Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 419
Filter
1.
J Biol Chem ; 299(12): 105446, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37949230

ABSTRACT

Increasing evidence suggests that aberrant regulation of sortilin ectodomain shedding can contribute to amyloid-ß pathology and frontotemporal dementia, although the mechanism by which this occurs has not been elucidated. Here, we probed for novel binding partners of sortilin using multiple and complementary approaches and identified two proteins of the neuron-specific gene (NSG) family, NSG1 and NSG2, that physically interact and colocalize with sortilin. We show both NSG1 and NSG2 induce subcellular redistribution of sortilin to NSG1- and NSG2-enriched compartments. However, using cell surface biotinylation, we found only NSG1 reduced sortilin cell surface expression, which caused significant reductions in uptake of progranulin, a molecular determinant for frontotemporal dementia. In contrast, we demonstrate NSG2 has no effect on sortilin cell surface abundance or progranulin uptake, suggesting specificity for NSG1 in the regulation of sortilin cell surface expression. Using metalloproteinase inhibitors and A disintegrin and metalloproteinase 10 KO cells, we further show that NSG1-dependent reduction of cell surface sortilin occurred via proteolytic processing by A disintegrin and metalloproteinase 10 with a concomitant increase in shedding of sortilin ectodomain to the extracellular space. This represents a novel regulatory mechanism for sortilin ectodomain shedding that is regulated in a neuron-specific manner. Furthermore, this finding has implications for the development of strategies for brain-specific regulation of sortilin and possibly sortilin-driven pathologies.


Subject(s)
Adaptor Proteins, Vesicular Transport , Carrier Proteins , Metalloproteases , Nerve Tissue Proteins , Neurons , Adaptor Proteins, Vesicular Transport/metabolism , Biotinylation , Brain/cytology , Brain/metabolism , Brain/pathology , Carrier Proteins/metabolism , Disintegrins/deficiency , Disintegrins/genetics , Disintegrins/metabolism , Frontotemporal Dementia/metabolism , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Progranulins/metabolism , Protein Binding , Proteolysis , Cell Membrane/metabolism , Amyloid beta-Peptides/metabolism
2.
Biomolecules ; 11(10)2021 09 28.
Article in English | MEDLINE | ID: mdl-34680049

ABSTRACT

BACKGROUND: Acute myocarditis often progresses to heart failure because there is no effective, etiology-targeted therapy of this disease. Simvastatin has been shown to be cardioprotective by decreasing matrix metalloproteinases' (MMPs) activity. The study was designed to determine whether simvastatin inhibits MMPs activity, decreases the severity of inflammation and contractile dysfunction of the heart in experimental autoimmune myocarditis (EAM). METHODS: Simvastatin (3 or 30 mg/kg/day) was given to experimental rats with EAM by gastric gavage for 21 days. Then transthoracic echocardiography was performed, MMPs activity and troponin I level were determined and tissue samples were assessed under a light and transmission electron microscope. RESULTS: Hearts treated with simvastatin did not show left ventricular enlargement. As a result of EAM, there was an enhanced activation of MMP-9, which was significantly reduced in the high-dose simvastatin group compared to the low-dose group. It was accompanied by prevention of myofilaments degradation and reduction of severity of inflammation. CONCLUSIONS: The cardioprotective effects of simvastatin in the acute phase of EAM are, at least in part, due to its ability to decrease MMP-9 activity and subsequent decline in myofilaments degradation and suppression of inflammation. These effects were achieved in doses equivalent to therapeutic doses in humans.


Subject(s)
Inflammation/drug therapy , Metalloproteases/genetics , Myocarditis/drug therapy , Simvastatin/pharmacology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Cardiotonic Agents/pharmacology , Echocardiography , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Metalloproteases/antagonists & inhibitors , Models, Animal , Myocarditis/genetics , Myocarditis/immunology , Myocarditis/pathology , Rats , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/prevention & control
3.
J Biochem Mol Toxicol ; 35(10): e22886, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34392581

ABSTRACT

The RECK gene, a tumor suppressor gene, inhibits angiogenesis, invasion, and tumor metastasis. Epigenetic regulation of the RECK gene constitutes a potent approach to the molecular basis of liver malignancy. This study aims to evaluate the promoter methylation status of the RECK gene and its serum level in patients with hepatitis C virus (HCV)-related hepatocellular carcinoma (HCC) and the potential association of RECK gene methylation with clinical criteria of HCC. One hundred and fifty-five subjects were included (healthy control [55], chronic HCV patients [55], HCV-related HCC patients [45]). The methylation status of the RECK gene promoter and serum RECK level were investigated by methylation-specific PCR and enzyme-linked immunosorbent assay techniques, respectively. RECK gene promoter hypermethylation was recorded in 46.7% of HCC patients, and 10.9% of HCV patients, but not in control subjects (0%). It was related to RECK protein level, varices, edema, ascites, lymph node metastasis, vascular invasion, and the largest diameter of focal lesions. Meanwhile, it was not associated with focal lesion number nor distant metastasis of HCC. In conclusion, RECK gene promoter hypermethylation is linked to HCV genotype-4-related HCC. Moreover, different degrees of RECK gene promoter methylation are associated with serum RECK level, lymph node metastasis, and vascular invasion, which could prove its pathogenic role in hepatocarcinogenesis in chronic HCV-infected patients.


Subject(s)
Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/genetics , DNA Methylation/genetics , GPI-Linked Proteins/genetics , Hepacivirus/genetics , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/genetics , Liver Neoplasms/complications , Liver Neoplasms/genetics , Metalloproteases/antagonists & inhibitors , Adult , Aged , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/blood , Case-Control Studies , Epigenesis, Genetic , Female , GPI-Linked Proteins/blood , Genes, Tumor Suppressor , Genotype , Hepacivirus/immunology , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/virology , Humans , Liver Neoplasms/blood , Lymphatic Metastasis/genetics , Male , Middle Aged , Promoter Regions, Genetic/genetics
4.
Bioorg Med Chem Lett ; 49: 128290, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34311087

ABSTRACT

While the biochemistry of rhomboid proteases has been extensively studied since their discovery two decades ago, efforts to define the physiological roles of these enzymes are ongoing and would benefit from chemical probes that can be used to manipulate the functions of these proteins in their native settings. Here, we describe the use of activity-based protein profiling (ABPP) technology to conduct a targeted screen for small-molecule inhibitors of the mitochondrial rhomboid protease PARL, which plays a critical role in regulating mitophagy and cell death. We synthesized a series of succinimide-containing sulfonyl esters and sulfonamides and discovered that these compounds serve as inhibitors of PARL with the most potent sulfonamides having submicromolar affinity for the enzyme. A counterscreen against the bacterial rhomboid protease GlpG demonstrates that several of these compounds display selectivity for PARL over GlpG by as much as two orders of magnitude. Both the sulfonyl ester and sulfonamide scaffolds exhibit reversible binding and are able to engage PARL in mammalian cells. Collectively, our findings provide encouraging precedent for the development of PARL-selective inhibitors and establish N-[(arylsulfonyl)oxy]succinimides and N-arylsulfonylsuccinimides as new molecular scaffolds for inhibiting members of the rhomboid protease family.


Subject(s)
Benzenesulfonates/pharmacology , Metalloproteases/antagonists & inhibitors , Mitochondrial Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Succinimides/pharmacology , Sulfonamides/pharmacology , Benzenesulfonates/chemical synthesis , DNA-Binding Proteins/antagonists & inhibitors , Endopeptidases , Escherichia coli/enzymology , Escherichia coli Proteins/antagonists & inhibitors , HEK293 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Protease Inhibitors/chemical synthesis , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Succinimides/chemical synthesis , Sulfonamides/chemical synthesis
5.
Toxins (Basel) ; 13(4)2021 03 31.
Article in English | MEDLINE | ID: mdl-33807363

ABSTRACT

Snake envenomation can result in hemorrhage, local necrosis, swelling, and if not treated properly can lead to adverse systemic effects such as coagulopathy, nephrotoxicity, neurotoxicity, and cardiotoxicity, which can result in death. As such, snake venom metalloproteinases (SVMPs) and disintegrins are two toxic components that contribute to hemorrhage and interfere with the hemostatic system. Administration of a commercial antivenom is the common antidote to treat snake envenomation, but the high-cost, lack of efficacy, side effects, and limited availability, necessitates the development of new strategies and approaches for therapeutic treatments. Herein, we describe the neutralization ability of anti-disintegrin polyclonal antibody on the activities of isolated disintegrins, P-II/P-III SVMPs, and crude venoms. Our results show disintegrin activity on platelet aggregation in whole blood and the migration of the SK-Mel-28 cells that can be neutralized with anti-disintegrin polyclonal antibody. We characterized a SVMP and found that anti-disintegrin was also able to inhibit its activity in an in vitro proteolytic assay. Moreover, we found that anti-disintegrin could neutralize the proteolytic and hemorrhagic activities from crude Crotalus atrox venom. Our results suggest that anti-disintegrin polyclonal antibodies have the potential for a targeted approach to neutralize SVMPs in the treatment of snakebite envenomations.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antivenins/pharmacology , Crotalid Venoms/antagonists & inhibitors , Crotalus , Disintegrins/antagonists & inhibitors , Metalloproteases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Snake Bites/drug therapy , Allosteric Regulation , Animals , Antibody Specificity , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cross Reactions , Crotalid Venoms/enzymology , Crotalid Venoms/immunology , Disease Models, Animal , Disintegrins/immunology , Disintegrins/metabolism , Hemorrhage/enzymology , Hemorrhage/etiology , Hemorrhage/prevention & control , Humans , Metalloproteases/immunology , Metalloproteases/metabolism , Mice, Inbred BALB C , Platelet Aggregation/drug effects , Snake Bites/blood , Snake Bites/enzymology , Snake Bites/immunology
6.
J Am Soc Mass Spectrom ; 32(4): 1105-1115, 2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33725446

ABSTRACT

The snake venom gland is the place for the synthesis, storage, and secretion of a complex mixture of proteins and peptides, i.e., the venom. The morphology of the gland has been revealed by classical histology and microscopic studies. However, knowledge about the gland's cellular secretory and functional processes is still incomplete and has so far been neglected by the omics disciplines. We used autofocusing atmospheric-pressure matrix-assisted laser desorption/ionization (AP-SMALDI) mass spectrometry imaging (MSI) to investigate endogenous biomolecular distributions in the venom glands of the saw-scaled viper, Echis carinatus sochureki, employing different sample preparation methods. Fresh-freezing and formalin-fixation were tested for the gland to obtain intact tissue sections. Subsequently, MSI was conducted with 12 µm pixel resolution for both types of preparations, and the lateral distributions of the metabolites were identified. Experiments revealed that lipids belonging to the classes of PC, SM, PE, PS, PA, and TG are present in the venom gland. PC (32:0) and SM (36:1) were found to be specifically located in the areas where cells are present. The snake venom metalloprotease inhibitor pEKW (m/z 444.2233) was identified in the venom by top-down LC-MS/MS and localized by MALDI-MSI in the gland across secretory epithelial cells. The peptide can inhibit the venom's enzymatic activity during long-term storage within the venom gland. With a high degree of spectral similarities, we concluded that formalin-fixed tissue, in addition to its high ability to preserve tissue morphology, can be considered as an alternative method to fresh-frozen tissue in the case of lipid and peptide MS imaging in venom gland tissues.


Subject(s)
Exocrine Glands/ultrastructure , Hyperspectral Imaging/methods , Lipids/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Viper Venoms/analysis , Viperidae/anatomy & histology , Animals , Chromatography, Liquid/methods , Exocrine Glands/chemistry , Formaldehyde , Freezing , Metalloproteases/analysis , Metalloproteases/antagonists & inhibitors , Tandem Mass Spectrometry/methods , Tissue Fixation/methods , Viper Venoms/enzymology
8.
PLoS Negl Trop Dis ; 15(2): e0008596, 2021 02.
Article in English | MEDLINE | ID: mdl-33529194

ABSTRACT

Echis carinatus (EC) is known as saw-scaled viper and it is endemic to the Indian subcontinent. Envenoming by EC represents a major cause of snakebite mortality and morbidity in the Indian subcontinent. Zinc (Zn++) dependent snake venom metalloproteases (SVMPs) present in Echis carinatus venom (ECV) is well known to cause systemic hemorrhage and coagulopathy in experimental animals. An earlier report has shown that ECV activates neutrophils and releases neutrophil extracellular traps (NETs) that blocks blood vessels leading to severe tissue necrosis. However, the direct involvement of SVMPs in the release of NETs is not clear. Here, we investigated the direct involvement of EC SVMPs in observed pathological symptoms in a preclinical setup using specific Zn++ metal chelator, Tetraethyl thiuram disulfide (TTD)/disulfiram. TTD potently antagonizes the activity of SVMPs-mediated ECM protein degradation in vitro and skin hemorrhage in mice. In addition, TTD protected mice from ECV-induced footpad tissue necrosis by reduced expression of citrullinated H3 (citH3) and myeloperoxidase (MPO) in footpad tissue. TTD also neutralized ECV-induced systemic hemorrhage and conferred protection against lethality in mice. Moreover, TTD inhibited ECV-induced NETosis in human neutrophils and decreased the expression of peptidyl arginine deiminase (PAD) 4, citH3, MPO, and p-ERK. Further, we demonstrated that ECV-induced NETosis and tissue necrosis are mediated via PAR-1-ERK axis. Overall, our results provide an insight into SVMPs-induced toxicities and the promising protective efficacy of TTD can be extrapolated to treat severe tissue necrosis complementing anti-snake venom (ASV).


Subject(s)
Disulfiram/pharmacology , Metalloproteases/antagonists & inhibitors , Neutrophils/drug effects , Snake Bites/physiopathology , Viper Venoms/metabolism , Viperidae/physiology , Animals , Antivenins/therapeutic use , Extracellular Traps/drug effects , Female , Hemorrhage/prevention & control , Humans , Metalloproteases/toxicity , Mice , Necrosis , Snake Bites/drug therapy , Viper Venoms/toxicity
9.
Int J Biol Macromol ; 169: 583-596, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33385454

ABSTRACT

Protealysin is a Serratia proteamaculans metalloproteinase of the M4 peptidase family and the prototype of a large group of protealysin-like proteases (PLPs). PLPs are likely involved in bacterial interaction with plants and animals as well as in bacterial pathogenesis. We demonstrated that the PLP genes in bacteria colocalize with the genes of putative conserved proteins. In S. proteamaculans, these two genes form a bicistronic operon. The putative S. proteamaculans protein that we called emfourin (M4in) was expressed in Escherichia coli and characterized. M4in forms a complex with protealysin with a 1:1 stoichiometry and is a potent slow-binding competitive inhibitor of protealysin (Ki = 52 ± 14 pM); besides, M4in is not secreted from S. proteamaculans constitutively. A comparison of amino acid sequences of M4in and its homologs with those of known inhibitors suggests that M4in is the prototype of a new family of protein inhibitors of proteases.


Subject(s)
Metalloproteases/antagonists & inhibitors , Metalloproteases/genetics , Serratia/enzymology , Serratia/genetics , Amino Acid Sequence , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli/genetics , Metalloproteases/chemistry , Metalloproteases/metabolism , Operon/genetics , Peptide Hydrolases/metabolism , Serratia/metabolism
10.
ChemMedChem ; 16(6): 976-988, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33369214

ABSTRACT

Astacin metalloproteinases, in particular meprins α and ß, as well as ovastacin, are emerging drug targets. Drug-discovery efforts have led to the development of the first potent and selective inhibitors in the last few years. However, the most recent compounds are based on a highly flexible tertiary amine scaffold that could cause metabolic liabilities or decreased potency due to the entropic penalty upon binding to the target. Thus, the aim of this study was to discover novel conformationally constrained scaffolds as starting points for further inhibitor optimization. Shifting from flexible tertiary amines to rigid heteroaromatic cores resulted in a boost in inhibitory activity. Moreover, some compounds already exhibited higher activity against individual astacin proteinases compared to recently reported inhibitors and also a favorable off-target selectivity profile, thus qualifying them as very suitable chemical probes for target validation.


Subject(s)
Amines/pharmacology , Antineoplastic Agents/pharmacology , Drug Discovery , Hydrocarbons, Aromatic/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Metalloproteases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Amines/chemical synthesis , Amines/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Hydrocarbons, Aromatic/chemical synthesis , Hydrocarbons, Aromatic/chemistry , Metalloendopeptidases/metabolism , Metalloproteases/metabolism , Molecular Structure , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Molecules ; 27(1)2021 Dec 22.
Article in English | MEDLINE | ID: mdl-35011288

ABSTRACT

Inhibition of bacterial virulence is believed to be a new treatment option for bacterial infections. In the present study, we tested dipicolylamine (DPA), tripicolylamine (TPA), tris pyridine ethylene diamine (TPED), pyridine and thiophene derivatives as putative inhibitors of the bacterial virulence factors thermolysin (TLN), pseudolysin (PLN) and aureolysin (ALN) and the human zinc metalloproteases, matrix metalloprotease-9 (MMP-9) and matrix metalloprotease-14 (MMP-14). These compounds have nitrogen or sulfur as putative donor atoms for zinc chelation. In general, the compounds showed stronger inhibition of MMP-14 and PLN than of the other enzymes, with Ki values in the lower µM range. Except for DPA, none of the compounds showed significantly stronger inhibition of the virulence factors than of the human zinc metalloproteases. TPA and Zn230 were the only compounds that inhibited all five zinc metalloproteinases with a Ki value in the lower µM range. The thiophene compounds gave weak or no inhibition. Docking indicated that some of the compounds coordinated zinc by one oxygen atom from a hydroxyl or carbonyl group, or by oxygen atoms both from a hydroxyl group and a carbonyl group, and not by pyridine nitrogen as in DPA and TPA.


Subject(s)
Chelating Agents/chemistry , Chelating Agents/pharmacology , Metalloproteases/antagonists & inhibitors , Metalloproteases/chemistry , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Zinc Compounds/chemistry , Zinc Compounds/pharmacology , Amino Acids , Bacteria/drug effects , Bacteria/enzymology , Catalytic Domain , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Humans , Models, Molecular , Molecular Conformation , Molecular Structure , Structure-Activity Relationship
12.
EBioMedicine ; 60: 102984, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32979835

ABSTRACT

BACKGROUND: Pulmonary damage by Pseudomonas aeruginosa during cystic fibrosis lung infection and ventilator-associated pneumonia is mediated both by pathogen virulence factors and host inflammation. Impaired immune function due to tissue damage and inflammation, coupled with pathogen multidrug resistance, complicates the management of these deep-seated infections. Pathological inflammation during infection is driven by interleukin-1ß (IL-1ß), but the molecular processes involved are not fully understood. METHODS: We examined IL-1ß activation in a pulmonary model infection of Pseudomonas aeruginosa and in vitro using genetics, specific inhibitors, recombinant proteins, and targeted reporters of protease activity and IL-1ß bioactivity. FINDINGS: Caspase-family inflammasome proteases canonically regulate maturation of this proinflammatory cytokine, but we report that plasticity in IL-1ß proteolytic activation allows for its direct maturation by the pseudomonal protease LasB. LasB promotes IL-1ß activation, neutrophilic inflammation, and destruction of lung architecture characteristic of severe P. aeruginosa pulmonary infection. INTERPRETATION: Preservation of lung function and effective immune clearance may be enhanced by selectively controlling inflammation. Discovery of this IL-1ß regulatory mechanism provides a distinct target for anti-inflammatory therapeutics, such as matrix metalloprotease inhibitors that inhibit LasB and limit inflammation and pathology during P. aeruginosa pulmonary infections. FUNDING: Full details are provided in the Acknowledgements section.


Subject(s)
Host-Pathogen Interactions , Interleukin-1beta/metabolism , Pseudomonas aeruginosa/enzymology , Serine Endopeptidases/metabolism , Animals , Biomarkers , Cystic Fibrosis/complications , Cystic Fibrosis/pathology , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Immunohistochemistry , Inflammasomes/metabolism , Inflammation Mediators , Metalloproteases/antagonists & inhibitors , Mice , Mice, Knockout , Models, Biological , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/pathology , Protein Binding , Pseudomonas Infections/etiology , Pseudomonas Infections/metabolism , Pseudomonas Infections/pathology
13.
ChemMedChem ; 15(16): 1499-1504, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32946206

ABSTRACT

Despite huge progress in hormonal therapy and improved in vitro fertilization methods, the success rates in infertility treatment are still limited. A recently discovered mechanism revealed the interplay between the plasma protein fetuin-B and the cortical granule-based proteinase ovastacin to be a novel key mechanism in the regulation of fertilization. Upon sperm-egg fusion, cleavage of a distinct zona pellucida component by ovastacin destroys the sperm receptor, enhances zona robustness, and eventually provides a definitive block against polyspermy. An untimely onset of this zona hardening prior to fertilization would consequently result in infertility. Physiologically, this process is controlled by fetuin-B, an endogenous ovastacin inhibitor. Here we aimed to discover small-molecule inhibitors of ovastacin that could mimic the effect of fetuin-B. These compounds could be useful lead structures for the development of specific ovastacin inhibitors that can be used in infertility treatment or in vitro fertilization.


Subject(s)
Amines/pharmacology , Hydroxamic Acids/pharmacology , Infertility, Female/drug therapy , Metalloproteases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Amines/chemistry , Animals , Biocatalysis , Dose-Response Relationship, Drug , Female , Hydroxamic Acids/chemistry , Infertility, Female/metabolism , Metalloproteases/metabolism , Mice , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemistry , Structure-Activity Relationship
15.
PLoS One ; 15(7): e0236175, 2020.
Article in English | MEDLINE | ID: mdl-32697798

ABSTRACT

Adenoviruses cause upper respiratory infections, conjunctivitis, keratitis, and gastrointestinal illness. These can be fatal in immunocompromised individuals. Adenoviruses have also been engineered into viral vectors to deliver therapeutic genes or induce immunity as vaccine carriers. The success of ocular gene therapy is driven partly by the immunologic and biochemical influences of the intraocular environment. We have shown that versican and hyaluronan modulate adenoviral vector transgene expression through CD44 signaling. Herein we explored the role of these pathways on virus replication and viral protein expression of wild type adenovirus. We report that the addition of vitreous humor (which contains both versican and hyaluronan) increases viral hexon protein levels. Vitreous humor also increased wild type adenovirus DNA replication in vitro. Metalloproteinase and γ-secretase inhibitors, which inhibit CD44 proteolytic activation, blocked adenoviral replication in vitro. Similarly, protein kinase C and RhoA kinase inhibitors, both proteins associated with CD44 mediated pathways, also inhibited wild type adenoviral replication in vitro. Application of metalloproteinase and γ-secretase inhibitors to human conjunctival explants sharply decreased adenoviral vector gene expression. Our results demonstrate that pharmacologic delivery of these inhibitors is easily achievable. The inhibition of these enzymes should be explored as potential therapies of wild type adenoviral infections.


Subject(s)
Adenoviridae Infections/drug therapy , Adenoviridae/drug effects , Antiviral Agents/pharmacology , Genetic Vectors/drug effects , Virus Replication/drug effects , Adenoviridae/physiology , Adenoviridae Infections/virology , Administration, Ophthalmic , Amides/pharmacology , Amides/therapeutic use , Antiviral Agents/therapeutic use , Conjunctiva/metabolism , DNA, Viral/genetics , DNA, Viral/isolation & purification , Diamines/pharmacology , Diamines/therapeutic use , Dipeptides/pharmacology , Dipeptides/therapeutic use , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/physiology , HeLa Cells , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Maleimides/pharmacology , Maleimides/therapeutic use , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , Permeability , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Proteolysis/drug effects , Pyridines/pharmacology , Pyridines/therapeutic use , Signal Transduction/drug effects , Thiazoles/pharmacology , Thiazoles/therapeutic use , Versicans/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Vitreous Body/metabolism , rho-Associated Kinases/metabolism
16.
mBio ; 11(4)2020 07 21.
Article in English | MEDLINE | ID: mdl-32694141

ABSTRACT

Cryptococcus neoformans (Cn) is the leading cause of fungal meningitis, a deadly disease with limited therapeutic options. Dissemination to the central nervous system hinges on the ability of Cn to breach the blood-brain barrier (BBB) and is considered an attribute of Cn virulence. Targeting virulence instead of growth for antifungal drug development has not been fully exploited despite the benefits of this approach. Mpr1 is a secreted fungal metalloprotease not required for fungal growth, but rather, it functions as a virulence factor by facilitating Cn migration across the BBB. This central role for Mpr1, its extracellular location, and lack of expression in mammalian cells make Mpr1 a high-value target for an antivirulence approach aimed at developing therapeutics for cryptococcal meningitis. To test this notion, we devised a large-scale screen to identify compounds that prohibited Cn from crossing the BBB by selectively blocking Mpr1 proteolytic activity, without inhibiting the growth of Cn A phytochemical natural product-derived library was screened to identify new molecular scaffolds of prototypes unique to a Cn microecosystem. Of the 240 pure natural products examined, 3 lead compounds, abietic acid, diosgenin, and lupinine inhibited Mpr1 proteolytic activity with 50% inhibitory concentration (IC50) values of <10 µM, displayed little to no mammalian cell toxicity, and did not affect Cn growth. Notably, the lead compounds blocked Cn from crossing the BBB, without damaging the barrier integrity, suggesting the bioactive molecules had no off-target effects. We propose that these new drug scaffolds are promising candidates for the development of antivirulence therapy against cryptococcal meningitis.IMPORTANCE Fungal infections like cryptococcal meningitis are difficult to resolve because of the limited therapies available. The small arsenal of antifungal drugs reflect the difficulty in finding available targets in fungi because like mammalian cells, fungi are eukaryotes. The limited efficacy, toxicity, and rising resistance of antifungals contribute to the high morbidity and mortality of fungal infections and further underscore the dire but unmet need for new antifungal drugs. The traditional approach in antifungal drug development has been to target fungal growth, but an attractive alternative is to target mechanisms of pathogenesis. An important attribute of Cryptococcus neoformans (Cn) pathogenesis is its ability to enter the central nervous system. Here, we describe a large-scale screen that identified three natural products that prevented Cn from crossing the blood-brain barrier by inhibiting the virulence factor Mpr1 without affecting the growth of Cn We propose that compounds identified here could be further developed as antivirulence therapy that would be administered preemptively or serve as a prophylactic in patients at high risk for developing cryptococcal meningitis.


Subject(s)
Antifungal Agents/pharmacology , Biological Products/pharmacology , Blood-Brain Barrier/microbiology , Cryptococcus neoformans/drug effects , Metalloproteases/antagonists & inhibitors , Brain/cytology , Brain/microbiology , Cell Line , Cryptococcus neoformans/enzymology , Fungal Proteins/antagonists & inhibitors , Humans , Inhibitory Concentration 50 , Meningitis, Cryptococcal/drug therapy , Meningitis, Cryptococcal/prevention & control , Phytochemicals/pharmacology , Small Molecule Libraries/pharmacology , Virulence
17.
Mol Biol (Mosk) ; 54(1): 146-152, 2020.
Article in Russian | MEDLINE | ID: mdl-32163398

ABSTRACT

Migration of cancer cells from the primary tumor site to nearby tissues is the starting point of the metastatic process. The invasive properties of cells are especially important for carcinomas, since tumor cells need to overcome the basement membrane and go beyond its boundaries to the underlying tissues. Substances that reduce the invasive ability of malignant cells are promising as antimetastatic agents. In the present work, the possibility of inhibiting the ability of different cancer cell lines to migrate under the influence of the Bacillus pumilus ribonuclease (binase) was analyzed using the scratch-wound assay. It was established that binase at non-toxic concentrations (10 µg/mL) reliably suppressed the migratory ability of HuTu 80 human duodenum adenocarcinoma cells incubated with RNase for 48-72 h. The antimetastatic potential of binase is confirmed by molecular modeling data demonstrating the ability of binase to inhibit cellular metalloproteinases that determine the migration of tumor cells.


Subject(s)
Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Cell Movement/drug effects , Duodenum/pathology , Ribonucleases/metabolism , Ribonucleases/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism
18.
Anticancer Agents Med Chem ; 20(9): 1038-1050, 2020.
Article in English | MEDLINE | ID: mdl-32067622

ABSTRACT

BACKGROUND: Melanoma is the most aggressive skin cancer, and BRAF (V600E) is the most frequent mutation that led to the development of BRAF inhibitors (BRAFi). However, patients treated with BRAFi usually present recidivism after 6-9 months. Curcumin is a turmeric substance, and it has been deeply investigated due to its anti-inflammatory and antitumoral effects. Still, the low bioavailability and biodisponibility encouraged the investigation of different analogs. DM-1 is a curcumin analog and has shown an antitumoral impact in previous studies. METHODS: Evaluated DM-1 stability and cytotoxic effects for BRAFi-sensitive and resistant melanomas, as well as the role in the metalloproteinases modulation. RESULTS: DM-1 showed growth inhibitory potential for melanoma cells, demonstrated by reduction of colony formation, migration and endothelial tube formation, and cell cycle arrest. Subtoxic doses were able to downregulate important Metalloproteinases (MMPs) related to invasiveness, such as MMP-1, -2 and -9. Negative modulations of TIMP-2 and MMP-14 reduced MMP-2 and -9 activity; however, the reverse effect is seen when increased TIMP-2 and MMP-14 resulted in raised MMP-2. CONCLUSION: These findings provide essential details into the functional role of DM-1 in melanomas, encouraging further studies in the development of combinatorial treatments for melanomas.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Melanoma/drug therapy , Metalloproteases/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Cycle Checkpoints/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Melanoma/metabolism , Melanoma/pathology , Metalloproteases/metabolism , Molecular Structure , Proto-Oncogene Proteins B-raf/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
19.
J Am Chem Soc ; 142(5): 2338-2345, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31918547

ABSTRACT

We describe a process for engineering a synthetic polymer nanoparticle (NP) that functions as an effective, broad-spectrum metalloproteinase inhibitor. Inhibition is achieved by incorporating three functional elements in the NP: a group that interacts with the catalytic zinc ion, functionality that enhances affinity to the substrate-binding pocket, and fine-tuning of the chemical composition of the polymer to strengthen NP affinity for the enzyme surface. The approach is validated by synthesis of a NP that sequesters and inhibits the proteolytic activity of snake venom metalloproteinases from five clinically relevant species of snakes. The mechanism of action of the NP mimics that of endogenous tissue inhibitors of metalloproteinases. The strategy provides a general design principle for synthesizing abiotic polymer inhibitors of enzymes.


Subject(s)
Biomimetics , Metalloproteases/antagonists & inhibitors , Nanoparticles/chemistry , Polymers/chemistry , Tissue Inhibitor of Metalloproteinases/pharmacology , Catalysis , Zinc/chemistry
20.
Eur J Med Chem ; 185: 111812, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31703818

ABSTRACT

Zinc metalloprotease 1 (Zmp1) is an extracellular enzyme, which has been found essential for the intracellular survival and pathogenesis of Mycobacterium tuberculosis. In this work, we designed and synthesized a series of novel thiazolidinedione-hydroxamates and evaluated in silico their drug-likeness behavior. Then, their inhibitory properties towards a recombinant Zmp1 from Mycobacterium tuberculosis were analyzed by MALDI-TOF MS. Nine of the tested compounds were found to inhibit the enzymatic reaction more effectively than the generic metalloprotease inhibitor phosphoramidon. Furthermore, the synthesized thiazolidinedione-hydroxamate hybrids were evaluated for their in vitro antimycobacterial activity and acute cytotoxicity using whole-cell assays. Results showed that none of the hybrids exhibited acute cytotoxicity against RAW264.7 macrophages. Whereas extracellular antimycobacterial activity was limited, RAW264.7 macrophage infection results showed that a majority of the hybrids inhibited the intracellular growth of Mycobacterium tuberculosis at a concentration of 100 and 10 µM. The thiazolidinedione-hydroxamate compound 2n was considered to be the best candidate of the evaluated library.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Hydroxamic Acids/pharmacology , Metalloproteases/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Thiazolidinediones/pharmacology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Bacterial Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Hydroxamic Acids/chemistry , Metalloproteases/metabolism , Molecular Docking Simulation , Molecular Structure , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Structure-Activity Relationship , Thiazolidinediones/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...