1
|
Ni T, Zhao RH, Wu JF, Li CY, Xue G, Lin X. KLK7, KLK10, and KLK11 in Papillary Thyroid Cancer: Bioinformatic Analysis and Experimental Validation. Biochem Genet 2024; 62:4446-4471. [PMID: 38316654 DOI: 10.1007/s10528-024-10679-8] [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: 05/30/2023] [Accepted: 01/02/2024] [Indexed: 02/07/2024]
Abstract
Despite many studies on papillary thyroid carcinoma (PTC) in the past few decades, some critical and significant genes remain undiscovered. To explore genes that may play crucial roles in PTC, a detailed analysis of the expression levels, mutations, and clinical significance of Kallikrein-related peptidases (KLKs) family genes in PTC was undertaken to provide new targets for the precise treatment of the disease. A comprehensive analysis of KLK family genes was performed using various online tools, such as GEPIA, Kaplan-Meier Plotter, LinkedOmics, GSCA, TIMER, and Cluego. KLK7, KLK10, and KLK11 were critical factors of KLK family genes. Then, functional assays were carried out on KLK7/10/11 to determine their proliferation, migration, and invasion capabilities in PTC. The mRNA expression levels of KLK7, KLK10, KLK11, and KLK13 were significantly elevated in thyroid carcinoma, while KLK1, KLK2, KLK3 and KLK4 mRNA levels were decreased compared to normal tissues. Correlations between KLK2/7-12/15 expression levels and tumor stage were also observed in thyroid carcinoma. Survival analysis demonstrated that KLK4/5/7/9-12/14 was associated with overall survival in patients with thyroid cancer. Not only were KLK genes strongly associated with cancer-related pathways, but also KLK7/10/11 was associated with immune-cell infiltration. Finally, silencing KLK7/10/11 impaired human papillary thyroid carcinoma cells' growth, migration ability, and invasiveness. The increased expression of KLK7, KLK10, and KLK11 may serve as molecular markers to identify PTC patients. KLK7, KLK10, and KLK11 could be potential prognostic indicators and targets for precision therapy against PTC.
Collapse
Affiliation(s)
- Tao Ni
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China
| | - Ru-Hua Zhao
- Department of Morphology Laboratory, Hebei North University, Zhangjiakou, 075000, China
| | - Jing-Fang Wu
- Department of Morphology Laboratory, Hebei North University, Zhangjiakou, 075000, China
| | - Chao-You Li
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China
| | - Gang Xue
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China.
| | - Xu Lin
- Department of Morphology Laboratory, Hebei North University, Zhangjiakou, 075000, China.
| |
Collapse
|
2
|
Wenta T, Nastaly P, Lipinska B, Manninen A. Remodeling of the extracellular matrix by serine proteases as a prerequisite for cancer initiation and progression. Matrix Biol 2024; 134:197-219. [PMID: 39500383 DOI: 10.1016/j.matbio.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024]
Abstract
The extracellular matrix (ECM) serves as a physical scaffold for tissues that is composed of structural proteins such as laminins, collagens, proteoglycans and fibronectin, forming a three dimensional network, and a wide variety of other matrix proteins with ECM-remodeling and signaling functions. The activity of ECM-associated signaling proteins is tightly regulated. Thus, the ECM serves as a reservoir for water and growth regulatory signals. The ECM architecture is dynamically modulated by multiple serine proteases that process both structural and signaling proteins to regulate physiological processes such as organogenesis and tissue homeostasis but they also contribute to pathological events, especially cancer progression. Here, we review the current literature regarding the role of ECM remodeling by serine proteases (KLKs, uPA, furin, HtrAs, granzymes, matriptase, hepsin) in tumorigenesis.
Collapse
Affiliation(s)
- Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland.
| | - Paulina Nastaly
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
3
|
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
|
4
|
Singh K, Oladipupo SS. An overview of CCN4 (WISP1) role in human diseases. J Transl Med 2024; 22:601. [PMID: 38937782 PMCID: PMC11212430 DOI: 10.1186/s12967-024-05364-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/01/2024] [Indexed: 06/29/2024] Open
Abstract
CCN4 (cellular communication network factor 4), a highly conserved, secreted cysteine-rich matricellular protein is emerging as a key player in the development and progression of numerous disease pathologies, including cancer, fibrosis, metabolic and inflammatory disorders. Over the past two decades, extensive research on CCN4 and its family members uncovered their diverse cellular mechanisms and biological functions, including but not limited to cell proliferation, migration, invasion, angiogenesis, wound healing, repair, and apoptosis. Recent studies have demonstrated that aberrant CCN4 expression and/or associated downstream signaling is key to a vast array of pathophysiological etiology, suggesting that CCN4 could be utilized not only as a non-invasive diagnostic or prognostic marker, but also as a promising therapeutic target. The cognate receptor of CCN4 remains elusive till date, which limits understanding of the mechanistic insights on CCN4 driven disease pathologies. However, as therapeutic agents directed against CCN4 begin to make their way into the clinic, that may start to change. Also, the pathophysiological significance of CCN4 remains underexplored, hence further research is needed to shed more light on its disease and/or tissue specific functions to better understand its clinical translational benefit. This review highlights the compelling evidence of overlapping and/or diverse functional and mechanisms regulated by CCN4, in addition to addressing the challenges, study limitations and knowledge gaps on CCN4 biology and its therapeutic potential.
Collapse
Affiliation(s)
- Kirti Singh
- Biotherapeutic Enabling Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46225, USA
| | - Sunday S Oladipupo
- Biotherapeutic Enabling Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46225, USA.
| |
Collapse
|
5
|
Morishima N, Kamada Y, Ota H, Iwagami Y, Takahashi H, Shimosaka M, Sakon D, Kondo J, Yamada M, Kumada T, Eguchi H, Miyoshi E. Serum levels of the N-terminal fragment of connective tissue growth factor is a novel biomarker for chronic pancreatitis. Pract Lab Med 2024; 40:e00402. [PMID: 38828385 PMCID: PMC11143898 DOI: 10.1016/j.plabm.2024.e00402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024] Open
Abstract
Chronic inflammation of the pancreas is considered to be one of the causes of pancreatic cancer. However, the diagnosis of chronic pancreatitis (CP) is very difficult in the pancreas, where biopsies are difficult to perform. The prevalence of CP is estimated to be many times more common than in patients with actual symptomatic CP. In recent years, abnormal cleavage of certain proteins has attracted attention as a biomarker for CP other than pancreatic enzymes. Connective tissue growth factor (CTGF) is one of the growth factors involved in tissue repair and other processes and is increased by stimulation of transforming growth factor-β, suggesting a relationship of CTGF with fibrosis. In this study, we measured the total length of CTGF in blood and N-terminal fragment CTGF in 48 cases of chronic pancreatitis, 64 cases of pancreatic cancer and 45 healthy volunteers (HV). Interestingly, we found that blood N-terminal fragment CTGF level was significantly increased in CP and pancreatic cancer patients. Multiple logistic regression analysis showed serum levels of N-terminal fragment CTGF, CRP and amylase were significant and independent variables for the differential diagnosis of CP from HV. Receiver operating characteristic analysis showed that area under the curve (AUC) value of serum N-terminal fragment CTGF level was 0.933, which can differentiate between CP and HV. Several factors would be involved in the increase in serum N-terminal fragment CTGF level. In conclusion, serum N-terminal fragment CTGF level is a promising new biomarker for CP.
Collapse
Affiliation(s)
- Naoki Morishima
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hiyori Ota
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshifumi Iwagami
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Munefumi Shimosaka
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Daisuke Sakon
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Jumpei Kondo
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | | | - Takashi Kumada
- Department of Nursing, Faculty of Nursing, Gifu Kyoritsu University, 5-50, Kitagata-cho, Ogaki, 503-8550, Gifu, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
6
|
Stuardo-Parada A, López-Muñoz R, Villarroel-Espindola F, Figueroa CD, Ehrenfeld P. Minireview: functional roles of tissue kallikrein, kinins, and kallikrein-related peptidases in lung cancer. Med Oncol 2023; 40:224. [PMID: 37405520 DOI: 10.1007/s12032-023-02090-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/17/2023] [Indexed: 07/06/2023]
Abstract
Despite campaigns and improvements in detection and treatment, lung cancer continues to increase worldwide and represents a major public health problem. One approach to treating patients suffering from lung cancer is to target surface receptors overexpressed on tumor cells, such as GPCR-family kinin receptors, and proteases that control tumor progression, such as kallikrein-related peptidases (KLKs). These proteases have been visualized in recent years due to their contribution to the progression of cancers, such as prostate and ovarian cancer, facilitating the invasive and metastatic capacity of tumor cells in these tissues. In fact, KLK3 is the specific prostate antigen, the only tissue-specific biomarker used to diagnose this malignancy. In lung cancer to date, evidence indicates that KLK5, KLK6, KLK8, KLK11, and KLK14 are the major peptidases regulated and involved in its progression. The expression levels of KLKs in this neoplasm are modulated by the secretome of the different cell types present in the tumor microenvironment, the cancer subtype and the tumor stage, among others. Considering the multiple functions of kinin receptors and KLKs, this review highlights their roles, even considering the SARS-CoV-2 effects. Since lung cancer is often diagnosed in advanced stages, our efforts should focus on early diagnosis, validating for example specific KLKs, especially in high-risk populations such as smokers and people exposed to carcinogenic fumes, oil fields, and contaminated workplaces, unexplored fields to investigate. Furthermore, their modulation could be considered as a promising approach in lung cancer therapeutics.
Collapse
Affiliation(s)
- Adriana Stuardo-Parada
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Rodrigo López-Muñoz
- Center for Interdisciplinary Studies on Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | | | - Carlos D Figueroa
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.
- Center for Interdisciplinary Studies on Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
7
|
Fibroblast Growth Factors and Cellular Communication Network Factors: Intimate Interplay by the Founding Members in Cartilage. Int J Mol Sci 2022; 23:ijms23158592. [PMID: 35955724 PMCID: PMC9369280 DOI: 10.3390/ijms23158592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
Fibroblast growth factors (FGFs) constitute a large family of signaling molecules that act in an autocrine/paracrine, endocrine, or intracrine manner, whereas the cellular communication network factors (CCN) family is composed of six members that manipulate extracellular signaling networks. FGFs and CCNs are structurally and functionally distinct, except for the common characteristics as matricellular proteins. Both play significant roles in the development of a variety of tissues and organs, including the skeletal system. In vertebrates, most of the skeletal parts are formed and grow through a process designated endochondral ossification, in which chondrocytes play the central role. The growth plate cartilage is the place where endochondral ossification occurs, and articular cartilage is left to support the locomotive function of joints. Several FGFs, including FGF-2, one of the founding members of this family, and all of the CCNs represented by CCN2, which is required for proper skeletal development, can be found therein. Research over a decade has revealed direct binding of CCN2 to FGFs and FGF receptors (FGFRs), which occasionally affect the biological outcome via FGF signaling. Moreover, a recent study uncovered an integrated regulation of FGF and CCN genes by FGF signaling. In this review, after a brief introduction of these two families, molecular and genetic interactions between CCN and FGF family members in cartilage, and their biological effects, are summarized. The molecular interplay represents the mutual involvement of the other in their molecular functions, leading to collaboration between CCN2 and FGFs during skeletal development.
Collapse
|
8
|
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
|
9
|
Zaykov V, Chaqour B. The CCN2/CTGF interactome: an approach to understanding the versatility of CCN2/CTGF molecular activities. J Cell Commun Signal 2021; 15:567-580. [PMID: 34613590 DOI: 10.1007/s12079-021-00650-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/23/2021] [Indexed: 01/16/2023] Open
Abstract
Cellular communication network 2 (CCN2), also known as connective tissue growth factor (CTGF) regulates diverse cellular processes, some at odds with others, including adhesion, proliferation, apoptosis, and extracellular matrix (ECM) protein synthesis. Although a cause-and-effect relationship between CCN2/CTGF expression and local fibrotic reactions has initially been established, CCN2/CTGF manifests cell-, tissue-, and context-specific functions and differentially affects developmental and pathological processes ranging from progenitor cell fate decisions and angiogenesis to inflammation and tumorigenesis. CCN2/CTGF multimodular structure, binding to and activation or inhibition of multiple cell surface receptors, growth factors and ECM proteins, and susceptibility for proteolytic cleavage highlight the complexity to CCN2/CTGF biochemical attributes. CCN2/CTGF expression and dosage in the local environment affects a defined community of its interacting partners, and this results in sequestration of growth factors, interference with or potentiation of ligand-receptor binding, cellular internalization of CCN2/CTGF, inhibition or activation of proteases, and generation of CCN2/CTGF degradome products that add molecular diversity and expand the repertoire of functional modules in the cells and their microenvironment. Through these interactions, different intracellular signals and cellular responses are elicited culminating into physiological or pathological reactions. Thus, the CCN2/CTGF interactome is a defining factor of its tissue- and context-specific effects. Mapping of new CCN2/CTGF binding partners might shed light on yet unknown roles of CCN2/CTGF and provide a solid basis for tissue-specific targeting this molecule or its interacting partners in a therapeutic context.
Collapse
Affiliation(s)
- Viktor Zaykov
- Department of Cell Biology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA
| | - Brahim Chaqour
- Department of Cell Biology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA. .,Department of Ophthalmology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA.
| |
Collapse
|
10
|
Sepúlveda V, Maurelia F, González M, Aguayo J, Caprile T. SCO-spondin, a giant matricellular protein that regulates cerebrospinal fluid activity. Fluids Barriers CNS 2021; 18:45. [PMID: 34600566 PMCID: PMC8487547 DOI: 10.1186/s12987-021-00277-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/11/2021] [Indexed: 12/28/2022] Open
Abstract
Cerebrospinal fluid is a clear fluid that occupies the ventricular and subarachnoid spaces within and around the brain and spinal cord. Cerebrospinal fluid is a dynamic signaling milieu that transports nutrients, waste materials and neuroactive substances that are crucial for the development, homeostasis and functionality of the central nervous system. The mechanisms that enable cerebrospinal fluid to simultaneously exert these homeostatic/dynamic functions are not fully understood. SCO-spondin is a large glycoprotein secreted since the early stages of development into the cerebrospinal fluid. Its domain architecture resembles a combination of a matricellular protein and the ligand-binding region of LDL receptor family. The matricellular proteins are a group of extracellular proteins with the capacity to interact with different molecules, such as growth factors, cytokines and cellular receptors; enabling the integration of information to modulate various physiological and pathological processes. In the same way, the LDL receptor family interacts with many ligands, including β-amyloid peptide and different growth factors. The domains similarity suggests that SCO-spondin is a matricellular protein enabled to bind, modulate, and transport different cerebrospinal fluid molecules. SCO-spondin can be found soluble or polymerized into a dynamic threadlike structure called the Reissner fiber, which extends from the diencephalon to the caudal tip of the spinal cord. Reissner fiber continuously moves caudally as new SCO-spondin molecules are added at the cephalic end and are disaggregated at the caudal end. This movement, like a conveyor belt, allows the transport of the bound molecules, thereby increasing their lifespan and action radius. The binding of SCO-spondin to some relevant molecules has already been reported; however, in this review we suggest more than 30 possible binding partners, including peptide β-amyloid and several growth factors. This new perspective characterizes SCO-spondin as a regulator of cerebrospinal fluid activity, explaining its high evolutionary conservation, its apparent multifunctionality, and the lethality or severe malformations, such as hydrocephalus and curved body axis, of knockout embryos. Understanding the regulation and identifying binding partners of SCO-spondin are crucial for better comprehension of cerebrospinal fluid physiology.
Collapse
Affiliation(s)
- Vania Sepúlveda
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Felipe Maurelia
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Maryori González
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Jaime Aguayo
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Teresa Caprile
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
11
|
Lenga Ma Bonda W, Lavergne M, Vasseur V, Brisson L, Roger S, Legras A, Guillon A, Guyétant S, Hiemstra PS, Si-Tahar M, Iochmann S, Reverdiau P. Kallikrein-related peptidase 5 contributes to the remodeling and repair of bronchial epithelium. FASEB J 2021; 35:e21838. [PMID: 34582061 DOI: 10.1096/fj.202002649r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 06/28/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022]
Abstract
Inflammation, oxidative stress, and protease/protease inhibitor imbalance with excessive production of proteases are factors associated with pathogenesis of the chronic obstructive pulmonary disease (COPD). In this study, we report that kallikrein-related peptidase 5 (KLK5) is a crucial protease involved in extracellular matrix (ECM) remodeling and bronchial epithelial repair after injury. First, we showed that KLK5 degrades the basal layer formed by culture of primary bronchial epithelial cells from COPD or non-COPD patients. Also, exogenous KLK5 acted differently on BEAS-2B cells already engaged in epithelial-to-mesenchymal transition (EMT) or on 16HBE 14o- cells harboring epithelial characteristics. Indeed, by inducing EMT, KLK5 reduced BEAS-2B cell adherence to the ECM. This effect, neutralized by tissue factor pathway inhibitor 2, a kunitz-type serine protease inhibitor, was due to a direct proteolytic activity of KLK5 on E-cadherin, β-catenin, fibronectin, and α5β1 integrin. Thus, KLK5 may strengthen EMT mechanisms and promote the migration of cells by activating the mitogen-activated protein kinase signaling pathway required for this function. In contrast, knockdown of endogenous KLK5 in 16HBE14o- cells, accelerated wound healing repair after injury, and exogenous KLK5 addition delayed the closure repair. These data suggest that among proteases, KLK5 could play a critical role in airway remodeling events associated with COPD during exposure of the pulmonary epithelium to inhaled irritants or smoking and the inflammation process.
Collapse
Affiliation(s)
- Woodys Lenga Ma Bonda
- Université de Tours, Tours, France.,Centre d'Etude des Pathologies Respiratoires (CEPR), INSERM, UMR 1100, Tours, France
| | - Marion Lavergne
- Université de Tours, Tours, France.,Centre d'Etude des Pathologies Respiratoires (CEPR), INSERM, UMR 1100, Tours, France
| | - Virginie Vasseur
- Université de Tours, Tours, France.,Centre d'Etude des Pathologies Respiratoires (CEPR), INSERM, UMR 1100, Tours, France
| | - Lucie Brisson
- Université de Tours, Tours, France.,Nutrition, Croissance et Cancer (N2C), INSERM, UMR 1069, Tours, France
| | - Sébastien Roger
- Université de Tours, Tours, France.,EA 4245 "Transplantation, Immunologie, Inflammation", Tours, France.,Institut Universitaire de France, Paris, France
| | - Antoine Legras
- Université de Tours, Tours, France.,Département de chirurgie thoracique, CHRU de Tours, Tours, France
| | - Antoine Guillon
- Université de Tours, Tours, France.,Centre d'Etude des Pathologies Respiratoires (CEPR), INSERM, UMR 1100, Tours, France.,Service de médecine intensive et réanimation, CHRU de Tours, Tours, France
| | - Serge Guyétant
- Université de Tours, Tours, France.,Département d'anatomie et cytologie pathologiques, CHRU de Tours, Tours, France
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mustapha Si-Tahar
- Université de Tours, Tours, France.,Centre d'Etude des Pathologies Respiratoires (CEPR), INSERM, UMR 1100, Tours, France
| | - Sophie Iochmann
- Université de Tours, Tours, France.,Centre d'Etude des Pathologies Respiratoires (CEPR), INSERM, UMR 1100, Tours, France.,Institut Universitaire de Technologie, Tours, France
| | - Pascale Reverdiau
- Université de Tours, Tours, France.,Centre d'Etude des Pathologies Respiratoires (CEPR), INSERM, UMR 1100, Tours, France.,Institut Universitaire de Technologie, Tours, France
| |
Collapse
|
12
|
Lavergne M, Guillon-Munos A, Lenga Ma Bonda W, Attucci S, Kryza T, Barascu A, Moreau T, Petit-Courty A, Sizaret D, Courty Y, Iochmann S, Reverdiau P. Tissue factor pathway inhibitor 2 is a potent kallikrein-related protease 12 inhibitor. Biol Chem 2021; 402:1257-1268. [PMID: 33977679 DOI: 10.1515/hsz-2020-0389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/30/2021] [Indexed: 11/15/2022]
Abstract
The protease activities are tightly regulated by inhibitors and dysregulation contribute to pathological processes such as cancer and inflammatory disorders. Tissue factor pathway inhibitor 2 (TFPI-2) is a serine proteases inhibitor, that mainly inhibits plasmin. This protease activated matrix metalloproteases (MMPs) and degraded extracellular matrix. Other serine proteases are implicated in these mechanisms like kallikreins (KLKs). In this study, we identified for the first time that TFPI-2 is a potent inhibitor of KLK5 and 12. Computer modeling showed that the first Kunitz domain of TFPI-2 could interact with residues of KLK12 near the catalytic triad. Furthermore, like plasmin, KLK12 was able to activate proMMP-1 and -3, with no effect on proMMP-9. Thus, the inhibition of KLK12 by TFPI-2 greatly reduced the cascade activation of these MMPs and the cleavage of cysteine-rich 61, a matrix signaling protein. Moreover, when TFPI-2 bound to extracellular matrix, its classical localisation, the KLK12 inhibition was retained. Finally, TFPI-2 was downregulated in human non-small-cell lung tumour tissue as compared with non-affected lung tissue. These data suggest that TFPI-2 is a potent inhibitor of KLK12 and could regulate matrix remodeling and cancer progression mediated by KLK12.
Collapse
Affiliation(s)
- Marion Lavergne
- Université de Tours, F-37032 Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
| | - Audrey Guillon-Munos
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
- Groupe IMT, Bio3 Institute, 15 rue du Plat D'Etain, F-37020 Tours Cedex 1, France
| | - Woodys Lenga Ma Bonda
- Université de Tours, F-37032 Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
| | - Sylvie Attucci
- Université de Tours, F-37032 Tours, France
- INSERM, Imagerie et Cerveau (iBrain), UMR 1253, F-37000 Tours, France
| | - Thomas Kryza
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
- Mater Research Institute, The University of Queensland, Woollongabba Brisbane, QLD, Australia
| | - Aurélia Barascu
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
- UMR 8226-CNRS/UPMC, Institut de Biologie Physico-chimique, Sorbonne Université, 13 rue Pierre et Marie Curie, F-75005 Paris, France
| | - Thierry Moreau
- Université de Tours, F-37032 Tours, France
- INRA, UMR INRA 0083 - Biologie des Oiseaux et Aviculture (BOA), F-37380 Nouzilly, France
| | - Agnès Petit-Courty
- Université de Tours, F-37032 Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
| | - Damien Sizaret
- Département d'Anatomie et Cytologie Pathologiques, Hôpital Trousseau, CHRU de Tours, F-37044 Tours, France
| | - Yves Courty
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
| | - Sophie Iochmann
- Université de Tours, F-37032 Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
- Institut Universitaire de Technologie, F-37082 Tours, France
| | - Pascale Reverdiau
- Université de Tours, F-37032 Tours, France
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, 10 boulevard Tonnellé, F-37032 Tours, France
- Institut Universitaire de Technologie, F-37082 Tours, France
| |
Collapse
|
13
|
Quesnel A, Karagiannis GS, Filippou PS. Extracellular proteolysis in glioblastoma progression and therapeutics. Biochim Biophys Acta Rev Cancer 2020; 1874:188428. [PMID: 32956761 DOI: 10.1016/j.bbcan.2020.188428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022]
Abstract
Gliomas encompass highly invasive primary central nervous system (CNS) tumours of glial cell origin with an often-poor clinical prognosis. Of all gliomas, glioblastoma is the most aggressive form of primary brain cancer. Current treatments in glioblastoma are insufficient due to the invasive nature of brain tumour cells, which typically results in local tumour recurrence following treatment. The latter represents the most important cause of mortality in glioblastoma and underscores the necessity for an in-depth understanding of the underlying mechanisms. Interestingly, increased synthesis and secretion of several proteolytic enzymes within the tumour microenvironment, such as matrix metalloproteinases, lysosomal proteases, cathepsins and kallikreins for extracellular-matrix component degradation may play a major role in the aforementioned glioblastoma invasion mechanisms. These proteolytic networks are key players in establishing and maintaining a tumour microenvironment that promotes tumour cell survival, proliferation, and migration. Indeed, the targeted inhibition of these proteolytic enzymes has been a promisingly useful therapeutic strategy for glioblastoma management in both preclinical and clinical development. We hereby summarize current advances on the biology of the glioblastoma tumour microenvironment, with a particular emphasis on the role of proteolytic enzyme families in glioblastoma invasion and progression, as well as on their subsequent prognostic value as biomarkers and their therapeutic targeting in the era of precision medicine.
Collapse
Affiliation(s)
- Agathe Quesnel
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, 38 John Dixon Ln, Darlington, DL1 1HG, United Kingdom
| | - George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA; Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, New York, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Panagiota S Filippou
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, 38 John Dixon Ln, Darlington, DL1 1HG, United Kingdom.
| |
Collapse
|
14
|
Andreae EA, Warejcka DJ, Twining SS. Thrombin alters the synthesis and processing of CYR61/CCN1 in human corneal stromal fibroblasts and myofibroblasts through multiple distinct mechanisms. Mol Vis 2020; 26:540-562. [PMID: 32818017 PMCID: PMC7406864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 07/27/2020] [Indexed: 11/08/2022] Open
Abstract
Purpose Previous research in our laboratory indicated that prothrombin and other coagulation enzymes required to activate prothrombin to thrombin are synthesized by the cornea and that apoptotic human corneal stromal cells can provide a surface for prothrombin activation through the intrinsic and extrinsic coagulation pathways. The purpose of the work reported here is to study the role of thrombin activity in the regulation of matricellular protein Cyr61 (CCN1) produced by wounded phenotype human corneal stromal fibroblasts and myofibroblasts. Methods Stromal cells from human donor corneas were converted to defined wounded phenotype fibroblasts and myofibroblasts with fetal bovine serum, followed by basic fibroblast growth factor (bFGF) and transforming growth factor beta-1 (TGFβ-1), respectively, and stimulated with varying concentrations (0-10.0 units (U)/ml) of thrombin from 1-7 h. Cyr61 transcript levels were determined using reverse transcriptase-PCR (RT-PCR) and quantitative PCR (qPCR) while protein forms were analyzed using western blot data. Protease activities were characterized via protease class-specific inhibitors and western blot analysis. Thrombin activity was quantified using the fluorogenic peptide Phe-Pro-Arg-AFC. Protease-activated receptor (PAR) agonist peptides-1 and -4 were used to determine whether cells increased Cyr61 through PAR signaling pathways. The PAR-1 antagonist SCH 79797 was used to block the thrombin cleavage of the receptor. PCR data were analyzed using MxPro software and western blot data were analyzed using Image Lab™ and Image J software. Student t test and one- and two-way ANOVA (with or without ranking, depending on sample distribution), together with Dunnett's test or Tukey comparison tests for post-hoc analysis, were used to determine statistical significance. Results: Full-length Cyr61 is expressed by human corneal stromal fibroblasts and myofibroblasts and is significantly upregulated by active thrombin stimulation at the message (p<0.03) and protein (p<0.03) levels for fibroblasts and myofibroblasts. Inhibition by the allosteric thrombin-specific inhibitor hirudin prevented the thrombin-associated increase in the Cyr61 protein expression, indicating that the proteolytic activity of thrombin is required for the increase of the Cyr61 protein level. PAR-1 agonist stimulation of fibroblasts and myofibroblasts significantly increased cell-associated Cyr61 protein levels (p<0.04), and PAR-1 antagonist SCH 79797 significantly inhibited the thrombin stimulated increase of Cyr61 in fibroblasts but not in myofibroblasts. In the fibroblast and myofibroblast conditioned media, Cyr61 was detected as the full-length 40 kDa protein in the absence of thrombin, and mainly at 24 kDa in the presence of thrombin at ≥0.5 U/ml, using an antibody directed toward the internal linker region between the von Willebrand factor type C and thrombospondin type-1 domains. Although known to undergo alternative splicing, Cyr61 that is synthesized by corneal fibroblasts and myofibroblasts is not alternatively spliced in response to thrombin stimulation nor is Cyr61 directly cleaved by thrombin to generate its 24 kDa form; instead, Cyr61 is proteolytically processed into 24 kDa N- and 16 kDa C-terminal fragments by a thrombin activated leupeptin-sensitive protease present in conditioned media with activity distinct from the proteolytic activity of thrombin. Conclusions In cultured human corneal stromal fibroblasts and myofibroblasts, thrombin regulates Cyr61 through two mechanisms: 1) thrombin increases the Cyr61 expression at the message and protein levels, and 2) thrombin increases the activation of a leupeptin-sensitive protease that stimulates the cleavage of Cyr61 into N- and C-terminal domain populations in or near the thrombospondin type-1 domain. Generation of Cyr61 peptides during corneal injury stimulation may reveal additional functions of the protein, which modulate corneal wound healing activities or decrease activities of the full-length Cyr61 form.
Collapse
Affiliation(s)
- Emily A Andreae
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
- Marshfield Clinic Research Institute, Marshfield, WI
| | - Debra J Warejcka
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - Sally S Twining
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
- Department of Ophthalmology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
15
|
Eyeing the Extracellular Matrix in Vascular Development and Microvascular Diseases and Bridging the Divide between Vascular Mechanics and Function. Int J Mol Sci 2020; 21:ijms21103487. [PMID: 32429045 PMCID: PMC7278940 DOI: 10.3390/ijms21103487] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
The extracellular matrix (ECM) is critical in all aspects of vascular development and health: supporting cell anchorage, providing structure, organization and mechanical stability, and serving as a sink for growth factors and sustained survival signals. Abnormal changes in ECM protein expression, organization, and/or properties, and the ensuing changes in vascular compliance affect vasodilator responses, microvascular pressure transmission, and collateral perfusion. The changes in microvascular compliance are independent factors initiating, driving, and/or exacerbating a plethora of microvascular diseases of the eye including diabetic retinopathy (DR) and vitreoretinopathy, retinopathy of prematurity (ROP), wet age-related macular degeneration (AMD), and neovascular glaucoma. Congruently, one of the major challenges with most vascular regenerative therapies utilizing localized growth factor, endothelial progenitor, or genetically engineered cell delivery, is the regeneration of blood vessels with physiological compliance properties. Interestingly, vascular cells sense physical forces, including the stiffness of their ECM, through mechanosensitive integrins, their associated proteins and the actomyosin cytoskeleton, which generates biochemical signals that culminate in a rapid expression of matricellular proteins such as cellular communication network 1 (CCN1) and CCN2 (aka connective tissue growth factor or CTGF). Loss or gain of function of these proteins alters genetic programs of cell growth, ECM biosynthesis, and intercellular signaling, that culminate in changes in cell behavior, polarization, and barrier function. In particular, the function of the matricellular protein CCN2/CTGF is critical during retinal vessel development and regeneration wherein new blood vessels form and invest a preformed avascular neural retina following putative gradients of matrix stiffness. These observations underscore the need for further in-depth characterization of the ECM-derived cues that dictate structural and functional properties of the microvasculature, along with the development of new therapeutic strategies addressing the ECM-dependent regulation of pathophysiological stiffening of blood vessels in ischemic retinopathies.
Collapse
|
16
|
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
|
17
|
Zhu N, Pauciulo MW, Welch CL, Lutz KA, Coleman AW, Gonzaga-Jauregui C, Wang J, Grimes JM, Martin LJ, He H, Shen Y, Chung WK, Nichols WC. Novel risk genes and mechanisms implicated by exome sequencing of 2572 individuals with pulmonary arterial hypertension. Genome Med 2019; 11:69. [PMID: 31727138 PMCID: PMC6857288 DOI: 10.1186/s13073-019-0685-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/06/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Group 1 pulmonary arterial hypertension (PAH) is a rare disease with high mortality despite recent therapeutic advances. Pathogenic remodeling of pulmonary arterioles leads to increased pulmonary pressures, right ventricular hypertrophy, and heart failure. Mutations in bone morphogenetic protein receptor type 2 and other risk genes predispose to disease, but the vast majority of non-familial cases remain genetically undefined. METHODS To identify new risk genes, we performed exome sequencing in a large cohort from the National Biological Sample and Data Repository for PAH (PAH Biobank, n = 2572). We then carried out rare deleterious variant identification followed by case-control gene-based association analyses. To control for population structure, only unrelated European cases (n = 1832) and controls (n = 12,771) were used in association tests. Empirical p values were determined by permutation analyses, and the threshold for significance defined by Bonferroni's correction for multiple testing. RESULTS Tissue kallikrein 1 (KLK1) and gamma glutamyl carboxylase (GGCX) were identified as new candidate risk genes for idiopathic PAH (IPAH) with genome-wide significance. We note that variant carriers had later mean age of onset and relatively moderate disease phenotypes compared to bone morphogenetic receptor type 2 variant carriers. We also confirmed the genome-wide association of recently reported growth differentiation factor (GDF2) with IPAH and further implicate T-box 4 (TBX4) with child-onset PAH. CONCLUSIONS We report robust association of novel genes KLK1 and GGCX with IPAH, accounting for ~ 0.4% and 0.9% of PAH Biobank cases, respectively. Both genes play important roles in vascular hemodynamics and inflammation but have not been implicated in PAH previously. These data suggest new genes, pathogenic mechanisms, and therapeutic targets for this lethal vasculopathy.
Collapse
Affiliation(s)
- Na Zhu
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Carrie L Welch
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Katie A Lutz
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
| | - Anna W Coleman
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
| | | | - Jiayao Wang
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Joseph M Grimes
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Lisa J Martin
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Hua He
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 7016, Cincinnati, OH, USA.
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
18
|
Chaqour B. Caught between a "Rho" and a hard place: are CCN1/CYR61 and CCN2/CTGF the arbiters of microvascular stiffness? J Cell Commun Signal 2019; 14:21-29. [PMID: 31376071 DOI: 10.1007/s12079-019-00529-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 07/26/2019] [Indexed: 12/18/2022] Open
Abstract
The extracellular matrix (ECM) is a deformable dynamic structure that dictates the behavior, function and integrity of blood vessels. The composition, density, chemistry and architecture of major globular and fibrillar proteins of the matrisome regulate the mechanical properties of the vasculature (i.e., stiffness/compliance). ECM proteins are linked via integrins to a protein adhesome directly connected to the actin cytoskeleton and various downstream signaling pathways that enable the cells to respond to external stimuli in a coordinated manner and maintain optimal tissue stiffness. However, cardiovascular risk factors such as diabetes, dyslipidemia, hypertension, ischemia and aging compromise the mechanical balance of the vascular wall. Stiffening of large blood vessels is associated with well-known qualitative and quantitative changes of fibrillar and fibrous macromolecules of the vascular matrisome. However, the mechanical properties of the thin-walled microvasculature are essentially defined by components of the subendothelial matrix. Cellular communication network (CCN) 1 and 2 proteins (aka Cyr61 and CTGF, respectively) of the CCN protein family localize in and act on the pericellular matrix of microvessels and constitute primary candidate markers and regulators of microvascular compliance. CCN1 and CCN2 bind various integrin and non-integrin receptors and initiate signaling pathways that regulate connective tissue remodeling and response to injury, the associated mechanoresponse of vascular cells, and the subsequent inflammatory response. The CCN1 and CCN2 genes are themselves responsive to mechanical stimuli in vascular cells, wherein mechanotransduction signaling converges into the common Rho GTPase pathway, which promotes actomyosin-based contractility and cellular stiffening. However, CCN1 and CCN2 each exhibit unique functional attributes in these processes. A better understanding of their synergistic or antagonistic effects on the maintenance (or loss) of microvascular compliance in physiological and pathological situations will assist more broadly based studies of their functional properties and translational value.
Collapse
Affiliation(s)
- Brahim Chaqour
- Department of Cell Biology and Department of Ophthalmology, State University of New York - SUNY Downstate Medical Center, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA.
| |
Collapse
|
19
|
Figueroa CD, Molina L, Bhoola KD, Ehrenfeld P. Overview of tissue kallikrein and kallikrein-related peptidases in breast cancer. Biol Chem 2019; 399:937-957. [PMID: 29885274 DOI: 10.1515/hsz-2018-0111] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/17/2018] [Indexed: 12/11/2022]
Abstract
The kallikrein family comprises tissue kallikrein and 14 kallikrein-related peptidases (KLKs) recognized as a subgroup of secreted trypsin- or chymotrypsin-like serine proteases. KLKs are expressed in many cellular types where they regulate important physiological activities such as semen liquefaction, immune response, neural development, blood pressure, skin desquamation and tooth enamel formation. Tissue kallikrein, the oldest member and kinin-releasing enzyme, and KLK3/PSA, a tumor biomarker for prostate cancer are the most prominent components of the family. Additionally, other KLKs have shown an abnormal expression in neoplasia, particularly in breast cancer. Thus, increased levels of some KLKs may increase extracellular matrix degradation, invasion and metastasis; other KLKs modulate cell growth, survival and angiogenesis. On the contrary, KLKs can also inhibit angiogenesis and produce tumor suppression. However, there is a lack of knowledge on how KLKs are regulated in tumor microenvironment by molecules present at the site, namely cytokines, inflammatory mediators and growth factors. Little is known about the signaling pathways that control expression/secretion of KLKs in breast cancer, and further how activation of PAR receptors may contribute to functional activity in neoplasia. A better understanding of these molecular events will allow us to consider KLKs as relevant therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Carlos D Figueroa
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Luis Molina
- Department of Science, Universidad San Sebastián, sede De la Patagonia, Puerto Montt, Chile
| | - Kanti D Bhoola
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Centro de Investigaciones del Sistema Nervioso (CISNe), Valdivia, Chile, e-mail:
| |
Collapse
|
20
|
Lenga Ma Bonda W, Iochmann S, Magnen M, Courty Y, Reverdiau P. Kallikrein-related peptidases in lung diseases. Biol Chem 2019; 399:959-971. [PMID: 29604204 DOI: 10.1515/hsz-2018-0114] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/26/2018] [Indexed: 02/06/2023]
Abstract
Human tissue kallikreins (KLKs) are 15 members of the serine protease family and are present in various healthy human tissues including airway tissues. Multiple studies have revealed their crucial role in the pathophysiology of a number of chronic, infectious and tumour lung diseases. KLK1, 3 and 14 are involved in asthma pathogenesis, and KLK1 could be also associated with the exacerbation of this inflammatory disease caused by rhinovirus. KLK5 was demonstrated as an influenza virus activating protease in humans, and KLK1 and 12 could also be involved in the activation and spread of these viruses. KLKs are associated with lung cancer, with up- or downregulation of expression depending on the KLK, cancer subtype, stage of tumour and also the microenvironment. Functional studies showed that KLK12 is a potent pro-angiogenic factor. Moreover, KLK6 promotes malignant-cell proliferation and KLK13 invasiveness. In contrast, KLK8 and KLK10 reduce proliferation and invasion of malignant cells. Considering the involvement of KLKs in various physiological and pathological processes, KLKs appear to be potential biomarkers and therapeutic targets for lung diseases.
Collapse
Affiliation(s)
- Woodys Lenga Ma Bonda
- Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Faculté de Médecine, 10 Boulevard Tonnellé, F-37032 Tours, France.,Université de Tours, F-37032 Tours, France
| | - Sophie Iochmann
- Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Faculté de Médecine, 10 Boulevard Tonnellé, F-37032 Tours, France.,Université de Tours, F-37032 Tours, France.,IUT de Tours, Université de Tours, F-37082 Tours, France
| | - Mélia Magnen
- Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Faculté de Médecine, 10 Boulevard Tonnellé, F-37032 Tours, France.,Université de Tours, F-37032 Tours, France
| | - Yves Courty
- Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Faculté de Médecine, 10 Boulevard Tonnellé, F-37032 Tours, France.,Université de Tours, F-37032 Tours, France
| | - Pascale Reverdiau
- Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Faculté de Médecine, 10 Boulevard Tonnellé, F-37032 Tours, France.,Université de Tours, F-37032 Tours, France.,IUT de Tours, Université de Tours, F-37082 Tours, France
| |
Collapse
|
21
|
Sabir N, Hussain T, Liao Y, Wang J, Song Y, Shahid M, Cheng G, Mangi MH, Yao J, Yang L, Zhao D, Zhou X. Kallikrein 12 Regulates Innate Resistance of Murine Macrophages against Mycobacterium bovis Infection by Modulating Autophagy and Apoptosis. Cells 2019; 8:cells8050415. [PMID: 31060300 PMCID: PMC6562459 DOI: 10.3390/cells8050415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 12/27/2022] Open
Abstract
Mycobacterium bovis (M. bovis) is a member of the Mycobacterium tuberculosis (Mtb) complex causing bovine tuberculosis (TB) and imposing a high zoonotic threat to human health. Kallikreins (KLKs) belong to a subgroup of secreted serine proteases. As their role is established in various physiological and pathological processes, it is likely that KLKs expression may mediate a host immune response against the M. bovis infection. In the current study, we report in vivo and in vitro upregulation of KLK12 in the M. bovis infection. To define the role of KLK12 in immune response regulation of murine macrophages, we produced KLK12 knockdown bone marrow derived macrophages (BMDMs) by using siRNA transfection. Interestingly, the knockdown of KLK12 resulted in a significant downregulation of autophagy and apoptosis in M. bovis infected BMDMs. Furthermore, we demonstrated that this KLK12 mediated regulation of autophagy and apoptosis involves mTOR/AMPK/TSC2 and BAX/Bcl-2/Cytochrome c/Caspase 3 pathways, respectively. Similarly, inflammatory cytokines IL-1β, IL-6, IL-12 and TNF-α were significantly downregulated in KLK12 knockdown macrophages but the difference in IL-10 and IFN-β expression was non-significant. Taken together, these findings suggest that upregulation of KLK12 in M. bovis infected murine macrophages plays a substantial role in the protective immune response regulation by modulating autophagy, apoptosis and pro-inflammatory pathways. To our knowledge, this is the first report on expression and the role of KLK12 in the M. bovis infection and the data may contribute to a new paradigm for diagnosis and treatment of bovine TB.
Collapse
Affiliation(s)
- Naveed Sabir
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Tariq Hussain
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Yi Liao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Jie Wang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Yinjuan Song
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Muhammad Shahid
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Guangyu Cheng
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Mazhar Hussain Mangi
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Jiao Yao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Lifeng Yang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Deming Zhao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Xiangmei Zhou
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
22
|
Kaasbøll OJ, Gadicherla AK, Wang JH, Monsen VT, Hagelin EMV, Dong MQ, Attramadal H. Connective tissue growth factor (CCN2) is a matricellular preproprotein controlled by proteolytic activation. J Biol Chem 2018; 293:17953-17970. [PMID: 30262666 DOI: 10.1074/jbc.ra118.004559] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/23/2018] [Indexed: 11/06/2022] Open
Abstract
Connective tissue growth factor (CTGF; now often referred to as CCN2) is a secreted protein predominantly expressed during development, in various pathological conditions that involve enhanced fibrogenesis and tissue fibrosis, and in several cancers and is currently an emerging target in several early-phase clinical trials. Tissues containing high CCN2 activities often display smaller degradation products of full-length CCN2 (FL-CCN2). Interpretation of these observations is complicated by the fact that a uniform protein structure that defines biologically active CCN2 has not yet been resolved. Here, using DG44 CHO cells engineered to produce and secrete FL-CCN2 and cell signaling and cell physiological activity assays, we demonstrate that FL-CCN2 is itself an inactive precursor and that a proteolytic fragment comprising domains III (thrombospondin type 1 repeat) and IV (cystine knot) appears to convey all biologically relevant activities of CCN2. In congruence with these findings, purified FL-CCN2 could be cleaved and activated following incubation with matrix metalloproteinase activities. Furthermore, the C-terminal fragment of CCN2 (domains III and IV) also formed homodimers that were ∼20-fold more potent than the monomeric form in activating intracellular phosphokinase cascades. The homodimer elicited activation of fibroblast migration, stimulated assembly of focal adhesion complexes, enhanced RANKL-induced osteoclast differentiation of RAW264.7 cells, and promoted mammosphere formation of MCF-7 mammary cancer cells. In conclusion, CCN2 is synthesized and secreted as a preproprotein that is autoinhibited by its two N-terminal domains and requires proteolytic processing and homodimerization to become fully biologically active.
Collapse
Affiliation(s)
- Ole Jørgen Kaasbøll
- From the Institute for Surgical Research, Oslo University Hospital and University of Oslo, NO-0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, NO-0316 Oslo, Norway
| | - Ashish K Gadicherla
- From the Institute for Surgical Research, Oslo University Hospital and University of Oslo, NO-0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, NO-0316 Oslo, Norway
| | - Jian-Hua Wang
- National Institute of Biological Sciences, 102206 Beijing, China
| | - Vivi Talstad Monsen
- From the Institute for Surgical Research, Oslo University Hospital and University of Oslo, NO-0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, NO-0316 Oslo, Norway
| | - Else Marie Valbjørn Hagelin
- From the Institute for Surgical Research, Oslo University Hospital and University of Oslo, NO-0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, NO-0316 Oslo, Norway
| | - Meng-Qiu Dong
- National Institute of Biological Sciences, 102206 Beijing, China
| | - Håvard Attramadal
- From the Institute for Surgical Research, Oslo University Hospital and University of Oslo, NO-0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, NO-0316 Oslo, Norway.
| |
Collapse
|
23
|
Papachristopoulou G, Tsapralis N, Michaelidou K, Ardavanis-Loukeris G, Griniatsos I, Scorilas A, Talieri M. Human kallikrein-related peptidase 12 (KLK12) splice variants discriminate benign from cancerous breast tumors. Clin Biochem 2018; 58:78-85. [PMID: 29807016 DOI: 10.1016/j.clinbiochem.2018.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/15/2018] [Accepted: 05/24/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVES As kallikrein-related peptidase 12 (KLK12) has been implicated in the cancer progression and alternative splicing plays significant role in this disease, the aim of this study was to examine the expression profile and the clinical impact of the KLK12 splice variants in breast cancer. DESIGN AND METHODS Total RNA was isolated and reverse transcripted from 141 tissues. Afterwards, quantitative real-time PCR were conducted, followed by the performance of the comparative CT (2-ΔΔCT) method for relative quantification, whilst their correlation with the clinicopathological features of breast malignancies were assessed by statistical analysis. RESULTS Both KLK12sv1/2 and KLK12sv3 showed higher expression in non-cancerous than in cancerous samples. KLKsv1/2 (P = 0.001) upregulated and KLK12sv3 (P < 0.001) downregulated in the malignant compared to the benign tumors and their discriminative ability was verified by ROC curve analysis. Moreover, KLK12sv3 was associated with grade (P = 0.012) and hormonal receptor status (P = 0.001). Furthermore, Kaplan-Meier and Cox regression analyses showed that patients with positive KLK12sv1/2 and KLK12sv3 levels presented a significantly longer disease-free survival (P = 0.014 and P = 0.013, respectively) and overall survival (P = 0.062 and P = 0.004, respectively). CONCLUSIONS Our results demonstrate the discriminative value of KLK12sv1/2 and KLK12sv3 between benign and malignant breast tumors as well as their potential favorable prognostic significance in breast adenocarcinoma.
Collapse
Affiliation(s)
- Georgia Papachristopoulou
- Department of Pathology, "Saint Savvas" Cancer Hospital of Athens, Athens GR-11522, Greece; Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens GR-15701, Greece
| | - Nikolaos Tsapralis
- Department of Breast Cancer Surgery, "Saint Savvas" Cancer Hospital of Athens, Athens GR-11522, Greece
| | - Kleita Michaelidou
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens GR-15701, Greece
| | - Gerasimos Ardavanis-Loukeris
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens GR-15701, Greece
| | - Ioannis Griniatsos
- First Department of Surgery, National and Kapodistrian University of Athens, "Laiko" Hospital, Athens GR-11527, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens GR-15701, Greece
| | - Maroulio Talieri
- Department of Cellular Physiology, G. Papanicolaou Research Center of Oncology, "Saint Savvas" Cancer Hospital of Athens, Athens GR-11522, Greece.
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Otani Y, Ishida J, Kurozumi K, Oka T, Shimizu T, Tomita Y, Hattori Y, Uneda A, Matsumoto Y, Michiue H, Tomida S, Matsubara T, Ichikawa T, Date I. PIK3R1Met326Ile germline mutation correlates with cysteine-rich protein 61 expression and poor prognosis in glioblastoma. Sci Rep 2017; 7:7391. [PMID: 28785028 PMCID: PMC5547066 DOI: 10.1038/s41598-017-07745-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 06/30/2017] [Indexed: 01/12/2023] Open
Abstract
Despite therapeutic advances, glioblastoma represents a lethal brain tumor. Recently, research to identify prognostic markers for glioblastoma has intensified. Our previous study demonstrated that median progression-free survival (PFS) and overall survival (OS) of patients with high cysteine-rich protein 61 (CCN1) expression was significantly shorter than that of patients with low CCN1 expression. To understand the molecular mechanisms that regulate CCN1 expression, we examined 147 tumour samples from 80 patients with glioblastoma and 67 patients with lower grade glioma. Next-generation and Sanger sequencing showed that PIK3R1Met326Ile was more frequent in the CCN1 high expression group (10/37 cases, 27.0%) than the CCN1 low expression group (3/38 cases, 7.9%) in glioblastoma. This mutation was also detected in corresponding blood samples. In multivariate analysis, high CCN1 expression and PIK3R1Met326Ile in glioblastoma patients were prognostic factors for OS [HR = 2.488 (1.298–4.769), p = 0.006] and [HR = 2.089 (1.020–4.277), p = 0.0439], respectively. Thus, the PIK3R1Met326Ile germline appears to be correlated with CCN1 expression and poor prognosis in glioblastoma.
Collapse
Affiliation(s)
- Yoshihiro Otani
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Joji Ishida
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiko Kurozumi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.
| | - Tetsuo Oka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Toshihiko Shimizu
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yusuke Tomita
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yasuhiko Hattori
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Atsuhito Uneda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yuji Matsumoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroyuki Michiue
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Shuta Tomida
- Okayama University Hospital Biobank, Okayama University Hospital, Okayama, Japan.,Department of Biobank, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takehiro Matsubara
- Okayama University Hospital Biobank, Okayama University Hospital, Okayama, Japan
| | - Tomotsugu Ichikawa
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
27
|
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
|
28
|
Thakur R, Mishra DP. Matrix reloaded: CCN, tenascin and SIBLING group of matricellular proteins in orchestrating cancer hallmark capabilities. Pharmacol Ther 2016; 168:61-74. [DOI: 10.1016/j.pharmthera.2016.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
29
|
Kallikrein in the Interstitial Space. Protein Sci 2016. [DOI: 10.1201/9781315374307-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
30
|
Butler GS, Connor AR, Sounni NE, Eckhard U, Morrison CJ, Noël A, Overall CM. Degradomic and yeast 2-hybrid inactive catalytic domain substrate trapping identifies new membrane-type 1 matrix metalloproteinase (MMP14) substrates: CCN3 (Nov) and CCN5 (WISP2). Matrix Biol 2016; 59:23-38. [PMID: 27471094 DOI: 10.1016/j.matbio.2016.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/19/2016] [Accepted: 07/19/2016] [Indexed: 12/20/2022]
Abstract
Members of the CCN family of matricellular proteins are cytokines linking cells to the extracellular matrix. We report that CCN3 (Nov) and CCN5 (WISP2) are novel substrates of MMP14 (membrane-type 1-matrix metalloproteinase, MT1-MMP) that we identified using MMP14 "inactive catalytic domain capture" (ICDC) as a yeast two-hybrid protease substrate trapping platform in parallel with degradomics mass spectrometry screens for MMP14 substrates. CCN3 and CCN5, previously unknown substrates of MMPs, were biochemically validated as substrates of MMP14 and other MMPs in vitro-CCN5 was processed in the variable region by MMP14 and MMP2, as well as by MMP1, 3, 7, 8, 9 and 15. CCN1, 2 and 3 are proangiogenic factors yet we found novel opposing activity of CCN5 that was potently antiangiogenic in an aortic ring vessel outgrowth model. MMP14, a known regulator of angiogenesis, cleaved CCN5 and abrogated the angiostatic activity. CCN3 was also processed in the variable region by MMP14 and MMP2, and by MMP1, 8 and 9. In addition to the previously reported cleavages of CCN1 and CCN2 by several MMPs we found that MMPs 8, 9, and 1 process CCN1, and MMP8 and MMP9 also process CCN2. Thus, our study reveals additional and pervasive family-wide processing of CCN matricellular proteins/cytokines by MMPs. Furthermore, CCN5 cleavage by proangiogenic MMPs results in removal of an angiogenic brake held by CCN5. This highlights the importance of thorough dissection of MMP substrates that is needed to reveal higher-level control mechanisms beyond type IV collagen and other extracellular matrix protein remodelling in angiogenesis. SUMMARY We find CCN family member cleavage by MMPs is more pervasive than previously reported and includes CCN3 (Nov) and CCN5 (WISP2). CCN5 is a novel antiangiogenic factor, whose function is abrogated by proangiogenic MMP cleavage. By processing CCN proteins, MMPs regulate cell responses angiogenesis in connective tissues.
Collapse
Affiliation(s)
- Georgina S Butler
- Centre for Blood Research, Departments of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Andrea R Connor
- Centre for Blood Research, Departments of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Nor Eddine Sounni
- Centre for Blood Research, Departments of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Ulrich Eckhard
- Centre for Blood Research, Departments of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Charlotte J Morrison
- Centre for Blood Research, Departments of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Agnès Noël
- Centre for Blood Research, Departments of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Christopher M Overall
- Centre for Blood Research, Departments of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
31
|
Filippou PS, Karagiannis GS, Musrap N, Diamandis EP. Kallikrein-related peptidases (KLKs) and the hallmarks of cancer. Crit Rev Clin Lab Sci 2016; 53:277-91. [PMID: 26886390 DOI: 10.3109/10408363.2016.1154643] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The kallikrein-related peptidases (KLKs) represent the largest family of serine proteases within the human genome and are expressed in various tissues. Although they regulate several important physiological functions, KLKs have also been implicated in numerous pathophysiological processes, including cancer. Growing evidence describing the deregulation of KLK expression and secretion, as well as activation in various malignancies, has uncovered their potential as mediators of cancer progression, biomarkers of disease and as candidate therapeutic targets. The diversity of signalling pathways and proteolytic cascades involving KLKs and their downstream targets appears to affect cancer biology through multiple mechanisms, including those related to the hallmarks of cancer. The aim of this review is to provide an update on the importance of KLK-driven molecular pathways in relation to cancer cell traits associated with the hallmarks of cancer and to highlight their potential in personalized therapeutics.
Collapse
Affiliation(s)
- Panagiota S Filippou
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada
| | - George S Karagiannis
- b Department of Anatomy & Structural Biology , Albert Einstein College of Medicine, Yeshiva University Bronx , New York , NY , USA
| | - Natasha Musrap
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada
| | - Eleftherios P Diamandis
- a Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , ON , Canada .,c Department of Clinical Biochemistry , University Health Network , Toronto , ON , Canada , and.,d Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
32
|
Chaqour B. Regulating the regulators of angiogenesis by CCN1 and taking it up a Notch. J Cell Commun Signal 2016; 10:259-261. [PMID: 27146903 DOI: 10.1007/s12079-016-0328-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/02/2016] [Indexed: 01/16/2023] Open
Abstract
CCN1 is encoded by an extracellular matrix protein-gene that is essential for the proper development of the cardiovascular system and the control of angiogenesis, inflammation, progenitor cell lineage commitment and extracellular matrix protein remodeling during the adult life. High-precision genetic models of tissue-specific gene deletion demonstrated a pivotal role of CCN1 in providing positional information to angiogenic endothelial cells (ECs) during the outgrowth and maturation of nascent blood vessel sprouts, fine-controlling Notch-dependent inter-endothelial cell communications and mediating interaction with inflammatory cells. Some of these pleiotropic activities of CCN1 are unique among proteins of the extracellular matrix. Thus, CCN1 represents a model molecule for investigating and unraveling novel aspects of extracellular protein signaling in vascular development and diseases.
Collapse
Affiliation(s)
- Brahim Chaqour
- Department of Cell Biology, State University of New York - SUNY Downstate Medical Center, 450 Clarkson Avenue, Box 5, Brooklyn, NY, 11203, USA. .,Department of Ophthalmology, State University of New York - SUNY Downstate Medical Center, 450 Clarkson Avenue, Box 5, Brooklyn, NY, 11203, USA. .,SUNY Eye Institute, 450 Clarkson Avenue, Box 5, Brooklyn, NY, 11203, USA.
| |
Collapse
|
33
|
Kallikrein-related peptidase 8 is expressed in myocardium and induces cardiac hypertrophy. Sci Rep 2016; 7:20024. [PMID: 26823023 PMCID: PMC4731818 DOI: 10.1038/srep20024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 12/22/2015] [Indexed: 12/13/2022] Open
Abstract
The tissue kallikrein-related peptidase family (KLK) is a group of trypsin- and chymotrypsin-like serine proteases that share a similar homology to parent tissue kallikrein (KLK1). KLK1 is identified in heart and has anti-hypertrophic effects. However, whether other KLK family members play a role in regulating cardiac function remains unknown. In the present study, we demonstrated for the first time that KLK8 was expressed in myocardium. KLK8 expression was upregulated in left ventricle of cardiac hypertrophy models. Both intra-cardiac adenovirus-mediated and transgenic-mediated KLK8 overexpression led to cardiac hypertrophy in vivo. In primary neonatal rat cardiomyocytes, KLK8 knockdown inhibited phenylephrine (PE)-induced cardiomyocyte hypertrophy, whereas KLK8 overexpression promoted cardiomyocyte hypertrophy via a serine protease activity-dependent but kinin receptor-independent pathway. KLK8 overexpression increased epidermal growth factor (EGF) production, which was blocked by the inhibitors of serine protease. EGF receptor (EGFR) antagonist and EGFR knockdown reversed the hypertrophy induced by KLK8 overexpression. KLK8-induced cardiomyocyte hypertrophy was also significantly decreased by blocking the protease-activated receptor 1 (PAR1) or PAR2 pathway. Our data suggest that KLK8 may promote cardiomyocyte hypertrophy through EGF signaling- and PARs-dependent but a kinin receptor-independent pathway. It is implied that different KLK family members can subtly regulate cardiac function and remodeling.
Collapse
|
34
|
Kallikreins - The melting pot of activity and function. Biochimie 2015; 122:270-82. [PMID: 26408415 DOI: 10.1016/j.biochi.2015.09.023] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/21/2015] [Indexed: 12/20/2022]
Abstract
The human tissue kallikrein and kallikrein-related peptidases (KLKs), encoded by the largest contiguous cluster of protease genes in the human genome, are secreted serine proteases with diverse expression patterns and physiological roles. Because of the broad spectrum of processes that are modulated by kallikreins, these proteases are the subject of extensive investigations. This review brings together basic information about the biochemical properties affecting enzymatic activity, with highlights on post-translational modifications, especially glycosylation. Additionally, we present the current state of knowledge regarding the physiological functions of KLKs in major human organs and outline recent discoveries pertinent to the involvement of kallikreins in cell signaling and in viral infections. Despite the current depth of knowledge of these enzymes, many questions regarding the roles of kallikreins in health and disease remain unanswered.
Collapse
|
35
|
The kallikrein-related peptidase family: Dysregulation and functions during cancer progression. Biochimie 2015; 122:283-99. [PMID: 26343558 DOI: 10.1016/j.biochi.2015.09.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/01/2015] [Indexed: 01/07/2023]
Abstract
Cancer is the second leading cause of death with 14 million new cases and 8.2 million cancer-related deaths worldwide in 2012. Despite the progress made in cancer therapies, neoplastic diseases are still a major therapeutic challenge notably because of intra- and inter-malignant tumour heterogeneity and adaptation/escape of malignant cells to/from treatment. New targeted therapies need to be developed to improve our medical arsenal and counter-act cancer progression. Human kallikrein-related peptidases (KLKs) are secreted serine peptidases which are aberrantly expressed in many cancers and have great potential in developing targeted therapies. The potential of KLKs as cancer biomarkers is well established since the demonstration of the association between KLK3/PSA (prostate specific antigen) levels and prostate cancer progression. In addition, a constantly increasing number of in vitro and in vivo studies demonstrate the functional involvement of KLKs in cancer-related processes. These peptidases are now considered key players in the regulation of cancer cell growth, migration, invasion, chemo-resistance, and importantly, in mediating interactions between cancer cells and other cell populations found in the tumour microenvironment to facilitate cancer progression. These functional roles of KLKs in a cancer context further highlight their potential in designing new anti-cancer approaches. In this review, we comprehensively review the biochemical features of KLKs, their functional roles in carcinogenesis, followed by the latest developments and the successful utility of KLK-based therapeutics in counteracting cancer progression.
Collapse
|
36
|
Chijiiwa M, Mochizuki S, Kimura T, Abe H, Tanaka Y, Fujii Y, Shimizu H, Enomoto H, Toyama Y, Okada Y. CCN1 (Cyr61) Is Overexpressed in Human Osteoarthritic Cartilage and Inhibits ADAMTS-4 (Aggrecanase 1) Activity. Arthritis Rheumatol 2015; 67:1557-67. [PMID: 25709087 DOI: 10.1002/art.39078] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/12/2015] [Indexed: 01/22/2023]
Abstract
OBJECTIVE ADAMTS-4, also called aggrecanase 1, is considered to play a key role in aggrecan degradation in human osteoarthritic (OA) cartilage, but information about regulators of ADAMTS-4 aggrecanase activity remains limited. We undertook this study to search for molecules that modulate ADAMTS-4 activity. METHODS Molecules copurified with ADAMTS-4 from ADAMTS-4-transfected chondrocytic cells were sequenced by nanoscale liquid chromatography tandem mass spectrometry. Binding activity was determined by immunoprecipitation and solid-phase binding assay. Effects on ADAMTS-4 activity were examined by aggrecan digestion assay. Expression of the binding molecule in OA cartilage and chondrocytes was examined by immunohistochemistry and reverse transcription-polymerase chain reaction. RESULTS We identified CCN1 (Cyr61) as an ADAMTS-4-binding protein and showed specific binding to the ADAMTS-4 cysteine-rich domain. Aggrecanase activity of ADAMTS-4 was inhibited by interaction with CCN1. Expression of messenger RNA for CCN1 was significantly higher in human OA cartilage than in normal cartilage. CCN1 was immunolocalized to chondrocytes in OA cartilage, showing direct correlations of immunoreactivity with the Mankin score of cartilage lesions and chondrocyte cloning. CCN1 and ADAMTS-4 were commonly coexpressed in clustered chondrocytes. CCN1 expression in OA chondrocytes was down-regulated by interleukin-1α (IL-1α) and up-regulated by transforming growth factor β (TGFβ). ADAMTS-4 expression was induced by treatment with IL-1α or TGFβ, but aggrecanase activity was detected only under stimulation with IL-1α. TGFβ-treated chondrocytes exhibited aggrecanase activity when CCN1 expression was knocked down. CONCLUSION Our findings provide the first evidence that CCN1 suppresses ADAMTS-4 activity and that CCN1 overexpression is directly correlated with chondrocyte cloning in OA cartilage. Our results suggest that the TGFβ/CCN1 axis plays a role in chondrocyte cluster formation through inhibition of ADAMTS-4.
Collapse
Affiliation(s)
| | | | - Tokuhiro Kimura
- Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Hitoshi Abe
- Keio University School of Medicine, Tokyo, Japan
| | - Yukie Tanaka
- Fukui University School of Medicine, Fukui, Japan
| | - Yutaka Fujii
- Fukui University School of Medicine, Fukui, Japan
| | | | | | | | | |
Collapse
|
37
|
Yu Y, Prassas I, Dimitromanolakis A, Diamandis EP. Novel Biological Substrates of Human Kallikrein 7 Identified through Degradomics. J Biol Chem 2015; 290:17762-17775. [PMID: 26032414 DOI: 10.1074/jbc.m115.643551] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Indexed: 01/03/2023] Open
Abstract
Kallikrein-related peptidases (KLKs) are a group of serine proteases widely expressed in various tissues and involved in a wide range of physiological and pathological processes. Although our understanding of the pathophysiological roles of most KLKs has blossomed in recent years, identification of the direct endogenous substrates of human KLKs remains an unmet objective. In this study we employed a degradomics approach to systemically investigate the endogenous substrates of KLK7 in an effort to understand the molecular pathways underlying KLK7 action in skin. We identified several previously known as well as novel protein substrates. Our most promising candidates were further validated with the use of targeted quantitative proteomics (selected reaction monitoring methods) and in vitro recombinant protein digestion assays. Our study revealed midkine, CYR61, and tenascin-C as endogenous substrates for KLK7. Interestingly, some of these substrates (e.g. midkine) were prone to proteolytic cleavage only by KLK7 (and not by other skin-associated KLKs), whereas others (e.g. CYR61 and tenascin-C) could be digested by several KLKs. Furthermore, using melanoma cell line, we show that KLK7-mediated cleavage of midkine results in an overall reduction in the pro-proliferative and pro-migratory effect of midkine. An inverse relation between KLK7 and midkine is also observed in human melanoma tissues. In summary, our degradomics approach revealed three novel endogenous substrates for KLK7, which may shed more light on the pathobiological roles of KLK7 in human skin. Similar substrate screening approaches could be applied for the discovery of biological substrates of other protease.
Collapse
Affiliation(s)
- Yijing Yu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario M5T 3L9, Canada
| | - Ioannis Prassas
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario M5T 3L9, Canada
| | | | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario M5T 3L9, Canada; Department of Clinical Biochemistry, University Health Network, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
38
|
Klaassen I, van Geest RJ, Kuiper EJ, van Noorden CJF, Schlingemann RO. The role of CTGF in diabetic retinopathy. Exp Eye Res 2015; 133:37-48. [PMID: 25819453 DOI: 10.1016/j.exer.2014.10.016] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 10/16/2014] [Accepted: 10/17/2014] [Indexed: 10/23/2022]
Abstract
Connective tissue growth factor (CTGF, CCN2) contributes to fibrotic responses in diabetic retinopathy, both before clinical manifestations occur in the pre-clinical stage of diabetic retinopathy (PCDR) and in proliferative diabetic retinopathy (PDR), the late clinical stage of the disease. CTGF is a secreted protein that modulates the actions of many growth factors and extracellular matrix (ECM) proteins, leading to tissue reorganization, such as ECM formation and remodeling, basal lamina (BL) thickening, pericyte apoptosis, angiogenesis, wound healing and fibrosis. In PCDR, CTGF contributes to thickening of the retinal capillary BL and is involved in loss of pericytes. In this stage, CTGF expression is induced by advanced glycation end products, and by growth factors such as vascular endothelial growth factor (VEGF) and transforming growth factor (TGF)-β. In PDR, the switch from neovascularization to a fibrotic phase - the angio-fibrotic switch - in PDR is driven by CTGF, in a critical balance with vascular endothelial growth factor (VEGF). We discuss here the roles of CTGF in the pathogenesis of DR in relation to ECM remodeling and wound healing mechanisms, and explore whether CTGF may be a potential novel therapeutic target in the clinical management of early as well as late stages of DR.
Collapse
Affiliation(s)
- Ingeborg Klaassen
- Ocular Angiogenesis Group, Department of Ophthalmology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Rob J van Geest
- Ocular Angiogenesis Group, Department of Ophthalmology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther J Kuiper
- Ocular Angiogenesis Group, Department of Ophthalmology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Cornelis J F van Noorden
- Ocular Angiogenesis Group, Department of Ophthalmology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Department of Ophthalmology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Netherlands Institute for Neuroscience, Royal Academy of Sciences, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Chintala H, Krupska I, Yan L, Lau L, Grant M, Chaqour B. The matricellular protein CCN1 controls retinal angiogenesis by targeting VEGF, Src homology 2 domain phosphatase-1 and Notch signaling. Development 2015; 142:2364-74. [PMID: 26002917 DOI: 10.1242/dev.121913] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/14/2015] [Indexed: 12/19/2022]
Abstract
Physiological angiogenesis depends on the highly coordinated actions of multiple angiogenic regulators. CCN1 is a secreted cysteine-rich and integrin-binding matricellular protein required for proper cardiovascular development. However, our understanding of the cellular origins and activities of this molecule is incomplete. Here, we show that CCN1 is predominantly expressed in angiogenic endothelial cells (ECs) at the leading front of actively growing vessels in the mouse retina. Endothelial deletion of CCN1 in mice using a Cre-Lox system is associated with EC hyperplasia, loss of pericyte coverage and formation of dense retinal vascular networks lacking the normal hierarchical arrangement of arterioles, capillaries and venules. CCN1 is a product of an immediate-early gene that is transcriptionally induced in ECs in response to stimulation by vascular endothelial growth factor (VEGF). We found that CCN1 activity is integrated with VEGF receptor 2 (VEGF-R2) activation and downstream signaling pathways required for tubular network formation. CCN1-integrin binding increased the expression of and association between Src homology 2 domain-containing protein tyrosine phosphatase-1 (SHP-1) and VEGF-R2, which leads to rapid dephosphorylation of VEGF-R2 tyrosine, thus preventing EC hyperproliferation. Predictably, CCN1 further brings receptors/signaling molecules into proximity that are otherwise spatially separated. Furthermore, CCN1 induces integrin-dependent Notch activation in cultured ECs, and its targeted gene inactivation in vivo alters Notch-dependent vascular specification and remodeling, suggesting that functional levels of Notch signaling requires CCN1 activity. These data highlight novel functions of CCN1 as a naturally optimized molecule, fine-controlling key processes in physiological angiogenesis and safeguarding against aberrant angiogenic responses.
Collapse
Affiliation(s)
- Hemabindu Chintala
- State University of New York (SUNY) Eye Institute and Department of Cell Biology, Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Izabela Krupska
- State University of New York (SUNY) Eye Institute and Department of Cell Biology, Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Lulu Yan
- State University of New York (SUNY) Eye Institute and Department of Cell Biology, Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Lester Lau
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, Chicago, IL 60607, USA
| | - Maria Grant
- Departments of Ophthalmology and Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brahim Chaqour
- State University of New York (SUNY) Eye Institute and Department of Cell Biology, Downstate Medical Center, Brooklyn, NY 11203, USA Department of Ophthalmology, Downstate Medical Center, Brooklyn, NY 11203, USA
| |
Collapse
|
40
|
Kubota S, Maeda-Uematsu A, Nishida T, Takigawa M. New functional aspects of CCN2 revealed by trans-omic approaches. J Oral Biosci 2015. [DOI: 10.1016/j.job.2014.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
41
|
Cellular and molecular actions of CCN2/CTGF and its role under physiological and pathological conditions. Clin Sci (Lond) 2014; 128:181-96. [DOI: 10.1042/cs20140264] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CCN family protein 2 (CCN2), also widely known as connective tissue growth factor (CTGF), is one of the founding members of the CCN family of matricellular proteins. Extensive investigation on CCN2 over decades has revealed the novel molecular action and functional properties of this unique signalling modulator. By its interaction with multiple molecular counterparts, CCN2 yields highly diverse and context-dependent biological outcomes in a variety of microenvironments. Nowadays, CCN2 is recognized to conduct the harmonized development of relevant tissues, such as cartilage and bone, in the skeletal system, by manipulating extracellular signalling molecules involved therein by acting as a hub through a web. However, on the other hand, CCN2 occasionally plays profound roles in major human biological disorders, including fibrosis and malignancies in major organs and tissues, by modulating the actions of key molecules involved in these clinical entities. In this review, the physiological and pathological roles of this unique protein are comprehensively summarized from a molecular network-based viewpoint of CCN2 functionalities.
Collapse
|
42
|
Moon HG, Kim SH, Gao J, Quan T, Qin Z, Osorio JC, Rosas IO, Wu M, Tesfaigzi Y, Jin Y. CCN1 secretion and cleavage regulate the lung epithelial cell functions after cigarette smoke. Am J Physiol Lung Cell Mol Physiol 2014; 307:L326-37. [PMID: 24973403 DOI: 10.1152/ajplung.00102.2014] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Despite extensive research, the pathogenesis of cigarette smoking (CS)-associated emphysema remains incompletely understood, thereby impeding development of novel therapeutics, diagnostics, and biomarkers. Here, we report a novel paradigm potentially involved in the development of epithelial death and tissue loss in CS-associated emphysema. After prolonged exposure of CS, CCN1 cleavage was detected both in vitro and in vivo. Full-length CCN1 (flCCN1) was secreted in an exosome-shuttled manner, and secreted plasmin converted flCCN1 to cleaved CCN1 (cCCN1) in extracellular matrix. Interestingly, exosome-shuttled flCCN1 facilitated the interleukin (IL)-8 and vascular endothelial growth factor (VEGF) release in response to cigarette smoke extract (CSE). Therefore, flCCN1 potentially promoted CS-induced inflammation via IL-8-mediated neutrophil recruitment and also maintained the lung homeostasis via VEGF secretion. Interestingly, cCCN1 abolished these functions. Furthermore, cCCN1 promoted protease and matrix metalloproteinase (MMP)-1 production after CSE. These effects were mainly mediated by the COOH-terminal fragments of CCN1 after cleavage. Both the decrease of VEGF and the elevation of MMPs favor the development of emphysema. cCCN1, therefore, likely contributes to the epithelial cell damage after CS. Additionally, CSE and cCCN1 both stimulated integrin-α7 expressions in lung epithelial cells. The integrin-α7 appeared to be the binding receptors of cCCN1 and, subsequently, mediated its cellular function by promoting MMP1. Consistent with our observation on the functional roles of cCCN1 in vitro, elevated cCCN1 level was found in the bronchoalveolar lavage fluid from mice with emphysematous changes after 6 mo CS exposure. Taken together, we hypothesize that cCCN1 promoted the epithelial cell death and tissue loss after prolonged CS exposure.
Collapse
Affiliation(s)
- Hyung-Geun Moon
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sang-Heon Kim
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Jinming Gao
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Zhaoping Qin
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Juan C Osorio
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Min Wu
- Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota; and
| | | | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
43
|
Fuhrman-Luck RA, Silva ML, Dong Y, Irving-Rodgers H, Stoll T, Hastie ML, Loessner D, Gorman JJ, Clements JA. Proteomic and other analyses to determine the functional consequences of deregulated kallikrein-related peptidase (KLK) expression in prostate and ovarian cancer. Proteomics Clin Appl 2014; 8:403-15. [PMID: 24535680 DOI: 10.1002/prca.201300098] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/23/2013] [Accepted: 11/30/2013] [Indexed: 02/06/2023]
Abstract
Rapidly developing proteomic tools are improving detection of deregulated kallikrein-related peptidase (KLK) expression, at the protein level, in prostate and ovarian cancer, as well as facilitating the determination of functional consequences downstream. MS-driven proteomics uniquely allows for the detection, identification, and quantification of thousands of proteins in a complex protein pool, and this has served to identify certain KLKs as biomarkers for these diseases. In this review, we describe applications of this technology in KLK biomarker discovery and elucidate MS-based techniques that have been used for unbiased, global screening of KLK substrates within complex protein pools. Although MS-based KLK degradomic studies are limited to date, they helped to discover an array of novel KLK substrates. Substrates identified by MS-based degradomics are reported with improved confidence over those determined by incubating a purified or recombinant substrate and protease of interest, in vitro. We propose that these novel proteomic approaches represent the way forward for KLK research, in order to correlate proteolysis of biological substrates with tissue-related consequences, toward clinical targeting of KLK expression and function for cancer diagnosis, prognosis, and therapies.
Collapse
Affiliation(s)
- Ruth Anna Fuhrman-Luck
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, Australia; Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kryza T, Achard C, Parent C, Marchand-Adam S, Guillon-Munos A, Iochmann S, Korkmaz B, Respaud R, Courty Y, Heuzé-Vourc'h N. Angiogenesis stimulated by human kallikrein-related peptidase 12 acting via a platelet-derived growth factor B-dependent paracrine pathway. FASEB J 2013; 28:740-51. [PMID: 24225148 DOI: 10.1096/fj.13-237503] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
KLK12, a kallikrein peptidase, is thought to take part in the control of angiogenesis. Our analysis of the secretome of endothelial cells (ECs) that had been treated with KLK12 showed that KLK12 converts the extracellular matrix- or membrane-bound precursor of platelet-derived growth factor B (PDGF-B) into a soluble form. Both PDGF-B and vascular endothelial growth factor A (VEGF-A) take part in the induction of angiogenesis by KLK12 in a coculture model of angiogenesis that mimics endothelial tubule formation. We used a cellular approach to analyze the interplay between KLK12, PDGF-B, and VEGF-A and showed that release of PDGF-B by KLK12 leads to the fibroblast-mediated secretion of VEGF-A. This then stimulates EC differentiation and the formation of capillary tube-like structures. Thus, KLK12 favors the interaction of ECs and stromal cells. The released PDGF-B acts as a paracrine factor that modulates VEGF-A secretion by stromal cells, which ultimately leads to angiogenesis. Moreover, the genes encoding KLK12 and PDGFB are both expressed in ECs and up-regulated in tumor cells kept under hypoxic conditions, which is consistent with the physiological involvement of KLK12 in PDGF-B maturation.
Collapse
Affiliation(s)
- Thomas Kryza
- 2CEPR INSERM U1100/EA 6305, Faculté de Médecine, 10 Blvd. Tonnellé, F-37032 Tours cedex, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Single-target RNA interference for the blockade of multiple interacting proinflammatory and profibrotic pathways in cardiac fibroblasts. J Mol Cell Cardiol 2013; 66:141-56. [PMID: 24239602 DOI: 10.1016/j.yjmcc.2013.11.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 10/18/2013] [Accepted: 11/04/2013] [Indexed: 12/14/2022]
Abstract
Therapeutic targets of broad relevance are likely located in pathogenic pathways common to disorders of various etiologies. Screening for targets of this type revealed CCN genes to be consistently upregulated in multiple cardiomyopathies. We developed RNA interference (RNAi) to silence CCN2 and found this single-target approach to block multiple proinflammatory and profibrotic pathways in activated primary cardiac fibroblasts (PCFBs). The RNAi-strategy was developed in murine PCFBs and then investigated in "individual" human PCFBs grown from human endomyocardial biopsies (EMBs). Screening of short hairpin RNA (shRNA) sequences for high silencing efficacy and specificity yielded RNAi adenovectors silencing CCN2 in murine or human PCFBs, respectively. Comparison of RNAi with CCN2-modulating microRNA (miR) vectors expressing miR-30c or miR-133b showed higher efficacy of RNAi. In murine PCFBs, CCN2 silencing resulted in strongly reduced expression of stretch-induced chemokines (Ccl2, Ccl7, Ccl8), matrix metalloproteinases (MMP2, MMP9), extracellular matrix (Col3a1), and a cell-to-cell contact protein (Cx43), suggesting multiple signal pathways to be linked to CCN2. Immune cell chemotaxis towards CCN2-depleted PCFBs was significantly reduced. We demonstrate here that this RNAi strategy is technically applicable to "individual" human PCFBs, too, but that these display individually strikingly different responses to CCN2 depletion. Either genomically encoded factors or stable epigenetic modification may explain different responses between individual PCFBs. The new RNAi approach addresses a key regulator protein induced in cardiomyopathies. Investigation of this and other molecular therapies in individual human PCBFs may help to dissect differential pathogenic processes between otherwise similar disease entities and individuals.
Collapse
|
46
|
Swarts DRA, Van Neste L, Henfling MER, Eijkenboom I, Eijk PP, van Velthuysen ML, Vink A, Volante M, Ylstra B, Van Criekinge W, van Engeland M, Ramaekers FCS, Speel EJM. An exploration of pathways involved in lung carcinoid progression using gene expression profiling. Carcinogenesis 2013; 34:2726-37. [PMID: 23929435 DOI: 10.1093/carcin/bgt271] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pulmonary carcinoids comprise a well-differentiated subset of neuroendocrine tumors usually associated with a favorable prognosis, but mechanisms underlying disease progression are poorly understood. In an explorative approach to identify pathways associated with progression, we compared gene expression profiles of tumors from five patients with a favorable and five with a poor disease outcome. Differentially expressed genes were validated using quantitative real-time PCR on 65 carcinoid tumors, in combination with survival analysis. One of the identified pathways was further examined using immunohistochemistry. As compared with other chromosomal locations, a significantly higher number of genes downregulated in carcinoids with a poor prognosis were located at chromosome 11q (P = 0.00017), a region known to be frequently lost in carcinoids. In addition, a number of upregulated genes were found involved in the mitotic spindle checkpoint, the chromosomal passenger complex (CPC), mitotic kinase CDC2 activity and the BRCA-Fanconi anemia pathway. At the individual gene level, BIRC5 (survivin), BUB1, CD44, IL20RA, KLK12 and OTP were independent predictors of patient outcome. For survivin, the number of positive nuclei was also related to poor prognosis within the group of carcinoids. Aurora B kinase and survivin, major components of the CPC, were particularly upregulated in high-grade carcinomas and may therefore comprise therapeutic targets for these tumors. To our knowledge, this is the first expression profiling study focusing specifically on pulmonary carcinoids and progression. We have identified novel pathways underlying malignant progression and validated several genes as being strong prognostic indicators, some of which could serve as putative therapeutic targets.
Collapse
Affiliation(s)
- Dorian R A Swarts
- Department of Molecular Cell Biology, GROW - School for Oncology & Developmental Biology, Maastricht University Medical Center, PO Box 616, 6200 MD Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Choi J, Lin A, Shrier E, Lau LF, Grant MB, Chaqour B. Degradome products of the matricellular protein CCN1 as modulators of pathological angiogenesis in the retina. J Biol Chem 2013; 288:23075-89. [PMID: 23798676 DOI: 10.1074/jbc.m113.475418] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
CCN1 is a matricellular protein involved in normal vascular development and tissue repair. CCN1 exhibits cell- and context-dependent activities that are reflective of its tetramodular structure phylogenetically linked to four domains found in various matrix proteins. Here, we show that vitreal fluids from patients with proliferative diabetic retinopathy (PDR) were enriched with a two-module form of CCN1 comprising completely or partially the insulin-like growth factor-binding protein (IGFBP) and von Willebrand factor type C (vWC) domains. The two- and three-module forms comprising, in addition to IGFBP and vWC, the thrombospondin type 1 (TSP1) repeats are CCN1 degradome products by matrix metalloproteinase-2 and -14. The functional significance of CCN1 and its truncated variants was determined in the mouse model of oxygen-induced retinopathy, which simulates neovascular growth associated with PDR and assesses treatment outcomes. In this model, lentivirus-mediated expression of either CCN1 or the IGFBP-vWC-TSP1 form reduced ischemia-induced neovascularization, whereas ectopic expression of the IGFBP-vWC variant exacerbated pathological angiogenesis. The IGFBP-vWC form has potent proangiogenic properties promoting retinal endothelial cell growth, migration, and three-dimensional tubular structure formation, whereas the IGFBP-vWC-TSP1 variant suppressed cell growth and angiogenic gene expression. Both IGFBP-vWC and IGFBP-vWC-TSP1 forms exhibited predictable variations of their domain folding that enhanced their functional potential. These data provide new insights into the formation and activities of CCN1-truncated variants and raise the predictive value of the form containing completely or partially the IGFBP and vWC domains as a surrogate marker of CCN1 activity in PDR distinguishing pathological from physiological angiogenesis.
Collapse
Affiliation(s)
- Jinok Choi
- Department of Cell Biology, State University of New York Eye Institute, Downstate Medical Center, Brooklyn, New York 11203, USA
| | | | | | | | | | | |
Collapse
|
48
|
Robinson PM, Smith TS, Patel D, Dave M, Lewin AS, Pi L, Scott EW, Tuli SS, Schultz GS. Proteolytic processing of connective tissue growth factor in normal ocular tissues and during corneal wound healing. Invest Ophthalmol Vis Sci 2012; 53:8093-8103. [PMID: 23139278 PMCID: PMC3522436 DOI: 10.1167/iovs.12-10419] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 10/11/2012] [Accepted: 11/02/2012] [Indexed: 01/23/2023] Open
Abstract
PURPOSE Connective tissue growth factor (CTGF) is a fibrogenic cytokine that is up-regulated by TGF-β and mediates most key fibrotic actions of TGF-β, including stimulation of synthesis of extracellular matrix and differentiation of fibroblasts into myofibroblasts. This study addresses the role of proteolytic processing of CTGF in human corneal fibroblasts (HCF) stimulated with TGF-β, normal ocular tissues and wounded corneas. METHODS Proteolytic processing of CTGF in HCF cultures, normal animal eyes, and excimer laser wounded rat corneas were examined by Western blot. The identity of a 21-kDa band was determined by tandem mass spectrometry, and possible alternative splice variants of CTGF were assessed by 5' Rapid Amplification of cDNA Ends (RACE). RESULTS HCF stimulated by TGF-β contained full length 38-kDa CTGF and fragments of 25, 21, 18, and 13 kDa, while conditioned medium contained full length 38- and a 21-kDa fragment of CTGF that contained the middle "hinge" region of CTGF. Fragmentation of recombinant CTGF incubated in HCF extracts was blocked by the aspartate protease inhibitor, pepstatin. Normal mouse, rat, and rabbit whole eyes and rabbit ocular tissues contained abundant amounts of C-terminal 25- and 21-kDa fragments and trace amounts of 38-kDa CTGF, although no alternative transcripts were detected. All forms of CTGF (38, 25, and 21 kDa) were detected during healing of excimer ablated rat corneas, peaking on day 11. CONCLUSIONS Proteolytic processing of 38-kDa CTGF occurs during corneal wound healing, which may have important implications in regulation of corneal scar formation.
Collapse
Affiliation(s)
| | | | - Dilan Patel
- From the Department of Obstetrics and Gynecology, the
| | - Meera Dave
- From the Department of Obstetrics and Gynecology, the
| | | | - Liya Pi
- Department of Molecular Genetics and Microbiology, and the
| | | | - Sonal S. Tuli
- Department of Ophthalmology, Institute for Wound Research, University of Florida, Gainesville, Florida
| | | |
Collapse
|
49
|
Ouellet V, Siegel PM. CCN3 modulates bone turnover and is a novel regulator of skeletal metastasis. J Cell Commun Signal 2012; 6:73-85. [PMID: 22427255 PMCID: PMC3368020 DOI: 10.1007/s12079-012-0161-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 02/15/2012] [Indexed: 12/16/2022] Open
Abstract
The CCN family of proteins is composed of six secreted proteins (CCN1-6), which are grouped together based on their structural similarity. These matricellular proteins are involved in a large spectrum of biological processes, ranging from development to disease. In this review, we focus on CCN3, a founding member of this family, and its role in regulating cells within the bone microenvironment. CCN3 impairs normal osteoblast differentiation through multiple mechanisms, which include the neutralization of pro-osteoblastogenic stimuli such as BMP and Wnt family signals or the activation of pathways that suppress osteoblastogenesis, such as Notch. In contrast, CCN3 is known to promote chondrocyte differentiation. Given these functions, it is not surprising that CCN3 has been implicated in the progression of primary bone cancers such as osteosarcoma, Ewing’s sarcoma and chondrosarcoma. More recently, emerging evidence suggests that CCN3 may also influence the ability of metastatic cancers to colonize and grow in bone.
Collapse
Affiliation(s)
- Véronique Ouellet
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 513, Montreal, Quebec Canada H3A 1A3
| | - Peter M. Siegel
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Room 513, Montreal, Quebec Canada H3A 1A3
- Departments of Anatomy and Cell Biology, Biochemistry and Medicine, McGill University, Montreal, Quebec Canada
| |
Collapse
|
50
|
Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets. Nat Rev Drug Discov 2011; 10:945-63. [PMID: 22129992 DOI: 10.1038/nrd3599] [Citation(s) in RCA: 513] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Members of the CCN family of matricellular proteins are crucial for embryonic development and have important roles in inflammation, wound healing and injury repair in adulthood. Deregulation of CCN protein expression or activities contributes to the pathobiology of various diseases - many of which may arise when inflammation or tissue injury becomes chronic - including fibrosis, atherosclerosis, arthritis and cancer, as well as diabetic nephropathy and retinopathy. Emerging studies indicate that targeting CCN protein expression or signalling pathways holds promise in the development of diagnostics and therapeutics for such diseases. This Review summarizes the biology of CCN proteins, their roles in various pathologies and their potential as therapeutic targets.
Collapse
|