Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Diabetes Obes Metab ; 25(11): 3152-3160, 2023 11.
Article in English | MEDLINE | ID: mdl-37417375

ABSTRACT

AIM: To evaluate the effect of four different drug classes on soluble urokinase plasminogen activator receptor (suPAR), a biomarker active in multiple inflammatory processes and a risk factor for complications, in people with type 1 and type 2 diabetes. METHODS: We conducted post hoc analyses of a randomized, open-label, crossover trial including 26 adults with type 1 and 40 with type 2 diabetes with urinary albumin-creatinine ratio ≥30 and ≤500 mg/g assigned to 4-week treatments with telmisartan 80 mg, empagliflozin 10 mg, linagliptin 5 mg and baricitinib 2 mg, separated by 4-week washouts. Plasma suPAR was measured before and after each treatment. SuPAR change after each treatment was calculated and, for each individual, the best suPAR-reducing drug was identified. Subsequently, the effect of the best individual drug was compared against the mean of the other three drugs. Repeated-measures linear mixed-effects models were employed. RESULTS: The baseline median (interquartile range) plasma suPAR was 3.5 (2.9, 4.3) ng/mL. No overall effect on suPAR levels was observed for any one drug. The individual best-performing drug varied, with baricitinib being selected for 20 participants (30%), followed by empagliflozin for 19 (29%), linagliptin for 16 (24%) and telmisartan for 11 (17%). The individual best-performing drug reduced suPAR by 13.3% (95% confidence interval [CI] 3.7, 22.8; P = 0.007). The difference in suPAR response between the individual best-performing drug and the other three was -19.7% (95% CI -23.1, -16.3; P < 0.001). CONCLUSIONS: We demonstrated no overall effect of 4-week treatment with telmisartan, empagliflozin, linagliptin or baricitinib on suPAR. However, individualization of treatment might significantly reduce suPAR levels.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Receptors, Urokinase Plasminogen Activator , Adult , Humans , Biomarkers , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Linagliptin/therapeutic use , Receptors, Urokinase Plasminogen Activator/drug effects , Telmisartan/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism
2.
Vascul Pharmacol ; 110: 49-54, 2018 11.
Article in English | MEDLINE | ID: mdl-30098417

ABSTRACT

BACKGROUND AND AIMS: Macrophages are versatile immune cells involved in tissue degradation and remodeling. Proinflammatory macrophages have the highest capacity of matrix degradation and proteolysis. Within atherosclerotic lesions, proinflammatory macrophages are associated with unstable plaques. Statins have been demonstrated to increase plaque stability. Possible changes of polarized macrophage tissue degradation behavior under statin treatment are currently unknown. METHODS: Polarized macrophages were tested in vitro for matrix degradation capacity with or without statin treatment. RESULTS: Proinflammatory macrophages show high matrix degradation capacity, which is lost after statin treatment. Statin concentrations were within a physiological range and did not influence overall macrophage polarization. Proinflammatory macrophages showed however a loss of filopodia where activators of MMPs are located. Loss of matrix degradation in proinflammatory macrophages was associated with changes of MMP14 activation and loss of uPAR localization at filopodia. Supplementation of mevalonate restored localization of uPAR to cellular protrusions and matrix degradation capacity. CONCLUSION: Statins reduce the matrix degradation potential of proinflammatory macrophages by reducing uPAR localization to cellular filopodia and reducing intracellular MMP14 activation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Atorvastatin/pharmacology , Cell Plasticity , Extracellular Matrix/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation/drug therapy , Macrophages/drug effects , Cells, Cultured , Extracellular Matrix/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Macrophages/metabolism , Matrix Metalloproteinase 14/metabolism , Phenotype , Proteolysis/drug effects , Pseudopodia/drug effects , Pseudopodia/metabolism , Receptors, Urokinase Plasminogen Activator/drug effects , Receptors, Urokinase Plasminogen Activator/metabolism
3.
Diabetes Obes Metab ; 19(6): 901-905, 2017 06.
Article in English | MEDLINE | ID: mdl-28105731

ABSTRACT

We assessed the effects of liraglutide treatment on five cardiovascular risk biomarkers, reflecting different pathophysiology: tumour necrosis factor (TNF)-α; soluble urokinase plasminogen activator receptor (suPAR); mid-regional pro-adrenomedullin (MR-proADM); mid-regional pro-atrial natriuretic peptide (MR-proANP); and copeptin, in people with type 2 diabetes with albuminuria. In a randomized, double-blind, placebo-controlled, crossover trial we enrolled people with type 2 diabetes and persistent albuminuria (urinary albumin-to-creatinine ratio [UACR] >30 mg/g) and estimated glomerular filtration rate (eGFR) ≥30 mL/min/1.73 m2 . Participants received liraglutide (1.8 mg/d) and matched placebo for 12 weeks, in random order. The primary endpoint was change in albuminuria; this was a prespecified sub-study. A total of 32 participants were randomized, of whom 27 completed the study. TNF-α level was 12% (95% confidence interval [CI] 3; 20) lower after liraglutide treatment compared with placebo (P = .012); MR-proADM level was 4% (95% CI 0; 8) lower after liraglutide treatment compared with placebo (P = .038), and MR-proANP level was 13% (95% CI 4; 21) lower after liraglutide treatment compared with placebo (P = .006). In the present study, we showed anti-inflammatory effects of liraglutide treatment, reflected in reductions in levels of TNF-α and MR-proADM, while the reduction in MR-proANP levels may represent a clinically relevant benefit with regard to heart failure.


Subject(s)
Albuminuria/blood , Cardiovascular Diseases/etiology , Diabetes Mellitus, Type 2/blood , Hypoglycemic Agents/therapeutic use , Liraglutide/therapeutic use , Adrenomedullin/blood , Adrenomedullin/drug effects , Aged , Albuminuria/drug therapy , Albuminuria/etiology , Atrial Natriuretic Factor/blood , Atrial Natriuretic Factor/drug effects , Biomarkers/blood , Cross-Over Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Double-Blind Method , Female , Glycopeptides/blood , Glycopeptides/drug effects , Humans , Male , Middle Aged , Peptide Fragments/blood , Peptide Fragments/drug effects , Protein Precursors/blood , Protein Precursors/drug effects , Receptors, Urokinase Plasminogen Activator/blood , Receptors, Urokinase Plasminogen Activator/drug effects , Risk Factors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/drug effects
4.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 41(11): 1124-1127, 2016 Nov 28.
Article in Chinese | MEDLINE | ID: mdl-27932755

ABSTRACT

OBJECTIVE: To investigate the effect of apigenin on self-renewal for sphere-forming cells in human small cell lung cancer cell line NCI-H446 and the underlying mechanisms.
 Methods: Sphere-forming cells from NCI-H446 cell line were cultured in stem cell-conditioned culture medium with ultra-low attachment surface plates. The rate of sphere-forming cells in the second passage sphere-forming cells was used to evaluate the inhibitory effects of apigenin on the self-renewal for sphere-forming cells. The protein level of urokinase-type plasminogen activator receptor (uPAR) in spheroids was analyzed by Western blot.
 Results: Apigenin signifcantly inhibited the self-renewal of the second passage sphere-forming cells [0, 5.0, 10.0, 20.0 µmol/L apigenin: (18.2±1.9)%, (13.6±1.7)%, (10.6±1.6)%, (6.9±1.3)%, respectively] and down-regulated uPAR expression in a concentration-dependent manner (P<0.05).
 Conclusion: Apigenin inhibits the self-renewal capacity of sphere-forming cells in NCI-H446 cells, which may be associated with down-regulation of uPAR expression.


Subject(s)
Apigenin/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/physiology , Receptors, Urokinase Plasminogen Activator/metabolism , Small Cell Lung Carcinoma/pathology , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/genetics , Humans , Lung Neoplasms , Neoplastic Stem Cells/pathology , Receptors, Cell Surface , Receptors, Urokinase Plasminogen Activator/drug effects , Receptors, Urokinase Plasminogen Activator/genetics , Signal Transduction , Small Cell Lung Carcinoma/drug therapy , Spheroids, Cellular/drug effects , Spheroids, Cellular/physiology , Stem Cells
5.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 41(12): 1297-1304, 2016 Dec 28.
Article in Chinese | MEDLINE | ID: mdl-28070042

ABSTRACT

OBJECTIVE: To investigate the effect of the jianpi-jiedu formula (JPJD) on the expression of angiogenesis-relevant genes in colon cancer.
 Methods: Crude extract was obtained from JPJD by water extract method. The effect of JPJD crude extract on colon cancer cell proliferation capacity was determined by MTT assays. The IC50 value was calculated by GraphPad Prism5 software. Affymetrix gene expression profiling chip was used to detect significant differences in expressions of genes after JPJD intervention, and pathway enrichment analysis was performed to analyze the differentially expressed genes. Ingenuity Pathway Analysis software was applied to analyze differentially expressed genes relevant to tumor angiogenesis based on mammalian target of rapamycin (mTOR) signaling pathway and then the network diagram was built. Western blot was used to verify the protein levels of key genes related to tumor angiogenesis.
 Results: JPJD crud extract inhibited the proliferation capacity in colon cancer cells. The IC50 values in 24, 48, and 72 hours after treatment were 13.060, 9.646 and 8.448 mg/mL, respectively. The results of chip showed that 218 genes significantly upgraded, and 252 genes significantly downgraded after JPJD treatment. Most of the genes were related to the function of biosynthesis, metabolism, cell apoptosis, antigen extraction, angiogenesis and so on. There were 12 differentially expressed angiogenesis genes. IPA software analysis showed that the JPJD downregulated expression of sphingomyelin phosphodiesterase 3 (SMPD3), VEGF, vascular endothelial growth factor A (VEGFA), integrin subunit alpha 1 (ITGA1), cathepsin B (CTSB), and cathepsin S (CTSS) genes, while upregulated expressions of GAB2 and plasminogen activator, urokinase receptor (PLAUR) genes in the colorectal cancer cell. Western blot results demonstrated that JPJD obviously downregulated expressions of phospho-mTOR (P-mTOR), signal transducer and activator of transcription 3 (STAT3), hypoxia inducible factor-1α (HIF-1α), and VEGF proteins, while obviously upregulated the level of phospho-P53 (P-P53) protein.
 Conclusion: JPJD may inhibit colorectal tumor angiogenesis through regulation of the mTOR-HIF-1α-VEGF signal pathway.


Subject(s)
Cell Line, Tumor/drug effects , Colorectal Neoplasms/genetics , Drugs, Chinese Herbal/pharmacology , Animals , Blotting, Western , Cathepsin B/drug effects , Cathepsin B/metabolism , Cathepsins/drug effects , Cathepsins/metabolism , Colorectal Neoplasms/blood supply , Down-Regulation , Gene Expression Profiling/methods , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Integrin alpha Chains/drug effects , Integrin alpha Chains/metabolism , Neovascularization, Pathologic/genetics , Receptors, Urokinase Plasminogen Activator/drug effects , Receptors, Urokinase Plasminogen Activator/metabolism , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction , Sphingomyelin Phosphodiesterase/drug effects , Sphingomyelin Phosphodiesterase/metabolism , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism
6.
Prog Brain Res ; 214: 229-62, 2014.
Article in English | MEDLINE | ID: mdl-25410361

ABSTRACT

Currently, there are about 20 antiepileptic drugs on market. Still, seizures in about 30% of patients with epilepsy are not adequately controlled, or the drugs cause quality-of-life-compromising adverse events. Importantly, there are no treatments to combat epileptogenesis, a process that leads to the development of epilepsy and its progression. To fill the gaps in the treatment of epilepsy, there is an urgent need for identification of novel treatment targets. Data emerging over the recent years have shown that different components of the extracellular matrix (ECM) contribute to many components of tissue reorganization during epileptogenesis and the ECM is also a major regulator of synaptic excitability. Here, we review the role of urokinase-type plasminogen activator receptor interactome, matrix metalloproteinases, tenascin-R, and LGI1 in epileptogenesis and ictogenesis. Moreover, the role of the ECM in epilepsy-related comorbidities is reviewed. As there is active development of new imaging methods, we also summarize the data available on imaging of the ECM in epilepsy.


Subject(s)
Epilepsy/pathology , Extracellular Matrix/physiology , Receptors, Urokinase Plasminogen Activator/metabolism , Animals , Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Epilepsy/metabolism , Extracellular Matrix/drug effects , Humans , Receptors, Urokinase Plasminogen Activator/drug effects
7.
Transplant Proc ; 46(4): 1226-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24815166

ABSTRACT

Conversion to rapamycin from calcineurin inhibitors may contribute to improvement of graft function in kidney transplant recipients, especially in patients with calcineurin inhibitor-related nephrotoxicity. The conversion from calcineurin inhibitors to rapamycin in kidney transplant recipients has been associated with a higher incidence of proteinuria. It could be explained by possible hemodynamic changes due to withdrawal of calcineurin inhibitors. Podocyte damage occurs commonly in rapamycin-related proteinuria. The vascular endothelial growth factor system has been suggested to be implicated in mammalian target of rapamycin inhibitor-associated proteinuria. However, induction of urokinase receptor (uPAR) signaling in podocytes leads to foot process effacement and urinary protein. In this study, we assessed the role of uPAR in primary cultured podocytes with rapamycin treatment. Our results indicate that 24-hour rapamycin treatment promotes podocyte migration on the wound scratch assay in a dose-dependent manner. Rapamycin treatment for 2 days does not increase the apoptosis of podocytes or affect the podocyte cell viability and morphology. The up-regulation of uPAR in podocytes was confirmed by immunofluorescence staining, real-time polymerase chain reaction (1.8 ± 0.3-fold increase of relative quantification; P < .01) and Western blot analysis. Rapamycin treatment also causes the activation of FAK and ILK in a dose-dependent manner. In summary, rapamycin could promote podocyte migration through the up-regulation of uPAR. This finding provides a new mechanism of rapamycin-associated proteinuria. It also suggests that pharmacologic inhibition of uPAR signaling cascade may have therapeutic potential in the setting of rapamycin-related proteinuria.


Subject(s)
Cell Movement/drug effects , Immunosuppressive Agents/toxicity , Podocytes/drug effects , Receptors, Urokinase Plasminogen Activator/drug effects , Sirolimus/toxicity , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation , Focal Adhesion Kinase 1/metabolism , Podocytes/metabolism , Podocytes/pathology , Primary Cell Culture , Protein Serine-Threonine Kinases/metabolism , Proteinuria/chemically induced , Proteinuria/metabolism , Proteinuria/pathology , Rats, Wistar , Receptors, Urokinase Plasminogen Activator/metabolism , Signal Transduction/drug effects , Time Factors , Up-Regulation
8.
Oral Oncol ; 48(12): 1202-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22818892

ABSTRACT

OBJECTIVES: To investigate the efficacy of the bispecific targeted toxin, dEGFATFKDEL, on head and neck carcinoma cell lines in vitro and in vivo. MATERIALS AND METHODS: A deimmunized bispecific anti-cancer agent was constructed to simultaneously target both the overexpressed EGF receptor on carcinomas and the urokinase receptor (uPAR), that is found on the endothelial cells of the neovasculature within tumors. Flow cytometry assays were performed to determine the level of EGFR expressed on a variety of carcinoma lines. These lines were then tested in tritiated leucine incorporation assays to determine the efficacy of dEGFATFKDEL. Human vein endothelial primary cells were also tested to determine the effectiveness of the ATF portion of the molecule that binds uPAR. Furthermore, mouse studies were performed to determine whether dEGFATFKDEL was effective at inhibiting tumor growth in vivo. RESULTS: UMSCC-11B and NA, two head and neck squamous cell carcinomas, highly expressed EGFR. Both the carcinoma lines and the human vein endothelial cells were inhibited at sub-nanomolar concentrations by dEGFATFKDEL. The tumor studies showed that the tumors treated with dEGFATFKDEL were significantly inhibited whereas the negative control and untreated tumors progressed. In a separate in vivo study involving another carcinoma line, MDA-MB-231, the effectiveness of dEGFATFKDEL was confirmed. No toxicity was seen at the doses used in either of these mouse studies. CONCLUSIONS: This bispecific agent is effective in a mouse model of head and neck squamous cell carcinoma. Further study of this reagent for use in the treatment of carcinomas is warranted.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Disease Models, Animal , ErbB Receptors/drug effects , Head and Neck Neoplasms/metabolism , Oligopeptides/pharmacology , Receptors, Urokinase Plasminogen Activator/drug effects , Animals , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , ErbB Receptors/metabolism , Flow Cytometry , Head and Neck Neoplasms/pathology , Humans , Male , Mice , Mice, Nude , Oligopeptides/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism
9.
Int J Cancer ; 126(5): 1055-66, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19551865

ABSTRACT

We previously demonstrated that the CNYYSNS peptide derived from tumstatin inhibited in vivo tumor progression. The YSNS motif formed a beta-turn crucial for biological activity. More recently, a YSNSG cyclopeptide with a constrained beta-turn on the YSNS residues was designed. Intraperitoneal administration of the YSNSG cyclopeptide inhibited in vivo melanoma progression more efficiently than the native linear peptide. In the present article, we showed that the YSNSG cyclopeptide also triggered an inhibition of in vivo tumor neovascularization and we further analyzed its in vitroantiangiogenic effect. The YSNSG cyclopeptide did not alter endothelial cell proliferation but inhibited cell migration by 83% in an in vitro wound healing model. The inhibition was mediated by a decrease in active MT1-MMP at the migration front as well as a decrease in u-PA and u-PAR expression. The cyclopeptide also altered beta1-integrin distribution in endothelial cell lamellipodia, induced a strong decrease in the phosphorylated focal adhesion kinase (p125(FAK)), disorganized F-actin stress fibers and decreased the number of lamellipodia, resulting in a non migratory phenotype. Our results confirm the YSNSG cyclopeptide as a potent antitumor agent, through both the inhibition of invasive properties of tumor cells and the antiangiogenic activity.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Endothelial Cells/drug effects , Neovascularization, Pathologic/drug therapy , Peptides, Cyclic/pharmacology , Animals , Autoantigens/chemistry , Blotting, Western , Cell Proliferation/drug effects , Collagen Type IV/chemistry , Down-Regulation , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Matrix Metalloproteinase 14/drug effects , Matrix Metalloproteinase 14/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Receptors, Urokinase Plasminogen Activator/drug effects , Receptors, Urokinase Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/drug effects , Urokinase-Type Plasminogen Activator/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...