Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Angew Chem Int Ed Engl ; 63(3): e202314028, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38029352

ABSTRACT

The caseinolytic protease is a highly conserved serine protease, crucial to prokaryotic and eukaryotic protein homeostasis, and a promising antibacterial and anticancer drug target. Herein, we describe the potent cystargolides as the first natural ß-lactone inhibitors of the proteolytic core ClpP. Based on the discovery of two clpP genes next to the cystargolide biosynthetic gene cluster in Kitasatospora cystarginea, we explored ClpP as a potential cystargolide target. We show the inhibition of Staphylococcus aureus ClpP by cystargolide A and B by different biochemical methods in vitro. Synthesis of semisynthetic derivatives and probes with improved cell penetration allowed us to confirm ClpP as a specific target in S. aureus cells and to demonstrate the anti-virulence activity of this natural product class. Crystal structures show cystargolide A covalently bound to all 14 active sites of ClpP from S. aureus, Aquifex aeolicus, and Photorhabdus laumondii, and reveal the molecular mechanism of ClpP inhibition by ß-lactones, the predominant class of ClpP inhibitors.


Subject(s)
Dipeptides , Staphylococcus aureus , Staphylococcus aureus/metabolism , Catalytic Domain , Dipeptides/metabolism , Virulence , Endopeptidase Clp/metabolism
2.
J Biol Chem ; 300(1): 105507, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38029966

ABSTRACT

Cystargolides are natural products originally isolated from Kitasatospora cystarginea NRRL B16505 as inhibitors of the proteasome. They are composed of a dipeptide backbone linked to a ß-lactone warhead. Recently, we identified the cystargolide biosynthetic gene cluster, but systematic genetic analyses had not been carried out because of the lack of a heterologous expression system. Here, we report the discovery of a homologous cystargolide biosynthetic pathway in Streptomyces durhamensis NRRL-B3309 by genome mining. The gene cluster was cloned via transformation-associated recombination and heterologously expressed in Streptomyces coelicolor M512. We demonstrate that it contains all genes necessary for the production of cystargolide A and B. Single gene deletion experiments reveal that only five of the eight genes from the initially proposed gene cluster are essential for cystargolide synthesis. Additional insights into the cystargolide pathway could be obtained from in vitro assays with CysG and chemical complementation of the respective gene knockout. This could be further supported by the in vitro investigation of the CysG homolog BelI from the belactosin biosynthetic gene cluster. Thereby, we confirm that CysG and BelI catalyze a cryptic SAM-dependent transfer of a methyl group that is critical for the construction of the cystargolide and belactosin ß-lactone warheads.


Subject(s)
Dipeptides , Methyltransferases , Streptomycetaceae , Biosynthetic Pathways , Dipeptides/metabolism , Lactones/metabolism , Methyltransferases/chemistry , Methyltransferases/genetics , Methyltransferases/metabolism , Multigene Family , Streptomyces coelicolor/genetics , Streptomycetaceae/enzymology , Streptomycetaceae/genetics
3.
Methods Mol Biol ; 2017: 149-163, 2019.
Article in English | MEDLINE | ID: mdl-31197775

ABSTRACT

During cytokine- or chemotherapy-induced hematopoietic stem cell (HSC) mobilization, a highly proteolytic microenvironment can be observed in the bone marrow that has a strong influence on adhesive and chemotactic interactions of HSC with their niches. The increase of proteases during mobilization goes along with a decrease of endogenous protease inhibitors. Prominent members of the proteases involved in HSC mobilization belong to the families of matrix metalloproteinases and cathepsins, which are able to degrade chemokines/cytokines, extracellular matrix components, and membrane-bound adhesion receptors. To determine the functional activity of different proteolytic enzymes, zymographic analyses with different substrates and pH conditions can be employed. An involvement of cysteine cathepsins can be determined by the "active site labeling" technique using a modified inhibitor irreversibly binding to the active center of the enzymes. Intact or degraded chemokines and cytokines, which fall into the range between 1000 and 20,000 Da, can readily be detected by MALDI-TOF analysis. These three methods can help to detect proteolytic activities directly involved in the mobilization process.


Subject(s)
Cytokines/chemistry , Hematopoietic Stem Cells/cytology , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Catalytic Domain , Cathepsins/chemistry , Cathepsins/metabolism , Cell Line , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Humans , Matrix Metalloproteinases/chemistry , Matrix Metalloproteinases/metabolism , Proteolysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stem Cell Niche
4.
Cancer Res ; 73(13): 3902-12, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23633492

ABSTRACT

Recruitment of monocytes into sites of inflammation is essential in the immune response. In cancer, recruited monocytes promote invasion, metastasis, and possibly angiogenesis. LDL receptor-related protein (LRP1) is an endocytic and cell-signaling receptor that regulates cell migration. In this study, we isografted PanO2 pancreatic carcinoma cells into mice in which LRP1 was deleted in myeloid lineage cells. Recruitment of monocytes into orthotopic and subcutaneous tumors was significantly increased in these mice, compared with control mice. LRP1-deficient bone marrow-derived macrophages (BMDM) expressed higher levels of multiple chemokines, including, most prominently, macrophage inflammatory protein-1α/CCL3, which is known to amplify inflammation. Increased levels of CCL3 were detected in LRP1-deficient tumor-associated macrophages (TAM), isolated from PanO2 tumors, and in RAW 264.7 macrophage-like cells in which LRP1 was silenced. LRP1-deficient BMDMs migrated more rapidly than LRP1-expressing cells in vitro. The difference in migration was reversed by CCL3-neutralizing antibody, by CCR5-neutralizing antibody, and by inhibiting NF-κB with JSH-23. Inhibiting NF-κB reversed the increase in CCL3 expression associated with LRP1 gene silencing in RAW 264.7 cells. Tumors formed in mice with LRP1-deficient myeloid cells showed increased angiogenesis. Although VEGF mRNA expression was not increased in LRP1-deficient TAMs, at the single-cell level, the increase in TAM density in tumors with LRP1-deficient myeloid cells may have allowed these TAMs to contribute an increased amount of VEGF to the tumor microenvironment. Our results show that macrophage density in tumors is correlated with cancer angiogenesis in a novel model system. Myeloid cell LRP1 may be an important regulator of cancer progression.


Subject(s)
Monocytes/immunology , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Receptors, LDL/deficiency , Tumor Suppressor Proteins/deficiency , Animals , Cell Line , Cell Movement , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Gene Expression Regulation, Neoplastic , Humans , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice , Mice, Transgenic , Monocytes/metabolism , Myeloid Cells , Neoplasm Transplantation , Neoplasms/immunology , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Receptors, LDL/genetics , Receptors, LDL/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
5.
Stem Cells Dev ; 21(11): 1924-35, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22066471

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) are known to reside in specialized niches at the endosteum in the trabecular bone. Osteoblasts are the major cell type of the endosteal niche. It is well established that secreted proteases are involved in cytokine-induced mobilization processes that release stem cell from their niches. However, migratory processes such as the regular trafficking of HSPCs between their niches and the periphery are not fully understood. In the current study we analyzed whether osteoblast-secreted cysteine cathepsins are able to reduce the direct interaction of HSPCs with bone-forming osteoblasts. Isolated human osteoblasts were shown to secrete proteolytically active cysteine cathepsins, such as cathepsins B, K, L, and X. All of these cathepsins were able to digest, although with different efficacy, the chemokine CXCL12, which is known to be important for retaining HSPCs in their niches. Of the 4 identified cathepsins, only cathepsin X was able to reduce binding of HSPCs to osteoblasts. Interestingly, nonactivated pro-cathepsin X and mature cathepsin X did not interfere with HSPC-osteoblast interactions. Only pro-cathepsin X treated with dithiothreitol, which unfolds but does not lead to full maturation of cathepsin X, significantly reduced HSPC adhesion to osteoblasts. These observations argue for a role of the accessible cathepsin X prodomain in diminishing cell binding. Our findings strongly suggest that the cysteine cathepsins B, K, and L constitutively secreted by osteoblasts are part of the fine-tuned regulation of CXCL12 in the bone marrow, whereas pro-cathepsin X with its prodomain can affect HSPC trafficking in the niche.


Subject(s)
Cathepsins/chemistry , Chemokine CXCL12/chemistry , Osteoblasts/chemistry , Protein Precursors/chemistry , Antigens, CD34/chemistry , Cathepsin B/chemistry , Cathepsin B/pharmacology , Cathepsin K/chemistry , Cathepsin K/pharmacology , Cathepsins/pharmacology , Cell Adhesion/drug effects , Cell Movement , Cellular Microenvironment , Culture Media, Conditioned/chemistry , Dithiothreitol/pharmacology , Enzyme Activation , Enzyme Assays , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Osteoblasts/cytology , Osteoblasts/drug effects , Primary Cell Culture , Protein Precursors/pharmacology , Proteolysis , Recombinant Proteins/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors
6.
Haematologica ; 95(9): 1452-60, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20494937

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.


Subject(s)
Cathepsin Z/metabolism , Cell Adhesion , Chemokine CXCL12/metabolism , Hematopoietic Stem Cells/cytology , Osteoblasts/cytology , Bone Marrow , Cell Movement , Cells, Cultured , Hematopoietic Stem Cells/metabolism , Humans , Osteoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...