Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Part Fibre Toxicol ; 18(1): 34, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34496918

ABSTRACT

BACKGROUND: Multiwalled carbon nanotubes (MWCNT) are an increasingly utilized engineered nanomaterial that pose the potential for significant risk of exposure-related health outcomes. The mechanism(s) underlying MWCNT-induced toxicity to extrapulmonary sites are still being defined. MWCNT-induced serum-borne bioactivity appears to dysregulate systemic endothelial cell function. The serum compositional changes after MWCNT exposure have been identified as a surge of fragmented endogenous peptides, likely derived from matrix metalloproteinase (MMP) activity. In the present study, we utilize a broad-spectrum MMP inhibitor, Marimastat, along with a previously described oropharyngeal aspiration model of MWCNT administration to investigate the role of MMPs in MWCNT-derived serum peptide generation and endothelial bioactivity. RESULTS: C57BL/6 mice were treated with Marimastat or vehicle by oropharyngeal aspiration 1 h prior to MWCNT treatment. Pulmonary neutrophil infiltration and total bronchoalveolar lavage fluid protein increased independent of MMP blockade. The lung cytokine profile similarly increased following MWCNT exposure for major inflammatory markers (IL-1ß, IL-6, and TNF-α), with minimal impact from MMP inhibition. However, serum peptidomic analysis revealed differential peptide compositional profiles, with MMP blockade abrogating MWCNT-derived serum peptide fragments. The serum, in turn, exhibited differential potency in terms of inflammatory bioactivity when incubated with primary murine cerebrovascular endothelial cells. Serum from MWCNT-treated mice led to inflammatory responses in endothelial cells that were significantly blunted with serum from Marimastat-treated mice. CONCLUSIONS: Thus, MWCNT exposure induced pulmonary inflammation that was largely independent of MMP activity but generated circulating bioactive peptides through predominantly MMP-dependent pathways. This MWCNT-induced lung-derived bioactivity caused pathological consequences of endothelial inflammation and barrier disruption.


Subject(s)
Nanotubes, Carbon , Pneumonia , Animals , Bronchoalveolar Lavage Fluid , Endothelial Cells , Hydroxamic Acids , Lung , Matrix Metalloproteinase Inhibitors/toxicity , Mice , Mice, Inbred C57BL , Nanotubes, Carbon/toxicity , Pneumonia/chemically induced
2.
Carbohydr Polym ; 271: 118452, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34364546

ABSTRACT

The present paper describes the functionalization of sodium hyaluronate (NaHA) with a small molecule (2-((N-(6-aminohexyl)-4-methoxyphenyl)sulfonamido)-N-hydroxyacetamide) (MMPI) having proven inhibitory activity against membrane metalloproteins involved in inflammatory processes (i.e. MMP12). The obtained derivative (HA-MMPI) demonstrated an increased resistance to the in-vitro degradation by hyaluronidase, viscoelastic properties close to those of healthy human synovial fluid, cytocompatibility towards human chondrocytes and nanomolar affinity towards MMP 12. Thus, HA-MMPI can be considered a good candidate as viscosupplement in the treatment of knee osteoarticular disease.


Subject(s)
Hyaluronic Acid/pharmacology , Hydroxamic Acids/pharmacology , Matrix Metalloproteinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Viscoelastic Substances/pharmacology , Catalytic Domain , Chondrocytes/drug effects , Hyaluronic Acid/chemical synthesis , Hyaluronic Acid/metabolism , Hyaluronic Acid/toxicity , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/metabolism , Hydroxamic Acids/toxicity , Matrix Metalloproteinase 12/chemistry , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase Inhibitors/chemical synthesis , Matrix Metalloproteinase Inhibitors/metabolism , Matrix Metalloproteinase Inhibitors/toxicity , Protein Binding , Sulfonamides/chemical synthesis , Sulfonamides/metabolism , Sulfonamides/toxicity , Viscoelastic Substances/chemical synthesis , Viscoelastic Substances/metabolism , Viscoelastic Substances/toxicity
3.
Mol Neurobiol ; 58(11): 5703-5721, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34390469

ABSTRACT

Dolutegravir (DTG) is a first-line antiretroviral drug (ARV) used in combination therapy for the treatment of human immunodeficiency virus type-1 (HIV-1) infection. The drug is effective, safe, and well tolerated. Nonetheless, concerns have recently emerged for its usage in pregnant women or those of child-bearing age. Notably, DTG-based ARV regimens have been linked to birth defects seen as a consequence of periconceptional usages. To this end, uncovering an underlying mechanism for DTG-associated adverse fetal development outcomes has gained clinical and basic research interest. We now report that DTG inhibits matrix metalloproteinases (MMPs) activities that could affect fetal neurodevelopment. DTG is a broad-spectrum MMPs inhibitor and binds to Zn++ at the enzyme's catalytic domain. Studies performed in pregnant mice show that DTG readily reaches the fetal central nervous system during gestation and inhibits MMP activity. Postnatal screenings of brain health in mice pups identified neuroinflammation and neuronal impairment. These abnormalities persist as a consequence of in utero DTG exposure. We conclude that DTG inhibition of MMPs activities during gestation has the potential to affect prenatal and postnatal neurodevelopment.


Subject(s)
Anti-Retroviral Agents/toxicity , Heterocyclic Compounds, 3-Ring/toxicity , Matrix Metalloproteinase Inhibitors/toxicity , Neural Tube Defects/chemically induced , Neurodevelopmental Disorders/chemically induced , Neuroinflammatory Diseases/chemically induced , Oxazines/toxicity , Piperazines/toxicity , Pyridones/toxicity , Animals , Anti-Retroviral Agents/pharmacokinetics , Anti-Retroviral Agents/pharmacology , Brain/embryology , Brain/enzymology , Catalytic Domain/drug effects , Female , Gene Expression Profiling , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/pharmacology , Male , Matrix Metalloproteinase Inhibitors/pharmacokinetics , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Mice, Inbred C3H , Molecular Docking Simulation , Neural Tube Defects/embryology , Neuroimaging , Neuroinflammatory Diseases/embryology , Oxazines/pharmacokinetics , Oxazines/pharmacology , Piperazines/pharmacokinetics , Piperazines/pharmacology , Placenta/chemistry , Pregnancy , Pyridones/pharmacokinetics , Pyridones/pharmacology , Tissue Distribution , Zinc/metabolism
4.
Reprod Sci ; 28(11): 3085-3093, 2021 11.
Article in English | MEDLINE | ID: mdl-34050522

ABSTRACT

Benzo(a)pyrene (BaP) is a ubiquitous environmental endocrine-disrupting chemical that is known to have toxic effects on reproduction. However, the underlying mechanisms describing how BaP and its metabolite benzo[a]pyrene-7, 8-diol-9, 10-epoxide (BPDE) induce recurrent pregnancy loss (RPL) are still largely unclear. In this study, we identified a novel long non-coding RNA (lnc-HZ07, NCBI MT936329) that was upregulated in trophoblast cells after exposure to BPDE, and lnc-HZ07 expression was significantly higher in RPL villous tissues than that in control villous tissues. Knockdown of lnc-HZ07 promoted trophoblast cell migration, whereas overexpression of lnc-HZ07 inhibited trophoblast cell migration. Further study showed that lnc-HZ07 inhibited trophoblast migration by downregulating matrix metalloproteinase 2 (MMP2) expression via dephosphorylation of AKT. These results demonstrated a novel regulatory pathway in which BaP downregulated AKT phosphorylation and inhibited MMP2 expression by upregulating lnc-HZ07, suggesting that lnc-HZ07 could be considered as a potential pathological marker of BaP-induced RPL and therapeutic target for this disease.


Subject(s)
Abortion, Habitual/metabolism , Benzo(a)pyrene/toxicity , Matrix Metalloproteinase 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Trophoblasts/drug effects , Abortion, Habitual/chemically induced , Adult , Animals , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Environmental Pollution/adverse effects , Female , Humans , Matrix Metalloproteinase Inhibitors/toxicity , Mice , Mice, Inbred C57BL , Phosphoinositide-3 Kinase Inhibitors/toxicity , Pregnancy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA, Long Noncoding/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Trophoblasts/metabolism , Trophoblasts/pathology , Up-Regulation/drug effects , Up-Regulation/physiology , Young Adult
5.
ChemMedChem ; 16(8): 1257-1267, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33506625

ABSTRACT

Microbial infections are a significant threat to public health, and resistance is on the rise, so new antibiotics with novel modes of action are urgently needed. The extracellular zinc metalloprotease collagenase H (ColH) from Clostridium histolyticum is a virulence factor that catalyses tissue damage, leading to improved host invasion and colonisation. Besides the major role of ColH in pathogenicity, its extracellular localisation makes it a highly attractive target for the development of new antivirulence agents. Previously, we had found that a highly selective and potent thiol prodrug (with a hydrolytically cleavable thiocarbamate unit) provided efficient ColH inhibition. We now report the synthesis and biological evaluation of a range of zinc-binding group (ZBG) variants of this thiol-derived inhibitor, with the mercapto unit being replaced by other zinc ligands. Among these, an analogue with a phosphonate motif as ZBG showed promising activity against ColH, an improved selectivity profile, and significantly higher stability than the thiol reference compound, thus making it an attractive candidate for future drug development.


Subject(s)
Acetanilides/pharmacology , Bacterial Proteins/antagonists & inhibitors , Chelating Agents/pharmacology , Matrix Metalloproteinase Inhibitors/pharmacology , Organophosphonates/pharmacology , Acetanilides/chemical synthesis , Acetanilides/toxicity , Animals , Bacillus cereus/enzymology , Bacterial Proteins/metabolism , Cell Line, Tumor , Chelating Agents/chemical synthesis , Chelating Agents/toxicity , Clostridium histolyticum/enzymology , Collagen/metabolism , Collagenases/metabolism , HEK293 Cells , Humans , Matrix Metalloproteinase Inhibitors/chemical synthesis , Matrix Metalloproteinase Inhibitors/toxicity , Organophosphonates/chemical synthesis , Organophosphonates/toxicity , Swine , Zebrafish , Zinc/chemistry
6.
Curr Eye Res ; 46(1): 7-13, 2021 01.
Article in English | MEDLINE | ID: mdl-32567381

ABSTRACT

PURPOSE: To investigate the efficacy of RSH-12, a novel selective matrix metalloproteinase 9 (MMP-9) inhibitor peptide in rabbit models of dry eye syndrome (DES). METHODS: In vitro toxicity of RSH-12 on cultured human corneal fibroblasts was investigated with MTT. Ocular toxicity of RSH-12 was investigated by clinical examinations, histology, and TUNEL assay. Experimental model of dry eye was induced by 1.0% atropine sulfate administration followed after 15 min by treatment with PBS, RSH-12, and Restasis in individual groups, three times a day for 7 days. In addition to performing Schirmer's test for evaluating basic tear secretion and tear break-up time test for investigating tear stability, the occurrence of superficial punctate keratopathy was also investigated in the study groups. RESULTS: MTT assay demonstrated that RSH-12 was not toxic to human corneal fibroblasts in different concentrations. During the administration of atropine, TBUT values and tear volume were decreased in vehicle group while these indices improved significantly in groups treated with RSH-12 in a promising manner. RSH-12 increased the mean value of tear volume from 4.85 to 10.75 mm (P = .0001) and mean of TBUT values from 20.3 s to 34.5 s (P = .0001) compared with the vehicle. In contrast to the presence of severe superficial punctate keratopathy in the controls, no significant dotted staining was observed in the RSH-12 and Restasis groups. CONCLUSIONS: These outcomes propose that RSH-12 has a therapeutic effect in the rabbit model of dry eye and might be a potential treatment for severe DES.


Subject(s)
Disease Models, Animal , Dry Eye Syndromes/drug therapy , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase Inhibitors/therapeutic use , Oligopeptides/therapeutic use , Animals , Cell Survival , Corneal Keratocytes/drug effects , Corneal Stroma/cytology , Dry Eye Syndromes/enzymology , Female , Humans , In Situ Nick-End Labeling , Matrix Metalloproteinase Inhibitors/toxicity , Oligopeptides/toxicity , Rabbits , Slit Lamp Microscopy , Tears/physiology
7.
Eur J Pharmacol ; 887: 173431, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32758568

ABSTRACT

Intraocular pressure (IOP) lowering in glaucomatous eyes is currently achieved mainly by improved aqueous outflow via alternate drainage pathways. However, the focus is now shifting to trabecular meshwork (TM), the site or major pathological changes including increased extracellular matrix (ECM) deposition and reduced matrix metalloproteinases (MMPs) secretion by TM cells. Trans-resveratrol was previously shown to lower IOP and reduce ECM deposition; however, the mechanisms of action remain unclear. Therefore, we determined the effect of trans-resveratrol on MMP-2 and -9 expression by human TM cells (HTMCs) in the presence of dexamethasone and whether it also affects adenosine A1 receptors (A1AR) expression and nuclear factor kappa B (NFkB) activation. We observed that trans-resveratrol, 12.5 µM, increased MMP-2 and -9 protein expression by HTMCs despite exposure to dexamethasone (1.89- and 1.53-fold, respectively; P < 0.001). Further it was observed that trans-resveratrol increases A1AR expression in HTMC in the presence of dexamethasone (1.55-fold; P < 0.01). Trans-resveratrol also increased NFkB activation in the presence of dexamethasone and A1AR antagonist (P < 0.01 versus dexamethasone group). These effects of trans-resveratrol were associated with increased MMP -2 and -9 expression. It could be concluded that trans-resveratrol prevents dexamethasone-induced reduction in MMP-2 and -9 secretion by NFkB activation in HTMCs. This effect of trans-resveratrol is likely to involve increased A1AR expression.


Subject(s)
Dexamethasone/toxicity , Matrix Metalloproteinases/biosynthesis , NF-kappa B/biosynthesis , Receptor, Adenosine A1/biosynthesis , Resveratrol/pharmacology , Trabecular Meshwork/metabolism , Antioxidants/pharmacology , Cells, Cultured , Gene Expression Regulation, Enzymologic , Humans , Matrix Metalloproteinase Inhibitors/toxicity , NF-kappa B/antagonists & inhibitors , Trabecular Meshwork/drug effects
8.
J Korean Med Sci ; 32(4): 666-671, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28244295

ABSTRACT

We evaluated the safety of matrix metalloproteinase (MMP) inhibitor in experimental glaucoma filtration surgery in an animal model. Fifteen New Zealand white rabbits underwent an experimental trabeculectomy and were randomly allocated into 3 groups according to the adjuvant agent: no treatment group (n = 5), 0.02% mitomycin C (MMC) soaking group (n = 5), and MMP inhibitor (ilomastat) subconjunctival injection group (n = 5). Slit lamp examination with Seidel testing, pachymetry, and specular microscopy was performed preoperatively and postoperatively. The conjunctiva and ciliary body toxicity were evaluated with scores according to the pathologic grading systems. Electron microscopy was used to examine the structural changes in cornea, conjunctiva, and ciliary body. In the ilomastat-treated group, there was no statistically significant change in central corneal thickness preoperatively and at 28 days postoperatively (P = 0.655). There were also no significant changes in specular microscopy findings over the duration of the study in the ilomastat-treated group. The conjunctival toxicity score was 1 in the control group, 1.5 in the ilomastat-treated group, and 2 in the MMC-treated group. When assessing ciliary body toxicity scores, the ilomastat-treated group score was 0.5 and the MMC-treated group score was 1.5. Transmission electron microscopy did not show structural changes in the cornea and ciliary body whereas the structural changes were noticed in MMC group. A single subconjunctival injection of MMP inhibitor during the experimental trabeculectomy showed a less toxic affect in the rabbit cornea, conjunctiva, and ciliary body compared to MMC.


Subject(s)
Conjunctiva/drug effects , Glaucoma/surgery , Indoles/toxicity , Matrix Metalloproteinase Inhibitors/toxicity , Animals , Ciliary Body/drug effects , Ciliary Body/physiology , Conjunctiva/physiology , Cornea/drug effects , Cornea/physiology , Hydroxamic Acids , Microscopy, Electron, Transmission , Mitomycin/toxicity , Rabbits , Trabeculectomy
9.
J Med Chem ; 59(1): 313-27, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26653735

ABSTRACT

Matrix metalloproteinase-13 (MMP-13) is a zinc-dependent protease responsible for the cleavage of type II collagen, the major structural protein of articular cartilage. Degradation of this cartilage matrix leads to the development of osteoarthritis. We previously have described highly potent and selective carboxylic acid containing MMP-13 inhibitors; however, nephrotoxicity in preclinical toxicology species precluded development. The accumulation of compound in the kidneys mediated by human organic anion transporter 3 (hOAT3) was hypothesized as a contributing factor for the finding. Herein we report our efforts to optimize the MMP-13 potency and pharmacokinetic properties of non-carboxylic acid leads resulting in the identification of compound 43a lacking the previously observed preclinical toxicology at comparable exposures.


Subject(s)
Matrix Metalloproteinase 13/drug effects , Matrix Metalloproteinase Inhibitors/chemical synthesis , Matrix Metalloproteinase Inhibitors/pharmacology , Osteoarthritis/drug therapy , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Tetrazoles/chemical synthesis , Tetrazoles/pharmacology , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Collagenases/drug effects , Dogs , Drug Design , Humans , Kidney/metabolism , Macaca fascicularis , Male , Matrix Metalloproteinase Inhibitors/toxicity , Models, Molecular , Organic Anion Transporters, Sodium-Independent/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
10.
J Dent Res ; 94(8): 1085-91, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26040283

ABSTRACT

Nanoparticles (NPs) are currently the focus of considerable attention for dental applications; however, their biological effects have not been fully elucidated. The long-term, slow release of matrix metalloproteases (MMPs) digests collagen fibrils within resin-dentin bonds. Therefore, MMP inhibitors can prolong the durability of resin-dentin bonds. However, there have been few reports evaluating the combined effect of MMP inhibition and the cytotoxic effects of NPs for dentin bonding. The aim of this study was to evaluate MMP inhibition and cytotoxic responses to gold (AuNPs) and platinum nanoparticles (PtNPs) stabilized by polyvinylpyrrolidone (PVP) in cultured murine macrophages (RAW264) by using MMP inhibition assays, measuring cell viability and inflammatory responses (quantitative reverse transcription polymerase chain reaction [RT-qPCR]), and conducting a micromorphological analysis by fluorescence and transmission electron microscopy. Cultured RAW264 cells were exposed to metal NPs at various concentrations (1, 10, 100, and 400 µg/mL). AuNPs and PtNPs markedly inhibited MMP-8 and MMP-9 activity. Although PtNPs were cytotoxic at high concentrations (100 and 400 µg/mL), no cytotoxic effects were observed for AuNPs at any concentration. Transmission electron microscopy images showed a significant nonrandom intercellular distribution for AuNPs and PtNPs, which were mostly observed to be localized in lysosomes but not in the nucleus. RT-qPCR analysis demonstrated inflammatory responses were not induced in RAW264 cells by AuNPs or PtNPs. The cytotoxicity of nanoparticles might depend on the core metal composition and arise from a "Trojan horse" effect; thus, MMP inhibition could be attributed to the surface charge of PVP, which forms the outer coating of NPs. The negative charge of the surface coating of PVP binds to Zn(2+) from the active center of MMPs by chelate binding and results in MMP inhibition. In summary, AuNPs are attractive NPs that effectively inhibit MMP activity without cytotoxicity or inflammatory responses.


Subject(s)
Gold/chemistry , Gold/toxicity , Macrophages/drug effects , Matrix Metalloproteinase Inhibitors/chemistry , Matrix Metalloproteinase Inhibitors/toxicity , Nanoparticles/chemistry , Nanoparticles/toxicity , Animals , Cell Survival/drug effects , Humans , Immunohistochemistry , Mice , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Platinum/chemistry , Platinum/toxicity , Povidone/chemistry , Povidone/toxicity , Reverse Transcriptase Polymerase Chain Reaction
11.
Biochem Biophys Res Commun ; 457(4): 538-41, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25600809

ABSTRACT

Matrix metalloproteinases (MMPs) are zinc (Zn(2+)) and calcium (Ca(2+)) dependant endopeptidases, capable of degradation of numerous components of the extracellular matrix. Cadmium (Cd(2+)) is a well known environmental contaminant which could impair the activity of MMPs. In this sense, this study was conducted to evaluate if Cd(2+) intake inhibits these endopeptidases activities at the rat prostate and testicles and if it directly inhibits the activity of MMP2 and MMP9 at gelatinolytic assays when present in the incubation buffer. To investigate this hypothesis, Wistar rats (5 weeks old), were given tap water (untreated, n = 9), or 15 ppm CdCl2 diluted in drinking water, during 10 weeks (n = 9) and 20 weeks (n = 9). The animals were euthanized and their ventral prostate, dorsal prostate, and testicles were removed. These tissue samples were processed for protein extraction and subjected to gelatin zymography evaluation. Additionally, we performed an experiment of gelatin zymography in which 5 µM or 2 mM cadmium chloride (CdCl2) was directly dissolved at the incubation buffer, using the prostatic tissue samples from untreated animals that exhibited the highest MMP2 and MMP9 activities in the previous experiment. We have found that CdCl2 intake in the drinking water led to the inhibition of 35% and 30% of MMP2 and MMP9 (p < 0.05) at the ventral prostate and testis, respectively, in Cd(2+) treated animals when compared to controls. Moreover, the activities of the referred enzymes were 80% and 100% inhibited by 5 µM and 2 mM of CdCl2, respectively, even in the presence of 10 mM of CaCl2 within the incubation buffer solution. These important findings demonstrate that environmental cadmium contamination may deregulate the natural balance in the extracellular matrix turnover, through MMPs downregulation, which could contribute to the toxic effects observed in prostatic and testicular tissue after its exposure.


Subject(s)
Cadmium/toxicity , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/toxicity , Prostate/enzymology , Testis/enzymology , Water Pollutants, Chemical/toxicity , Animals , Male , Prostate/drug effects , Rats, Wistar , Testis/drug effects
12.
J Periodontol ; 85(6): 868-75, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24215203

ABSTRACT

BACKGROUND: Recent studies have shown that epigallocatechin-3-gallate (EGCG), a major constituent of green tea extract, exhibits effects of anti-inflammation and antioxidation on periodontal inflammation. The present in vitro study examines the effect of EGCG on Porphyromonas gingivalis (Pg) lipopolysaccharide (LPS)-enhanced expression of interleukin (IL)-6 and matrix metalloproteinase (MMP)-1, as well as the activation of nuclear factor-kappa B (NF-κB). Furthermore, the role of IL-6 on LPS-enhanced MMP-1 production is evaluated using human gingival fibroblasts (HGFs). METHODS: HGFs were primary cultured from human gingiva specimens. The cytotoxicities of EGCG and LPS were tested by cell viability tests. The cellular mRNA expression of IL-6 was determined by reverse-transcription polymerase chain reaction, and the protein expression of MMP-1 and IL-6 was examined by enzyme-linked immunosorbent assay. The cytosol expression and nuclear translocation of NF-κB was evaluated by immunocytochemistry followed by confocal laser scanning microscopy. RESULTS: Pg LPS significantly increased MMP-1 production in HGFs, whereas adding EGCG significantly attenuated this enhanced production of MMP-1. LPS treatment also increased the mRNA and protein expression of IL-6 and stimulated NF-κB activation in HGFs. However, the addition of EGCG significantly attenuated the IL-6 expression and NF-κB activation. Supplemental addition of IL-6 significantly enhanced cellular MMP-1 production, whereas anti-IL-6 antibody inhibited LPS-enhanced MMP-1 production. CONCLUSION: EGCG could attenuate Pg LPS-enhanced production of MMP-1 in HGFs, whereas this attenuation might be due to the inhibition of IL-6 by EGCG.


Subject(s)
Antioxidants/pharmacology , Catechin/analogs & derivatives , Fibroblasts/drug effects , Gingiva/drug effects , Interleukin-6/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase Inhibitors/pharmacology , Porphyromonas gingivalis/drug effects , Adult , Antioxidants/toxicity , Catechin/pharmacology , Catechin/toxicity , Cell Culture Techniques , Cell Survival/drug effects , Cells, Cultured , Female , Gingiva/cytology , Humans , Interleukin-6/analysis , Male , Matrix Metalloproteinase 1/analysis , Matrix Metalloproteinase Inhibitors/toxicity , Microscopy, Confocal , NF-kappa B/drug effects , Young Adult
13.
Bioorg Med Chem ; 21(21): 6456-65, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24071448

ABSTRACT

The complexity of matrix metalloproteinase inhibitors (MMPIs) design derives from the difficulty in carefully addressing their inhibitory activity towards the MMP isoforms involved in many pathological conditions. In particular, specific metalloproteinases, such as MMP-2 and MMP-9, are key regulators of the 'vicious cycle' occurring between tumor metastases growth and bone remodeling. In an attempt to devise new approaches to selective inhibitor derivatives, we describe novel bisphosphonate bone seeking MMP inhibitors (BP-MMPIs), capable to be selectively targeted and to overcome undesired side effects of broad spectrum MMPIs. In vitro activity (IC50 values) for each inhibitor was determined against MMP-2, -8, -9 and -14, because of their relevant role in skeletal development and renewal. The results show that BP-MMPIs reached IC50 values of enzymatic inhibition in the low micromolar range. Computational studies, used to rationalize some trends in the observed inhibitory profiles, suggest a possible differential binding mode in MMP-2 that explains the selective inhibition of this isoform. In addition, survival assay was conducted on J774 cell line, a well known model system used to evaluate the structure-activity relationship of BPs for inhibiting bone resorption. The resulting data, confirming the specific activity of BP-MMPIs, and their additional proved propensity to bind hydroxyapatite powder in vitro, suggest a potential use of BP-MMPIs in skeletal malignancies.


Subject(s)
Diphosphonates/chemistry , Matrix Metalloproteinase Inhibitors/chemistry , Matrix Metalloproteinases/chemistry , Animals , Binding Sites , Cell Line , Cell Survival/drug effects , Diphosphonates/metabolism , Diphosphonates/toxicity , Durapatite/chemistry , Durapatite/metabolism , Hep G2 Cells , Humans , Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 8/chemistry , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/metabolism , Matrix Metalloproteinase Inhibitors/toxicity , Matrix Metalloproteinases/metabolism , Mice , Molecular Docking Simulation , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
14.
J Med Chem ; 56(11): 4357-73, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23631440

ABSTRACT

New series of pyrrolidine mercaptosulfide, 2-mercaptocyclopentane arylsulfonamide, and 3-mercapto-4-arylsulfonamidopyrrolidine matrix metalloproteinase inhibitors (MMPIs) were designed, synthesized, and evaluated. Exhibiting unique properties over other MMPIs (e.g., hydroxamates), these newly reported compounds are capable of modulating activities of several MMPs in the low nanomolar range, including MMP-2 (~2 to 50 nM), MMP-13 (~2 to 50 nM), and MMP-14 (~4 to 60 nM). Additionally these compounds are selective to intermediate- and deep-pocket MMPs but not shallow-pocketed MMPs (e.g., MMP-1, ~850 to >50,000 nM; MMP-7, ~4000 to >25,000 nM). Our previous work with the mercaptosulfide functionality attached to both cyclopentane and pyrrolidine frameworks demonstrated that the cis-(3S,4R)-stereochemistry was optimal for all of the MMPs tested. However, in our newest compounds an interesting shift of preference to the trans form of the mercaptosulfonamides was observed with increased oxidative stability and biological compatibility. We also report several kinetic and biological characteristics showing that these compounds may be used to probe the mechanistic activities of MMPs in disease.


Subject(s)
Cyclopentanes/chemical synthesis , Matrix Metalloproteinase Inhibitors/chemical synthesis , Pyrrolidines/chemical synthesis , Sulfhydryl Compounds/chemical synthesis , Sulfonamides/chemical synthesis , Cells, Cultured , Cyclopentanes/chemistry , Cyclopentanes/toxicity , Drug Stability , Humans , Kinetics , Matrix Metalloproteinase Inhibitors/chemistry , Matrix Metalloproteinase Inhibitors/toxicity , Matrix Metalloproteinases/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Pyrrolidines/chemistry , Pyrrolidines/toxicity , Stereoisomerism , Structure-Activity Relationship , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/toxicity , Sulfonamides/chemistry , Sulfonamides/toxicity
15.
Biochem Pharmacol ; 85(12): 1770-82, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23603294

ABSTRACT

Infliximab, a monoclonal antibody directed against human tumor necrosis factor-alpha (TNF-α), effectively treats anterior uveitis, which can accompany Behçet's disease. Here, we investigated the underlying mechanism of this action. We examined human, non-pigmented ciliary epithelial cells (HNPCECs), which make up the blood-aqueous barrier (BAB) in the uvea. We measured the expression levels of matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs in the presence or absence of TNF-α using quantitative, real-time polymerase chain reaction and enzyme-linked immunosorbent assays. The expression of MMP-1, MMP-3, and MMP-9 increased in the presence of TNF-α, and the addition of infliximab reversed the increase. The TNF-α effects were more attenuated when infliximab was added before than when it was added after TNF-α exposure. Gelatin zymography demonstrated that the protease activity of these MMPs was also increased in the presence of TNF-α and attenuated with infliximab. Immunostaining showed that MMP-1, MMP-3, and MMP-9 degraded claudin-1 and occludin in HNPCECs and in non-pigmented ciliary epithelial cells of the swine ciliary body. In a monolayer of HNPCECs, we found that permeability was significantly increased with MMP treatment. Thus, TNF-α increased levels of MMPs in cells that form the BAB, and MMPs degraded components of the tight junctions in the BAB, which increased permeability through the cellular barrier. Furthermore, infliximab effectively attenuated the TNF-α-induced increases in MMP expression in cells that make up the BAB. These findings might suggest a basis for the clinical prevention of anterior uveitis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Ciliary Body/metabolism , Claudin-1/antagonists & inhibitors , Matrix Metalloproteinases/metabolism , Occludin/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/toxicity , Animals , Cells, Cultured , Ciliary Body/drug effects , Claudin-1/metabolism , Down-Regulation/drug effects , Down-Regulation/immunology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Infliximab , Matrix Metalloproteinase Inhibitors/metabolism , Matrix Metalloproteinase Inhibitors/toxicity , Matrix Metalloproteinases/biosynthesis , Occludin/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...