1
|
Fujii Y, Asadi Z, Mehla K. Cathepsins: Emerging targets in the tumor ecosystem to overcome cancers. Semin Cancer Biol 2025; 112:150-166. [PMID: 40228591 DOI: 10.1016/j.semcancer.2025.04.001] [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: 08/09/2024] [Revised: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Cathepsins, a group of lysosomal peptidases, have traditionally been recognized as tumor facilitators. Recent research, however, highlights their critical role in orchestrating cancer and the tumor microenvironment (TME). Primality, cathepsins degrade extracellular matrix, enabling cancer cells to invade and metastasize, while also promoting vascular endothelial infiltration and subsequent angiogenesis. Additionally, cathepsins boost fibroblast growth, thereby supporting tumor progression. More importantly, cathepsins are pivotal in modulating immune cells within the TME by regulating their recruitment, antigen processing and presentation, differentiation, and cell death, primarily contributing to immune suppression. Given their overexpression in tumors and elevated levels in the circulation of cancer patients, it is crucial to consider the systemic effects of cathepsins. Although the comprehensive role of cathepsins in cancer patients' bodies remains underexplored, they likely influence systemic immunity and inflammation, cellular metabolism, muscle wasting, and distant metastasis through their unique proteolytic functions. Notably, cathepsins also confer resistance to chemoradiotherapy by rewriting the cellular profile within the TME. In this context, promising results are emerging from studies combining cathepsin inhibitors with conventional therapies to suppress tumor development effectively. This review aims to decipher the cathepsin-driven networks within cancer cells and the TME, detailing their contribution to chemoradioresistance by reshaping both micro- and macroenvironments. Furthermore, we explore current and future perspectives on therapies targeting cathepsins' interactions, offering insights into innovative treatment strategies.
Collapse
Affiliation(s)
- Yuki Fujii
- Department of Oncology Science, University of Oklahoma Health Sciences, Oklahoma City, OK 73014, USA
| | - Zahra Asadi
- Department of Oncology Science, University of Oklahoma Health Sciences, Oklahoma City, OK 73014, USA; Department of Pathology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Kamiya Mehla
- Department of Oncology Science, University of Oklahoma Health Sciences, Oklahoma City, OK 73014, USA; Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA; Department of Pathology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA.
| |
Collapse
|
2
|
Meade RK, Adefisayo OO, Gontijo MTP, Harris SJ, Pyle CJ, Wilburn KM, Ecker AMV, Hughes EJ, Garcia PD, Ivie J, McHenry ML, Benchek PH, Mayanja-Kizza H, Neff JL, Ko DC, Stout JE, Stein CM, Hawn TR, Tobin DM, Smith CM. Cathepsin Z is a conserved susceptibility factor underlying tuberculosis severity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.644622. [PMID: 40236047 PMCID: PMC11996505 DOI: 10.1101/2025.04.01.644622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Tuberculosis (TB) outcomes vary widely, from asymptomatic infection to mortality, yet most animal models do not recapitulate human phenotypic and genotypic variation. The genetically diverse Collaborative Cross mouse panel models distinct facets of TB disease that occur in humans and allows identification of genomic loci underlying clinical outcomes. We previously mapped a TB susceptibility locus on mouse chromosome 2. Here, we identify cathepsin Z ( Ctsz ) as a lead candidate underlying this TB susceptibility and show that Ctsz ablation leads to increased bacterial burden, CXCL1 overproduction, and decreased survival in mice. Ctsz disturbance within murine macrophages enhances production of CXCL1, a known biomarker of TB severity. From a Ugandan household contact study, we identify significant associations between CTSZ variants and TB disease severity. Finally, we examine patient-derived TB granulomas and report CTSZ localization within granuloma-associated macrophages, placing human CTSZ at the host-pathogen interface. These findings implicate a conserved CTSZ-CXCL1 axis in humans and genetically diverse mice that mediates TB disease severity.
Collapse
|
3
|
Wang Y, Wang Y, Jiang Y, Qin Q, Wei S. The essential function of cathepsin X of the orange-spotted grouper, Epinephelus coioides during SGIV infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105278. [PMID: 39395685 DOI: 10.1016/j.dci.2024.105278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
Cathepsin X, a class of cysteine proteases in the lysosome, involved in intracellular protein degradation processes. Numerous reports revealed that many kinds of cysteine proteases played a crucial role in pathogen invasion. To investigate the relationship between cathepsin X of teleost fish and virus infection, EcCX was cloned and characterized in the orange-spotted grouper, Epinephelus coioides. The open reading frame (ORF) of EcCX included 909 nucleotides and encoded a protein consisting of 302 amino acids, which shared 75% and 56% identity with zebrafish and humans, respectively. The protein EcCX mainly consisted of a signal peptide (1-19 aa), a pro-pre-peptide region (20-55 aa), and a mature cysteine protease region (56-302 aa). Subcellular localization analysis showed that EcCX was mainly distributed in the cytoplasm, but EcCX ectoped to the vicinity of apoptotic vesicles in FHM cells during SGIV infection. Following stimulation with SGIV or Poly (dA:dT), there was a notable rise in the expression levels of EcCX. EcCX overexpression facilitated virus infection, upregulated the production of inflammatory factors, and induced the activation of the NF-κB promoter. Furthermore, the overexpression of EcCX also accelerated the process of SGIV-induced apoptosis, potentially by enhancing the promoter activity of P53 and AP-1. Overall, our findings demonstrated a correlation between the function of EcCX and SGIV infection, providing a new understanding of the mechanisms involved in fish virus infection.
Collapse
Affiliation(s)
- Yuexuan Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Yewen Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Yunxiang Jiang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, 511457, China.
| | - Shina Wei
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, 511457, China.
| |
Collapse
|
4
|
Xu B, Anderson BM, Mountford SJ, Thompson PE, Mintern JD, Edgington-Mitchell LE. Cathepsin X deficiency alters the processing and localisation of cathepsin L and impairs cleavage of a nuclear cathepsin L substrate. Biol Chem 2024; 405:351-365. [PMID: 38410910 DOI: 10.1515/hsz-2023-0355] [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: 11/29/2023] [Accepted: 02/12/2024] [Indexed: 02/28/2024]
Abstract
Proteases function within sophisticated networks. Altering the activity of one protease can have sweeping effects on other proteases, leading to changes in their activity, structure, specificity, localisation, stability, and expression. Using a suite of chemical tools, we investigated the impact of cathepsin X, a lysosomal cysteine protease, on the activity and expression of other cysteine proteases and their inhibitors in dendritic cells. Among all proteases examined, cathepsin X gene deletion specifically altered cathepsin L levels; pro-cathepsin L and its single chain accumulated while the two-chain form was unchanged. This effect was recapitulated by chemical inhibition of cathepsin X, suggesting a dependence on its catalytic activity. We demonstrated that accumulation of pro- and single chain cathepsin L was not due to a lack of direct cleavage by cathepsin X or altered glycosylation, secretion, or mRNA expression but may result from changes in lysosomal oxidative stress or pH. In the absence of active cathepsin X, nuclear cathepsin L and cleavage of the known nuclear cathepsin L substrate, Lamin B1, were diminished. Thus, cathepsin X activity selectively regulates cathepsin L, which has the potential to impact the degree of cathepsin L proteolysis, the nature of substrates that it cleaves, and the location of cleavage.
Collapse
Affiliation(s)
- Bangyan Xu
- Department of Biochemistry & Pharmacology, 2281 Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne , Parkville, VIC 3052, Australia
| | - Bethany M Anderson
- Department of Biochemistry & Pharmacology, 2281 Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne , Parkville, VIC 3052, Australia
| | - Simon J Mountford
- Medicinal Chemistry, 2541 Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - Philip E Thompson
- Medicinal Chemistry, 2541 Monash Institute of Pharmaceutical Sciences, Monash University , Parkville, VIC 3052, Australia
| | - Justine D Mintern
- Department of Biochemistry & Pharmacology, 2281 Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne , Parkville, VIC 3052, Australia
| | - Laura E Edgington-Mitchell
- Department of Biochemistry & Pharmacology, 2281 Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne , Parkville, VIC 3052, Australia
| |
Collapse
|
5
|
Liu H, Peng J, Huang L, Ruan D, Li Y, Yuan F, Tu Z, Huang K, Zhu X. The role of lysosomal peptidases in glioma immune escape: underlying mechanisms and therapeutic strategies. Front Immunol 2023; 14:1154146. [PMID: 37398678 PMCID: PMC10311646 DOI: 10.3389/fimmu.2023.1154146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
Glioblastoma is the most common primary malignant tumor of the central nervous system, which has the characteristics of strong invasion, frequent recurrence, and rapid progression. These characteristics are inseparable from the evasion of glioma cells from immune killing, which makes immune escape a great obstacle to the treatment of glioma, and studies have confirmed that glioma patients with immune escape tend to have poor prognosis. The lysosomal peptidase lysosome family plays an important role in the immune escape process of glioma, which mainly includes aspartic acid cathepsin, serine cathepsin, asparagine endopeptidases, and cysteine cathepsins. Among them, the cysteine cathepsin family plays a prominent role in the immune escape of glioma. Numerous studies have confirmed that glioma immune escape mediated by lysosomal peptidases has something to do with autophagy, cell signaling pathways, immune cells, cytokines, and other mechanisms, especially lysosome organization. The relationship between protease and autophagy is more complicated, and the current research is neither complete nor in-depth. Therefore, this article reviews how lysosomal peptidases mediate the immune escape of glioma through the above mechanisms and explores the possibility of lysosomal peptidases as a target of glioma immunotherapy.
Collapse
Affiliation(s)
- Hao Liu
- Department of Neurosurgery, The Second Affifiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Jie Peng
- Department of Neurosurgery, The Second Affifiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Linzhen Huang
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Dong Ruan
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Yuguang Li
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Fan Yuan
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Zewei Tu
- Department of Neurosurgery, The Second Affifiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
- Jiangxi Health Commission (JXHC) Key Laboratory of Neurological Medicine, Nanchang, China
| | - Kai Huang
- Department of Neurosurgery, The Second Affifiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
- Jiangxi Health Commission (JXHC) Key Laboratory of Neurological Medicine, Nanchang, China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affifiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
- Jiangxi Health Commission (JXHC) Key Laboratory of Neurological Medicine, Nanchang, China
| |
Collapse
|
6
|
Senjor E, Kos J, Nanut MP. Cysteine Cathepsins as Therapeutic Targets in Immune Regulation and Immune Disorders. Biomedicines 2023; 11:biomedicines11020476. [PMID: 36831012 PMCID: PMC9953096 DOI: 10.3390/biomedicines11020476] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Cysteine cathepsins, as the most abundant proteases found in the lysosomes, play a vital role in several processes-such as protein degradation, changes in cell signaling, cell morphology, migration and proliferation, and energy metabolism. In addition to their lysosomal function, they are also secreted and may remain functional in the extracellular space. Upregulation of cathepsin expression is associated with several pathological conditions including cancer, neurodegeneration, and immune-system dysregulation. In this review, we present an overview of cysteine-cathepsin involvement and possible targeting options for mitigation of aberrant function in immune disorders such as inflammation, autoimmune diseases, and immune response in cancer.
Collapse
Affiliation(s)
- Emanuela Senjor
- Department of Biotechnology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Janko Kos
- Department of Biotechnology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Milica Perišić Nanut
- Department of Biotechnology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
7
|
Anes E, Pires D, Mandal M, Azevedo-Pereira JM. Spatial localization of cathepsins: Implications in immune activation and resolution during infections. Front Immunol 2022; 13:955407. [PMID: 35990632 PMCID: PMC9382241 DOI: 10.3389/fimmu.2022.955407] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022] Open
Abstract
Cathepsins were first described, as endolysosomal proteolytic enzymes in reference to the organelles where they degrade the bulk of endogenous and exogenous substrates in a slightly acidic environment. These substrates include pathogens internalized via endocytosis and/or marked for destruction by autophagy. However, the role of cathepsins during infection far exceeds that of direct digestion of the pathogen. Cathepsins have been extensively investigated in the context of tumour associated immune cells and chronic inflammation. Several cathepsin-dependent immune responses develop in the endocytic pathway while others take place in the cytosol, the nucleus, or in the extracellular space. In this review we highlight the spatial localization of cathepsins and their implications in immune activation and resolution pathways during infection.
Collapse
|
8
|
Abadijoo H, Khayamian MA, Faramarzpour M, Ghaderinia M, Simaee H, Shalileh S, Yazdanparast SM, Ghabraie B, Makarem J, Sarrami-Forooshani R, Abdolahad M. Healing Field: Using Alternating Electric Fields to Prevent Cytokine Storm by Suppressing Clonal Expansion of the Activated Lymphocytes in the Blood Sample of the COVID-19 Patients. Front Bioeng Biotechnol 2022; 10:850571. [PMID: 35721862 PMCID: PMC9201910 DOI: 10.3389/fbioe.2022.850571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
In the case of the COVID-19 early diagnosis, numerous tech innovations have been introduced, and many are currently employed worldwide. But, all of the medical procedures for the treatment of this disease, up to now, are just limited to chemical drugs. All of the scientists believe that the major challenge toward the mortality of the COVID-19 patients is the out-of-control immune system activation and the subsequent cytokine production. During this process, the adaptive immune system is highly activated, and many of the lymphocytes start to clonally expand; hence many cytokines are also released. So, any attempt to harness this cytokine storm and calm down the immune outrage is appreciated. While the battleground for the immune hyperactivation is the lung ambient of the infected patients, the only medical treatment for suppressing the hypercytokinemia is based on the immunosuppressor drugs that systemically dampen the immunity with many unavoidable side effects. Here, we applied the alternating electric field to suppress the expansion of the highly activated lymphocytes, and by reducing the number of the renewed cells, the produced cytokines were also decreased. Applying this method to the blood of the COVID-19 patients in vitro showed ∼33% reduction in the average concentration of the three main cytokines after 4 days of stimulation. This method could carefully be utilized to locally suppress the hyperactivated immune cells in the lung of the COVID-19 patients without any need for systemic suppression of the immune system by the chemical drugs.
Collapse
Affiliation(s)
- Hamed Abadijoo
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Center of Excellence, Thin Film and Nano Electronics Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Institute of Cancer, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Khayamian
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Center of Excellence, Thin Film and Nano Electronics Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Institute of Cancer, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Mohammad Ali Khayamian, , ; Mohammad Abdolahad, ,
| | - Mahsa Faramarzpour
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Center of Excellence, Thin Film and Nano Electronics Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Institute of Cancer, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Ghaderinia
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Center of Excellence, Thin Film and Nano Electronics Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Institute of Cancer, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Simaee
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Center of Excellence, Thin Film and Nano Electronics Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Institute of Cancer, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahriar Shalileh
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Center of Excellence, Thin Film and Nano Electronics Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Institute of Cancer, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mojtaba Yazdanparast
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Center of Excellence, Thin Film and Nano Electronics Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Institute of Cancer, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Ghabraie
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Center of Excellence, Thin Film and Nano Electronics Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Institute of Cancer, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Jalil Makarem
- UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Sarrami-Forooshani
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Mohammad Abdolahad
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Center of Excellence, Thin Film and Nano Electronics Lab, School of Electrical and Computer Engineering, University of Tehran, Tehran, Iran
- Institute of Cancer, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
- UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Mohammad Ali Khayamian, , ; Mohammad Abdolahad, ,
| |
Collapse
|
9
|
Cathepsin X Activity Does Not Affect NK-Target Cell Synapse but Is Rather Distributed to Cytotoxic Granules. Int J Mol Sci 2021; 22:ijms222413495. [PMID: 34948293 PMCID: PMC8707301 DOI: 10.3390/ijms222413495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022] Open
Abstract
Cathepsin X is a lysosomal peptidase that is involved in tumour progression and represents a potential target for therapeutic interventions. In addition, it regulates important functions of immune cells and is implicated in the modulation of tumour cell–immune cell crosstalk. Selective cathepsin X inhibitors have been proposed as prospective antitumour agents to prevent cancer progression; however, their impact on the antitumour immune response has been overlooked. Previous studies indicate that the migration and adhesion of T cells and dendritic cells are affected by diminished cathepsin X activity. Meanwhile, the influence of cathepsin X inhibition on natural killer (NK) cell function has not yet been explored. Here, we examined the localization patterns of cathepsin X and the role of its inhibitors on the cytotoxicity of cell line NK-92, which is used for adoptive cellular immunotherapy in cancer patients. NK-92 cells depend on lymphocyte function-associated antigen 1 (LFA-1) to form stable immunoconjugates with target cells, providing, in this way, optimal cytotoxicity. Since LFA-1 is a substrate for cathepsin X activity in other types of cells, we hypothesized that cathepsin X could disturb the formation of NK-92 immunoconjugates. Thus, we employed cathepsin X reversible and irreversible inhibitors and evaluated their effects on the NK-92 cell interactions with target cells and on the NK-92 cell cytotoxicity. We show that cathepsin X inhibition does not impair stable conjugate formation or the lytic activity of NK-92 cells. Similarly, the conjugate formation between Jurkat T cells and target cells was not affected by cathepsin X activity. Unlike in previous migration and adhesion studies on T cells, in NK-92 cells cathepsin X was not co-localized with LFA-1 at the plasma membrane but was, rather, redistributed to the cytotoxic granules and secreted during degranulation.
Collapse
|
10
|
Perišić Nanut M, Pečar Fonović U, Jakoš T, Kos J. The Role of Cysteine Peptidases in Hematopoietic Stem Cell Differentiation and Modulation of Immune System Function. Front Immunol 2021; 12:680279. [PMID: 34335582 PMCID: PMC8322073 DOI: 10.3389/fimmu.2021.680279] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/01/2021] [Indexed: 01/21/2023] Open
Abstract
Cysteine cathepsins are primarily involved in the degradation and recycling of proteins in endo-lysosomal compartments but are also gaining recognition as pivotal proteolytic contributors to various immune functions. Through their extracellular proteolytic activities within the hematopoietic stem cell niche, they are involved in progenitor cell mobilization and differentiation. Cysteine cathepsins, such as cathepsins L and S contribute to antigen-induced adaptive immunity through major histocompatibility complex class II antigen presentation whereas cathepsin X regulates T-cell migration. By regulating toll-like receptor signaling and cytokine secretion cysteine cathepsins activate innate immune cells and affect their functional differentiation. Cathepsins C and H are expressed in cytotoxic T lymphocytes and natural killer cells and are involved in processing of pro-granzymes into proteolytically active forms. Cytoplasmic activities of cathepsins B and L contribute to the maintenance of homeostasis of the adaptive immune response by regulating cell death of T and B lymphocytes. The expression pattern, localization, and activity of cysteine cathepsins is tightly connected to their function in immune cells. Furthermore, cysteine cathepsins together with their endogenous inhibitors, serve as mediators in the interplay between cancer and immune cells that results in immune cell anergy. The aim of the present article is to review the mechanisms of dysregulation of cysteine cathepsins and their inhibitors in relation to immune dysfunction to address new possibilities for regulation of their function.
Collapse
Affiliation(s)
| | | | - Tanja Jakoš
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Janko Kos
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
11
|
Liu YJ, Zhang T, Cheng D, Yang J, Chen S, Wang X, Li X, Duan D, Lou H, Zhu L, Luo J, Ho MS, Wang XD, Duan S. Late endosomes promote microglia migration via cytosolic translocation of immature protease cathD. SCIENCE ADVANCES 2020; 6:6/50/eaba5783. [PMID: 33298434 PMCID: PMC7725477 DOI: 10.1126/sciadv.aba5783] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 10/26/2020] [Indexed: 06/12/2023]
Abstract
Organelle transport requires dynamic cytoskeleton remodeling, but whether cytoskeletal dynamics are, in turn, regulated by organelles remains elusive. Here, we demonstrate that late endosomes, a type of prelysosomal organelles, facilitate actin-cytoskeleton remodeling via cytosolic translocation of immature protease cathepsin D (cathD) during microglia migration. After cytosolic translocation, late endosome-derived cathD juxtaposes actin filaments at the leading edge of lamellipodia. Suppressing cathD expression or blocking its cytosolic translocation impairs the maintenance but not the initiation of lamellipodial extension. Moreover, immature cathD balances the activity of the actin-severing protein cofilin to maintain globular-actin (G-actin) monomer pool for local actin recycling. Our study identifies cathD as a key lysosomal molecule that unconventionally contributes to actin cytoskeleton remodeling via cytosolic translocation during adenosine triphosphate-evoked microglia migration.
Collapse
Affiliation(s)
- Yi-Jun Liu
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Ting Zhang
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Daxiao Cheng
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Junhua Yang
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Sicong Chen
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xingyue Wang
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xia Li
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Duo Duan
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Huifang Lou
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liya Zhu
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jianhong Luo
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Margaret S Ho
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Xiao-Dong Wang
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China.
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Shumin Duan
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China.
- Mental Health Center, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Dolenc I, Štefe I, Turk D, Taler-Verčič A, Turk B, Turk V, Stoka V. Human cathepsin X/Z is a biologically active homodimer. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1869:140567. [PMID: 33227497 DOI: 10.1016/j.bbapap.2020.140567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022]
Abstract
Human cathepsin X belongs to the cathepsin family of 11 lysosomal cysteine proteases. We expressed recombinant procathepsin X in Pichia pastoris in vitro and cleaved it into its active mature form using aspartic cathepsin E. We found, using size exclusion chromatography, X-ray crystallography, and small-angle X-ray scattering, that cathepsin X is a biologically active homodimer with a molecular weight of ~53 kDa. The novel finding that cathepsin X is a dimeric protein opens new horizons in the understanding of its function and the underlying pathophysiological mechanisms of various diseases including neurodegenerative disorders in humans.
Collapse
Affiliation(s)
- Iztok Dolenc
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | - Ivica Štefe
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Dušan Turk
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Ajda Taler-Verčič
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Vito Turk
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | - Veronika Stoka
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
13
|
Fuchs N, Meta M, Schuppan D, Nuhn L, Schirmeister T. Novel Opportunities for Cathepsin S Inhibitors in Cancer Immunotherapy by Nanocarrier-Mediated Delivery. Cells 2020; 9:E2021. [PMID: 32887380 PMCID: PMC7565055 DOI: 10.3390/cells9092021] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022] Open
Abstract
Cathepsin S (CatS) is a secreted cysteine protease that cleaves certain extracellular matrix proteins, regulates antigen presentation in antigen-presenting cells (APC), and promotes M2-type macrophage and dendritic cell polarization. CatS is overexpressed in many solid cancers, and overall, it appears to promote an immune-suppressive and tumor-promoting microenvironment. While most data suggest that CatS inhibition or knockdown promotes anti-cancer immunity, cell-specific inhibition, especially in myeloid cells, appears to be important for therapeutic efficacy. This makes the design of CatS selective inhibitors and their targeting to tumor-associated M2-type macrophages (TAM) and DC an attractive therapeutic strategy compared to the use of non-selective immunosuppressive compounds or untargeted approaches. The selective inhibition of CatS can be achieved through optimized small molecule inhibitors that show good pharmacokinetic profiles and are orally bioavailable. The targeting of these inhibitors to TAM is now more feasible using nanocarriers that are functionalized for a directed delivery. This review discusses the role of CatS in the immunological tumor microenvironment and upcoming possibilities for a nanocarrier-mediated delivery of potent and selective CatS inhibitors to TAM and related APC to promote anti-tumor immunity.
Collapse
Affiliation(s)
- Natalie Fuchs
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University of Mainz, Staudingerweg 5, D, 55128 Mainz, Germany; (N.F.); (M.M.)
| | - Mergim Meta
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University of Mainz, Staudingerweg 5, D, 55128 Mainz, Germany; (N.F.); (M.M.)
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 63, 55131 Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Lutz Nuhn
- Max Planck Institute for Polymer Research Ackermannweg 10, 55128 Mainz, Germany
| | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University of Mainz, Staudingerweg 5, D, 55128 Mainz, Germany; (N.F.); (M.M.)
| |
Collapse
|
14
|
Majc B, Sever T, Zarić M, Breznik B, Turk B, Lah TT. Epithelial-to-mesenchymal transition as the driver of changing carcinoma and glioblastoma microenvironment. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118782. [PMID: 32554164 DOI: 10.1016/j.bbamcr.2020.118782] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/05/2020] [Accepted: 06/07/2020] [Indexed: 02/07/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is an essential molecular and cellular process that is part of normal embryogenesis and wound healing, and also has a ubiquitous role in various types of carcinoma and glioblastoma. EMT is activated and regulated by specific microenvironmental endogenous triggers and a complex network of signalling pathways. These mostly include epigenetic events that affect protein translation-controlling factors and proteases, altogether orchestrated by the switching on and off of oncogenes and tumour-suppressor genes in cancer cells. The hallmark of cancer-linked EMT is that the process is incomplete, as it is opposed by the reverse process of mesenchymal-to-epithelial transition, which results in a hybrid epithelial/mesenchymal phenotype that shows notable cell plasticity. This is a characteristic of cancer stem cells (CSCs), and it is of the utmost importance in their niche microenvironment, where it governs CSC migratory and invasive properties, thereby creating metastatic CSCs. These cells have high resistance to therapeutic treatments, in particular in glioblastoma.
Collapse
Affiliation(s)
- Bernarda Majc
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia
| | - Tilen Sever
- Jožef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia; Department of Biochemistry, Molecular and Structural Biology, Josef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia
| | - Miki Zarić
- Jožef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia; Department of Biochemistry, Molecular and Structural Biology, Josef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Boris Turk
- Department of Biochemistry, Molecular and Structural Biology, Josef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, Večna pot 113, 1000 Ljubljana, Slovenia; Institute of Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Bol'shaya Pirogovskaya Ulitsa, 19с1, Moscow 119146, Russia
| | - Tamara T Lah
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, Večna pot 113, 1000 Ljubljana, Slovenia.
| |
Collapse
|
15
|
Dheer D, Nicolas J, Shankar R. Cathepsin-sensitive nanoscale drug delivery systems for cancer therapy and other diseases. Adv Drug Deliv Rev 2019; 151-152:130-151. [PMID: 30690054 DOI: 10.1016/j.addr.2019.01.010] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 01/23/2019] [Indexed: 12/26/2022]
Abstract
Cathepsins are an important category of enzymes that have attracted great attention for the delivery of drugs to improve the therapeutic outcome of a broad range of nanoscale drug delivery systems. These proteases can be utilized for instance through actuation of polymer-drug conjugates (e.g., triggering the drug release) to bypass limitations of many drug candidates. A substantial amount of work has been witnessed in the design and the evaluation of Cathepsin-sensitive drug delivery systems, especially based on the tetra-peptide sequence (Gly-Phe-Leu-Gly, GFLG) which has been extensively used as a spacer that can be cleaved in the presence of Cathepsin B. This Review Article will give an in-depth overview of the design and the biological evaluation of Cathepsin-sensitive drug delivery systems and their application in different pathologies including cancer before discussing Cathepsin B-cleavable prodrugs under clinical trials.
Collapse
|
16
|
Choi KM, Joo MS, Cho DH, Han HJ, Kim MS, Cho MY, Jung SH, Kim DH, Park CI. Functional analysis and gene expression profiling of extracellular cathepsin Z in red sea bream, Pagrus major. FISH & SHELLFISH IMMUNOLOGY 2019; 93:208-215. [PMID: 31306760 DOI: 10.1016/j.fsi.2019.07.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/02/2019] [Accepted: 07/11/2019] [Indexed: 06/10/2023]
Abstract
Cathepsin Z (CTSZ) is a lysosomal cysteine protease that is known to be involved in the maintenance of homeostasis and the biological mechanisms of immune cells. In this study, we have confirmed the tissue specific expression of the cathepsin Z (PmCTSZ) gene in Pagrus major, and confirmed its biological function after producing recombinant protein using Escherichia coli (E. coli). Multiple sequence alignment analysis revealed that the active site of the cysteine proteases and three N-glycosylation sites of the deduced protein sequence were highly conserved among all of the organisms. Phylogenetic analysis revealed that PmCTSZ was included in the clusters of CTSZ and the cysteine proteases of other bony fish and is most closely related to Japanese flounder CTSZ. PmCTSZ was distributed in all of the tissues from healthy red sea bream that were used in the experiment and was most abundantly found in the spleen and gill. Analysis of mRNA expression after bacterial (Edwardsiella piscicida: E. piscicida and Streptococcus iniae: S. iniae) or viral (red seabream iridovirus: RSIV) challenge showed significant gene expression regulation in immune-related tissues, but they maintained relatively normal levels of expression. We produced recombinant PmCTSZ (rPmCTSZ) using an E. coli expression system and confirmed the biological function of extracellular rPmCTSZ in vitro. We found that bacterial proliferation was significantly inhibited by rPmCTSZ, and the leukocytes of red sea bream also induced apoptosis and viability reduction.
Collapse
Affiliation(s)
- Kwang-Min Choi
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Min-Soo Joo
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Dong-Hee Cho
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Hyun-Ja Han
- Pathology Research Division, National Institute of Fisheries Science, 408-1 Sirang-ri, Gijang-up, Gijang-gun, Busan, 46083, Republic of Korea
| | - Myoung Sug Kim
- Pathology Research Division, National Institute of Fisheries Science, 408-1 Sirang-ri, Gijang-up, Gijang-gun, Busan, 46083, Republic of Korea
| | - Mi Young Cho
- Pathology Research Division, National Institute of Fisheries Science, 408-1 Sirang-ri, Gijang-up, Gijang-gun, Busan, 46083, Republic of Korea
| | - Sung Hee Jung
- Pathology Research Division, National Institute of Fisheries Science, 408-1 Sirang-ri, Gijang-up, Gijang-gun, Busan, 46083, Republic of Korea
| | - Do-Hyung Kim
- Department of Aquatic Life Medicine, College of Fisheries Science, Pukyong National University, 45, Yongso-ro, Nam-Gu., Busan, Republic of Korea.
| | - Chan-Il Park
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea.
| |
Collapse
|
17
|
Jakoš T, Pišlar A, Jewett A, Kos J. Cysteine Cathepsins in Tumor-Associated Immune Cells. Front Immunol 2019; 10:2037. [PMID: 31555270 PMCID: PMC6724555 DOI: 10.3389/fimmu.2019.02037] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/12/2019] [Indexed: 12/23/2022] Open
Abstract
Cysteine cathepsins are key regulators of the innate and adaptive arms of the immune system. Their expression, activity, and subcellular localization are associated with the distinct development and differentiation stages of immune cells. They promote the activation of innate myeloid immune cells since they contribute to toll-like receptor signaling and to cytokine secretion. Furthermore, they control lysosomal biogenesis and autophagic flux, thus affecting innate immune cell survival and polarization. They also regulate bidirectional communication between the cell exterior and the cytoskeleton, thus influencing cell interactions, morphology, and motility. Importantly, cysteine cathepsins contribute to the priming of adaptive immune cells by controlling antigen presentation and are involved in cytotoxic granule mediated killing in cytotoxic T lymphocytes and natural killer cells. Cathepins'aberrant activity can be prevented by their endogenous inhibitors, cystatins. However, dysregulated proteolysis contributes significantly to tumor progression also by modulation of the antitumor immune response. Especially tumor-associated myeloid cells, such as tumor-associated macrophages and myeloid-derived suppressor cells, which are known for their tumor promoting and immunosuppressive functions, constitute the major source of excessive cysteine cathepsin activity in cancer. Since they are enriched in the tumor microenvironment, cysteine cathepsins represent exciting targets for development of new diagnostic and therapeutic moieties.
Collapse
Affiliation(s)
- Tanja Jakoš
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Anja Pišlar
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Anahid Jewett
- UCLA School of Dentistry and Medicine, Los Angeles, CA, United States
| | - Janko Kos
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.,Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| |
Collapse
|
18
|
Cathepsin Z as a novel potential biomarker for osteoporosis. Sci Rep 2019; 9:9752. [PMID: 31278293 PMCID: PMC6611782 DOI: 10.1038/s41598-019-46068-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/18/2019] [Indexed: 12/31/2022] Open
Abstract
Osteoporosis, one of the most prevalent chronic ageing-related bone diseases, often goes undetected until the first fragility fracture occurs, causing patient suffering and cost to health/social care services. Osteoporosis arises from imbalanced activity of osteoclasts and osteoblasts. Since these cell lineages produce the protease, cathepsin Z, the aim of this study was to investigate whether altered cathepsin Z mRNA levels are associated with osteoporosis in clinical samples. Cathepsin Z mRNA in human peripheral blood mononuclear cells was significantly differentially-expressed among non-osteoporotic controls, osteopenia and osteoporosis patients (p < 0.0001) and in female osteoporosis patients over the age of 50 years (P = 0.0016). Cathepsin Z mRNA level strongly correlated with low bone mineral density (BMD) (g/cm2), lumbar spine L2-L4 and femoral neck (T-scores) (P = 0.0149, 0.0002 and 0.0139, respectively). Importantly, cathepsin Z mRNA was significantly associated with fragility fracture in osteoporosis patients (P = 0.0018). The levels of cathepsin Z mRNA were not significantly higher in patients with chronic inflammatory disorders in these two groups compared to those without (P = 0.774 and 0.666, respectively). ROC analysis showed that cathepsin Z mRNA has strong diagnostic value for osteoporosis and osteoporotic fracture. The results show for the first time that cathepsin Z could be a future diagnostic biomarker for osteoporosis including female osteoporosis patients over the age of 50 years.
Collapse
|
19
|
Fibroblasts stimulate macrophage migration in interconnected extracellular matrices through tunnel formation and fiber alignment. Biomaterials 2019; 209:88-102. [PMID: 31030083 DOI: 10.1016/j.biomaterials.2019.03.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 02/07/2023]
Abstract
In vivo, macrophages and fibroblasts navigate through and remodel the three-dimensional (3D) extra-cellular matrix (ECM). The orientation of fibers, the porosity, and degree of cross-linking can change the interconnectivity of the ECM and affect cell migration. In turn, migrating cells can alter their microenvironment. To study the relationships between ECM interconnectivity and migration of cells, we assembled collagen hydrogels with dense (DCN) or with loosely interconnected networks (LCN). We find that in DCNs, RAW 264.7 macrophages in monocultures were virtually stationary. In DCN co-cultures, Balb/c 3T3 fibroblasts created tunnels that provided conduits for macrophage migration. In LCNs, fibroblasts aligned fibers up to a distance of 100 μm, which provided tracks for macrophages. Intra-cellular and extra-cellular fluorescent fragments of internalized and degraded collagen were detected inside both cell types as well as around their cell peripheries. Macrophages expressed higher levels of urokinase-type plasminogen activator receptor associated protein (uPARAP)/mannose receptor 1 (CD206) compared to α2β1 indicating that collagen internalization in these cells occurred primarily via integrin-independent mechanisms. Network remodeling indicated by higher Young's modulus was observed in fibroblast monocultures as a result of TGF-β secretion. This work unveils new roles for fibroblasts in forming tunnels in networked ECM to modulate macrophage migration.
Collapse
|
20
|
Abstract
Cathepsins (CTS) are mainly lysosomal acid hydrolases extensively involved in the prognosis of different diseases, and having a distinct role in tumor progression by regulating cell proliferation, autophagy, angiogenesis, invasion, and metastasis. As all these processes conjunctively lead to cancer progression, their site-specific regulation might be beneficial for cancer treatment. CTS regulate activation of the proteolytic cascade and protein turnover, while extracellular CTS is involved in promoting extracellular matrix degradation and angiogenesis, thereby stimulating invasion and metastasis. Despite cancer regulation, the involvement of CTS in cellular adaptation toward chemotherapy and radiotherapy augments their therapeutic potential. However, lysosomal permeabilization mediated cytosolic translocation of CTS induces programmed cell death. This complex behavior of CTS generates the need to discuss the different aspects of CTS associated with cancer regulation. In this review, we mainly focused on the significance of each cathepsin in cancer signaling and their targeting which would provide noteworthy information in the context of cancer biology and therapeutics.
Collapse
Affiliation(s)
- Tejinder Pal Khaket
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Dalseo-Gu, Daegu 704-701, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea.
| |
Collapse
|
21
|
Kos J, Nanut MP, Prunk M, Sabotič J, Dautović E, Jewett A. Cystatin F as a regulator of immune cell cytotoxicity. Cancer Immunol Immunother 2018; 67:1931-1938. [PMID: 29748898 PMCID: PMC11028163 DOI: 10.1007/s00262-018-2165-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 04/30/2018] [Indexed: 01/08/2023]
Abstract
Cysteine cathepsins are lysosomal peptidases involved in the regulation of innate and adaptive immune responses. Among the diverse processes, regulation of granule-dependent cytotoxicity of cytotoxic T-lymphocytes (CTLs) and natural killer (NK) cells during cancer progression has recently gained significant attention. The function of cysteine cathepsins is regulated by endogenous cysteine protease inhibitors-cystatins. Whereas other cystatins are generally cytosolic or extracellular proteins, cystatin F is present in endosomes and lysosomes and is thus able to regulate the activity of its target directly. It is delivered to endosomal/lysosomal vesicles as an inactive, disulphide-linked dimer. Proteolytic cleavage of its N-terminal part leads to the monomer, the only form that is a potent inhibitor of cathepsins C, H and L, involved in the activation of granzymes and perforin. In NK cells and CTLs the levels of active cathepsin C and of granzyme B are dependent on the concentration of monomeric, active cystatin F. In tumour microenvironment, inactive dimeric cystatin F can be secreted from tumour cells or immune cells and further taken up by the cytotoxic cells. Subsequent monomerization and inhibition of cysteine cathepsins within the endosomal/lysosomal vesicles impairs granzyme and perforin activation, and provokes cell anergy. Further, the glycosylation pattern has been shown to be important in controlling secretion of cystatin F from target cells, as well as internalization by cytotoxic cells and trafficking to endosomal/lysosomal vesicles. Cystatin F is therefore an important mediator used by bystander cells to reduce NK and T-cell cytotoxicity.
Collapse
Affiliation(s)
- Janko Kos
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia.
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia.
| | | | - Mateja Prunk
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Jerica Sabotič
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | | | - Anahid Jewett
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California-Los Angeles, Los Angeles, USA
| |
Collapse
|
22
|
Pišlar A, Jewett A, Kos J. Cysteine cathepsins: Their biological and molecular significance in cancer stem cells. Semin Cancer Biol 2018; 53:168-177. [DOI: 10.1016/j.semcancer.2018.07.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022]
|
23
|
Li W, Yu X, Ma X, Xie L, Xia Z, Liu L, Yu X, Wang J, Zhou H, Zhou X, Yang Y, Liu H. Deguelin attenuates non-small cell lung cancer cell metastasis through inhibiting the CtsZ/FAK signaling pathway. Cell Signal 2018; 50:131-141. [PMID: 30018008 DOI: 10.1016/j.cellsig.2018.07.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/09/2018] [Accepted: 07/05/2018] [Indexed: 01/13/2023]
Abstract
Lung cancer is the leading cause of cancer-related death among both men and women every year, mainly due to metastasis. Although natural compound deguelin has been reported to inhibited cell migration and invasion in various cancer cells, the details of this regulation progress remain to be fully elucidated. In this study, we investigated the underlying mechanism of deguelin-suppressed metastasis of non-small cell lung cancer (NSCLC) cells. Our results demonstrate that deguelin inhibits NSCLC cell migration, invasion, and metastasis both in vitro and in vivo. These inhibitory effects of deguelin were mediated by suppressing of Cathepsin Z (CtsZ) expression and interrupting the interaction of CtsZ with integrin β3. Moreover, deguelin inhibits the activation of CtsZ downstream FAK/Src/Paxillin signaling. Knockdown of CtsZ mimicked the effect of deguelin on NSCLC cells migration and invasion. Our study reveals that deguelin exerts its anti-metastatic effect both in vitro and in vivo is partly dependent on the suppression of CtsZ signaling. Deguelin would be a potential anti-metastasis agent against NSCLC.
Collapse
Affiliation(s)
- Wei Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Xinfang Yu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Xiaolong Ma
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Li Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhenkun Xia
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Lijun Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xinyou Yu
- Shangdong Lvdu Bio-Industry Co., Ltd., Binzhou, Shangdong 256600, China
| | - Jian Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Huiling Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xinmin Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Haidan Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
24
|
Tuo H, Shu F, She S, Yang M, Zou XQ, Huang J, Hu HD, Hu P, Ren H, Peng SF, Yang YX. Sorcin induces gastric cancer cell migration and invasion contributing to STAT3 activation. Oncotarget 2017; 8:104258-104271. [PMID: 29262638 PMCID: PMC5732804 DOI: 10.18632/oncotarget.22208] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is a globally occurring malignancy that is characterized by a high mortality rate due to a high tendency to metastasize and poor prognoses. Sorcin, as known as SRI, a soluble resistance-related calcium-binding protein, plays a significant role in multidrug resistance. Sorcin is related to the migration and invasion of cancer cells. However, the mechanism remains unclear. Here, we used immunohistochemistry to confirm that the expression of sorcin in cancer tissues is higher than that in the adjacent normal tissues. The wound healing and transwell results indicate that sorcin can induce migration and invasion of GC cells. To explore the role of sorcin in GC metastasis, isobaric tags for relative and absolutely quantitation (iTRAQ) were used to examine cells with and without sorcin knockdown to identify the differentially expressed proteins (DEPs). The results were evaluated via RT-PCR and western blot to confirm the ITRAQ data. Inhibition of sorcin expression can down- regulate the expression of CTSZ, MMP2, MMP9 and p-STAT3 followed by suppression of tumor growth and metastasis. Together, we concluded that sorcin has a oncogenic activity via inducing tumor growth and metastasis, leading to development of therapeutic treatments for GC.
Collapse
Affiliation(s)
- Huan Tuo
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Feng Shu
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Sha She
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Min Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiao Qin Zou
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Juan Huang
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Huai Dong Hu
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Institute for Viral Hepatitis of Chongqing Medical University, Chongqing 400016, China
| | - Peng Hu
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Institute for Viral Hepatitis of Chongqing Medical University, Chongqing 400016, China.,Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Hong Ren
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Institute for Viral Hepatitis of Chongqing Medical University, Chongqing 400016, China.,Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Shi Fang Peng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Hunan 410008, China.,Department of Health Management Center, Xiangya Hospital, Central South University, Hunan 410008, China
| | - Yi Xuan Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Institute for Viral Hepatitis of Chongqing Medical University, Chongqing 400016, China.,Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
25
|
Fonović UP, Mitrović A, Knez D, Jakoš T, Pišlar A, Brus B, Doljak B, Stojan J, Žakelj S, Trontelj J, Gobec S, Kos J. Identification and characterization of the novel reversible and selective cathepsin X inhibitors. Sci Rep 2017; 7:11459. [PMID: 28904354 PMCID: PMC5597618 DOI: 10.1038/s41598-017-11935-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/31/2017] [Indexed: 01/21/2023] Open
Abstract
Cathepsin X is a cysteine peptidase involved in the progression of cancer and neurodegenerative diseases. Targeting this enzyme with selective inhibitors opens a new possibility for intervention in several therapeutic areas. In this study triazole-based reversible and selective inhibitors of cathepsin X have been identified. Their selectivity and binding is enhanced when the 2,3-dihydrobenzo[b][1,4]dioxine moiety is present as the R1 substituent. Of a series of selected triazole-benzodioxine derivatives, compound 22 is the most potent inhibitor of cathepsin X carboxypeptidase activity (Ki = 2.45 ± 0.05 μM) with at least 100-fold greater selectivity in comparison to cathepsin B or other related cysteine peptidases. Compound 22 is not cytotoxic to prostate cancer cells PC-3 or pheochromocytoma PC-12 cells at concentrations up to 10 μM. It significantly inhibits the migration of tumor cells and increases the outgrowth of neurites, both processes being under the control of cathepsin X carboxypeptidase activity. Compound 22 and other characterized triazole-based inhibitors thus possess a great potential for further development resulting in several in vivo applications.
Collapse
Affiliation(s)
- Urša Pečar Fonović
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia.
| | - Ana Mitrović
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia
| | - Damijan Knez
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia
| | - Tanja Jakoš
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia
| | - Anja Pišlar
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia
| | - Boris Brus
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia
| | - Bojan Doljak
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia
| | - Jure Stojan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Simon Žakelj
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia
| | - Jurij Trontelj
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia
| | - Stanislav Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia
| | - Janko Kos
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana, Slovenia.,Department of Biotechnology, Jožef Stefan Institute, Jamova 39, Ljubljana, Slovenia
| |
Collapse
|
26
|
Cysteine cathepsins B and X promote epithelial-mesenchymal transition of tumor cells. Eur J Cell Biol 2017; 96:622-631. [DOI: 10.1016/j.ejcb.2017.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 01/28/2023] Open
|
27
|
Pečar Fonović U, Kos J. Cathepsin X Cleaves Profilin 1 C-Terminal Tyr139 and Influences Clathrin-Mediated Endocytosis. PLoS One 2015; 10:e0137217. [PMID: 26325675 PMCID: PMC4567178 DOI: 10.1371/journal.pone.0137217] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/18/2015] [Indexed: 11/19/2022] Open
Abstract
Cathepsin X, a cysteine carboxypeptidase, is upregulated in several types of cancer. Its molecular target in tumor cells is profilin 1, a known tumor suppressor and regulator of actin cytoskeleton dynamics. Cathepsin X cleaves off the C-terminal Tyr139 of profilin 1, affecting binding of poly-L-proline ligands and, consequently, tumor cell migration and invasion. Profilin 1 with mutations at the C-terminus, transiently expressed in prostate cancer cells PC-3, showed that Tyr139 is important for proper function of profilin 1 as a tumor suppressor. Cleaving off Tyr139 prevents the binding of clathrin, a poly-L-proline ligand involved in endocytosis. More profilin 1—clathrin complexes were present in PC-3 cells when cathepsin X was inhibited by its specific inhibitor AMS36 or silenced by siRNA. As a consequence, the endocytosis of FITC-labeled dextran and transferrin conjugate was significantly increased. These results constitute the first report of the regulation of clathrin-mediated endocytosis in tumor cells through proteolytic processing of profilin 1.
Collapse
Affiliation(s)
| | - Janko Kos
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|
28
|
Löser R, Pietzsch J. Cysteine cathepsins: their role in tumor progression and recent trends in the development of imaging probes. Front Chem 2015; 3:37. [PMID: 26157794 PMCID: PMC4477214 DOI: 10.3389/fchem.2015.00037] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/29/2015] [Indexed: 12/16/2022] Open
Abstract
Papain-like cysteine proteases bear an enormous potential as drug discovery targets for both infectious and systemic human diseases. The considerable progress in this field over the last two decades has also raised interest in the visualization of these enzymes in their native context, especially with regard to tumor imaging. After a short introduction to structure and general functions of human cysteine cathepsins, we highlight their importance for drug discovery and development and provide a critical update on the current state of knowledge toward their involvement in tumor progression, with a special emphasis on their role in therapy response. In accordance with a radiopharmaceutical point of view, the main focus of this review article will be the discussion of recently developed fluorescence and radiotracer-based imaging agents together with related molecular probes.
Collapse
Affiliation(s)
- Reik Löser
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf Dresden, Germany ; Department of Chemistry and Food Chemistry, Technische Universität Dresden Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf Dresden, Germany ; Department of Chemistry and Food Chemistry, Technische Universität Dresden Dresden, Germany
| |
Collapse
|
29
|
Intracellular signaling by cathepsin X: Molecular mechanisms and diagnostic and therapeutic opportunities in cancer. Semin Cancer Biol 2015; 31:76-83. [DOI: 10.1016/j.semcancer.2014.05.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/27/2014] [Accepted: 05/05/2014] [Indexed: 01/27/2023]
|
30
|
Pseudorabies virus triggers glycoprotein gE-mediated ERK1/2 activation and ERK1/2-dependent migratory behavior in T cells. J Virol 2014; 89:2149-56. [PMID: 25473050 DOI: 10.1128/jvi.02549-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED The interaction between viruses and immune cells of the host may lead to modulation of intracellular signaling pathways and to subsequent changes in cellular behavior that are of benefit for either virus or host. ERK1/2 (extracellular signal regulated kinase 1/2) signaling represents one of the key cellular signaling axes. Here, using wild-type and gE null virus, recombinant gE, and gE-transfected cells, we show that the gE glycoprotein of the porcine Varicellovirus pseudorabies virus (PRV) triggers ERK1/2 phosphorylation in Jurkat T cells and primary porcine T lymphocytes. PRV-induced ERK1/2 signaling resulted in homotypic T cell aggregation and increased motility of T lymphocytes. Our study reveals a new function of the gE glycoprotein of PRV and suggests that PRV, through activation of ERK1/2 signaling, has a substantial impact on T cell behavior. IMPORTANCE Herpesviruses are known to be highly successful in evading the immune system of their hosts, subverting signaling pathways of the host to their own advantage. The ERK1/2 signaling pathway, being involved in many cellular processes, represents a particularly attractive target for viral manipulation. Glycoprotein E (gE) is an important virulence factor of alphaherpesviruses, involved in viral spread. In this study, we show that gE has the previously uncharacterized ability to trigger ERK1/2 phosphorylation in T lymphocytes. We also show that virus-induced ERK1/2 signaling leads to increased migratory behavior of T cells and that migratory T cells can spread the infection to susceptible cells. In conclusion, our results point to a novel function for gE and suggest that virus-induced ERK1/2 activation may trigger PRV-carrying T lymphocytes to migrate and infect other cells susceptible to PRV replication.
Collapse
|
31
|
Akkari L, Gocheva V, Kester JC, Hunter KE, Quick ML, Sevenich L, Wang HW, Peters C, Tang LH, Klimstra DS, Reinheckel T, Joyce JA. Distinct functions of macrophage-derived and cancer cell-derived cathepsin Z combine to promote tumor malignancy via interactions with the extracellular matrix. Genes Dev 2014; 28:2134-50. [PMID: 25274726 PMCID: PMC4180975 DOI: 10.1101/gad.249599.114] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
During the process of tumor progression, cancer cells can produce the requisite growth- and invasion-promoting factors and can also rely on noncancerous cells in the tumor microenvironment as an alternative, cell-extrinsic source. However, whether the cellular source influences the function of such tumor-promoting factors remains an open question. Here, we examined the roles of the cathepsin Z (CtsZ) protease, which is provided by both cancer cells and macrophages in pancreatic neuroendocrine tumors in humans and mice. We found that tumor proliferation was exclusively regulated by cancer cell-intrinsic functions of CtsZ, whereas tumor invasion required contributions from both macrophages and cancer cells. Interestingly, several of the tumor-promoting functions of CtsZ were not dependent on its described catalytic activity but instead were mediated via the Arg-Gly-Asp (RGD) motif in the enzyme prodomain, which regulated interactions with integrins and the extracellular matrix. Together, these results underscore the complexity of interactions within the tumor microenvironment and indicate that cellular source can indeed impact molecular function.
Collapse
Affiliation(s)
- Leila Akkari
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York,10065, USA
| | - Vasilena Gocheva
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York,10065, USA
| | - Jemila C Kester
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York,10065, USA
| | - Karen E Hunter
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York,10065, USA
| | - Marsha L Quick
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York,10065, USA
| | - Lisa Sevenich
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York,10065, USA
| | - Hao-Wei Wang
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York,10065, USA
| | - Christoph Peters
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs University, D-79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, D-79104 Freiburg, Germany; German Cancer Consortium (DKTK), D-79104 Freiburg, Germany
| | - Laura H Tang
- Pathology Department, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | - David S Klimstra
- Pathology Department, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs University, D-79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, D-79104 Freiburg, Germany; German Cancer Consortium (DKTK), D-79104 Freiburg, Germany
| | - Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York,10065, USA;
| |
Collapse
|
32
|
Dysregulation of apoptotic signaling pathways by interaction of RPLP0 and cathepsin X/Z in gastric cancer. Pathol Res Pract 2014; 211:62-70. [PMID: 25433997 DOI: 10.1016/j.prp.2014.09.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/16/2014] [Accepted: 09/16/2014] [Indexed: 01/30/2023]
Abstract
Cathepsin X (CTSX, also called cathepsin Z/P) is a cysteine protease that still plays an unknown role in human cancer. It has been shown to bind cell surface heparin sulphate proteoglycans and integrins, indicating possible functions of CTSX in cellular adhesion, phagocytosis, and immune response. Our previous studies have shown an association between Helicobacter pylori (H. pylori) infection, a strong up-regulation of CTSX, and development of gastric cancer. In this study, yeast two-hybrid analysis revealed that RPLP0, a ribosomal protein P0, interacts with the human CTSX protein in gastric cancer. The CTSX/RPLP0 interaction was confirmed by co-immunoprecipitation assays. In addition, co-localization studies in cancer cell line N87 and gastric cancer tissue samples were performed. Laserscan microscopy revealed a shuttling of RPLP0 (and CTSX) from cytoplasm to the nucleus after CTSX knockdown. Down-regulation of RPLP0 resulted in G1 arrest of gastric cancer cells, whereas knockdown of CTSX led to G1 arrest and apoptosis after 48 h. Knockdown of both proteins caused increased apoptosis. RPLP0 deficiency could suppress cell growth and cell cycle progression by down-regulating CDK2. It was further demonstrated that RPLP0 affected p21 expression, but did not change the expression of Cyclin E. Down-regulation of both proteins at least through CDK2 suggests an anti-apoptotic effect on gastric cancer cells and opens up new possibilities for apoptotic immune modulation and gastric cancer therapy.
Collapse
|
33
|
Differences in the antigens of Helicobacter pylori strains influence on the innate immune response in the in vitro experiments. Mediators Inflamm 2014; 2014:287531. [PMID: 24616553 PMCID: PMC3927579 DOI: 10.1155/2014/287531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/07/2013] [Indexed: 02/06/2023] Open
Abstract
The immune response to Helicobacter pylori importantly determines the pathogenesis of infection as well as the success of antibiotic eradication of the bacteria. Strains of H. pylori were gathered from 14 patients who failed to eradicate H. pylori infection with antibiotics—therapy resistant strains (TRS)—or from patients who were able to eradicate H. pylori infection—therapy susceptible strains (TSS). The THP-1 cells were stimulated with H. pylori antigens. Cathepsin X expression on THP-1 cells and concentration of cytokines in the supernatant of THP-1 cells were measured with a flow cytometer.
TSS H. pylori antigens increased the proportion of cathepsin X positive cells compared to TRS H. pylori antigens. TSS H. pylori antigens induced higher secretion of IL-12 and IL-6 compared to TRS H. pylori antigens (P < 0.001; 0.02). Polymyxin B, a lipid A inhibitor, lowered the secretion of IL-12 and IL-6 in TRS and TSS.
We demonstrated a H. pylori strain-dependent cathepsin X and cytokine expression that can be associated with H. pylori resistance to eradication due to lack of effective immune response. Differences in lipid A of H. pylori might have an influence on the insufficient immune response, especially on phagocytosis.
Collapse
|
34
|
Huynh JL, Garg P, Thin TH, Yoo S, Dutta R, Trapp BD, Haroutunian V, Zhu J, Donovan MJ, Sharp AJ, Casaccia P. Epigenome-wide differences in pathology-free regions of multiple sclerosis-affected brains. Nat Neurosci 2014; 17:121-30. [PMID: 24270187 PMCID: PMC3934491 DOI: 10.1038/nn.3588] [Citation(s) in RCA: 188] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 10/29/2013] [Indexed: 02/08/2023]
Abstract
Using the Illumina 450K array and a stringent statistical analysis with age and gender correction, we report genome-wide differences in DNA methylation between pathology-free regions derived from human multiple sclerosis-affected and control brains. Differences were subtle, but widespread and reproducible in an independent validation cohort. The transcriptional consequences of differential DNA methylation were further defined by genome-wide RNA-sequencing analysis and validated in two independent cohorts. Genes regulating oligodendrocyte survival, such as BCL2L2 and NDRG1, were hypermethylated and expressed at lower levels in multiple sclerosis-affected brains than in controls, while genes related to proteolytic processing (for example, LGMN, CTSZ) were hypomethylated and expressed at higher levels. These results were not due to differences in cellular composition between multiple sclerosis and controls. Thus, epigenomic changes in genes affecting oligodendrocyte susceptibility to damage are detected in pathology-free areas of multiple sclerosis-affected brains.
Collapse
Affiliation(s)
- Jimmy L Huynh
- 1] Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA. [2] Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paras Garg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Tin Htwe Thin
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Seungyeul Yoo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael J Donovan
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrew J Sharp
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Patrizia Casaccia
- 1] Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA. [2] Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA. [3] Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
35
|
O'Grada CM, Morine MJ, Morris C, Ryan M, Dillon ET, Walsh M, Gibney ER, Brennan L, Gibney MJ, Roche HM. PBMCs reflect the immune component of the WAT transcriptome--implications as biomarkers of metabolic health in the postprandial state. Mol Nutr Food Res 2013; 58:808-20. [PMID: 24170299 DOI: 10.1002/mnfr.201300182] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 08/19/2013] [Accepted: 08/21/2013] [Indexed: 12/23/2022]
Abstract
SCOPE Food and nutrition studies often require accessing metabolically active tissues, including adipose tissue. This can involve invasive biopsy procedures that can be a limiting factor in study design. In contrast, peripheral blood mononuclear cells (PBMCs) are a population of circulating immune cells that are easily accessible through venipuncture. As transcriptomics is of growing importance in food and metabolism research, understanding the transcriptomic relationship between these tissue types can provide insight into the utility of PBMCs in this field. METHODS AND RESULTS We examine this relationship within eight subjects, in two postprandial states (following oral lipid tolerance test and oral glucose tolerance test). Multivariate analysis techniques were used to examine variation between tissues, samples, and subjects in order to define which genes havecommon/disparate expression profiles associated with highly defined metabolic phenotypes. We demonstrate global similarities in gene expression between PBMCs and white adipose tissue, irrespective of the metabolic challenge type. Closer examination of individual genes revealed this similarity to be strongest in pathways related to immune response/inflammation. Notably, the expression of metabolism-related nuclear receptors, including PPARs, LXR, etc. was discordant between tissues CONCLUSION The PBMC transcriptome may therefore provide a unique insight into the inflammatory component of metabolic health, as opposed to directly reflecting the metabolic component of the adipose tissue transcriptome.
Collapse
Affiliation(s)
- Colm M O'Grada
- Nutrigenomics Research Group, UCD Conway Institute of Biomolecular and Biomedical Research, School of Public Health and Population Science, University College Dublin, Belfield, Dublin, Ireland; Institute of Food and Health, University College Dublin, Belfield, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Binder BJ, Simpson MJ. Quantifying spatial structure in experimental observations and agent-based simulations using pair-correlation functions. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2013; 88:022705. [PMID: 24032862 DOI: 10.1103/physreve.88.022705] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 05/15/2013] [Indexed: 06/02/2023]
Abstract
We define a pair-correlation function that can be used to characterize spatiotemporal patterning in experimental images and snapshots from discrete simulations. Unlike previous pair-correlation functions, the pair-correlation functions developed here depend on the location and size of objects. The pair-correlation function can be used to indicate complete spatial randomness, aggregation, or segregation over a range of length scales, and quantifies spatial structures such as the shape, size, and distribution of clusters. Comparing pair-correlation data for various experimental and simulation images illustrates their potential use as a summary statistic for calibrating discrete models of various physical processes.
Collapse
Affiliation(s)
- Benjamin J Binder
- School of Mathematical Sciences, University of Adelaide, South Australia 5005, Australia
| | | |
Collapse
|
37
|
Inhibition of cathepsin X enzyme influences the immune response of THP-1 cells and dendritic cells infected with Helicobacter pylori. Radiol Oncol 2013; 47:258-65. [PMID: 24133391 PMCID: PMC3794882 DOI: 10.2478/raon-2013-0043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 05/11/2013] [Indexed: 12/30/2022] Open
Abstract
Background The immune response to Helicobacter pylori importantly determines the outcome of infection as well as the success of eradication therapy. We demonstrate the role of a cysteine protease cathepsin X in the immune response to H. pylori infection. Materials and methods We analysed how the inhibition of cathepsin X influenced the immune response in experiments when THP-1 cells or dendritic cells isolated from patients were stimulated with 48 strains of H. pylori isolated from gastric biopsy samples of patients which had problems with the eradication of bacteria. Results The experiments, performed with the help of a flow cytometer, showed that the expression of Toll-like receptors (TLRs), especially TLR-4 molecules, on the membranes of THP-1 cells or dendritic cells was higher when we stimulated cells with H. pylori together with inhibitor of cathepsin X 2F12 compared to THP-1 cells or dendritic cells stimulated with H. pylori only, and also in comparison with negative control samples. We also demonstrated that when we inhibited the action of cathepsin X in THP-1 cells, the concentrations of pro-inflammatory cytokines were lower than when THP-1 cell were stimulated with H. pylori only. Conclusions We demonstrated that inhibition of cathepsin X influences the internalization of TLR-2 and TLR-4. TLR-2 and TLR-4 redistribution to intra-cytoplasmic compartments is hampered if cathepsin X is blocked. The beginning of a successful immune response against H. pylori in the case of inhibition of cathepsin X is delayed.
Collapse
|
38
|
Pečar Fonović U, Jevnikar Z, Rojnik M, Doljak B, Fonović M, Jamnik P, Kos J. Profilin 1 as a target for cathepsin X activity in tumor cells. PLoS One 2013; 8:e53918. [PMID: 23326535 PMCID: PMC3542269 DOI: 10.1371/journal.pone.0053918] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 12/04/2012] [Indexed: 12/14/2022] Open
Abstract
Cathepsin X has been reported to be a tumor promotion factor in various types of cancer; however, the molecular mechanisms linking its activity with malignant processes are not understood. Here we present profilin 1, a known tumor suppressor, as a target for cathepsin X carboxypeptidase activity in prostate cancer PC-3 cells. Profilin 1 co-localizes strongly with cathepsin X intracellularly in the perinuclear area as well as at the plasma membrane. Selective cleavage of C-terminal amino acids was demonstrated on a synthetic octapeptide representing the profilin C-terminal region, and on recombinant profilin 1. Further, intact profilin 1 binds its poly-L-proline ligand clathrin significantly better than it does the truncated one, as shown using cathepsin X specific inhibitor AMS-36 and immunoprecipitation of the profilin 1/clathrin complex. Moreover, the polymerization of actin, which depends also on the binding of poly-L-proline ligands to profilin 1, was promoted by AMS-36 treatment of cells and by siRNA cathepsin X silencing. Our results demonstrate that increased adhesion, migration and invasiveness of tumor cells depend on the inactivation of the tumor suppressive function of profilin 1 by cathepsin X. The latter is thus designated as a target for development of new antitumor strategies.
Collapse
Affiliation(s)
| | - Zala Jevnikar
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Matija Rojnik
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Bojan Doljak
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Marko Fonović
- Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute and Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Ljubljana, Slovenia
| | | | - Janko Kos
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
- Department of Biotechnology, Jozef Stefan Institute, Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|
39
|
Jevnikar Z, Mirković B, Fonović UP, Zidar N, Švajger U, Kos J. Three-dimensional invasion of macrophages is mediated by cysteine cathepsins in protrusive podosomes. Eur J Immunol 2012; 42:3429-41. [DOI: 10.1002/eji.201242610] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Revised: 08/08/2012] [Accepted: 09/24/2012] [Indexed: 01/13/2023]
Affiliation(s)
- Zala Jevnikar
- Faculty of Pharmacy; University of Ljubljana; Aškerčeva 7, SI-1000 Ljubljana Slovenia
| | - Bojana Mirković
- Faculty of Pharmacy; University of Ljubljana; Aškerčeva 7, SI-1000 Ljubljana Slovenia
| | - Urša Pečar Fonović
- Faculty of Pharmacy; University of Ljubljana; Aškerčeva 7, SI-1000 Ljubljana Slovenia
| | - Nace Zidar
- Faculty of Pharmacy; University of Ljubljana; Aškerčeva 7, SI-1000 Ljubljana Slovenia
| | - Urban Švajger
- Blood Transfusion Center of Slovenia; Ljubljana Slovenia
| | - Janko Kos
- Faculty of Pharmacy; University of Ljubljana; Aškerčeva 7, SI-1000 Ljubljana Slovenia
- Department of Biotechnology; Jožef Stefan Institute; Jamova 39, SI-1000 Ljubljana Slovenia
| |
Collapse
|
40
|
Microbial and fungal protease inhibitors--current and potential applications. Appl Microbiol Biotechnol 2012; 93:1351-75. [PMID: 22218770 PMCID: PMC7080157 DOI: 10.1007/s00253-011-3834-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Revised: 12/07/2011] [Accepted: 12/09/2011] [Indexed: 01/18/2023]
Abstract
Proteolytic enzymes play essential metabolic and regulatory functions in many biological processes and also offer a wide range of biotechnological applications. Because of their essential roles, their proteolytic activity needs to be tightly regulated. Therefore, small molecules and proteins that inhibit proteases can be versatile tools in the fields of medicine, agriculture and biotechnology. In medicine, protease inhibitors can be used as diagnostic or therapeutic agents for viral, bacterial, fungal and parasitic diseases as well as for treating cancer and immunological, neurodegenerative and cardiovascular diseases. They can be involved in crop protection against plant pathogens and herbivorous pests as well as against abiotic stress such as drought. Furthermore, protease inhibitors are indispensable in protein purification procedures to prevent undesired proteolysis during heterologous expression or protein extraction. They are also valuable tools for simple and effective purification of proteases, using affinity chromatography. Because there are such a large number and diversity of proteases in prokaryotes, yeasts, filamentous fungi and mushrooms, we can expect them to be a rich source of protease inhibitors as well.
Collapse
|
41
|
Cathepsin H indirectly regulates morphogenetic protein-4 (BMP-4) in various human cell lines. Radiol Oncol 2011; 45:259-66. [PMID: 22933963 PMCID: PMC3423750 DOI: 10.2478/v10019-011-0034-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 09/13/2011] [Indexed: 11/23/2022] Open
Abstract
Background Cathepsin H is a cysteine protease considered to play a major role in tumor progression, however, its precise function in tumorigenesis is unclear. Cathepsin H was recently proposed to be involved in processing of bone morphogenetic protein 4 (BMP-4) in mice. In order to clarify whether cathepsin H also regulates BMP-4 in humans, its impact on BMP-4 expression, processing and degradation was investigated in prostate cancer (PC-3), osteosarcoma (HOS) and pro-monocytic (U937) human cell lines. Materials and methods BMP-4 expression was founded to be regulated by cathepsin H using PCR array technology and confirmed by real time PCR. Immunoassays including Western blot and confocal microscopy were used to evaluate the influence of cathepsin H on BMP-4 processing. Results In contrast to HOS, the expression of BMP-4 mRNA in U937 and PC3 cells was significantly decreased by cathepsin H. The different regulation of BMP-4 synthesis could be associated with the absence of the mature 28 kDa cathepsin H form in HOS cells, where only the intermediate 30 kDa form was observed. No co-localization of BMP-4 and cathepsin H was observed in human cell lines and the multistep processing of BMP-4 was not altered in the presence of specific cathepsin H inhibitor. Isolated cathepsin H does not cleave mature recombinant BMP-4, neither with its amino- nor its endopeptidase activity. Conclusions Our results exclude direct proteolytic processing of BMP-4 by cathepsin H, however, they provide support for its involvement in the regulation of BMP-4 expression.
Collapse
|
42
|
Jevnikar Z, Obermajer N, Kos J. LFA-1 fine-tuning by cathepsin X. IUBMB Life 2011; 63:686-93. [PMID: 21796748 DOI: 10.1002/iub.505] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 04/20/2011] [Indexed: 01/14/2023]
Abstract
The adhesion molecule lymphocyte function-associated antigen (LFA)-1 plays a key role in immune surveillance and response. Its conformation is spatially and temporally regulated, enabling adhesion and deadhesion during T-cell migration. LFA-1 adhesion to its major ligand intercellular adhesion molecule 1 is controlled by adaptor proteins which bind the cytoplasmic tail of the β (2) subunit. Cathepsin X, a cysteine carboxypeptidase, promotes T-cell migration and morphological changes by cleaving the β (2) cytoplasmic tail of LFA-1. In this way, it modulates the affinity of LFA-1 for structural adaptors talin-1 and α-actinin-1 and enables the stepwise transition between intermediate and high-affinity conformations of LFA-1, an event that is necessary for effective T-cell function. Cathepsin X regulation that would allow precise modulation of LFA-1 affinity has a great potential for anti-LFA-1 therapy.
Collapse
Affiliation(s)
- Zala Jevnikar
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.
| | | | | |
Collapse
|
43
|
A proteomic and cellular analysis of uropods in the pathogen Entamoeba histolytica. PLoS Negl Trop Dis 2011; 5:e1002. [PMID: 21483708 PMCID: PMC3071361 DOI: 10.1371/journal.pntd.0001002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 03/07/2011] [Indexed: 11/19/2022] Open
Abstract
Exposure of Entamoeba histolytica to specific ligands induces cell polarization via the activation of signalling pathways and cytoskeletal elements. The process leads to formation of a protruding pseudopod at the front of the cell and a retracting uropod at the rear. In the present study, we show that the uropod forms during the exposure of trophozoites to serum isolated from humans suffering of amoebiasis. To investigate uropod assembly, we used LC-MS/MS technology to identify protein components in isolated uropod fractions. The galactose/N-acetylgalactosamine lectin, the immunodominant antigen M17 (which is specifically recognized by serum from amoeba-infected persons) and a few other cells adhesion-related molecules were primarily involved. Actin-rich cytoskeleton components, GTPases from the Rac and Rab families, filamin, α-actinin and a newly identified ezrin-moesin-radixin protein were the main factors found to potentially interact with capped receptors. A set of specific cysteine proteases and a serine protease were enriched in isolated uropod fractions. However, biological assays indicated that cysteine proteases are not involved in uropod formation in E. histolytica, a fact in contrast to the situation in human motile immune cells. The surface proteins identified here are testable biomarkers which may be either recognized by the immune system and/or released into the circulation during amoebiasis.
Collapse
|
44
|
Staun-Ram E, Miller A. Cathepsins (S and B) and their inhibitor Cystatin C in immune cells: modulation by interferon-β and role played in cell migration. J Neuroimmunol 2010; 232:200-6. [PMID: 21112096 DOI: 10.1016/j.jneuroim.2010.10.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 09/13/2010] [Accepted: 10/14/2010] [Indexed: 01/20/2023]
Abstract
We investigated the modulation of Cathepsin S, Cathepsin B and Cystatin C expression in immune cells by interferon (IFN)-β, and their role in cell migration. Cathepsin levels were increased in monocytic and T line cells upon activation. IFN-β abolished this increase of Cathepsin B in monocytes and of Cathepsin S in T cells, while increased Cystatin C. Neutralizing Cystatin C increased cell migration, while migration of monocytes and T cells was inhibited by IFN-β, by Cystatin C, and T cell migration was suppressed by Cathepsin S and B inhibitors. These findings support further studies in the importance of Cathepsins and Cystatins in immunomodulation.
Collapse
Affiliation(s)
- Elsebeth Staun-Ram
- Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
45
|
Staudt ND, Aicher WK, Kalbacher H, Stevanovic S, Carmona AK, Bogyo M, Klein G. Cathepsin X is secreted by human osteoblasts, digests CXCL-12 and impairs adhesion of hematopoietic stem and progenitor cells to osteoblasts. Haematologica 2010; 95:1452-60. [PMID: 20494937 DOI: 10.3324/haematol.2009.018671] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Hematopoietic stem cells are retained within discrete bone marrow niches through the effects of cell adhesion molecules and chemokine gradients. However, a small proportion of hematopoietic stem cells can also be found trafficking in the peripheral blood. During induced stem cell mobilization a proteolytic microenvironment is generated, but whether proteases are also involved in physiological trafficking of hematopoietic stem cells is not known. In the present study we examined the expression, secretion and function of the cysteine protease cathepsin X by cells of the human bone marrow. DESIGN AND METHODS Human osteoblasts, bone marrow stromal cells and hematopoietic stem and progenitor cells were analyzed for the secretion of cathepsin X by western blotting, active site labeling, immunofluorescence staining and activity assays. A possible involvement of cathepsin X in cell adhesion and CXCL-12-mediated cell migration was studied in functional assays. Matrix-assisted laser desorption and ionization time-of-flight (MALDI-TOF) analysis revealed the digestion mechanism of CXCL-12 by cathepsin X. RESULTS Osteoblasts and stromal cells secrete cathepsin X, whereas hematopoietic stem and progenitor cells do not. Using a cathepsin X-selective substrate, we detected the catalytic activity of cathepsin X in cell culture supernatants of osteoblasts. Activated cathepsin X is able to reduce cellular adhesive interactions between CD34(+) hematopoietic stem and progenitor cells and adherent osteoblasts. The chemokine CXCL-12, a highly potent chemoattractant for hematopoietic stem cells secreted by osteoblasts, is readily digested by cathepsin X. CONCLUSIONS The exo-peptidase cathepsin X has been identified as a new member of the group of CXCL-12-degrading enzymes secreted by non-hematopoietic bone marrow cells. Functional data indicate that cathepsin X can influence hematopoietic stem and progenitor cell trafficking in the bone marrow.
Collapse
Affiliation(s)
- Nicole D Staudt
- Center for Medical Research, Section for Transplantation Immunology and Immunohematology, Waldhörnlestrasse 22, 72072 Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Jevnikar Z, Obermajer N, Kos J. Cysteine protease-mediated cytoskeleton interactions with LFA-1 promote T-cell morphological changes. ACTA ACUST UNITED AC 2010; 66:1030-40. [PMID: 19670215 DOI: 10.1002/cm.20413] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
T cells migrate through restrictive barriers in a protease-independent, amoeboid fashion that is characterized by morphological cell polarization. The interaction of cysteine-dependent carboxypeptidase cathepsin X with beta(2) integrin LFA-1 (lymphocyte function associated antigen 1) induces T-cell morphological changes, displaying into a 3D extracellular matrix a cytoplasmic projection termed a uropod. In the present study we show that inhibition of cathepsin X and a cysteine-dependent endopeptidase, cathepsin L, markedly inhibits T-cell actin polymerization, shape polarization, and chemotaxis. We propose that cathepsin L promotes T-cell migration associated processes by activating procathepsin X in the endolysosomal vesicles near the cell membrane and at the peak of the uropod, where both proteases were colocalized. We show that active cathepsin X modifies the beta(2) cytoplasmic tail of LFA-1 in the uropod, promoting its high affinity conformation. We suggest that LFA-1 cleavage contributes to the conformational change in the cytoplasmic tail, promoting the binding of the cytoskeletal protein talin. This interaction is restricted to the uropod and results in the stabilization of this region, promoting LFA-1-mediated cell uropod elongation.
Collapse
Affiliation(s)
- Zala Jevnikar
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | | |
Collapse
|
47
|
Jevnikar Z, Obermajer N, Pecar-Fonović U, Karaoglanovic-Carmona A, Kos J. Cathepsin X cleaves the beta2 cytoplasmic tail of LFA-1 inducing the intermediate affinity form of LFA-1 and alpha-actinin-1 binding. Eur J Immunol 2010; 39:3217-27. [PMID: 19750481 DOI: 10.1002/eji.200939562] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The motility of T cells depends on the dynamic spatial regulation of integrin-mediated adhesion and de-adhesion. Cathepsin X, a cysteine protease, has been shown to regulate T-cell migration by interaction with lymphocyte function associated antigen-1 (LFA-1). LFA-1 adhesion to the ICAM-1 is controlled by the association of actin-binding proteins with the cytoplasmic tail of the beta(2) chain of LFA-1. Cleavage by cathepsin X of the amino acid residues S(769), E(768) and A(767) from the C-terminal of the beta(2) cytoplasmic tail of LFA-1 is shown to promote binding of the actin-binding protein alpha-actinin-1. Furthermore, cathepsin X overexpression reduced LFA-1 clustering and induced an intermediate affinity LFA-1 conformation that is known to associate with alpha-actinin-1. Increased levels of intermediate affinity LFA-1 resulted in augmented cell spreading due to reduced attachment of T cells to the ICAM-1-coated surface. Gradual cleavage of LFA-1 by cathepsin X enables the transition between intermediate and high affinity LFA-1, an event that is crucial for effective T-cell migration.
Collapse
Affiliation(s)
- Zala Jevnikar
- Faculty of Pharmacy, University of Ljubljana, Askerceva, Ljubljana, Slovenia.
| | | | | | | | | |
Collapse
|
48
|
Colbert JD, Matthews SP, Miller G, Watts C. Diverse regulatory roles for lysosomal proteases in the immune response. Eur J Immunol 2010; 39:2955-65. [PMID: 19637232 DOI: 10.1002/eji.200939650] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The innate and adaptive immune system utilise endocytic protease activity to promote functional immune responses. Cysteine and aspartic proteases (cathepsins) constitute a subset of endocytic proteases, the immune function of which has been described extensively. Although historically these studies have focused on their role in processes such as antigen presentation and zymogen processing within the endocytic compartment, recent discoveries have demonstrated a critical role for these proteases in other intracellular compartments, and within the extracellular milieu. It has also become clear that their pattern of expression and substrate specificities are more diverse than was first envisaged. Here, we discuss recent advances addressing the role of lysosomal proteases in various aspects of the immune response. We pay attention to reports demonstrating cathepsin activity outside of its canonical endosome/lysosome microenvironment.
Collapse
Affiliation(s)
- Jeff D Colbert
- Division of Cell biology & Immunology, College of Life Sciences, University of Dundee, Dundee, UK
| | | | | | | |
Collapse
|
49
|
Hailfinger S, Rebeaud F, Thome M. Adapter and enzymatic functions of proteases in T-cell activation. Immunol Rev 2009; 232:334-47. [DOI: 10.1111/j.1600-065x.2009.00830.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
50
|
Cathepsin X prevents an effective immune response against Helicobacter pylori infection. Eur J Cell Biol 2009; 88:461-71. [PMID: 19446361 DOI: 10.1016/j.ejcb.2009.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2009] [Revised: 03/16/2009] [Accepted: 03/31/2009] [Indexed: 01/24/2023] Open
Abstract
Cathepsin X, a cysteine protease, has been shown to regulate an immune response by activating beta-2 integrin receptors. In this study we demonstrate its role in regulating the immune response to infection with H. pylori. The level of cathepsin X was determined in THP-1 monocyte cells primed with H. pylori antigens isolated from subjects suffering from gastritis, who had either eradicated or not the disease after the antibiotic therapy. We show that the specific clinical outcome of H. pylori eradication therapy correlates strongly with the membrane expression of cathepsin X in stimulated THP-1 cells, being significantly higher after stimulation with H. pylori strains from those subjects who did not respond to antibiotic therapy. The same antigens elicit a more vigorous immune response, increased expression of MHC II, however trigger inadequate cytokine profile (IFN-gamma and IL-4) to eradicate the pathogen. We propose that cathepsin X mediated activation of beta-2 integrin receptor Mac-1 suppresses the stimulatory signal in the form of cytokines. Cathepsin X co-localizes on the membrane of THP-1 cells with Mac-1 integrin receptor and its inhibition increases homotypic aggregation and mononuclear cell proliferation, events that are associated with low Mac-1 activity. Our study highlights the diversity of the innate immune response to H. pylori antigens leading to either successful eradication of the infection or maintenance of chronic inflammation, revealing cathepsin X location and activity as a regulator of the effectiveness of H. pylori eradication.
Collapse
|