Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 81(1): 8, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38092995

ABSTRACT

Cystatin F, a cysteine peptidase inhibitor, is a potent modulator of NK cytotoxicity. By inhibiting granule-mediated cytotoxicity pathway, cystatin F induces formation of non-functional NK cell stage, called split-anergy. We show that N-glycosylation determines the localization and cellular function of cystatin F. Cystatin F mostly exhibited high-mannose glycosylation in U-937 cells, both high-mannose and complex glycosylation in NK-92 and primary NKs, and predominantly complex glycosylation in super-charged NKs. Manipulating N-glycosylation with kifunensine increased high-mannose glycosylation of cystatin F and lysosome localisation, which decreased cathepsin C activity and reduced NK cytotoxicity. Mannose-6-phosphate could significantly reduce the internalization of extracellular cystatin F. By comparing NK cells with different cytotoxic potentials, we found that high-mannose cystatin F was strongly associated with lysosomes and cathepsin C in NK-92 cell line. In contrast, in highly cytotoxic super-charged NKs, cystatin F with complex glycosylation was associated with the secretory pathway and less prone to inhibit cathepsin C. Modulating glycosylation to alter cystatin F localisation could increase the cytotoxicity of NK cells, thereby enhancing their therapeutic potential for treating cancer patients.


Subject(s)
Antineoplastic Agents , Cystatins , Humans , Glycosylation , Mannose , Cathepsin C/metabolism , Killer Cells, Natural/metabolism
3.
Biomater Sci ; 10(17): 4933-4948, 2022 Aug 24.
Article in English | MEDLINE | ID: mdl-35861487

ABSTRACT

Cell stimulation using piezoelectric polymers, which is known as piezostimulation, is an innovative approach for designing antimicrobial protection. As an antibiotic-free and inorganic nanoparticle-free approach, it uses physical stimuli to target bacterial cells in a non-specific manner, which may be of great importance, particularly in the context of avoiding resistant bacterial strains. In this study, we prepared fully organic piezoelectric biodegradable films composed of poly-L-lactide (PLLA) and demonstrated their antimicrobial effect on S. epidermidis as a model of Gram-positive and E. coli as a model of Gram-negative bacteria. The PLLA films were either smooth and fabricated using simple melt- drawing or nanotextured, as self-standing nanotubes formed using the template-assisted method. The morphological differences between nanotextured and smooth films resulted in a larger surface area and better surface contact in nanotextured films, together with improved structural properties and better crystallinity, which were the main reasons for their better piezoelectric properties, and consequently stronger bactericidal effect. The comparison between the nanotextured surfaces with and without piezoelectric nature excluded the main role of morphology and directly confirmed piezoelectricity as the main reason for the observed antimicrobial affect. We also confirmed that piezo-stimulation using the antibacterial nanotextured film could damage the bacterial membrane as the main mechanism of action, while the contribution of pH changes and ROS generation was negligible. More importantly, the effect was selective toward the bacterial membrane and the same damage was not observed in human red blood cells, making the therapeutic use of these films possible.


Subject(s)
Anti-Infective Agents , Polymers , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Escherichia coli , Humans , Polymers/chemistry , Polymers/pharmacology , Staphylococcus epidermidis
4.
Int J Mol Sci ; 23(9)2022 Apr 28.
Article in English | MEDLINE | ID: mdl-35563308

ABSTRACT

We introduce a new family of fungal protease inhibitors with ß-trefoil fold from the mushroom Coprinopsis cinerea, named cocaprins, which inhibit both cysteine and aspartic proteases. Two cocaprin-encoding genes are differentially expressed in fungal tissues. One is highly transcribed in vegetative mycelium and the other in the stipes of mature fruiting bodies. Cocaprins are small proteins (15 kDa) with acidic isoelectric points that form dimers. The three-dimensional structure of cocaprin 1 showed similarity to fungal ß-trefoil lectins. Cocaprins inhibit plant C1 family cysteine proteases with Ki in the micromolar range, but do not inhibit the C13 family protease legumain, which distinguishes them from mycocypins. Cocaprins also inhibit the aspartic protease pepsin with Ki in the low micromolar range. Mutagenesis revealed that the ß2-ß3 loop is involved in the inhibition of cysteine proteases and that the inhibitory reactive sites for aspartic and cysteine proteases are located at different positions on the protein. Their biological function is thought to be the regulation of endogenous proteolytic activities or in defense against fungal antagonists. Cocaprins are the first characterized aspartic protease inhibitors with ß-trefoil fold from fungi, and demonstrate the incredible plasticity of loop functionalization in fungal proteins with ß-trefoil fold.


Subject(s)
Agaricales , Aspartic Acid Proteases , Cysteine Proteases , Lotus , Agaricales/chemistry , Aspartic Acid Endopeptidases , Aspartic Acid Proteases/genetics , Cysteine , Cysteine Proteases/genetics , Lotus/metabolism , Peptide Hydrolases/metabolism , Protease Inhibitors/chemistry
5.
FASEB J ; 36(4): e22215, 2022 04.
Article in English | MEDLINE | ID: mdl-35224765

ABSTRACT

Clitocybe nebularis lectin (CNL) is a GalNAcß1-4GlcNAc-binding lectin that exhibits an antiproliferative effect exclusively on the Jurkat leukemic T cell line by provoking homotypic aggregation and dose-dependent cell death. Cell death of Jurkat cells exhibited typical features of early apoptosis, but lacked the activation of initiating and executing caspases. None of the features of CNL-induced cell death were effectively blocked with the pan-caspase inhibitor or different cysteine peptidase inhibitors. Furthermore, CNL binding induced Jurkat cells to release the endogenous damage-associated molecular pattern molecule high-mobility group box 1 (HMGB1). A plant lectin with similar glycan-binding specificity, Wisteria floribunda agglutinin (WFA) showed less selective toxicity and induced cell death in Jurkat, Tall-104, and Hut-87 cell lines. HMGB1 release was also detected when Jurkat cells were treated with WFA. We identified the CD45 and CD43 cell surface glycoproteins on Jurkat cells as the main targets for CNL binding. However, the blockade of CD45 phosphatase activity failed to block either CNL-induced homotypic agglutination or cell death. Overall, our results indicate that CNL triggers atypical cell death selectively on Jurkat cells, suggesting the potential applicability of CNL in novel strategies for treating and/or detecting acute T cell leukemia.


Subject(s)
Agaricales/physiology , Cell Death , Lectins/metabolism , Membrane Glycoproteins/metabolism , Plant Lectins/metabolism , Receptors, N-Acetylglucosamine/metabolism , Humans , Jurkat Cells
6.
FEBS Open Bio ; 12(4): 708-738, 2022 04.
Article in English | MEDLINE | ID: mdl-35067006

ABSTRACT

Lysosomal peptidases are hydrolytic enzymes capable of digesting waste proteins that are targeted to lysosomes via endocytosis and autophagy. Besides intracellular protein catabolism, they play more specific roles in several other cellular processes and pathologies, either within lysosomes, upon secretion into the cell cytoplasm or extracellular space, or bound to the plasma membrane. In cancer, lysosomal peptidases are generally associated with disease progression, as they participate in crucial processes leading to changes in cell morphology, signaling, migration, and invasion, and finally metastasis. However, they can also enhance the mechanisms resulting in cancer regression, such as apoptosis of tumor cells or antitumor immune responses. Lysosomal peptidases have also been identified as hallmarks of aging and neurodegeneration, playing roles in oxidative stress, mitochondrial dysfunction, abnormal intercellular communication, dysregulated trafficking, and the deposition of protein aggregates in neuronal cells. Furthermore, deficiencies in lysosomal peptidases may result in other pathological states, such as lysosomal storage disease. The aim of this review was to highlight the role of lysosomal peptidases in particular pathological processes of cancer and neurodegeneration and to address the potential of lysosomal peptidases in diagnosing and treating patients.


Subject(s)
Neoplasms , Peptide Hydrolases , Apoptosis/physiology , Humans , Lysosomes/metabolism , Neoplastic Processes , Peptide Hydrolases/metabolism
7.
Front Immunol ; 12: 680279, 2021.
Article in English | MEDLINE | ID: mdl-34335582

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.


Subject(s)
Cell Differentiation/immunology , Cysteine Proteases/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunomodulation , Animals , Cell Differentiation/genetics , Clonal Anergy/immunology , Cysteine Proteases/chemistry , Cysteine Proteases/genetics , Cysteine Proteinase Inhibitors/pharmacology , Humans , Immune Tolerance , Immunomodulation/drug effects , Immunosenescence/drug effects , Multigene Family , Organogenesis/genetics , Organogenesis/immunology , Structure-Activity Relationship , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
8.
Microb Biotechnol ; 14(5): 2227-2240, 2021 09.
Article in English | MEDLINE | ID: mdl-34347360

ABSTRACT

Development of targeted treatment for colorectal cancer is crucial to avoid side effects. To harness the possibilities offered by microbiome engineering, we prepared safe multifunctional cancer cell-targeting bacteria Lactococcus lactis. They displayed, on their surface, binding proteins for cancer-associated transmembrane receptors epithelial cell adhesion molecule (EpCAM) and human epidermal growth factor receptor 2 (HER2) and co-expressed an infrared fluorescent protein for imaging. Binding of engineered L. lactis to tumour antigens EpCAM and HER2 was confirmed and characterised in vitro using soluble receptors. The proof-of-principle of targeting was demonstrated on human cell lines HEK293, HT-29 and Caco-2 with fluorescent microscopy and flow cytometry. The highest L. lactis adhesion was seen for the HEK293 cells with the overexpressed tumour antigens, where colocalisation with their tumour antigens was seen for 39% and 67% of EpCAM-targeting and HER2-targeting bacteria, respectively. On the other hand, no binding was observed to HEK293 cells without tumour antigens, confirming the selectivity of the engineered L. lactis. Apart from cell targeting in static conditions, targeting ability of engineered L. lactis was also shown in conditions of constant flow of bacterial suspension over the HEK293 cells. Successful targeting by engineered L. lactis support the future use of these bacteria in biopharmaceutical delivery for the treatment of colorectal cancer.


Subject(s)
Colorectal Neoplasms , Lactococcus lactis , Antigens, Neoplasm/genetics , Caco-2 Cells , Carrier Proteins , Colorectal Neoplasms/therapy , HEK293 Cells , Humans , Lactococcus lactis/genetics
9.
Int J Mol Sci ; 22(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34445118

ABSTRACT

The existence of CD4+ cytotoxic T cells (CTLs) at relatively high levels under different pathological conditions in vivo suggests their role in protective and/or pathogenic immune functions. CD4+ CTLs utilize the fundamental cytotoxic effector mechanisms also utilized by CD8+ CTLs and natural killer cells. During long-term cultivation, CD4+ T cells were also shown to acquire cytotoxic functions. In this study, CD4+ human T-cell clones derived from activated peripheral blood lymphocytes of healthy young adults were examined for the expression of cytotoxic machinery components. Cystatin F is a protein inhibitor of cysteine cathepsins, synthesized by CD8+ CTLs and natural killer cells. Cystatin F affects the cytotoxic efficacy of these cells by inhibiting the major progranzyme convertases cathepsins C and H as well as cathepsin L, which is involved in perforin activation. Here, we show that human CD4+ T-cell clones express the cysteine cathepsins that are involved in the activation of granzymes and perforin. CD4+ T-cell clones contained both the inactive, dimeric form as well as the active, monomeric form of cystatin F. As in CD8+ CTLs, cysteine cathepsins C and H were the major targets of cystatin F in CD4+ T-cell clones. Furthermore, CD4+ T-cell clones expressed the active forms of perforin and granzymes A and B. The levels of the cystatin F decreased with time in culture concomitantly with an increase in the activities of granzymes A and B. Therefore, our results suggest that cystatin F plays a role in regulating CD4+ T cell cytotoxicity. Since cystatin F can be secreted and taken up by bystander cells, our results suggest that CD4+ CTLs may also be involved in regulating immune responses through cystatin F secretion.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cysteine/metabolism , Protease Inhibitors/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cathepsin C/metabolism , Cathepsin L/metabolism , Cell Line, Tumor , Clone Cells , Granzymes/metabolism , Humans , Jurkat Cells , Killer Cells, Natural/metabolism , Lymphocyte Activation/physiology , T-Lymphocytes, Cytotoxic/metabolism
10.
Cell Oncol (Dordr) ; 44(5): 1051-1063, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34189679

ABSTRACT

PURPOSE: Glioblastoma, the most aggressive type of brain cancer, is composed of heterogeneous populations of differentiated cells, cancer stem cells and immune cells. Cystatin F, an endogenous inhibitor of lysosomal cysteine peptidases, regulates the function of cytotoxic immune cells. The aim of this study was to determine which type of cells expresses cystatin F in glioblastoma and to determine the role of cystatin F during disease progression. METHODS: RT-qPCR and immunohistochemistry were used to determine cystatin F mRNA and protein levels in glioblastoma tissue samples. The internalization of cystatin F was analyzed by Western blotting. Enzyme kinetics, real time invasion and calcein release cytotoxicity assays were used to assess the role of internalized cystatin F. RESULTS: We found that cystatin F was not expressed in non-cancer brain tissues, but that its expression increased with glioma progression. In tumor tissues, extensive staining was observed in cancer stem-like cells and microglia/monocytes, which secrete cystatin F into their microenvironment. In trans activity of cystatin F was confirmed using an in vitro glioblastoma cell model. Internalized cystatin F affected cathepsin L activity in glioblastoma cells and decreased their invasiveness. In addition, we found that cystatin F decreased the susceptibility of glioblastoma cells to the cytotoxic activity of natural killer (NK) cells. CONCLUSIONS: Our data implicate cystatin F as a mediator of immune suppression in glioblastoma. Increased cystatin F mRNA and protein levels in immune, glioblastoma and glioblastoma stem-like cells or trans internalized cystatin F may have an impact on decreased susceptibility of glioblastoma cells to NK cytotoxicity.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Cystatins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Killer Cells, Natural/metabolism , Neoplastic Stem Cells/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Line, Tumor , Cells, Cultured , Cystatins/metabolism , Cytotoxicity, Immunologic/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunohistochemistry , Microglia/metabolism , Monocytes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
11.
Cancers (Basel) ; 12(12)2020 Dec 06.
Article in English | MEDLINE | ID: mdl-33291222

ABSTRACT

Cystatin F is a protein inhibitor of cysteine cathepsins, peptidases involved in the activation of the effector molecules of the perforin/granzyme pathway. Cystatin F was previously shown to regulate natural killer cell cytotoxicity. Here, we show that extracellular cystatin F has a role in regulating the killing efficiency of cytotoxic T lymphocytes (CTLs). Extracellular cystatin F was internalised into TALL-104 cells, a cytotoxic T cell line, and decreased their cathepsin C and H activity. Correspondingly, granzyme A and B activity was also decreased and, most importantly, the killing efficiency of TALL-104 cells as well as primary human CTLs was reduced. The N-terminally truncated form of cystatin F, which can directly inhibit cathepsin C (unlike the full-length form), was more effective than the full-length inhibitor. Furthermore, cystatin F decreased cathepsin L activity, which, however, did not affect perforin processing. Cystatin F derived from K-562 target cells could also decrease the cytotoxicity of TALL-104 cells. These results clearly show that, by inhibiting cysteine cathepsin proteolytic activity, extracellular cystatin F can decrease the cytotoxicity of CTLs and thus compromise their function.

12.
PLoS Pathog ; 16(11): e1009013, 2020 11.
Article in English | MEDLINE | ID: mdl-33137165

ABSTRACT

Over the last 2 decades, several coronaviruses (CoVs) have crossed the species barrier into humans, causing highly prevalent and severe respiratory diseases, often with fatal outcomes. CoVs are a large group of enveloped, single-stranded, positive-sense RNA viruses, which encode large replicase polyproteins that are processed by viral peptidases to generate the nonstructural proteins (Nsps) that mediate viral RNA synthesis. Papain-like peptidases (PLPs) and chymotrypsin-like cysteine 3C-like peptidase are essential for coronaviral replication and represent attractive antiviral drug targets. Furthermore, CoVs utilize the activation of their envelope spike glycoproteins by host cell peptidases to gain entry into cells. CoVs have evolved multiple strategies for spike protein activation, including the utilization of lysosomal cysteine cathepsins. In this review, viral and host peptidases involved in CoV cell entry and replication are discussed in depth, with an emphasis on papain-like cysteine cathepsins. Furthermore, important findings on cysteine peptidase inhibitors with regard to virus attenuation are highlighted as well as the potential of such inhibitors for future treatment strategies for CoV-related diseases.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/virology , Coronavirus/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Virus Internalization/drug effects , Animals , Coronavirus Infections/drug therapy , Humans , Virus Replication/drug effects
13.
Acta Chim Slov ; 66(1): 58-61, 2019 Feb.
Article in English | MEDLINE | ID: mdl-33855476

ABSTRACT

Cysteine cathepsins are peptidases with housekeeping functions that play different specific roles in different tissues. Endogenous peptidase inhibitors, such as cystatins and thyropins are the ultimate way of controlling their activity. It appears, however, that cathepsin X, a monocarboxypeptidase, whose overexpression is associated with several pathological processes, is not under the control of endogenous inhibitors. Inhibitors belonging to various groups inhibit other cathepsins tested, but none decrease the carboxypeptidase activity of cathepsin X. This absence of inhibitor control is another feature that distinguishes cathepsin X from other members of the cysteine peptidases.

14.
Appl Microbiol Biotechnol ; 102(23): 10103-10117, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30191288

ABSTRACT

Lactic acid bacteria (LAB) are attractive hosts for the expression of heterologous proteins and can be engineered to deliver therapeutic proteins or peptides to mucosal surfaces. The gastric stable pentadecapeptide BPC-157 is able to prevent and treat gastrointestinal inflammation by reducing the production of reactive oxygen species (ROS). In this study, we used LAB Lactococcus lactis as a vector to deliver BPC-157 by surface display and trypsin shedding or by secretion to the growth medium. Surface display of BPC-157 was achieved by fusing it with basic membrane protein A (BmpA) or with the peptidoglycan binding domain of AcmA and Usp45 secretion signal. While the expression of BmpA-fusion proteins was higher than that of AcmA/Usp45-fusion protein, the surface display ability of BPC-157 was approximately 14-fold higher with AcmA/Usp45-fusion protein. Release of BPC-157 from the bacterial surface or from isolated fusion proteins by trypsinization was demonstrated with anti-BPC-157 antibodies or by mass spectrometry. The concentration of BPC-157 delivered by surface display via AcmA/Usp45-fusion was 30 ng/ml. This increased to 117 ng/ml by Usp45 signal-mediated secretion, making the latter the most effective lactococcal delivery approach for BPC-157. Secreted BPC-157 significantly decreased ROS production in 149BR fibroblast cell model, suggesting its potential benefit in the treatment of intestinal inflammations. Additionally, a comparison of different modes of small peptide delivery by L. lactis, performed in the present study, will facilitate the future use of L. lactis as peptide delivery vehicle.


Subject(s)
Drug Delivery Systems , Lactococcus lactis , Peptide Fragments/administration & dosage , Proteins/administration & dosage , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Inflammatory Bowel Diseases/therapy , Microorganisms, Genetically-Modified , Oxidative Stress , Peptide Fragments/pharmacology , Plasmids , Protein Engineering , Proteins/pharmacology , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
15.
Semin Cancer Biol ; 53: 178-188, 2018 12.
Article in English | MEDLINE | ID: mdl-30081230

ABSTRACT

We have recently shown that natural killer (NK) cells select and differentiate cancer stem cells (CSCs)/undifferentiated tumors via secreted and membrane bound IFN-gamma (IFN-γ) and TNF-alpha (TNF-α), preventing tumor growth and inducing remodeling of the tumor microenvironment. Since many conventional therapeutic strategies, including chemotherapy and radiotherapy remain fairly unsuccessful in treating CSCs/poorly differentiated tumors, there has been an increasing interest in NK cell-targeted immunotherapy for the treatment of aggressive tumors. In our recent studies, we used humanized-BLT (hu-BLT) mouse model with transplanted human bone marrow, liver and thymus to demonstrate the efficacy of adoptive transfer of ex vivo expanded, super-charged NK cells in selection and differentiation of stem-like tumors within the context of a fully reconstituted human immune system. Furthermore, we have demonstrated that CSCs differentiated with split-anergized NK cells prior to implantation in hu-BLT mice were not able to grow or metastasize. However, when NK cell-mediated tumor differentiation was blocked by the addition of antibodies to IFN-γ and TNF-α, tumors grew and metastasized. In this review, we present current advances in NK cell expansion and therapeutic delivery, and discuss the utility of allogeneic super-charged NK cells in treatment of cancer patients. In addition, NK suppression occurs not only at the stage of overt cancer, but also at the pre-neoplastic stage. Therefore, due to the indispensable role of NK cells in targeting CSCs/undifferentiated tumors and their role in differentiation of the tumors, NK cells should be placed high in the armamentarium of tumor immunotherapy.


Subject(s)
Killer Cells, Natural/immunology , Mouth Neoplasms/immunology , Neoplastic Stem Cells/immunology , Pancreatic Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Humans , Immunotherapy , Mouth Neoplasms/pathology , Mouth Neoplasms/therapy , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Tumor Burden/immunology
16.
Eur J Cell Biol ; 97(7): 463-473, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30033148

ABSTRACT

Cystatin F is an inhibitor of cysteine peptidases expressed solely in immune cells. It is the only type II cystatin able to enter endosomal/lysosomal vesicles and to regulate directly the activity of intracellular cysteine cathepsins. Its expression in promonocytic U937 and promyeloblastic HL-60 cells is highly upregulated but, after differentiation with phorbol 12-myristate 13-acetate - PMA, its levels drop significantly. In contrast, the activities of intracellular cysteine cathepsins C, L and S are higher in differentiated cells than in non-differentiated ones due, presumably, to the lower inhibitory capacity of cystatin F. Using immunofluorescence confocal microscopy, proximity ligation assay and co-immunoprecipitation, cathepsins C, L and S were confirmed to be the main interacting partners of cystatin F in U937 and HL-60 cells. The promoter region of the cystatin F gene, CST7, contains a unique binding site for transcription factor C/EBP α, one of the main myeloid differentiation instructors. Using the chromatin immunoprecipitation assay, C/EBP α was shown to bind to CST7 gene in U937 cells. Following cell differentiation with PMA, the binding of C/EBP α was decreased significantly. The protein level of C/EBP α was also significantly lower in differentiated than in non-differentiated cells. It was shown that, during monocyte to macrophage differentiation, the endosomal/lysosomal proteolytic activity can be regulated by cystatin F whose expression is under the control of transcriptional factor C/EBP α.


Subject(s)
Biomarkers, Tumor/metabolism , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation , Cystatins/metabolism , Macrophages/cytology , Monocytes/cytology , Cathepsins/metabolism , Cell Extracts , HL-60 Cells , Humans , Macrophages/metabolism , Monocytes/metabolism , Protein Binding/drug effects , Tetradecanoylphorbol Acetate/pharmacology , U937 Cells
17.
Front Immunol ; 8: 1459, 2017.
Article in English | MEDLINE | ID: mdl-29180998

ABSTRACT

Cystatin F is a cysteine peptidase inhibitor which, unlike other cystatin family members, is targeted to endosomal/lysosomal compartments. It is synthesized as an inactive disulfide-linked dimer which is then converted to an active monomer by proteolytic cleavage of 15 N-terminal residues. Cystatin F has been suggested to regulate the cytotoxicity of natural killer (NK) cells by inhibiting the major granzyme convertases, cathepsins C and H. To test this hypothesis, we prepared variants of cystatin F and analyzed their uptake, subcellular trafficking, and peptidase inhibition, as well as their impact on the cytotoxicity of NK-92 cells and primary NK cells. The N-glycosylation pattern is responsible for the secretion, uptake, and subcellular sorting of cystatin F in HeLa and Hek293 cells, whereas the legumain binding site had no effect on these processes. Active, N-terminally truncated, monomeric cystatin F can also be internalized by recipient cells and targeted to endo/lysosomes, affecting also cells lacking the activating peptidase. Cystatin F mutants capable of cell internalization and trafficking through the endo/lysosomal pathway significantly decreased cathepsin C and H activities, both in situ, following transfection and in trans, using conditioned media. Further, incubation of IL-2 stimulated NK-92 and primary NK cells with full-length and N-terminally truncated cystatin F mutants led to suppression of their granule-mediated cytotoxicity. This effect was most significant with the N-terminally truncated mutants. These results suggest that cystatin F can be an important mediator within tumor microenvironment affecting the cytotoxicity of NK cells and consequently antitumor immune response.

18.
PLoS One ; 11(9): e0162625, 2016.
Article in English | MEDLINE | ID: mdl-27606705

ABSTRACT

Infections with shiga toxin-producing bacteria, like enterohemorrhagic Escherichia coli and Shigella dysenteriae, represent a serious medical problem. No specific and effective treatment is available for patients with these infections, creating a need for the development of new therapies. Recombinant lactic acid bacterium Lactococcus lactis was engineered to bind Shiga toxin by displaying novel designed albumin binding domains (ABD) against Shiga toxin 1 B subunit (Stx1B) on their surface. Functional recombinant Stx1B was produced in Escherichia coli and used as a target for selection of 17 different ABD variants (named S1B) from the ABD scaffold-derived high-complex combinatorial library in combination with a five-round ribosome display. Two most promising S1Bs (S1B22 and S1B26) were characterized into more details by ELISA, surface plasmon resonance and microscale thermophoresis. Addition of S1Bs changed the subcellular distribution of Stx1B, completely eliminating it from Golgi apparatus most likely by interfering with its retrograde transport. All ABD variants were successfully displayed on the surface of L. lactis by fusing to the Usp45 secretion signal and to the peptidoglycan-binding C terminus of AcmA. Binding of Stx1B by engineered lactococcal cells was confirmed using flow cytometry and whole cell ELISA. Lactic acid bacteria prepared in this study are potentially useful for the removal of Shiga toxin from human intestine.


Subject(s)
Albumins/metabolism , Lactococcus lactis/metabolism , Protein Subunits/metabolism , Recombination, Genetic/genetics , Shiga Toxin 1/chemistry , Shiga Toxin 1/metabolism , Cell Surface Display Techniques , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , HeLa Cells , Humans , Immobilized Proteins/metabolism , Protein Binding , Protein Domains , Protein Transport , Recombinant Proteins/metabolism , Ribosomes/metabolism , Sequence Homology, Amino Acid , Surface Plasmon Resonance
19.
Semin Cancer Biol ; 35: 168-79, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26255843

ABSTRACT

Lysosomal cysteine peptidases - cysteine cathepsins - are general intracellular protein-degrading enzymes that control also a variety of specific physiological processes. They can trigger irreversible events leading to signal transduction and activation of signaling pathways, resulting in cell survival and proliferation or cell death. In cancer cells, lysosomal cysteine peptidases are involved in multiple processes during malignant progression. Their translocation from the endosomal/lysosomal pathway to nucleus, cytoplasm, plasma membrane and extracellular space enables the activation and remodeling of a variety of tumor promoting proteins. Thus, lysosomal cysteine peptidases interfere with cytokine/chemokine signaling, regulate cell adhesion and migration and endocytosis, are involved in the antitumor immune response and apoptosis, and promote cell invasion, angiogenesis and metastasis. Further, lysosomal cysteine peptidases modify growth factors and receptors involved in tyrosine kinase dependent pathways such as MAPK, Akt and JNK, thus representing key signaling tools for the activation of tumor cell growth and proliferation.


Subject(s)
Cysteine Endopeptidases/metabolism , Lysosomes/metabolism , Neoplasms/metabolism , Signal Transduction , Animals , Cathepsins/genetics , Cathepsins/metabolism , Cell Death/genetics , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cysteine Endopeptidases/genetics , Disease Progression , Endosomes/metabolism , Humans , Neoplasms/etiology , Neoplasms/pathology , Prognosis
20.
Front Immunol ; 5: 616, 2014.
Article in English | MEDLINE | ID: mdl-25520721

ABSTRACT

Cysteine cathepsins are lysosomal peptidases involved at different levels in the processes of the innate and adaptive immune responses. Some, such as cathepsins B, L, and H are expressed constitutively in most immune cells. In cells of innate immunity they play a role in cell adhesion and phagocytosis. Other cysteine cathepsins are expressed more specifically. Cathepsin X promotes dendritic cell maturation, adhesion of macrophages, and migration of T cells. Cathepsin S is implicated in major histocompatibility complex class II antigen presentation, whereas cathepsin C, expressed in cytotoxic T lymphocytes and natural killer (NK) cells, is involved in processing pro-granzymes into proteolytically active forms, which trigger cell death in their target cells. The activity of cysteine cathepsins is controlled by endogenous cystatins, cysteine protease inhibitors. Of these, cystatin F is the only cystatin that is localized in endosomal/lysosomal vesicles. After proteolytic removal of its N-terminal peptide, cystatin F becomes a potent inhibitor of cathepsin C with the potential to regulate pro-granzyme processing and cell cytotoxicity. This review is focused on the role of cysteine cathepsins and their inhibitors in the molecular mechanisms leading to the cytotoxic activity of T lymphocytes and NK cells in order to address new possibilities for regulation of their function in pathological processes.

SELECTION OF CITATIONS
SEARCH DETAIL
...