847] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 10/09/2017] [Indexed: 11/19/2022] Open
Abstract
KLK9, 10, 11, and 15 may represent potential cancer biomarkers for evaluating ovarian cancer prognosis. In the present study, we selected a homogeneous cohort including 139 patients of advanced high-grade serous ovarian cancer (FIGO stage III/IV) and assessed the mRNA levels of KLK9, 10, 11, and 15 in tumor tissue by quantitative PCR. No significant associations of KLK9, 10, 11, and 15 mRNA with established clinical parameters (residual tumor mass, ascitic fluid volume) were found. Pronounced correlations between KLK10/KLK11 (rs = 0.647) and between KLK9/KLK15 (rs = 0.716) mRNA, but not between other combinations, indicate coordinate expression of distinct pairs of peptidases. In univariate Cox regression analysis, elevated KLK11 mRNA levels were significantly linked with prolonged overall survival (OS; p = 0.021) and progression-free survival (PFS; p = 0.008). KLK15 mRNA levels showed a trend towards significance in case of OS (p = 0.06); KLK9 and KLK10 mRNA expression levels were not associated with patients' outcome. In multivariable Cox analysis, KLK11 mRNA expression levels, apart from residual tumor mass, remained an independent predictive marker for OS (p = 0.007) and PFS (p = 0.015). Here, elevated KLK15 mRNA expression levels turned out to be significantly related to prolonged OS (p = 0.025) as well. High KLK11 but not the other KLK mRNA levels can be considered as strong independent favorable prognostic factor in this major ovarian cancer subtype.
Collapse
|
41
|
Silva LM, Clements JA. Mass spectrometry based proteomics analyses in kallikrein-related peptidase research: implications for cancer research and therapy. Expert Rev Proteomics 2017; 14:1119-1130. [PMID: 29025353 DOI: 10.1080/14789450.2017.1389637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Kallikrein-related peptidases (KLKs) are a family of serine peptidases that are deregulated in numerous pathological conditions, with a multitude of KLK-mediated functional roles implicated in the progression of cancer. Advances in multidimensional mass spectrometry (MS)-based proteomics have facilitated the quantitative measurement of deregulated KLK expression in cancer, identifying certain KLKs, as well as their substrates, as potential cancer biomarkers. Areas covered: In this review, we discuss how these approaches have been utilized for KLK biomarker discovery and unbiased substrate determination in complex protein pools that mimic the in vivo extracellular microenvironment. Expert commentary: Although a limited number of studies have been performed, the quantity of information generated has greatly improved our understanding of the functional roles of KLKs in cancer progression. In addition, these data suggest additional means through which deregulated KLK expression may be targeted in cancer treatment, highlighting the potential therapeutic value of these state-of-the-art MS-based studies.
Collapse
Affiliation(s)
- Lakmali Munasinghage Silva
- a Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch , National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda , MD , USA
| | - Judith Ann Clements
- b School of Biomedical Sciences , Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Translational Research Institute , Woolloongabba , Australia
| |
Collapse
|
42
|
Kryza T, Silva LM, Bock N, Fuhrman-Luck RA, Stephens CR, Gao J, Samaratunga H, Lawrence MG, Hooper JD, Dong Y, Risbridger GP, Clements JA. Kallikrein-related peptidase 4 induces cancer-associated fibroblast features in prostate-derived stromal cells. Mol Oncol 2017; 11:1307-1329. [PMID: 28510269 PMCID: PMC5623815 DOI: 10.1002/1878-0261.12075] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 04/11/2017] [Accepted: 04/27/2017] [Indexed: 01/09/2023] Open
Abstract
The reciprocal communication between cancer cells and their microenvironment is critical in cancer progression. Although involvement of cancer‐associated fibroblasts (CAF) in cancer progression is long established, the molecular mechanisms leading to differentiation of CAFs from normal fibroblasts are poorly understood. Here, we report that kallikrein‐related peptidase‐4 (KLK4) promotes CAF differentiation. KLK4 is highly expressed in prostate epithelial cells of premalignant (prostatic intraepithelial neoplasia) and malignant lesions compared to normal prostate epithelia, especially at the peristromal interface. KLK4 induced CAF‐like features in the prostate‐derived WPMY1 normal stromal cell line, including increased expression of alpha‐smooth muscle actin, ESR1 and SFRP1. KLK4 activated protease‐activated receptor‐1 in WPMY1 cells increasing expression of several factors (FGF1, TAGLN, LOX, IL8, VEGFA) involved in prostate cancer progression. In addition, KLK4 induced WPMY1 cell proliferation and secretome changes, which in turn stimulated HUVEC cell proliferation that could be blocked by a VEGFA antibody. Importantly, the genes dysregulated by KLK4 treatment of WPMY1 cells were also differentially expressed between patient‐derived CAFs compared to matched nonmalignant fibroblasts and were further increased by KLK4 treatment. Taken together, we propose that epithelial‐derived KLK4 promotes tumour progression by actively promoting CAF differentiation in the prostate stromal microenvironment.
Collapse
Affiliation(s)
- Thomas Kryza
- Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Queensland University of Technology (QUT), Woolloongabba, Australia.,Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Kelvin Grove, Australia
| | - Lakmali M Silva
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Kelvin Grove, Australia
| | - Nathalie Bock
- Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Queensland University of Technology (QUT), Woolloongabba, Australia.,Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Kelvin Grove, Australia
| | - Ruth A Fuhrman-Luck
- Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Queensland University of Technology (QUT), Woolloongabba, Australia.,Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Kelvin Grove, Australia
| | - Carson R Stephens
- Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Queensland University of Technology (QUT), Woolloongabba, Australia.,Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Kelvin Grove, Australia
| | - Jin Gao
- Regenerative Dentistry and Oral Biology, Oral Health Centre, University of Queensland, Herston, Australia
| | - Hema Samaratunga
- Aquesta Pathology, Toowong, Australia.,School of Medicine, University of Queensland, Herston, Australia
| | -
- Australian Prostate Cancer BioResource, The Prostate Cancer Research Program, Monash University, Clayton, Australia
| | - Mitchell G Lawrence
- Prostate Research Group, Cancer Program - Biomedicine Discovery Institute Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash University, Clayton, Australia
| | - John D Hooper
- Cancer Biology and Care Program, Translational Research Institute, Mater Research Institute - The University of Queensland, Woolloongabba, Australia
| | - Ying Dong
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Kelvin Grove, Australia
| | - Gail P Risbridger
- Prostate Research Group, Cancer Program - Biomedicine Discovery Institute Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash University, Clayton, Australia.,Prostate Cancer Translational Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Parkville, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia
| | - Judith A Clements
- Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Queensland University of Technology (QUT), Woolloongabba, Australia.,Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Kelvin Grove, Australia
| |
Collapse
|
43
|
Silva LM, Stoll T, Kryza T, Stephens CR, Hastie ML, Irving-Rodgers HF, Dong Y, Gorman JJ, Clements JA. Mass spectrometry-based determination of Kallikrein-related peptidase 7 (KLK7) cleavage preferences and subsite dependency. Sci Rep 2017; 7:6789. [PMID: 28754951 PMCID: PMC5533777 DOI: 10.1038/s41598-017-06680-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 06/15/2017] [Indexed: 11/23/2022] Open
Abstract
The cleavage preferences of Kallikrein-related peptidase 7 (KLK7) have previously been delineated using synthetic peptide libraries of fixed length, or single protein chains and have suggested that KLK7 exerts a chymotryptic-like cleavage preference. Due to the short length of the peptides utilised, only a limited number of subsites have however been assessed. To determine the subsite preferences of KLK7 in a global setting, we used a mass spectrometry (MS)-based in-depth proteomics approach that utilises human proteome-derived peptide libraries of varying length, termed Proteomic Identification of protease Cleavage Sites (PICS). Consistent with previous findings, KLK7 was found to exert chymotryptic-like cleavage preferences. KLK7 subsite preferences were also characterised in the P2-P2′ region, demonstrating a preference for hydrophobic residues in the non-prime and hydrophilic residues in the prime subsites. Interestingly, single catalytic triad mutant KLK7 (mKLK7; S195A) also showed residual catalytic activity (kcat/KM = 7.93 × 102 s−1M−1). Catalytic inactivity of KLK7 was however achieved by additional mutation in this region (D102N). In addition to characterising the cleavage preferences of KLK7, our data thereby also suggests that the use of double catalytic triad mutants should be employed as more appropriate negative controls in future investigations of KLK7, especially when highly sensitive MS-based approaches are employed.
Collapse
Affiliation(s)
- Lakmali Munasinghage Silva
- Queensland University of Technology (QUT), Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences at the Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia.,Proteases and Tissue Remodelling Section, National Institute of Dental and Craniofacial Research, National Institutes of Science, 30 Convent Drive, Bethesda, Maryland, 20892, USA
| | - Thomas Stoll
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, 4006, Australia
| | - Thomas Kryza
- Queensland University of Technology (QUT), Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences at the Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Carson Ryan Stephens
- Queensland University of Technology (QUT), Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences at the Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Marcus Lachlan Hastie
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, 4006, Australia
| | - Helen Frances Irving-Rodgers
- School of Medical Science, Griffith University Gold Coast Campus, Parklands Drive, Southport, Queensland, 4215, Australia
| | - Ying Dong
- Queensland University of Technology (QUT), Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences at the Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Jeffrey John Gorman
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland, 4006, Australia
| | - Judith Ann Clements
- Queensland University of Technology (QUT), Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences at the Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia.
| |
Collapse
|
44
|
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
|
45
|
Debela M, Magdolen V, Bode W, Brandstetter H, Goettig P. Structural basis for the Zn2+ inhibition of the zymogen-like kallikrein-related peptidase 10. Biol Chem 2017; 397:1251-1264. [PMID: 27611765 PMCID: PMC5551965 DOI: 10.1515/hsz-2016-0205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/04/2016] [Indexed: 12/18/2022]
Abstract
Although kallikrein-related peptidase 10 (KLK10) is expressed in a variety of human tissues and body fluids, knowledge of its physiological functions is fragmentary. Similarly, the pathophysiology of KLK10 in cancer is not well understood. In some cancer types, a role as tumor suppressor has been suggested, while in others elevated expression is associated with poor patient prognosis. Active human KLK10 exhibits a unique, three residue longer N-terminus with respect to other serine proteases and an extended 99-loop nearly as long as in tissue kallikrein KLK1. Crystal structures of recombinant ligand-free KLK10 and a Zn2+ bound form explain to some extent the mixed trypsin- and chymotrypsin-like substrate specificity. Zn2+-inhibition of KLK10 appears to be based on a unique mechanism, which involves direct binding and blocking of the catalytic triad. Since the disordered N-terminus and several loops adopt a zymogen-like conformation, the active protease conformation is very likely induced by interaction with the substrate, in particular at the S1 subsite and at the unusual Ser193 as part of the oxyanion hole. The KLK10 structures indicate that the N-terminus, the nearby 75-, 148-, and the 99-loops are connected in an allosteric network, which is present in other trypsin-like serine proteases with several variations.
Collapse
Affiliation(s)
| | - Viktor Magdolen
- Klinische Forschergruppe der Frauenklinik, Klinikum rechts der Isar der TU München, Ismaninger Str. 22, D-81675 München, Germany
| | - Wolfram Bode
- Max-Planck-Institut für Biochemie, Proteinase Research Group, Am Klopferspitz 18, D-82152 Martinsried, Germany
| | - Hans Brandstetter
- Division of Structural Biology, Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, A-5020 Salzburg, Austria
| | | |
Collapse
|
46
|
Cheong KA, Lee TR, Lee AY. Complementary effect of hydroquinone and retinoic acid on corneocyte desquamation with their combination use. J Dermatol Sci 2017; 87:192-200. [PMID: 28433430 DOI: 10.1016/j.jdermsci.2017.03.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/09/2017] [Accepted: 03/29/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Retinoic acid (RA) enhances skin-lightening capabilities of hydroquinone (HQ), at least in part, by facilitating desquamation which leads to increase penetration of HQ. The desquamation also affects skin irritation levels. The mechanism of RA-induced desquamation, however, has not been completely explored and no such data has been available for HQ uses. OBJECTIVE To examine the role of HQ, RA, and their combination in the desquamation. METHODS Primary cultured normal human keratinocytes, which were treated with HQ and/or RA in presence or absence of serine-specific inhibitor Kazal type5 (SPINK5)/lympho-epithelial Kazal-type-related inhibitor (LEKTI) knockdown or recombinant human SPINK5/LEKTI, and biopsied skin samples applied with HQ or RA were examined. Expression levels of corneodesmosin (CDSN), desmocollin1 (DSC1), kallikrein5 (KLK5), KLK7, and SPINK5/LEKTI, and proteolysis activity against extracted human skin epidermal protein were determined using time-course real-time PCR, Western blotting, ELISA, and immunofluorescence staining. RESULTS HQ increased but RA decreased the synthesis of CDSN and DSC1. HQ reduced corneodesmosome degradation by the upregulation of SPINK5/LEKTI, whereas RA showed opposite results without upregulation of SPINK5/LEKTI. The combination of HQ and RA was close to the sum of the individual components. CONCLUSIONS HQ reduced corneocyte desquamation. However, RA enhanced desquamation. The combination induced more desquamation than HQ but less than RA.
Collapse
Affiliation(s)
- Kyung Ah Cheong
- Department of Dermatology, Dongguk University Ilsan Hospital, 814 Siksa-dong, Ilsandong-gu, Goyang-si, Gyeonggi-do 410-773, Republic of Korea
| | - Tae Ryong Lee
- Bioscience Institute, AmorePacific Corporation R&D Center, Yongin-si, Gyeonggi-do, 446-729, Republic of Korea
| | - Ai-Young Lee
- Department of Dermatology, Dongguk University Ilsan Hospital, 814 Siksa-dong, Ilsandong-gu, Goyang-si, Gyeonggi-do 410-773, Republic of Korea.
| |
Collapse
|
47
|
Swedberg JE, Li CY, de Veer SJ, Wang CK, Craik DJ. Design of Potent and Selective Cathepsin G Inhibitors Based on the Sunflower Trypsin Inhibitor-1 Scaffold. J Med Chem 2017; 60:658-667. [PMID: 28045523 DOI: 10.1021/acs.jmedchem.6b01509] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neutrophils are directly responsible for destroying invading pathogens via reactive oxygen species, antimicrobial peptides, and neutrophil serine proteases (NSPs). Imbalance between NSP activity and endogenous protease inhibitors is associated with chronic inflammatory disorders, and engineered inhibitors of NSPs are a potential therapeutic pathway. In this study we characterized the extended substrate specificity (P4-P1) of the NSP cathepsin G using a peptide substrate library. Substituting preferred cathepsin G substrate sequences into sunflower trypsin inhibitor-1 (SFTI-1) produced a potent cathepsin G inhibitor (Ki = 0.89 nM). Cathepsin G's P2' preference was determined by screening against a P2' diverse SFTI-based library, and the most preferred residue at P2' was combined in SFTI-1 with a preferred substrate sequence (P4-P2) and a nonproteinogenic P1 residue (4-guanidyl-l-phenylalanine) to produce a potent (Ki = 1.6 nM) and the most selective (≥360-fold) engineered cathepsin G inhibitor reported to date. This compound is a promising lead for further development of cathepsin G inhibitors targeting chronic inflammatory disorders.
Collapse
Affiliation(s)
- Joakim E Swedberg
- Institute for Molecular Bioscience, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Choi Yi Li
- Institute for Molecular Bioscience, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Simon J de Veer
- Institute for Molecular Bioscience, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Conan K Wang
- Institute for Molecular Bioscience, The University of Queensland , Brisbane, QLD 4072, Australia
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland , Brisbane, QLD 4072, Australia
| |
Collapse
|
48
|
Ramachandran R, Altier C, Oikonomopoulou K, Hollenberg MD. Proteinases, Their Extracellular Targets, and Inflammatory Signaling. Pharmacol Rev 2016; 68:1110-1142. [PMID: 27677721 DOI: 10.1124/pr.115.010991] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Given that over 2% of the human genome codes for proteolytic enzymes and their inhibitors, it is not surprising that proteinases serve many physiologic-pathophysiological roles. In this context, we provide an overview of proteolytic mechanisms regulating inflammation, with a focus on cell signaling stimulated by the generation of inflammatory peptides; activation of the proteinase-activated receptor (PAR) family of G protein-coupled receptors (GPCR), with a mechanism in common with adhesion-triggered GPCRs (ADGRs); and by proteolytic ion channel regulation. These mechanisms are considered in the much wider context that proteolytic mechanisms serve, including the processing of growth factors and their receptors, the regulation of matrix-integrin signaling, and the generation and release of membrane-tethered receptor ligands. These signaling mechanisms are relevant for inflammatory, neurodegenerative, and cardiovascular diseases as well as for cancer. We propose that the inflammation-triggering proteinases and their proteolytically generated substrates represent attractive therapeutic targets and we discuss appropriate targeting strategies.
Collapse
Affiliation(s)
- Rithwik Ramachandran
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Christophe Altier
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Katerina Oikonomopoulou
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Morley D Hollenberg
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| |
Collapse
|
49
|
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
|
50
|
Sangster-Guity N, Tu-Sekine B, Raben DM, Denmeade SR, Williams SA. Mutational Analysis of Prostate-Specific Antigen Defines the Intrinsic Proteolytic Activity of the proPSA Zymogen. Prostate 2016; 76:1203-17. [PMID: 27273171 DOI: 10.1002/pros.23216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 05/09/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Prostate-specific antigen (PSA) is an important prostate cancer biomarker. It is also a protease expressed at high concentrations by the normal and malignant prostate. PSA is secreted as a zymogen (proPSA) with an inhibitory prodomain that must be removed for full activity. ProPSA variants, assumed to be inactive, are found in the blood of prostate cancer patients, and are indicative of poor clinical outcome. Despite the abundance of clinical reports, our understanding of PSA's enzymology is limited, in part due to a lack of appropriate experimental systems. We sought to develop a series of PSA-derived mutants that would help to enhance our understanding of the gene. METHODS Sixteen rPSA variants were generated and characterized by a variety of biochemical methods. RESULTS The wildtype cDNA (WT) provided the template for generating a panel of recombinants. These included variants that abolished removal of the prodomain (R24A), disabled its enzymatic activity (S213A), and/or facilitated a cell-based conversion to the active conformation (FR). The purified variants' proteolytic activity was examined using a fluorogenic substrate, known PSA-cleavable proteins, and physiologically relevant inhibitors. Upon demonstrating our successful generation and purification of the PSA variants, we characterized proPSA activity, describing cleavage of synthetic and biologic substrates, but not serum protease inhibitors. This finding was exploited in the development of a self-activating mutant (PSA_QY) that exhibited the greatest enzymatic activity of all the variants. CONCLUSIONS The system described herein will prove useful for varied applications. ProPSA is partially functional with relatively high activity compared to the mature enzyme. In demonstrating the zymogen's intrinsic activity, we suggest that the proPSA in prostate cancer patient serum is not inert. This may have implications for our understanding of the disease. Prostate 76:1203-1217, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Niquiche Sangster-Guity
- Brady Urological Research Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Becky Tu-Sekine
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel M Raben
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Samuel R Denmeade
- Brady Urological Research Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Simon A Williams
- Brady Urological Research Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|