Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 806
Filter
1.
J Med Chem ; 65(3): 1933-1945, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34898192

ABSTRACT

The urokinase plasminogen activator (uPA) plays a critical role in tumor cell invasion and migration and is a promising antimetastasis target. 6-Substituted analogues of 5-N,N-(hexamethylene)amiloride (HMA) are potent and selective uPA inhibitors that lack the diuretic and antikaliuretic properties of the parent drug amiloride. However, the compounds display pronounced selectivity for human over mouse uPA, thus confounding interpretation of data from human xenograft mouse models of cancer. Here, computational and experimental findings reveal that residue 99 is a key contributor to the observed species selectivity, whereby enthalpically unfavorable expulsion of a water molecule by the 5-N,N-hexamethylene ring occurs when residue 99 is Tyr (as in mouse uPA). Analogue 7 lacking the 5-N,N-hexamethylene ring maintained similar water networks when bound to human and mouse uPA and displayed reduced selectivity, thus supporting this conclusion. The study will guide further optimization of dual-potent human/mouse uPA inhibitors from the amiloride class as antimetastasis drugs.


Subject(s)
Amiloride/analogs & derivatives , Enzyme Inhibitors/metabolism , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Water/chemistry , Amiloride/chemistry , Amiloride/metabolism , Animals , Enzyme Inhibitors/chemistry , Humans , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Mutation , Protein Binding , Species Specificity , Thermodynamics , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
2.
Phytomedicine ; 90: 153642, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34265701

ABSTRACT

BACKGROUND: Metastasis caused a decline in the 5-years survival rate of osteosarcoma. Therefore, developing new targeted therapeutics for osteosarcoma treatment is imperative. Dihydromyricetin (DHM) has several physiological functions: it counteracts inflammation, oxidation, and antitumor properties. However, the effects of DHM on osteosarcoma and its underlying mechanisms are still not well understood. PURPOSE: In this study, we investigated the antimetastatic properties of DHM in human osteosarcoma U-2 OS and HOS cells. METHODS: The effects of DHM (0, 25, 50, 75, and 100 µM) on cell viability, migration, and invasion were examined. Western blotting, RT-PCR, and quantitative real-time PCR (QPCR) were determined urokinase plasminogen activator (uPA) expression. The expression of transcriptional factor SP-1 and NF-κB was determined by using immunofluorescence assay, chromatin immunoprecipitation assay, and site-directed mutagenesis luciferase reporter. RESULTS: We observed that DHM suppresses cell migration and invasion in osteosarcoma cell lines. In addition, DHM inhibits metastasis by downregulating urokinase plasminogen activator (uPA) expression. Moreover, real-time polymerase chain reaction and promoter activity assays revealed that DHM decreased uPA expression at transcription levels. Furthermore, the inhibition of uPA expression was associated with the suppression of SP-1 and NF-κB, which bind to the uPA promoter. Regardless of blocking or inducing the extracellular signal-regulated kinase (ERK) pathway, we verified that the DHM-related suppression of uPA and cell metastasis occurred through the p-ERK pathway. CONCLUSION: We are the first study to propose that DHM suppresses osteosarcoma metastasis through the ERK pathway and through the suppression of SP-1 and NF-κB to inhibit downstream uPA expression. DHM is a potential therapeutic agent for antimetastatic therapy against osteosarcoma.


Subject(s)
Bone Neoplasms , Flavonols/pharmacology , Neoplasm Metastasis/drug therapy , Osteosarcoma , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Bone Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement , Humans , NF-kappa B/metabolism , Neoplasm Invasiveness , Osteosarcoma/drug therapy , Sp1 Transcription Factor/metabolism
3.
Int J Mol Sci ; 22(6)2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33804289

ABSTRACT

The K+-sparing diuretic amiloride shows off-target anti-cancer effects in multiple rodent models. These effects arise from the inhibition of two distinct cancer targets: the trypsin-like serine protease urokinase-type plasminogen activator (uPA), a cell-surface mediator of matrix degradation and tumor cell invasiveness, and the sodium-hydrogen exchanger isoform-1 (NHE1), a central regulator of transmembrane pH that supports carcinogenic progression. In this study, we co-screened our library of 5- and 6-substituted amilorides against these two targets, aiming to identify single-target selective and dual-targeting inhibitors for use as complementary pharmacological probes. Closely related analogs substituted at the 6-position with pyrimidines were identified as dual-targeting (pyrimidine 24 uPA IC50 = 175 nM, NHE1 IC50 = 266 nM, uPA selectivity ratio = 1.5) and uPA-selective (methoxypyrimidine 26 uPA IC50 = 86 nM, NHE1 IC50 = 12,290 nM, uPA selectivity ratio = 143) inhibitors, while high NHE1 potency and selectivity was seen with 5-morpholino (29 NHE1 IC50 = 129 nM, uPA IC50 = 10,949 nM; NHE1 selectivity ratio = 85) and 5-(1,4-oxazepine) (30 NHE1 IC50 = 85 nM, uPA IC50 = 5715 nM; NHE1 selectivity ratio = 67) analogs. Together, these amilorides comprise a new toolkit of chemotype-matched, non-cytotoxic probes for dissecting the pharmacological effects of selective uPA and NHE1 inhibition versus dual-uPA/NHE1 inhibition.


Subject(s)
Amiloride/pharmacology , Breast Neoplasms/drug therapy , Sodium-Hydrogen Exchanger 1/genetics , Urokinase-Type Plasminogen Activator/genetics , Amiloride/chemical synthesis , Amiloride/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Diuretics/chemical synthesis , Diuretics/chemistry , Diuretics/pharmacology , Female , Humans , Models, Molecular , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Structure-Activity Relationship , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
4.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805784

ABSTRACT

Hepatocellular carcinoma (HCC) frequently shows early invasion into blood vessels as well as intrahepatic metastasis. Innovations of novel small-molecule agents to block HCC invasion and subsequent metastasis are urgently needed. Moscatilin is a bibenzyl derivative extracted from the stems of a traditional Chinese medicine, orchid Dendrobium loddigesii. Although moscatilin has been reported to suppress tumor angiogenesis and growth, the anti-metastatic property of moscatilin has not been elucidated. The present results revealed that moscatilin inhibited metastatic behavior of HCC cells without cytotoxic fashion in highly invasive human HCC cell lines. Furthermore, moscatilin significantly suppressed the activity of urokinase plasminogen activator (uPA), but not matrix metalloproteinase (MMP)-2 and MMP-9. Interestingly, moscatilin-suppressed uPA activity was through down-regulation the protein level of uPA, and did not impair the uPA receptor and uPA inhibitory molecule (PAI-1) expressions. Meanwhile, the mRNA expression of uPA was inhibited via moscatilin in a concentration-dependent manner. In addition, the expression of phosphorylated Akt, rather than ERK1/2, was inhibited by moscatilin treatment. The expression of phosphor-IκBα, and -p65, as well as κB-luciferase activity were also repressed after moscatilin treatment. Transfection of constitutively active Akt (Myr-Akt) obviously restored the moscatilin-inhibited the activation of NF-κB and uPA, and cancer invasion in HCC cells. Taken together, these results suggest that moscatilin impedes HCC invasion and uPA expression through the Akt/NF-κB signaling pathway. Moscatilin might serve as a potential anti-metastatic agent against the disease progression of human HCC.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzyl Compounds/pharmacology , Cell Movement/drug effects , NF-kappa B/genetics , Proto-Oncogene Proteins c-akt/genetics , Urokinase-Type Plasminogen Activator/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Neoplastic , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/metabolism
5.
ChemMedChem ; 16(2): 377-387, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33107192

ABSTRACT

The urokinase receptor (uPAR) is a cell surface receptor that binds to the serine protease urokinase-type plasminogen activator (uPA) with high affinity. This interaction is beneficial for extravascular fibrin clearance, but it has also been associated with a broad range of pathological conditions including cancer, atherosclerosis, and kidney disease. Here, starting with a small molecule that we previously discovered by virtual screening and cheminformatics analysis, we design and synthesize several derivatives that were tested for binding and inhibition of the uPAR ⋅ uPA interaction. To confirm the binding site and establish a binding mode of the compounds, we carried out biophysical studies using uPAR mutants, among them uPARH47C-N259C , a mutant previously developed to mimic the structure of uPA-bound uPAR. Remarkably, a substantial increase in potency is observed for inhibition of uPARH47C-N259C binding to uPA compared to wild-type uPAR, consistent with our use of the structure of uPAR in its uPA-bound state to design small-molecule uPAR ⋅ uPA antagonists. Combined with the biophysical studies, molecular docking followed by extensive explicit-solvent molecular dynamics simulations and MM-GBSA free energy calculations yielded the most favorable binding pose of the compound. Collectively, these results suggest that potent inhibition of uPAR binding to uPA with small molecules will likely only be achieved by developing small molecules that exhibit high-affinity to solution apo structures of uPAR, rather than uPA-bound structures of the receptor.


Subject(s)
Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Binding Sites/drug effects , Cheminformatics , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Conformation , Receptors, Urokinase Plasminogen Activator/metabolism , Small Molecule Libraries/chemistry , Urokinase-Type Plasminogen Activator/metabolism
6.
Biochim Biophys Acta Proteins Proteom ; 1869(2): 140562, 2021 02.
Article in English | MEDLINE | ID: mdl-33221341

ABSTRACT

Affinity maturation of U33, a recombinant Fab inhibitor of uPA, was used to improve the affinity and the inhibitory effect compared to the parental Fab. Arginine scanning of the six CDR loops of U33 was done to identify initial binding determinants since uPA prefers arginine in its primary substrate binding pocket. Two CDR loops were selected to create an engineered affinity maturation library of U33 that was diversified around ArgL91 (CDR L3) and ArgH52 (CDR H2). Biopanning of the randomized U33 library under stringent conditions resulted in eight Fabs with improved binding properties. One of the most potent inhibitors, AB2, exhibited a 13-fold decrease in IC50 when compared to U33 largely due to a decrease in its off rate. To identify contributions of interfacial residues that might undergo structural rearrangement upon interface formation we used X-ray footprinting and mass spectrometry (XFMS). Four residues showed a pronounced decrease in solvent accessibility, and their clustering suggests that AB2 targets the active site and also engages residues in an adjacent pocket unique to human uPA. The 2.9 Å resolution crystal structure of AB2-bound to uPA shows a binding mode in which the CDR L1 loop inserts into the active site cleft and acts as a determinant of inhibition. The selectivity determinant of this binding mode is unlike previously identified inhibitory Fabs against uPA related serine proteases, MTSP-1, HGFA and FXIa. CDRs H2 and L3 loops aid in interface formation and provide critical salt-bridges to remodel loops surrounding the active site of uPA providing specificity and further evidence that antibodies can be potent and selective inhibitors of proteolytic enzymes.


Subject(s)
Recombinant Proteins/ultrastructure , Serine Proteases/chemistry , Serine Proteinase Inhibitors/chemistry , Urokinase-Type Plasminogen Activator/chemistry , Amino Acid Sequence/genetics , Humans , Quinuclidines/chemistry , Quinuclidines/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/ultrastructure , Serine Proteases/genetics , Serine Proteinase Inhibitors/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/genetics
7.
Molecules ; 26(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374783

ABSTRACT

Osteosarcoma, the most prevalent malignant bone tumor in the pediatric age group, is responsible for the great majority of cancer-associated deaths owing to its highly metastatic potential. The anti-metastatic effects of the new curcumin analogue L48H37 in human osteosarcoma are still unknown; hence, we investigated whether L48H37 represses human osteosarcoma cells' biological behavior of migratory potential and invasive activities and attempted to delve into its underlying mechanisms. L48H37 up to 5 µM inhibited, without cytotoxicity, the motility, migration, and invasion of human osteosarcoma U2OS and MG-63 cells. In U2OS cells, the human protease array revealed an obvious decrease in urokinase plasminogen activator (uPA) expression after L48H37 treatment, and L48H37 actually reduced the level, protein and mRNA expression, and promoter activity of uPA dose-dependently. L48H37 decreased the phosphorylation of STAT3, JAK1, JAK2, and JAK3 in U2OS cells, but did not affect the phosphorylation of ERK, JNK, p38, and Akt. Using colivelin, an activator of STAT3, the L48H37-induced decrease in uPA and migratory potential could be countered as expected. Collectively, L48H37 represses the invasion and migration capabilities of U2OS and MG-63 cells by the suppression of uPA expression and the inhibition of JAK/STAT signaling. These results suggest that L48H37 may be a potential candidate for anti-metastatic treatment of human osteosarcoma.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Janus Kinase 1/metabolism , Osteosarcoma/drug therapy , STAT3 Transcription Factor/metabolism , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Antineoplastic Agents/chemistry , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Movement , Cell Proliferation , Curcumin/analogs & derivatives , Humans , Janus Kinase 1/genetics , Neoplasm Invasiveness , Osteosarcoma/metabolism , Osteosarcoma/pathology , Phosphorylation , STAT3 Transcription Factor/genetics , Signal Transduction , Tumor Cells, Cultured
8.
Oncol Rep ; 44(6): 2345-2352, 2020 12.
Article in English | MEDLINE | ID: mdl-33125140

ABSTRACT

Hepatocellular carcinoma (HCC) is different from other solid tumors because it is commonly associated with the occurrence of intrahepatic metastasis. Additionally, the liver, unlike other organs, is the main site of coagulation and fibrinolytic factor production. Therefore, it was speculated that coagulation and fibrinolytic factors could be associated with intrahepatic metastasis of HCC. Do the coagulation and fibrinolytic systems protect HCC cells against anoikis during infiltration and metastasis? Conversely, do the coagulation and fibrinolytic systems lead to immune escape of HCC cells by affecting the immune microenvironment of patients? The current review aimed to present a number of novel hypotheses for the treatment of HCC by exploring the mechanisms of coagulation and fibrinolytic factors in the regulation of cancer growth.


Subject(s)
Blood Coagulation/immunology , Carcinoma, Hepatocellular/secondary , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Liver/pathology , Anoikis/drug effects , Anoikis/immunology , Biomarkers/metabolism , Blood Coagulation/drug effects , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carcinogenesis/pathology , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Humans , Liver/immunology , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Protein Precursors/metabolism , Prothrombin/metabolism , Tumor Escape/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/metabolism
9.
J Mol Graph Model ; 100: 107710, 2020 11.
Article in English | MEDLINE | ID: mdl-32829149

ABSTRACT

The emergence of SARS-CoV-2 has prompted a worldwide health emergency. There is an urgent need for therapeutics, both through the repurposing of approved drugs and the development of new treatments. In addition to the viral drug targets, a number of human drug targets have been suggested. In theory, targeting human proteins should provide an advantage over targeting viral proteins in terms of drug resistance, which is commonly a problem in treating RNA viruses. This paper focuses on the human protein TMPRSS2, which supports coronavirus life cycles by cleaving viral spike proteins. The three-dimensional structure of TMPRSS2 is not known and so we have generated models of the TMPRSS2 in the apo state as well as in complex with a peptide substrate and putative inhibitors to aid future work. Importantly, many related human proteases have 80% or higher identity with TMPRSS2 in the S1-S1' subsites, with plasminogen and urokinase-type plasminogen activator (uPA) having 95% identity. We highlight 376 approved, investigational or experimental drugs targeting S1A serine proteases that may also inhibit TMPRSS2. Whilst the presence of a relatively uncommon lysine residue in the S2/S3 subsites means that some serine protease inhibitors will not inhibit TMPRSS2, this residue is likely to provide a handle for selective targeting in a focused drug discovery project. We discuss how experimental drugs targeting related serine proteases might be repurposed as TMPRSS2 inhibitors to treat coronaviruses.


Subject(s)
Antiviral Agents/chemistry , Betacoronavirus/chemistry , Protease Inhibitors/chemistry , Serine Endopeptidases/chemistry , Small Molecule Libraries/chemistry , Amino Acid Sequence , Betacoronavirus/enzymology , COVID-19 , Catalytic Domain , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Drug Repositioning , Host-Pathogen Interactions , Humans , Ligands , Molecular Dynamics Simulation , Pandemics , Plasminogen/antagonists & inhibitors , Plasminogen/chemistry , Plasminogen/metabolism , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , SARS-CoV-2 , Sequence Alignment , Serine Endopeptidases/metabolism , Structural Homology, Protein , Structure-Activity Relationship , Thermodynamics , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/metabolism
10.
J Mol Med (Berl) ; 98(4): 585-593, 2020 04.
Article in English | MEDLINE | ID: mdl-32108909

ABSTRACT

Blocking the proteolytic capacity of urokinase-type plasminogen activator (uPA) with a monoclonal antibody (mAb) reduces arthritis progression in the collagen-induced mouse arthritis model to an extent that is on par with the effect of blocking tumor necrosis factor-alpha by etanercept. Seeking to develop a novel therapy for rheumatoid arthritis, a humanized mAb, NNC0266-0043, was selected for its dual inhibition of both the zymogen activation and the proteolytic capacity of human uPA. The antibody revealed nonlinear elimination kinetics in cynomolgus monkeys consistent with binding to and turnover of endogenous uPA. At a dose level of 20.6 mg kg-1, the antibody had a plasma half-life of 210 h. Plasma uPA activity, a pharmacodynamic marker of anti-uPA therapy, was reduced to below the detection limit during treatment, indicating that an efficacious plasma concentration was reached. Pharmacokinetic modeling predicted that sufficient antibody levels can be sustained in arthritis patients dosed subcutaneously once weekly. The anti-uPA mAb was also well tolerated in cynomolgus monkeys at weekly doses up to 200 mg kg-1 over 4 weeks. The data from cynomolgus monkeys and from human material presented here indicates that anti-uPA mAb NNC0266-0043 is suitable for clinical testing as a novel therapeutic for rheumatic diseases. KEY MESSAGES: Background: Anti-uPA therapy is on par with etanercept in a mouse arthritis model. A new humanized antibody blocks activation and proteolytic activity of human uPA. The antibody represents a radically novel mode-of-action in anti-rheumatic therapy. The antibody has PK/PD properties in primates consistent with QW clinical dosing.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/etiology , Drug Development , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Immunohistochemistry , Macaca fascicularis , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Monocytes/metabolism
11.
N Engl J Med ; 382(5): 416-426, 2020 01 30.
Article in English | MEDLINE | ID: mdl-31995687

ABSTRACT

BACKGROUND: Acute kidney injury is common, with a major effect on morbidity and health care utilization. Soluble urokinase plasminogen activator receptor (suPAR) is a signaling glycoprotein thought to be involved in the pathogenesis of kidney disease. We investigated whether a high level of suPAR predisposed patients to acute kidney injury in multiple clinical contexts, and we used experimental models to identify mechanisms by which suPAR acts and to assess it as a therapeutic target. METHODS: We measured plasma levels of suPAR preprocedurally in patients who underwent coronary angiography and patients who underwent cardiac surgery and at the time of admission to the intensive care unit in critically ill patients. We assessed the risk of acute kidney injury at 7 days as the primary outcome and acute kidney injury or death at 90 days as a secondary outcome, according to quartile of suPAR level. In experimental studies, we used a monoclonal antibody to urokinase plasminogen activator receptor (uPAR) as a therapeutic strategy to attenuate acute kidney injury in transgenic mice receiving contrast material. We also assessed cellular bioenergetics and generation of reactive oxygen species in human kidney proximal tubular (HK-2) cells that were exposed to recombinant suPAR. RESULTS: The suPAR level was assessed in 3827 patients who were undergoing coronary angiography, 250 who were undergoing cardiac surgery, and 692 who were critically ill. Acute kidney injury developed in 318 patients (8%) who had undergone coronary angiography. The highest suPAR quartile (vs. the lowest) had an adjusted odds ratio of 2.66 (95% confidence interval [CI], 1.77 to 3.99) for acute kidney injury and 2.29 (95% CI, 1.71 to 3.06) for acute kidney injury or death at 90 days. Findings were similar in the surgical and critically ill cohorts. The suPAR-overexpressing mice that were given contrast material had greater functional and histologic evidence of acute kidney injury than wild-type mice. The suPAR-treated HK-2 cells showed heightened energetic demand and mitochondrial superoxide generation. Pretreatment with a uPAR monoclonal antibody attenuated kidney injury in suPAR-overexpressing mice and normalized bioenergetic changes in HK-2 cells. CONCLUSIONS: High suPAR levels were associated with acute kidney injury in various clinical and experimental contexts. (Funded by the National Institutes of Health and others.).


Subject(s)
Acute Kidney Injury/blood , Cardiac Surgical Procedures/adverse effects , Coronary Angiography/adverse effects , Receptors, Urokinase Plasminogen Activator/blood , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Aged , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biomarkers/blood , Critical Illness , Disease Models, Animal , Female , Humans , Intensive Care Units , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Odds Ratio , Podocytes/drug effects , Podocytes/metabolism , Postoperative Complications/blood , Postoperative Complications/etiology , Risk Assessment/methods , Urokinase-Type Plasminogen Activator/pharmacology
12.
Cell Signal ; 65: 109432, 2020 01.
Article in English | MEDLINE | ID: mdl-31693876

ABSTRACT

Pirfenidone (PFD) is an orally available synthetic drug which has been approved for the treatment of idiopathic pulmonary fibrosis. In addition to its anti-fibrotic properties, PFD also exerts anti-tumor effects in cancer models by inducing alterations in the tumor microenvironment. Here, we demonstrate that PFD reduces proliferation, 2D- and 3D-migration as well as colony formation of the non-small-cell lung carcinoma (NSCLC) cells. On a molecular level, we show that PFD on the one hand interacts with plasminogen activator inhibitor-1 (PAI-1; Kd of 46.2±11.3nM) and affects its inhibitory potency, but on the other hand it also increases PAI-1 expression; in both cases consequently leading to the reduction of urokinase (uPA) activity. Finally, we report that the effect of PFD on 2D-migration of NSCLC cells depends on PAI-1 expression and thus on the activity of the uPA system whereas the PFD-induced changes in cancer cell proliferation, 3D-migration and colony formation are PAI-1 independent. To conclude, a direct interference of PFD with the uPA-PAI-1 system may deregulate pericellular proteolytic activity and thereby influence the stability of the tumor blood vessels and the matrix architecture within tumor stroma.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement/drug effects , Lung Neoplasms/metabolism , Pyridones/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/metabolism , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Indoleacetic Acids/pharmacology , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Membrane Proteins/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Tumor Microenvironment/drug effects
13.
Bioorg Med Chem Lett ; 29(24): 126753, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31679971

ABSTRACT

The oral K+-sparing diuretic amiloride shows anti-cancer side-activities in multiple rodent models. These effects appear to arise, at least in part, through moderate inhibition of the urokinase-type plasminogen activator (uPA, Ki = 2.4 µM), a pro-metastatic trypsin-like serine protease that is upregulated in many aggressive solid malignancies. In applying the selective optimization of side-activity (SOSA) approach, a focused library of twenty two 6-substituted amiloride derivatives were prepared, with multiple examples displaying uPA inhibitory potencies in the nM range. X-ray co-crystal structures revealed that the potency increases relative to amiloride arise from increased occupancy of uPA's S1ß subsite by the appended 6-substituents. Leading compounds were shown to have high selectivity over related trypsin-like serine proteases and no diuretic or anti-kaliuretic effects in rats. Compound 15 showed anti-metastatic effects in a xenografted mouse model of late-stage lung metastasis.


Subject(s)
Amiloride/analogs & derivatives , Amiloride/therapeutic use , Diuretics/therapeutic use , Neoplasm Metastasis/drug therapy , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Amiloride/pharmacology , Diuretics/pharmacology , Humans , Structure-Activity Relationship
14.
Bioorg Chem ; 93: 103332, 2019 12.
Article in English | MEDLINE | ID: mdl-31593885

ABSTRACT

A series of some new tetrahydroindolocarbazole derivatives has been synthesized. The structure of the synthesized compounds has been confirmed by different spectroscopic techniques such as IR, NMR, elemental analysis and mass spectrometry. The target compounds were evaluated for their antitumor activity against breast cancer cell line MCF-7, their GI% and their LC50 have been determined. Six of the synthesized compounds exhibited GI% values against MCF-7 cell lines exceeding 70% ranging from 71.9 to 85.0% in addition that compound 11 expressed GI% values of 99.9% and considered the most active derivatives among the synthesized ones. Compound 11 showed a remarkable decrease of u PA level to 3.5 ng/ml compared to DOX. Compound 5, 11 and 15 showed significant decrease in expression of MTAP and CDKN2A, in addition to a remarkable decrease in DNA damage comet assay method. Molecular modeling studies were performed to interpretate the behavior of active ligands as uPA inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Doxorubicin/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Binding Sites , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Female , Humans , MCF-7 Cells , Models, Molecular , Urokinase-Type Plasminogen Activator/metabolism
15.
Cells ; 8(8)2019 08 18.
Article in English | MEDLINE | ID: mdl-31426601

ABSTRACT

Dysregulation of vascular networks is characteristic of eye diseases associated with retinal cell degeneration and visual loss. Visual impairment is also the consequence of photoreceptor degeneration in inherited eye diseases with a major inflammatory component, but without angiogenic profile. Among the pathways with high impact on vascular/degenerative diseases of the eye, a central role is played by a system formed by the ligand urokinase-type plasminogen activator (uPA) and its receptor uPAR. The uPAR system, although extensively investigated in tumors, still remains a key issue in vascular diseases of the eye and even less studied in inherited retinal pathologies such as retinitis pigmantosa (RP). Its spectrum of action has been extended far beyond a classical pro-angiogenic function and has emerged as a central actor in inflammation. Preclinical studies in more prevalent eye diseases characterized by neovascular formation, as in retinopathy of prematurity, wet macular degeneration and rubeosis iridis or vasopermeability excess as in diabetic retinopathy, suggest a critical role of increased uPAR signaling indicating the potentiality of its modulation to counteract neovessel formation and microvascular dysfunction. The additional observation that the uPAR system plays a major role in RP by limiting the inflammatory cascade triggered by rod degeneration rises further questions about its role in the diseased eye.


Subject(s)
Inflammation/drug therapy , Neovascularization, Pathologic/drug therapy , Receptors, Urokinase Plasminogen Activator , Retinal Diseases , Urokinase-Type Plasminogen Activator , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Receptors, Urokinase Plasminogen Activator/physiology , Retinal Diseases/drug therapy , Retinal Diseases/metabolism , Retinal Diseases/pathology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/physiology
16.
J Cell Mol Med ; 23(8): 5176-5192, 2019 08.
Article in English | MEDLINE | ID: mdl-31251468

ABSTRACT

Retinitis pigmentosa (RP) is characterized by progressive loss of vision due to photoreceptor degeneration leading to secondary inflammation. The urokinase-type plasminogen activator (uPA) system contributes to retinal inflammation, but its role in RP is unknown. In the rd10 mouse model of RP, we addressed this question with the use of the peptide UPARANT designed to interact with the uPA system. UPARANT was systemically administered from post-natal day (PD) 10 to PD30 when its efficacy in RP rescue was investigated using electroretinographic recordings, Western blot and immunocytochemistry. Temporal profile of protein expression in the uPA system was also investigated. UPARANT reduced both Müller cell gliosis and up-regulated levels of inflammatory markers and exerted major anti-apoptotic effects without influencing the autophagy cascade. Rescue from retinal cell degeneration was accompanied by improved retinal function. No scotopic phototransduction was rescued in the UPARANT-treated animals as determined by the kinetic analysis of rod-mediated a-waves and confirmed by rod photoreceptor markers. In contrast, the cone photopic b-wave was recovered and its rescue was confirmed in the whole mounts using cone arrestin antibody. Investigation of the uPA system regulation over RP progression revealed extremely low levels of uPA and its receptor uPAR both of which were recovered by HIF-1α stabilization indicating that HIF-1 regulates the expression of the uPA/uPAR gene in the retina. Ameliorative effects of UPARANT were likely to occur through an inhibitory action on up-regulated activity of the αvß3 integrin/Rac1 pathway that was suggested as a novel target for the development of therapeutic approaches against RP.


Subject(s)
Oligopeptides/pharmacology , Retinal Degeneration/drug therapy , Retinitis Pigmentosa/drug therapy , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Disease Models, Animal , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Mice , Receptors, Urokinase Plasminogen Activator/genetics , Retina/drug effects , Retina/pathology , Retinal Cone Photoreceptor Cells/drug effects , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Urokinase-Type Plasminogen Activator/genetics
17.
Invest Ophthalmol Vis Sci ; 60(5): 1571-1580, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30995314

ABSTRACT

Purpose: To understand the role and further dissect pathways downstream of tissue plasminogen activator (tPA) and the fibrinolytic pathway in modulating outflow facility. Methods: Outflow facility of tissue plasminogen activator (Plat) knockout (KO) mice was determined and compared to that of wild-type (WT) littermates. Gene expression of urokinase plasminogen activator (Plau), plasminogen activator inhibitor (Pai-1), plasminogen (Plg), and matrix metalloproteinases (Mmp-2, -9, and -13) was measured in angle tissues. Expression of the same genes and outflow facility were measured in KO and WT mice treated with triamcinolone acetonide (TA). Amiloride was used to inhibit urokinase plasminogen activator (uPA) in Plat KO mice, and outflow facility was measured. Results: Plat deletion resulted in outflow facility reduction and decreased Mmp-9 expression in angle tissues. Plasminogen expression was undetectable in both KO and WT mice. TA led to further reduction in outflow facility and decreases in expression of Plau and Mmp-13 in plat KO mice. Amiloride inhibition of uPA activity prevented the TA-induced outflow facility reduction in Plat KO mice. Conclusions: tPA deficiency reduced outflow facility in mice and was associated with reduced MMP expression. The mechanism of action of tPA is unlikely to involve plasminogen activation. tPA is not the only mediator of TA-induced outflow facility change, as TA caused reduction in outflow facility of Plat KO mice. uPA did not substitute for tPA in outflow facility regulation but abrogated the effect of TA in the absence of tPA, suggesting a complex role of components of the fibrinolytic system in outflow regulation.


Subject(s)
Fibrinolysis/physiology , Plasminogen Activator Inhibitor 1/physiology , Plasminogen/physiology , Tissue Plasminogen Activator/physiology , Trabecular Meshwork/metabolism , Urokinase-Type Plasminogen Activator/physiology , Amiloride/pharmacology , Animals , Diuretics/pharmacology , Gene Expression Regulation/physiology , Glucocorticoids/pharmacology , Injections, Intraocular , Intraocular Pressure/physiology , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Trabecular Meshwork/drug effects , Triamcinolone Acetonide/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
18.
Pharmacol Res ; 143: 73-85, 2019 05.
Article in English | MEDLINE | ID: mdl-30862605

ABSTRACT

Advanced hepatocellular carcinoma (HCC) is a highly aggressive malignancy that is a serious threat to the public health system of China. Urokinase-plasminogen activator (uPA) can promote the invasive growth and metastasis of HCC cells by activating matrix metalloproteinases (MMPs), leading to the breakage of the extra-cellular matrix. uPA is a promising target for advanced HCC treatment. In this stuy the expression of uPA was examined by quantitative polymerase chain reaction in hepatic cell lines. Protein interaction between uPA and SPINK13 was identified by immunoprecipitation. In vitro biochemical assay was used to examine the inhibitory effect of the SPINK13 on the direct cleaving of the recombinant pro-MMP9 by uPA. The antitumor effect of SPINK13 was examined by transwell assay or the nude mice tumor model.The expression of uPA was much higher in highly aggressive HCC cell lines than in lowly aggressive HCC cell lines or non-tumor hepatic cell lines. SPINK13 interacted with uPA in HCC cells and directly inhibited the cleaving of MMP9 by uPA. Treatment of the recombinant SPINK13 protein inhibited the invasion of HCC cells in several experiments, such as transwell experiments or the intrahepatic growth model. The results of the study indicated that SPINK13 could function as a promising therapeutic approach for patients with advanced HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Serine Peptidase Inhibitors, Kazal Type/therapeutic use , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Nude , Molecular Targeted Therapy , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Serine Peptidase Inhibitors, Kazal Type/genetics , Serine Peptidase Inhibitors, Kazal Type/metabolism , Serine Peptidase Inhibitors, Kazal Type/pharmacology , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Wound Healing/drug effects
20.
J Med Chem ; 62(4): 2172-2183, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30707839

ABSTRACT

Urokinase-type plasminogen activator (uPA) is a diagnostic marker for breast and prostate cancers recommended by American Society for Clinical Oncology and German Breast Cancer Society. Inhibition of uPA was proposed as an efficient strategy for cancer treatments. In this study, we report peptide-based uPA inhibitors with high potency and specificity comparable to monoclonal antibodies. We revealed the binding and inhibitory mechanisms by combining crystallography, molecular dynamic simulation, and other biophysical and biochemical approaches. Besides, we showed that our peptides efficiently inhibited the invasion of cancer cells via intervening with the processes of the degradation of extracellular matrices. Furthermore, our peptides significantly suppressed the tumor growth and the cancer metastases in tumor-bearing mice. This study demonstrates that these uPA peptides are highly potent anticancer agents and reveals the mechanistic insights of these uPA inhibitors, which can be useful for developing other serine protease inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Peptides, Cyclic/therapeutic use , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Carcinoma/pathology , Cell Line, Tumor , Colon/pathology , Colonic Neoplasms/pathology , Lung Neoplasms/secondary , Male , Mice, Inbred BALB C , Molecular Dynamics Simulation , Neoplasm Invasiveness , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL