Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Life Sci ; 323: 121713, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37088412

ABSTRACT

Organ fibrosis is a pathological process of fibroblast activation and excessive deposition of extracellular matrix after persistent tissue injury and therefore is a common endpoint of many organ pathologies. Multiple cellular types and soluble mediators, including chemokines, cytokines and non-peptidic factors, are implicated in fibrogenesis and the remodeling of tissue architecture. The molecular basis of the fibrotic process is complex and consists of closely intertwined signaling networks. Research has strived for a better understanding of these pathological mechanisms to potentially reveal novel therapeutic targets for fibrotic diseases. In light of new knowledge, the receptor for advanced glycation end products (RAGE) emerged as an important candidate for the regulation of a wide variety of cellular functions related to fibrosis, including inflammation, cell proliferation, apoptosis, and angiogenesis. RAGE is a pattern recognition receptor that binds a broad range of ligands such as advanced glycation end products, high mobility group box-1, S-100 calcium-binding protein and amyloid beta protein. Although the link between RAGE and fibrosis has been established, the exact mechanisms need be investigated in further studies. The aim of this review is to collect all available information about the intricate function of RAGE and its signaling cascades in the pathogenesis of fibrotic diseases within different organs. In addition, to the major ligands and signaling pathways, we discuss potential strategies for targeting RAGE in fibrosis. We emphasize the functional links between RAGE, inflammation and fibrosis that may guide further studies and the development of improved therapeutic drugs.


Subject(s)
Amyloid beta-Peptides , Glycation End Products, Advanced , Humans , Receptor for Advanced Glycation End Products/metabolism , Glycation End Products, Advanced/metabolism , Inflammation/metabolism , Fibrosis
2.
Int Immunopharmacol ; 113(Pt B): 109393, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36375323

ABSTRACT

Hepatocellular carcinoma (HCC) accounts for ∼90 % of all liver cancer cases, which was the third most common cause of cancer death worldwide in 2020. Glypican-3 (GPC3) is highly and specifically expressed in HCC, which makes it a promising therapeutic target. We discovered novel antibody sequences against GPC3 from a phage display library and ranked the candidates by their binding affinity and epitope bins. Candidates with single- to double-digit nanomolar affinity were selected and expressed in Fab format and linked to a deimmunized bacterial exotoxin moiety via an intein trans-splicing reaction. The resulting immunotoxins were evaluated for their in vitro binding specificity and affinity, cell surface binding on the HepG2 or Huh7, rate of internalization, and potency of cytotoxicity. The immunotoxin called GT5 exhibited strong antigen binding and cell surface binding, as well as high internalization efficiency. The molecule GT5 was further evaluated for cytotoxicity in HepG2 and Huh7 cell-based assay and assessed for its pharmacokinetics and antitumor activity in a murine tumor xenograft model. GT5 significantly inhibited tumor growth and showed stronger potency than the chemotherapeutic drug sorafenib. In conclusion, GT5, a novel GPC3 targeting immunotoxin, was shown to have a high affinity towards GPC3 and effectively inhibit hepatocellular tumor growth in vitro and in vivo, thus providing the basis for further development of GT5 immunotoxin as a novel therapeutic modality for the treatment of liver cancer.


Subject(s)
Carcinoma, Hepatocellular , Immunotoxins , Liver Neoplasms , Humans , Mice , Animals , Carcinoma, Hepatocellular/therapy , Glypicans/chemistry , Glypicans/metabolism , Immunotoxins/pharmacology , Immunotoxins/therapeutic use , Liver Neoplasms/therapy , Cell Surface Display Techniques
3.
Biomedicines ; 10(8)2022 Jul 30.
Article in English | MEDLINE | ID: mdl-36009390

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly aggressive subset of breast cancer with limited therapeutic options. However, its immune evasion mechanisms, characterized by the over-expression of the immune checkpoint molecules PD-L1 and CD47, can be targeted in order to facilitate cancer elimination by cells of innate and adaptive immunity. In this paper, we describe the design, preparation, and evaluation of three novel dual-targeting fusion proteins that were based on the structure frame of prototype IAB (innate and adaptive dependent bispecific fusion protein) and the "Orcutt-type IgG-scFv" molecular model. Three molecules with different spatial conformations were designed to improve antigen-antibody affinity by the addition of Ag-Ab binding sites from the variable region sequences of the anti-PD-L1 monoclonal antibody (mAb) atezolizumab and CV1, a high-affinity receptor of CD47. The results showed that the best-performing among the three proteins designed in this study was protein Pro3; its CV1 N-terminus and Fc domain C-terminus were not sterically hindered. Pro3 was better at boosting T cell proliferation and the engulfment of macrophages than the IAB prototype and, at the same time, retained a level of ADCC activity similar to that of IAB. Through improved design, the novel constructed dual-targeting immunomodulatory protein Pro3 was superior at activating the anti-tumor immune response and has thus shown potential for use in clinical applications.

4.
Int J Mol Sci ; 23(13)2022 Jun 27.
Article in English | MEDLINE | ID: mdl-35806136

ABSTRACT

Activating transcription factor 5 (ATF5) belongs to the activating transcription factor/cyclic adenosine monophosphate (cAMP) response element-binding protein family of basic region leucine zipper transcription factors. ATF5 plays an important role in cell stress regulation and is involved in cell differentiation and survival, as well as centrosome maintenance and development. Accumulating evidence demonstrates that ATF5 plays an oncogenic role in cancer by regulating gene expressions involved in tumorigenesis and tumor survival. Recent studies have indicated that ATF5 may also modify the gene expressions involved in other diseases. This review explores in detail the regulation of ATF5 expression and signaling pathways and elucidates the role of ATF5 in cancer biology. Furthermore, an overview of putative therapeutic strategies that can be used for restoring aberrant ATF5 activity in different cancer types is provided.


Subject(s)
Activating Transcription Factors , Neoplasms , Activating Transcription Factors/genetics , Activating Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Humans , Neoplasms/genetics
5.
Mol Neurobiol ; 59(4): 2258-2276, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35066760

ABSTRACT

Microglia are resident macrophages in the central nervous system that are involved in immune responses driven by Toll-like receptors (TLRs). Microglia-mediated inflammation can lead to central nervous system disorders, and more than one TLR might be involved in these pathological processes. The cysteine peptidase cathepsin X has been recognized as a pathogenic factor for inflammation-induced neurodegeneration. Here, we hypothesized that simultaneous TLR3 and TLR4 activation induces synergized microglia responses and that these phenotype changes affect cathepsin X expression and activity. Murine microglia BV2 cells and primary murine microglia were exposed to the TLR3 ligand polyinosinic-polycytidylic acid (poly(I:C)) and the TLR4 ligand lipopolysaccharide (LPS), individually and simultaneously. TLR3 and TLR4 co-activation resulted in increased inflammatory responses compared to individual TLR activation, where poly(I:C) and LPS induced distinct patterns of proinflammatory factors together with different patterns of cathepsin X expression and activity. TLR co-activation decreased intracellular cathepsin X activity and increased cathepsin X localization at the plasma membrane with concomitant increased extracellular cathepsin X protein levels and activity. Inhibition of cathepsin X in BV2 cells by AMS36, cathepsin X inhibitor, significantly reduced the poly(I:C)- and LPS-induced production of proinflammatory cytokines as well as apoptosis. Additionally, inhibiting the TLR3 and TLR4 common signaling pathway, PI3K, with LY294002 reduced the inflammatory responses of the poly(I:C)- and LPS-activated microglia and recovered cathepsin X activity. We here provide evidence that microglial cathepsin X strengthens microglia activation and leads to subsequent inflammation-induced neurodegeneration. As such, cathepsin X represents a therapeutic target for treating neurodegenerative diseases related to excess inflammation.


Subject(s)
Microglia , Toll-Like Receptor 3 , Animals , Cysteine/metabolism , Inflammation/metabolism , Ligands , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , Poly I-C/adverse effects , Poly I-C/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism
6.
Int J Mol Sci ; 22(24)2021 Dec 16.
Article in English | MEDLINE | ID: mdl-34948293

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.


Subject(s)
Cathepsins/pharmacology , Cytoplasmic Granules/drug effects , Cytotoxicity, Immunologic/drug effects , Killer Cells, Natural/drug effects , Synapses/drug effects , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Membrane/drug effects , Humans , Immunotherapy, Adoptive/methods , Jurkat Cells , K562 Cells , Neoplasms/drug therapy , T-Lymphocytes/drug effects
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.
Crit Rev Immunol ; 41(2): 77-99, 2021.
Article in English | MEDLINE | ID: mdl-34348003

ABSTRACT

Natural killer (NK) cells represent critical effectors of anti-tumor immune responses due to their ability to target tumor cells that escape recognition by the adaptive arm of the immune system. NK cell efficacy depends on multiple factors, including their propensity to infiltrate tumors, to reach activation threshold, and to differentiate into mature cytotoxic cells. The tumor microenvironment counteracts protective immunity by delivering anti-inflammatory signals, which stimulate the development of myeloid-derived suppressor cells (MDSC). MDSCs utilize numerous proximity-dependent and independent mechanisms to suppress functions of cytotoxic T lymphocytes and NK cells. Importantly, substantial part of their suppressive activity depends on peptidases. MDSC-derived peptidases incapacitate NK cells by shedding ligands for their activating receptors and by processing key cytokines involved in regulation of immune responses. Moreover, they are needed for sustaining the immunosuppressive loop through promotion of MDSC accumulation, expansion, and enhancement of their survival. Peptidases are at the forefront of cancer progression. However, their disparate roles in immune cells have only recently become appreciated in orchestration of the cancer immune responses. Studies that focused on elucidating the potential of peptidase inhibitors in regulation of the anti-tumor immune responses have led to renewed interest in clinical development of peptidase inhibitors. In parallel, they inspired the development of novel theranostics, that exploit increased activity of peptidases in infiltrating immune cells for targeted drug release or activation of imaging probes.


Subject(s)
Myeloid-Derived Suppressor Cells , Neoplasms , Humans , Killer Cells, Natural , Neoplasms/therapy , Peptide Hydrolases , Tumor Microenvironment
9.
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.

10.
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
11.
Cancer Immunol Immunother ; 69(9): 1869-1880, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32372139

ABSTRACT

Increased proteolytic activity of cysteine cathepsins has long been known to facilitate malignant progression, and it has also been associated with tumor-promoting roles of myeloid-derived suppressor cells (MDSCs). Consequently, cysteine cathepsins have gained much attention as potential targets for cancer therapies. However, cross-talk between tumor cells and MDSCs needs to be taken into account when studying the efficacy of cathepsin inhibitors as anti-cancer agents. Here, we demonstrate the potential of the MDA-MB-231 breast cancer cell line to generate functional MDSCs from CD14+ cells of healthy human donors. During this transition to MDSCs, the overall levels of cysteine cathepsins increased, with the largest responses for cathepsins L and X. We used small-molecule inhibitors of cathepsins L and X (i.e., CLIK-148, Z9, respectively) to investigate their functional impact on tumor cells and immune cells in this co-culture system. Interactions with peripheral blood mononuclear cells reduced MDA-MB-231 cell invasion, while inhibition of cathepsin X activity by Z9 restored invasion. Inhibition of cathepsin L activity using CLIK-148 resulted in significantly increased CD8+ cytotoxicity. Of note, inhibition of cathepsins L and X in separate immune or tumor cells did not promote these functional changes. Together, our findings underlie the importance of tumor cell-immune cell interactions in the evaluation of the anti-cancer potential of cysteine cathepsin inhibitors.


Subject(s)
Cathepsin L/metabolism , Cysteine/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Humans , Leukocytes, Mononuclear/metabolism , Neoplasm Invasiveness/pathology , Neoplasms/pathology , PC-3 Cells
12.
Cancer Immunol Immunother ; 69(2): 275-283, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31813053

ABSTRACT

Cathepsins are lysosomal peptidases involved in intracellular protein catabolism as well as in various other physiological and pathological processes. Several members of the family, most notably cathepsins B, S, K and L, are frequently overexpressed in cancer and have been associated with remodeling of the proteins of the extracellular matrix, a process leading to tumor cell migration, invasion and metastasis. In addition, lysosomal cathepsins play a role in innate and adaptive immunity, regulation of antigen presentation, Toll-like receptor signaling, cytokine secretion, apoptosis, autophagy, differentiation, migration and cytotoxicity. In cancer, the cells of innate immunity, such as myeloid cells, are often subverted to the regulatory immunosuppressive phenotype. Most studies indicate that lysosomal cathepsins reinforce the pro-tumoral activity of myeloid-derived suppressor cells and tumor-associated macrophages as well as of neutrophils. On the other hand, in cytotoxic natural killer cells, tumor cells suppress lysosomal peptidases in their activation of perforin and granzymes, thus diminishing their killing ability. With multifaceted actions, lysosomal peptidases constitute an important regulatory mechanism for fine-tuning the anti-tumor immune response.


Subject(s)
Immunity, Innate , Lysosomes/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Peptide Hydrolases/metabolism , Animals , Biomarkers , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunity , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Macrophages/immunology , Macrophages/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/pathology , Signal Transduction , Toll-Like Receptors/metabolism
13.
Front Immunol ; 10: 2037, 2019.
Article in English | MEDLINE | ID: mdl-31555270

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.


Subject(s)
Cysteine Proteases/metabolism , Immune System/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Autophagy , Cysteine Proteases/chemistry , Cysteine Proteinase Inhibitors/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytokines/metabolism , Cytotoxicity, Immunologic , Enzyme Activation , Humans , Immunomodulation/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/physiology , Neoplasms/pathology , Protein Transport , Toll-Like Receptors/metabolism , Tumor Microenvironment/immunology
14.
Sci Rep ; 7(1): 11459, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28904354

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.


Subject(s)
Cathepsin K/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Drug Discovery , Animals , Cathepsin K/chemistry , Cysteine Proteinase Inhibitors/chemistry , Drug Discovery/methods , Kinetics , Magnetic Resonance Spectroscopy , Molecular Structure , Neuronal Outgrowth/drug effects , PC12 Cells , Protein Binding , Rats , Small Molecule Libraries , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...