21 research outputs found

    Structural and functional characterization of HMGB1 protein in rat liver during experimentally-induced diabetes type 1

    No full text
    ΠžΠΊΡΠΈΠ΄Π°Ρ‚ΠΈΠ²Π½ΠΈ стрСс ΠΈ Ρ…Ρ€ΠΎΠ½ΠΈΡ‡Π½Π° ΠΈΠ½Ρ„Π»Π°ΠΌΠ°Ρ†ΠΈΡ˜Π° ΡΠΌΠ°Ρ‚Ρ€Π°Ρ˜Ρƒ сС Π³Π»Π°Π²Π½ΠΈΠΌ ΡƒΠ·Ρ€ΠΎΡ†ΠΈΠΌΠ° појавС Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚ΠΈΡ‡Π½ΠΈΡ… ΠΊΠΎΠΌΠΏΠ»ΠΈΠΊΠ°Ρ†ΠΈΡ˜Π°, ΠΌΠ΅Ρ’Ρƒ којима су ΠΈ ΠΎΡˆΡ‚Π΅Ρ›Π΅ΡšΠ° Ρ˜Π΅Ρ‚Ρ€Π΅. Π’Π°ΠΆΠ½Ρƒ ΡƒΠ»ΠΎΠ³Ρƒ ΠΌΠ΅Π΄ΠΈΡ˜Π°Ρ‚ΠΎΡ€Π° ΠΎΠ²ΠΈΡ… процСса ΠΌΠΎΠΆΠ΅ ΠΈΠΌΠ°Ρ‚ΠΈ Π΅Π½Π΄ΠΎΠ³Π΅Π½ΠΈ ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½ HMGB1, који Ρƒ Π²Π°Π½Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΡƒ срСдину доспСва ΠΈΠ· Π½Π΅ΠΊΡ€ΠΎΡ‚ΠΈΡ‡Π½ΠΈΡ…, ΠΎΡˆΡ‚Π΅Ρ›Π΅Π½ΠΈΡ… ΠΈ Π°ΠΊΡ‚ΠΈΠ²ΠΈΡ€Π°Π½ΠΈΡ… Ρ›Π΅Π»ΠΈΡ˜Π°. Како јС Ρƒ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚Π΅ΡΡƒ понашањС HMGB1 ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½Π° слабо ΠΈΠ·ΡƒΡ‡Π°Π²Π°Π½ΠΎ, Ρƒ овој Π΄ΠΎΠΊΡ‚ΠΎΡ€ΡΠΊΠΎΡ˜ Π΄ΠΈΡΠ΅Ρ€Ρ‚Π°Ρ†ΠΈΡ˜ΠΈ испитиван јС допринос HMGB1 ΠΎΡˆΡ‚Π΅Ρ›Π΅ΡšΠΈΠΌΠ° Ρ˜Π΅Ρ‚Ρ€Π΅ ΠΏΠ°Ρ†ΠΎΠ²Π° са стрСптозотоцином-ΠΈΠ·Π°Π·Π²Π°Π½ΠΈΠΌ Π”Π’1. Показано јС Π΄Π° Π½ΠΈΠ²ΠΎ ΠΎΡˆΡ‚Π΅Ρ›Π΅ΡšΠ° Ρ˜Π΅Ρ‚Ρ€Π΅ Ρ‚ΠΎΠΊΠΎΠΌ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚Π΅Ρa ΠΊΠΎΡ€Π΅Π»ΠΈΡˆΠ΅ са присуством Π²Π°Π½Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΠΎΠ³ HMGB1. Овај ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½, Ρƒ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚ΠΈΡ‡Π½ΠΎΡ˜ Ρ˜Π΅Ρ‚Ρ€ΠΈ, Π±ΠΈΠ²Π° структурно ΠΌΠΎΠ΄ΠΈΡ„ΠΈΠΊΠΎΠ²Π°Π½ Π°Ρ†Π΅Ρ‚ΠΈΠ»Π°Ρ†ΠΈΡ˜ΠΎΠΌ, Ρ„ΠΎΡΡ„ΠΎΡ€ΠΈΠ»Π°Ρ†ΠΈΡ˜ΠΎΠΌ ΠΈ O-GlcNAc Π³Π»ΠΈΠΊΠΎΠ·ΠΈΠ»Π°Ρ†ΠΈΡ˜ΠΎΠΌ ΡˆΡ‚ΠΎ ΠΊΠΎΡ€Π΅Π»ΠΈΡˆΠ΅ са њСговим изласком ΠΈΠ· Ρ˜Π΅Π΄Ρ€Π° Ρ›Π΅Π»ΠΈΡ˜Π° Ρƒ Ρ†ΠΈΡ‚ΠΎΠΏΠ»Π°Π·ΠΌΡƒ ΠΈ ΠΏΠΎΠ²Π΅Ρ›Π°ΡšΠ΅ΠΌ њСговог присуства Ρƒ Ρ˜Π΅Ρ‚Ρ€ΠΈ ΠΈ сСруму. Π Π΅Π·ΡƒΠ»Ρ‚Π°Ρ‚ΠΈ Ρƒ Π²Π΅Π·ΠΈ са сниТавањeΠΌ Π½ΠΈΠ²ΠΎΠ° Π²Π°Π½Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΠΎΠ³ HMGB1 Ρ‚Ρ€Π΅Ρ‚ΠΌΠ°Π½ΠΎΠΌ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚ΠΈΡ‡Π½ΠΈΡ… ΠΏΠ°Ρ†ΠΎΠ²Π° ΠΌΠ΅Π»Π°Ρ‚ΠΎΠ½ΠΈΠ½ΠΎΠΌ ΠΈΠ»ΠΈ Π΅Ρ‚ΠΈΠ» ΠΏΠΈΡ€ΡƒΠ²Π°Ρ‚ΠΎΠΌ, ΡƒΠΊΠ°Π·ΡƒΡ˜Ρƒ Π΄Π° HMGB1 доприноси ΠΎΡˆΡ‚Π΅Ρ›Π΅ΡšΡƒ Ρ˜Π΅Ρ‚Ρ€Π΅ Ρƒ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚Π΅ΡΡƒ ΠΎΠ΄Ρ€ΠΆΠ°Π²Π°ΡšΠ΅ΠΌ ΡΡ‚Π°ΡšΠ° Ρ…Ρ€ΠΎΠ½ΠΈΡ‡Π½Π΅ ΠΈΠ½Ρ„Π»Π°ΠΌΠ°Ρ†ΠΈΡ˜Π΅, ΡΡ‚ΠΈΡˆΠ°Π²Π°ΡšΠ΅ΠΌ антиоксидативнС ΠΎΠ΄Π±Ρ€Π°Π½Π΅ ΠΈ ΡΡ‚ΠΈΡˆΠ°Π²Π°ΡšΠ΅ΠΌ Ρ€Π΅Π³Π΅Π½Π΅Ρ€Π°Ρ†ΠΈΡ˜Π΅. Π’Π°Π½Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΠΈ HMGB1 ΠΊΡ€ΠΎΠ· ΠΈΠ½Ρ‚Π΅Ρ€Π°ΠΊΡ†ΠΈΡ˜Π΅ са TLR4 Ρ€Π΅Ρ†Π΅ΠΏΡ‚ΠΎΡ€ΠΎΠΌ Π°ΠΊΡ‚ΠΈΠ²ΠΈΡ€Π° MAPК/NF-ΞΊB p65 ΠΈ ЈАК1/STAT3 сигналнС ΠΏΡƒΡ‚Π΅Π²Π΅, доприносСћи ΠΏΠΎΠ²Π΅Ρ›Π°ΡšΡƒ ΠΏΡ€ΠΎΠ΄ΡƒΠΊΡ†ΠΈΡ˜Π΅ ΠΏΡ€ΠΎΠΈΠ½Ρ„Π»Π°ΠΌΠ°Ρ†ΠΈΡ˜ΡΠΊΠΈΡ… Ρ†ΠΈΡ‚ΠΎΠΊΠΈΠ½Π° TNF-Ξ± ΠΈ IL-6 ΠΈ Π°ΠΊΡƒΡ‚Π½ΠΎ-Ρ„Π°Π·Π½ΠΎΠ³ ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½Π° Ρ…Π°ΠΏΡ‚ΠΎΠ³Π»ΠΎΠ±ΠΈΠ½Π°. ΠŸΠΎΠ΄ΡΡ‚ΠΈΡ†Π°ΡšΠ΅ΠΌ NF-ΞΊB p65 ΠΈΠ½Ρ„Π»Π°ΠΌΠ°Ρ†ΠΈΡ˜ΡΠΊΠΎΠ³ ΠΏΡƒΡ‚Π°, HMGB1 Π΄Π΅Π»ΡƒΡ˜Π΅ Π½Π΅Π³Π°Ρ‚ΠΈΠ²Π½ΠΎ Π½Π° Ρ†ΠΈΡ‚ΠΎΠΏΡ€ΠΎΡ‚Π΅ΠΊΡ‚ΠΈΠ²Π½ΠΈ ΠΎΠ΄Π³ΠΎΠ²ΠΎΡ€ Ρƒ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚ΠΈΡ‡Π½ΠΎΡ˜ Ρ˜Π΅Ρ‚Ρ€ΠΈ Ρ‚Π°ΠΊΠΎ ΡˆΡ‚ΠΎ ΠΎΠ½Π΅ΠΌΠΎΠ³ΡƒΡ›Π°Π²Π° активност Nrf2 ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½Π°, ΠΎΠ΄Π³ΠΎΠ²ΠΎΡ€Π½ΠΎΠ³ Π·Π° ΡΡ‚ΠΈΡˆΠ°Π²Π°ΡšΠ΅ ΠΈΠ½Ρ„Π»Π°ΠΌΠ°Ρ†ΠΈΡ˜Π΅ ΠΈ ΠΏΡ€ΠΎΠ΄ΡƒΠΊΡ†ΠΈΡ˜Ρƒ антиоксидативних Π΅Π½Π·ΠΈΠΌΠ°. На ΡΡ‚ΠΈΡˆΠ°Π²Π°ΡšΠ΅ Ρ€Π΅Π³Π΅Π½Π΅Ρ€Π°Ρ‚ΠΈΠ²Π½ΠΎΠ³ ΠΏΠΎΡ‚Π΅Π½Ρ†ΠΈΡ˜Π°Π»Π° Ρ˜Π΅Ρ‚Ρ€Π΅, Π°ΠΊΡ‚ΠΈΠ²ΠΈΡ€Π°Π½Π° HMGB1/TLR4 оса ΡƒΡ‚ΠΈΡ‡Π΅ ΠΏΡ€Π΅ΠΊΠΎ ΡƒΠ²Π΅Ρ›Π°ΡšΠ° присуства Π½Π΅Π³Π°Ρ‚ΠΈΠ²Π½ΠΈΡ… Ρ€Π΅Π³ΡƒΠ»Π°Ρ‚ΠΎΡ€Π° Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΠΎΠ³ циклуса - ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½Π° p53 ΠΈ p21, ΠΈ смањСњСм Π½ΠΈΠ²ΠΎΠ° Ρ†ΠΈΠΊΠ»ΠΈΠ½Π° D1. Π”ΠΎΠ±ΠΈΡ˜Π΅Π½ΠΈ Ρ€Π΅Π·ΡƒΠ»Ρ‚Π°Ρ‚ΠΈ ΡƒΠΊΠ°Π·ΡƒΡ˜Ρƒ Π½Π° слоТСност дСловања HMGB1 ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½Π° Ρƒ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚Π΅ΡΡƒ ΠΈ Π½Π° Π·Π½Π°Ρ‡Π°Ρ˜ ΡΠΏΡ€Π΅Ρ‡Π°Π²Π°ΡšΠ° ΠΎΡΠ»ΠΎΠ±Π°Ρ’Π°ΡšΠ° HMGB1 ΠΈΠ»ΠΈ Π±Π»ΠΎΠΊΠ°Π΄Π΅ HMGB1/TLR4 осС Ρƒ Ρ†ΠΈΡ™Ρƒ одлагања настанка ΠΎΡˆΡ‚Π΅Ρ›Π΅ΡšΠ° Ρ˜Π΅Ρ‚Ρ€Π΅.Oxidative stress and chronic inflammation are considered to be the main causes of diabetic complications, one of which is liver damage. An important mediator of these processes may be the endogenous HMGB1 protein, when released into the extracellular environment from the necrotic, damaged or activated cells. As the HMGB1 role in diabetes was insufficiently studied, in this doctoral dissertation the contribution of HMGB1 to liver damage of streptozotocin-induced diabetic rats was investigated. It has been shown that the level of liver damage in diabetes correlates with the presence of extracellular HMGB1. In diabetic liver, this protein is structurally modified by acetylation, phosphorylation, and O-GlcNAc glycosylation, which correlates with its translocation from the nucleus to the cytoplasm and an increase in its presence in the liver and serum. Reduction of the level of extracellular HMGB1 by melatonin or ethyl pyruvate treatment of diabetic rats, shows that HMGB1 contributes to diabetic liver damage by maintaining a chronic inflammation, by lowering antioxidant defense and by reducing regeneration. Extracellular HMGB1 activates MAPK/NF-ΞΊB p65 and JAK1/STAT3 signaling pathways through interactions with the TLR4 receptor, thus contributing increased production of proinflammatory cytokines TNF-Ξ± and IL-6 and the acute-phase protein, haptoglobin. By stimulating the NF-ΞΊB p65 inflammatory pathway, HMGB1 acts negatively on the cytoprotective response of the diabetic liver, by disabling Nrf2 protein activity, which is responsible for reduction of inflammation and antioxidant enzymes production. Activated HMGB1/TLR4 axis reduces regenerative potential of the liver by increasing the presence of negative cell cycle regulators - proteins p53 and p21, and also by decreasing the level of cyclin D1. The obtained results indicate the complexity of HMGB1 protein action in diabetes and underlines the importance of preventing the release of HMGB1 or blockage of HMGB1/TLR4 axis in order to delay the occurrence of liver damage

    Structural and functional characterization of HMGB1 protein in rat liver during experimentally-induced diabetes type 1

    Get PDF
    ΠžΠΊΡΠΈΠ΄Π°Ρ‚ΠΈΠ²Π½ΠΈ стрСс ΠΈ Ρ…Ρ€ΠΎΠ½ΠΈΡ‡Π½Π° ΠΈΠ½Ρ„Π»Π°ΠΌΠ°Ρ†ΠΈΡ˜Π° ΡΠΌΠ°Ρ‚Ρ€Π°Ρ˜Ρƒ сС Π³Π»Π°Π²Π½ΠΈΠΌ ΡƒΠ·Ρ€ΠΎΡ†ΠΈΠΌΠ° појавС Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚ΠΈΡ‡Π½ΠΈΡ… ΠΊΠΎΠΌΠΏΠ»ΠΈΠΊΠ°Ρ†ΠΈΡ˜Π°, ΠΌΠ΅Ρ’Ρƒ којима су ΠΈ ΠΎΡˆΡ‚Π΅Ρ›Π΅ΡšΠ° Ρ˜Π΅Ρ‚Ρ€Π΅. Π’Π°ΠΆΠ½Ρƒ ΡƒΠ»ΠΎΠ³Ρƒ ΠΌΠ΅Π΄ΠΈΡ˜Π°Ρ‚ΠΎΡ€Π° ΠΎΠ²ΠΈΡ… процСса ΠΌΠΎΠΆΠ΅ ΠΈΠΌΠ°Ρ‚ΠΈ Π΅Π½Π΄ΠΎΠ³Π΅Π½ΠΈ ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½ HMGB1, који Ρƒ Π²Π°Π½Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΡƒ срСдину доспСва ΠΈΠ· Π½Π΅ΠΊΡ€ΠΎΡ‚ΠΈΡ‡Π½ΠΈΡ…, ΠΎΡˆΡ‚Π΅Ρ›Π΅Π½ΠΈΡ… ΠΈ Π°ΠΊΡ‚ΠΈΠ²ΠΈΡ€Π°Π½ΠΈΡ… Ρ›Π΅Π»ΠΈΡ˜Π°. Како јС Ρƒ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚Π΅ΡΡƒ понашањС HMGB1 ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½Π° слабо ΠΈΠ·ΡƒΡ‡Π°Π²Π°Π½ΠΎ, Ρƒ овој Π΄ΠΎΠΊΡ‚ΠΎΡ€ΡΠΊΠΎΡ˜ Π΄ΠΈΡΠ΅Ρ€Ρ‚Π°Ρ†ΠΈΡ˜ΠΈ испитиван јС допринос HMGB1 ΠΎΡˆΡ‚Π΅Ρ›Π΅ΡšΠΈΠΌΠ° Ρ˜Π΅Ρ‚Ρ€Π΅ ΠΏΠ°Ρ†ΠΎΠ²Π° са стрСптозотоцином-ΠΈΠ·Π°Π·Π²Π°Π½ΠΈΠΌ Π”Π’1. Показано јС Π΄Π° Π½ΠΈΠ²ΠΎ ΠΎΡˆΡ‚Π΅Ρ›Π΅ΡšΠ° Ρ˜Π΅Ρ‚Ρ€Π΅ Ρ‚ΠΎΠΊΠΎΠΌ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚Π΅Ρa ΠΊΠΎΡ€Π΅Π»ΠΈΡˆΠ΅ са присуством Π²Π°Π½Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΠΎΠ³ HMGB1. Овај ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½, Ρƒ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚ΠΈΡ‡Π½ΠΎΡ˜ Ρ˜Π΅Ρ‚Ρ€ΠΈ, Π±ΠΈΠ²Π° структурно ΠΌΠΎΠ΄ΠΈΡ„ΠΈΠΊΠΎΠ²Π°Π½ Π°Ρ†Π΅Ρ‚ΠΈΠ»Π°Ρ†ΠΈΡ˜ΠΎΠΌ, Ρ„ΠΎΡΡ„ΠΎΡ€ΠΈΠ»Π°Ρ†ΠΈΡ˜ΠΎΠΌ ΠΈ O-GlcNAc Π³Π»ΠΈΠΊΠΎΠ·ΠΈΠ»Π°Ρ†ΠΈΡ˜ΠΎΠΌ ΡˆΡ‚ΠΎ ΠΊΠΎΡ€Π΅Π»ΠΈΡˆΠ΅ са њСговим изласком ΠΈΠ· Ρ˜Π΅Π΄Ρ€Π° Ρ›Π΅Π»ΠΈΡ˜Π° Ρƒ Ρ†ΠΈΡ‚ΠΎΠΏΠ»Π°Π·ΠΌΡƒ ΠΈ ΠΏΠΎΠ²Π΅Ρ›Π°ΡšΠ΅ΠΌ њСговог присуства Ρƒ Ρ˜Π΅Ρ‚Ρ€ΠΈ ΠΈ сСруму. Π Π΅Π·ΡƒΠ»Ρ‚Π°Ρ‚ΠΈ Ρƒ Π²Π΅Π·ΠΈ са сниТавањeΠΌ Π½ΠΈΠ²ΠΎΠ° Π²Π°Π½Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΠΎΠ³ HMGB1 Ρ‚Ρ€Π΅Ρ‚ΠΌΠ°Π½ΠΎΠΌ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚ΠΈΡ‡Π½ΠΈΡ… ΠΏΠ°Ρ†ΠΎΠ²Π° ΠΌΠ΅Π»Π°Ρ‚ΠΎΠ½ΠΈΠ½ΠΎΠΌ ΠΈΠ»ΠΈ Π΅Ρ‚ΠΈΠ» ΠΏΠΈΡ€ΡƒΠ²Π°Ρ‚ΠΎΠΌ, ΡƒΠΊΠ°Π·ΡƒΡ˜Ρƒ Π΄Π° HMGB1 доприноси ΠΎΡˆΡ‚Π΅Ρ›Π΅ΡšΡƒ Ρ˜Π΅Ρ‚Ρ€Π΅ Ρƒ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚Π΅ΡΡƒ ΠΎΠ΄Ρ€ΠΆΠ°Π²Π°ΡšΠ΅ΠΌ ΡΡ‚Π°ΡšΠ° Ρ…Ρ€ΠΎΠ½ΠΈΡ‡Π½Π΅ ΠΈΠ½Ρ„Π»Π°ΠΌΠ°Ρ†ΠΈΡ˜Π΅, ΡΡ‚ΠΈΡˆΠ°Π²Π°ΡšΠ΅ΠΌ антиоксидативнС ΠΎΠ΄Π±Ρ€Π°Π½Π΅ ΠΈ ΡΡ‚ΠΈΡˆΠ°Π²Π°ΡšΠ΅ΠΌ Ρ€Π΅Π³Π΅Π½Π΅Ρ€Π°Ρ†ΠΈΡ˜Π΅. Π’Π°Π½Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΠΈ HMGB1 ΠΊΡ€ΠΎΠ· ΠΈΠ½Ρ‚Π΅Ρ€Π°ΠΊΡ†ΠΈΡ˜Π΅ са TLR4 Ρ€Π΅Ρ†Π΅ΠΏΡ‚ΠΎΡ€ΠΎΠΌ Π°ΠΊΡ‚ΠΈΠ²ΠΈΡ€Π° MAPК/NF-ΞΊB p65 ΠΈ ЈАК1/STAT3 сигналнС ΠΏΡƒΡ‚Π΅Π²Π΅, доприносСћи ΠΏΠΎΠ²Π΅Ρ›Π°ΡšΡƒ ΠΏΡ€ΠΎΠ΄ΡƒΠΊΡ†ΠΈΡ˜Π΅ ΠΏΡ€ΠΎΠΈΠ½Ρ„Π»Π°ΠΌΠ°Ρ†ΠΈΡ˜ΡΠΊΠΈΡ… Ρ†ΠΈΡ‚ΠΎΠΊΠΈΠ½Π° TNF-Ξ± ΠΈ IL-6 ΠΈ Π°ΠΊΡƒΡ‚Π½ΠΎ-Ρ„Π°Π·Π½ΠΎΠ³ ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½Π° Ρ…Π°ΠΏΡ‚ΠΎΠ³Π»ΠΎΠ±ΠΈΠ½Π°. ΠŸΠΎΠ΄ΡΡ‚ΠΈΡ†Π°ΡšΠ΅ΠΌ NF-ΞΊB p65 ΠΈΠ½Ρ„Π»Π°ΠΌΠ°Ρ†ΠΈΡ˜ΡΠΊΠΎΠ³ ΠΏΡƒΡ‚Π°, HMGB1 Π΄Π΅Π»ΡƒΡ˜Π΅ Π½Π΅Π³Π°Ρ‚ΠΈΠ²Π½ΠΎ Π½Π° Ρ†ΠΈΡ‚ΠΎΠΏΡ€ΠΎΡ‚Π΅ΠΊΡ‚ΠΈΠ²Π½ΠΈ ΠΎΠ΄Π³ΠΎΠ²ΠΎΡ€ Ρƒ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚ΠΈΡ‡Π½ΠΎΡ˜ Ρ˜Π΅Ρ‚Ρ€ΠΈ Ρ‚Π°ΠΊΠΎ ΡˆΡ‚ΠΎ ΠΎΠ½Π΅ΠΌΠΎΠ³ΡƒΡ›Π°Π²Π° активност Nrf2 ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½Π°, ΠΎΠ΄Π³ΠΎΠ²ΠΎΡ€Π½ΠΎΠ³ Π·Π° ΡΡ‚ΠΈΡˆΠ°Π²Π°ΡšΠ΅ ΠΈΠ½Ρ„Π»Π°ΠΌΠ°Ρ†ΠΈΡ˜Π΅ ΠΈ ΠΏΡ€ΠΎΠ΄ΡƒΠΊΡ†ΠΈΡ˜Ρƒ антиоксидативних Π΅Π½Π·ΠΈΠΌΠ°. На ΡΡ‚ΠΈΡˆΠ°Π²Π°ΡšΠ΅ Ρ€Π΅Π³Π΅Π½Π΅Ρ€Π°Ρ‚ΠΈΠ²Π½ΠΎΠ³ ΠΏΠΎΡ‚Π΅Π½Ρ†ΠΈΡ˜Π°Π»Π° Ρ˜Π΅Ρ‚Ρ€Π΅, Π°ΠΊΡ‚ΠΈΠ²ΠΈΡ€Π°Π½Π° HMGB1/TLR4 оса ΡƒΡ‚ΠΈΡ‡Π΅ ΠΏΡ€Π΅ΠΊΠΎ ΡƒΠ²Π΅Ρ›Π°ΡšΠ° присуства Π½Π΅Π³Π°Ρ‚ΠΈΠ²Π½ΠΈΡ… Ρ€Π΅Π³ΡƒΠ»Π°Ρ‚ΠΎΡ€Π° Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΠΎΠ³ циклуса - ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½Π° p53 ΠΈ p21, ΠΈ смањСњСм Π½ΠΈΠ²ΠΎΠ° Ρ†ΠΈΠΊΠ»ΠΈΠ½Π° D1. Π”ΠΎΠ±ΠΈΡ˜Π΅Π½ΠΈ Ρ€Π΅Π·ΡƒΠ»Ρ‚Π°Ρ‚ΠΈ ΡƒΠΊΠ°Π·ΡƒΡ˜Ρƒ Π½Π° слоТСност дСловања HMGB1 ΠΏΡ€ΠΎΡ‚Π΅ΠΈΠ½Π° Ρƒ Π΄ΠΈΡ˜Π°Π±Π΅Ρ‚Π΅ΡΡƒ ΠΈ Π½Π° Π·Π½Π°Ρ‡Π°Ρ˜ ΡΠΏΡ€Π΅Ρ‡Π°Π²Π°ΡšΠ° ΠΎΡΠ»ΠΎΠ±Π°Ρ’Π°ΡšΠ° HMGB1 ΠΈΠ»ΠΈ Π±Π»ΠΎΠΊΠ°Π΄Π΅ HMGB1/TLR4 осС Ρƒ Ρ†ΠΈΡ™Ρƒ одлагања настанка ΠΎΡˆΡ‚Π΅Ρ›Π΅ΡšΠ° Ρ˜Π΅Ρ‚Ρ€Π΅.Oxidative stress and chronic inflammation are considered to be the main causes of diabetic complications, one of which is liver damage. An important mediator of these processes may be the endogenous HMGB1 protein, when released into the extracellular environment from the necrotic, damaged or activated cells. As the HMGB1 role in diabetes was insufficiently studied, in this doctoral dissertation the contribution of HMGB1 to liver damage of streptozotocin-induced diabetic rats was investigated. It has been shown that the level of liver damage in diabetes correlates with the presence of extracellular HMGB1. In diabetic liver, this protein is structurally modified by acetylation, phosphorylation, and O-GlcNAc glycosylation, which correlates with its translocation from the nucleus to the cytoplasm and an increase in its presence in the liver and serum. Reduction of the level of extracellular HMGB1 by melatonin or ethyl pyruvate treatment of diabetic rats, shows that HMGB1 contributes to diabetic liver damage by maintaining a chronic inflammation, by lowering antioxidant defense and by reducing regeneration. Extracellular HMGB1 activates MAPK/NF-ΞΊB p65 and JAK1/STAT3 signaling pathways through interactions with the TLR4 receptor, thus contributing increased production of proinflammatory cytokines TNF-Ξ± and IL-6 and the acute-phase protein, haptoglobin. By stimulating the NF-ΞΊB p65 inflammatory pathway, HMGB1 acts negatively on the cytoprotective response of the diabetic liver, by disabling Nrf2 protein activity, which is responsible for reduction of inflammation and antioxidant enzymes production. Activated HMGB1/TLR4 axis reduces regenerative potential of the liver by increasing the presence of negative cell cycle regulators - proteins p53 and p21, and also by decreasing the level of cyclin D1. The obtained results indicate the complexity of HMGB1 protein action in diabetes and underlines the importance of preventing the release of HMGB1 or blockage of HMGB1/TLR4 axis in order to delay the occurrence of liver damage

    Anti-melanoma effects of ingenanes isolated from Euphorbia species

    Get PDF
    In this research, from two species, E. palustris and E. lucida, four ingenane derivatives were isolated. Their anticancer effects were evaluated in the human melanoma – 518A2 cell line and compared with the effects of ingenolmebutate. Selectivity towards human melanoma cells was determined using normal human keratinocytes – HaCaT.11th Conference on Medicinal and Aromatic Plants of Southeast European Countries, (11th CMAPSEEC), Ohrid, North Macedonia, 6–10 October 202

    Π˜Π½Ρ…ΠΈΠ±ΠΈΡ†ΠΈΡ˜Π° Π°ΡƒΡ‚ΠΎΡ„Π°Π³ΠΈΡ˜Π΅ ΡΠ΅Π½Π·ΠΈΡ‚ΠΈΠ·ΡƒΡ˜Π΅ Ρ›Π΅Π»ΠΈΡ˜Π΅ глиобластома Π½Π° ΠΈΠ½Ρ…ΠΈΠ±ΠΈΡ‚ΠΎΡ€Π΅ Π‘Ρ€Ρ† Ρ‚ΠΈΡ€ΠΎΠ·ΠΈΠ½-ΠΊΠΈΠ½Π°Π·Π΅, Π΄Π΅Ρ€ΠΈΠ²Π°Ρ‚Π΅ ΠΏΠΈΡ€Π°Π·ΠΎΠ»ΠΎ[3,4- Π΄]ΠΏΠΈΡ€ΠΈΠΌΠΈΠ΄ΠΈΠ½Π° Si306 ΠΈ pro-Si306

    Get PDF
    Glioblastom je jedan od najagresivnijih tumora mozga koji karakteriΕ‘e infiltrirajuΔ‡a priroda, intenzivna proliferacija i rezistencija na terapiju. Δ†elije glioblastoma imaju visoku ekspresiju Src tirozin-kinaze koja reguliΕ‘e proliferaciju, preΕΎivljavanje i invazivnost tumorskih Δ‡elija čineΔ‡i je potencijalnom metom za terapiju. Inhibitori tirozin-kinaza mogu indukovati autofagiju koja deluje protektivno na tumorske Δ‡elije. Sposobnost inhibitora Src tirozin-kinaze, derivata pirazolo[3,4-d]pirimidina Si306 i njegovog proleka pro-Si306, da indukuju autofagiju ispitana je na Δ‡elijskoj liniji humanog glioblastoma U87 i njenoj varijanti sa viΕ‘estrukom rezistencijom na lekove U87-TxR. Tretman ovim jedinjenjima uzrokovao je pojavu autofagozoma u Δ‡elijama nakon 3 sata, a efekat na indukciju autofagije opstao je i nakon 48 sati Ε‘to je utvrΔ‘eno analizom markera autofagije LC3 i p62. Inhibicija autofagnog fluksa bafilomicinom A1 značajno je uveΔ‡ala postojeΔ‡e anti-proliferativno dejstvo Si306 i pro-Si306. TakoΔ‘e, kombinovani tretmani Src inhibitora sa bafilomicinom A1 doveli su do nekroze nakon 48 sati. Dobijeni rezultati sugeriΕ‘u da autofagija indukovana ovim jedinjenjima ima zaΕ‘titnu ulogu u Δ‡elijama glioblastoma i da se modulacija autofagije moΕΎe koristiti za senzitizaciju Δ‡elija glioblastoma na inhibitore Src tirozin-kinaze. Pored toga, pomenuti efekti Si306 i pro-Si306 nisu umanjeni prisustvom viΕ‘estruko- rezistentnog fenotipa, Ε‘to ovim jedinjenjima daje potencijal za lečenje rezistentnih tumora.Глиобластом јС јСдан ΠΎΠ΄ Π½Π°Ρ˜Π°Π³Ρ€Π΅ΡΠΈΠ²Π½ΠΈΡ˜ΠΈΡ… Ρ‚ΡƒΠΌΠΎΡ€Π° ΠΌΠΎΠ·Π³Π° који ΠΊΠ°Ρ€Π°ΠΊΡ‚Π΅Ρ€ΠΈΡˆΠ΅ ΠΈΠ½Ρ„ΠΈΠ»Ρ‚Ρ€ΠΈΡ€Π°Ρ˜ΡƒΡ›Π° ΠΏΡ€ΠΈΡ€ΠΎΠ΄Π°, ΠΈΠ½Ρ‚Π΅Π½Π·ΠΈΠ²Π½Π° ΠΏΡ€ΠΎΠ»ΠΈΡ„Π΅Ρ€Π°Ρ†ΠΈΡ˜Π° ΠΈ Ρ€Π΅Π·ΠΈΡΡ‚Π΅Π½Ρ†ΠΈΡ˜Π° Π½Π° Ρ‚Π΅Ρ€Π°ΠΏΠΈΡ˜Ρƒ. Π‹Π΅Π»ΠΈΡ˜Π΅ глиобластома ΠΈΠΌΠ°Ρ˜Ρƒ високу Π΅ΠΊΡΠΏΡ€Π΅ΡΠΈΡ˜Ρƒ Π‘Ρ€Ρ† Ρ‚ΠΈΡ€ΠΎΠ·ΠΈΠ½-ΠΊΠΈΠ½Π°Π·Π΅ која Ρ€Π΅Π³ΡƒΠ»ΠΈΡˆΠ΅ ΠΏΡ€ΠΎΠ»ΠΈΡ„Π΅Ρ€Π°Ρ†ΠΈΡ˜Ρƒ, ΠΏΡ€Π΅ΠΆΠΈΠ²Ρ™Π°Π²Π°ΡšΠ΅ ΠΈ инвазивност туморских Ρ›Π΅Π»ΠΈΡ˜Π° Ρ‡ΠΈΠ½Π΅Ρ›ΠΈ јС ΠΏΠΎΡ‚Π΅Π½Ρ†ΠΈΡ˜Π°Π»Π½ΠΎΠΌ ΠΌΠ΅Ρ‚ΠΎΠΌ Π·Π° Ρ‚Π΅Ρ€Π°ΠΏΠΈΡ˜Ρƒ. Π˜Π½Ρ…ΠΈΠ±ΠΈΡ‚ΠΎΡ€ΠΈ Ρ‚ΠΈΡ€ΠΎΠ·ΠΈΠ½-ΠΊΠΈΠ½Π°Π·Π° ΠΌΠΎΠ³Ρƒ ΠΈΠ½Π΄ΡƒΠΊΠΎΠ²Π°Ρ‚ΠΈ Π°ΡƒΡ‚ΠΎΡ„Π°Π³ΠΈΡ˜Ρƒ која Π΄Π΅Π»ΡƒΡ˜Π΅ ΠΏΡ€ΠΎΡ‚Π΅ΠΊΡ‚ΠΈΠ²Π½ΠΎ Π½Π° туморскС Ρ›Π΅Π»ΠΈΡ˜Π΅. Бпособност ΠΈΠ½Ρ…ΠΈΠ±ΠΈΡ‚ΠΎΡ€Π° Π‘Ρ€Ρ† Ρ‚ΠΈΡ€ΠΎΠ·ΠΈΠ½-ΠΊΠΈΠ½Π°Π·Π΅, Π΄Π΅Ρ€ΠΈΠ²Π°Ρ‚Π° ΠΏΠΈΡ€Π°Π·ΠΎΠ»ΠΎ[3,4-Π΄]ΠΏΠΈΡ€ΠΈΠΌΠΈΠ΄ΠΈΠ½Π° Si306 ΠΈ њСговог ΠΏΡ€ΠΎΠ»Π΅ΠΊΠ° pro-Si306, Π΄Π° ΠΈΠ½Π΄ΡƒΠΊΡƒΡ˜Ρƒ Π°ΡƒΡ‚ΠΎΡ„Π°Π³ΠΈΡ˜Ρƒ испитана јС Π½Π° Ρ›Π΅Π»ΠΈΡ˜ΡΠΊΠΎΡ˜ линији Ρ…ΡƒΠΌΠ°Π½ΠΎΠ³ глиобластома U87 ΠΈ њСној Π²Π°Ρ€ΠΈΡ˜Π°Π½Ρ‚ΠΈ са Π²ΠΈΡˆΠ΅ΡΡ‚Ρ€ΡƒΠΊΠΎΠΌ Ρ€Π΅Π·ΠΈΡΡ‚Π΅Π½Ρ†ΠΈΡ˜ΠΎΠΌ Π½Π° Π»Π΅ΠΊΠΎΠ²Π΅ U87-TxR. Π’Ρ€Π΅Ρ‚ΠΌΠ°Π½ ΠΎΠ²ΠΈΠΌ јСдињСњима ΡƒΠ·Ρ€ΠΎΠΊΠΎΠ²Π°ΠΎ јС ΠΏΠΎΡ˜Π°Π²Ρƒ Π°ΡƒΡ‚ΠΎΡ„Π°Π³ΠΎΠ·ΠΎΠΌΠ° Ρƒ Ρ›Π΅Π»ΠΈΡ˜Π°ΠΌΠ° Π½Π°ΠΊΠΎΠ½ 3 сата, Π° Π΅Ρ„Π΅ΠΊΠ°Ρ‚ Π½Π° ΠΈΠ½Π΄ΡƒΠΊΡ†ΠΈΡ˜Ρƒ Π°ΡƒΡ‚ΠΎΡ„Π°Π³ΠΈΡ˜Π΅ опстао јС ΠΈ Π½Π°ΠΊΠΎΠ½ 48 сати ΡˆΡ‚ΠΎ јС ΡƒΡ‚Π²Ρ€Ρ’Π΅Π½ΠΎ Π°Π½Π°Π»ΠΈΠ·ΠΎΠΌ ΠΌΠ°Ρ€ΠΊΠ΅Ρ€Π° Π°ΡƒΡ‚ΠΎΡ„Π°Π³ΠΈΡ˜Π΅ LC3 ΠΈ p62. Π˜Π½Ρ…ΠΈΠ±ΠΈΡ†ΠΈΡ˜Π° Π°ΡƒΡ‚ΠΎΡ„Π°Π³Π½ΠΎΠ³ флукса Π±Π°Ρ„ΠΈΠ»ΠΎΠΌΠΈΡ†ΠΈΠ½ΠΎΠΌ А1 Π·Π½Π°Ρ‡Π°Ρ˜Π½ΠΎ јС ΡƒΠ²Π΅Ρ›Π°Π»Π° ΠΏΠΎΡΡ‚ΠΎΡ˜Π΅Ρ›Π΅ Π°Π½Ρ‚ΠΈ-ΠΏΡ€ΠΎΠ»ΠΈΡ„Π΅Ρ€Π°Ρ‚ΠΈΠ²Π½ΠΎ Π΄Π΅Ρ˜ΡΡ‚Π²ΠΎ Si306 ΠΈ pro-Si306. Π’Π°ΠΊΠΎΡ’Π΅, ΠΊΠΎΠΌΠ±ΠΈΠ½ΠΎΠ²Π°Π½ΠΈ Ρ‚Ρ€Π΅Ρ‚ΠΌΠ°Π½ΠΈ Π‘Ρ€Ρ† ΠΈΠ½Ρ…ΠΈΠ±ΠΈΡ‚ΠΎΡ€Π° са Π±Π°Ρ„ΠΈΠ»ΠΎΠΌΠΈΡ†ΠΈΠ½ΠΎΠΌ А1 Π΄ΠΎΠ²Π΅Π»ΠΈ су Π΄ΠΎ Π½Π΅ΠΊΡ€ΠΎΠ·Π΅ Π½Π°ΠΊΠΎΠ½ 48 сати. Π”ΠΎΠ±ΠΈΡ˜Π΅Π½ΠΈ Ρ€Π΅Π·ΡƒΠ»Ρ‚Π°Ρ‚ΠΈ ΡΡƒΠ³Π΅Ρ€ΠΈΡˆΡƒ Π΄Π° Π°ΡƒΡ‚ΠΎΡ„Π°Π³ΠΈΡ˜Π° ΠΈΠ½Π΄ΡƒΠΊΠΎΠ²Π°Π½Π° ΠΎΠ²ΠΈΠΌ јСдињСњима ΠΈΠΌΠ° Π·Π°ΡˆΡ‚ΠΈΡ‚Π½Ρƒ ΡƒΠ»ΠΎΠ³Ρƒ Ρƒ Ρ›Π΅Π»ΠΈΡ˜Π°ΠΌΠ° глиобластома ΠΈ Π΄Π° сС ΠΌΠΎΠ΄ΡƒΠ»Π°Ρ†ΠΈΡ˜Π° Π°ΡƒΡ‚ΠΎΡ„Π°Π³ΠΈΡ˜Π΅ ΠΌΠΎΠΆΠ΅ користити Π·Π° ΡΠ΅Π½Π·ΠΈΡ‚ΠΈΠ·Π°Ρ†ΠΈΡ˜Ρƒ Ρ›Π΅Π»ΠΈΡ˜Π° глиобластома Π½Π° ΠΈΠ½Ρ…ΠΈΠ±ΠΈΡ‚ΠΎΡ€Π΅ Π‘Ρ€Ρ† Ρ‚ΠΈΡ€ΠΎΠ·ΠΈΠ½-ΠΊΠΈΠ½Π°Π·Π΅. ΠŸΠΎΡ€Π΅Π΄ Ρ‚ΠΎΠ³Π°, ΠΏΠΎΠΌΠ΅Π½ΡƒΡ‚ΠΈ Π΅Ρ„Π΅ΠΊΡ‚ΠΈ Si306 ΠΈ pro-Si306 нису ΡƒΠΌΠ°ΡšΠ΅Π½ΠΈ присуством Π²ΠΈΡˆΠ΅ΡΡ‚Ρ€ΡƒΠΊΠΎ- рСзистСнтног Ρ„Π΅Π½ΠΎΡ‚ΠΈΠΏΠ°, ΡˆΡ‚ΠΎ ΠΎΠ²ΠΈΠΌ јСдињСњима дајС ΠΏΠΎΡ‚Π΅Π½Ρ†ΠΈΡ˜Π°Π» Π·Π° Π»Π΅Ρ‡Π΅ΡšΠ΅ рСзистСнтних Ρ‚ΡƒΠΌΠΎΡ€Π°.Knjiga saΕΎetaka: TreΔ‡i Kongres biologa Srbije, Zlatibor, Srbija 21 - 25. 9. 2022

    Unveiling Anticancer Potential of COX-2 and 5-LOX Inhibitors: Cytotoxicity, Radiosensitization Potential and Antimigratory Activity against Colorectal and Pancreatic Carcinoma

    Get PDF
    Apart from cytotoxicity, inhibitors of the COX-2 enzyme have demonstrated additional effects important for cancer treatment (such as radiosensitization of tumor cells and cell antimigratory effects); however, the relationship between the inhibition of other inflammation-related enzyme 5-LOX inhibitors and anticancer activity is still not well understood. In our study, the cytotoxicity of thirteen COX-2 and 5-LOX inhibitors previously presented by our group (1–13) was tested on three cancer cell lines (HCT 116, HT-29 and BxPC-3) and one healthy cell line (MRC-5). Compounds 3, 5, 6 and 7 showed moderate cytotoxicity, but good selectivity towards cancer cell lines. IC50 values were in the range of 22.99–51.66 Β΅M (HCT 116 cell line), 8.63–41.20 Β΅M (BxPC-3 cell line) and 24.78–81.60 Β΅M (HT-29 cell line; compound 7 > 100 Β΅M). In comparison to tested, commercially available COX-2 and 5-LOX inhibitors, both cytotoxicity and selectivity were increased. The addition of compounds 6 and 7 to irradiation treatment showed the most significant decrease in cell proliferation of the HT-29 cell line (p < 0.001). The antimigratory potential of the best dual COX-2 and 5-LOX inhibitors (compounds 1, 2, 3 and 5) was tested by a wound-healing assay using the SW620 cell line. Compounds 1 and 3 were singled out as compounds with the most potent effect (relative wound closure was 3.20% (24 h), 5,08% (48 h) for compound 1 and 3.86% (24 h), 7.68% (48 h) for compound 3). Considering all these results, compound 3 stood out as the compound with the most optimal biological activity, with the best dual COX-2 and 5-LOX inhibitory activity, good selectivity towards tested cancer cell lines, significant cell antimigratory potential and a lack of toxic effects at therapeutic doses

    Autophagy Inhibition Enhances Anti-Glioblastoma Effects of Pyrazolo[3,4-d]pyrimidine Tyrosine Kinase Inhibitors

    Get PDF
    Drug resistance presents a major obstacle to the successful treatment of glioblastoma. Autophagy plays a key role in drug resistance, particularly in relation to targeted therapy, which has prompted the use of autophagy inhibitors to increase the effectiveness of targeted therapeutics. The ability of two Src tyrosine kinase inhibitors, Si306 and its prodrug pro-Si306, to induce autophagy was evaluated in the human glioblastoma cell line U87 and its multidrug-resistant counterpart U87-TxR. Autophagy markers were assessed by flow cytometry, microscopy, and Western blot, and induction of autophagy by these compounds was demonstrated after 3 h as well as 48 h. The effects of Si306 and pro-Si306 on cell proliferation and cell death were examined in the presence or absence of autophagy inhibition by bafilomycin A1. Combined treatments of Si306 and pro-Si306 with bafilomycin A1 were synergistic in nature, and the inhibition of autophagy sensitized glioblastoma cells to Src tyrosine kinase inhibitors. Si306 and pro-Si306 more strongly inhibited cell proliferation and triggered necrosis in combination with bafilomycin A1. Our findings suggest that modulation of Si306- and pro-Si306-induced autophagy can be used to enhance the anticancer effects of these Src tyrosine kinase inhibitors and overcome the drug-resistant phenotype in glioblastoma cells

    Anti-invasive effects of CXCR4 and FAK inhibitors in non-small cell lung carcinomas with mutually inactivated p53 and PTEN tumor suppressors

    Get PDF
    Non-small cell lung carcinoma (NSCLC) is the most common type of lung cancer. At the time of diagnosis, a large percentage of NSCLC patients have already developed metastasis, responsible for extremely high mortality rates. CXCR4 receptor and focal adhesion kinase (FAK) are known to regulate such invasive cancer behavior. Their expression is downregulated by p53 and PTEN tumor suppressors which are commonly co-inactivated in NSCLC patients and contribute to metastasis. Therefore, targeting CXCR4 or FAK seems to be a promising strategy in suppressing metastatic spread of p53/PTEN deficient NSCLCs. In this study, we first examined the invasive characteristics of NSCLC cells with suppressed p53 and PTEN activity using wound healing, gelatin degradation and invasion assays. Further, changes in the expression of CXCR4 and FAK were evaluated by RT-qPCR and Western Blot analysis. Finally, we tested the ability of CXCR4 and FAK inhibitors (WZ811 and PF-573228, respectively) to suppress the migratory and invasive potential of p53/PTEN deficient NSCLC cells, in vitro and in vivo using metastatic models of human NSCLC. Our results showed that cells with mutually inactive p53 and PTEN have significantly increased invasive potential associated with hyperactivation of CXCR4 and FAK signaling pathways. Treatments with WZ811 and PF-573228 inhibitors significantly reduced migratory and invasive capacity in vitro and showed a trend of improved survival in vivo. Accordingly, we demonstrated that p53/PTEN deficient NSCLCs have extremely invasive phenotype and provided a rationale for the use of CXCR4 or FAK inhibitors for the suppression of NSCLC dissemination.This is a post-peer-review, pre-copyedit version of an article published in Investigational New Drugs. The final authenticated version is available online at: [http://dx.doi.org/10.1007/s10637-017-0494-4

    Modulation of diabetes-related liver injury by the HMGB1/TLR4 inflammatory pathway.

    Get PDF
    Chronic inflammation plays an essential role in the development of diabetic complications. Understanding the molecular mechanisms that support inflammation is a prerequisite for the design of novel anti-inflammatory therapies. These would take into consideration circulating levels of cytokines and damage-associated molecular patterns (DAMPs) that include the high mobility group box 1 (HMGB1) protein which, in part, promotes the inflammatory response through TLR4 signaling. The liver, as the source of circulating cytokines and acute-phase proteins, contributes to the control of systemic inflammation. We previously found that liver injury in streptozotocin-induced diabetic rats correlated with the level of oxidative stress, increased expression of HMGB1, and with the activation of TLR4-mediated cell death pathways. In the present work, we examined the effects of ethyl pyruvate (EP), an inhibitor of HMGB1 release/expression, on the modulation of activation of the HMGB1/TLR4 inflammatory cascade in diabetic liver. We observed that increased expression of inflammatory markers, TNF-Ξ±, IL-6, and haptoglobin in diabetic liver was associated with increased HMGB1/TLR4 interaction, activation of MAPK (p38, ERK, JNK)/NF-ΞΊB p65 and JAK1/STAT3 signaling pathways, and with decreased expression of Nrf2-regulated antioxidative enzymes. The reduction in HMGB1 expression as the result of EP administration reduced the pro-inflammatory activity of HMGB1 and exerted a protective effect on diabetic liver, which was observed as improved liver histology and antioxidant and inflammatory statuses. Our results suggest that prevention of HMGB1 release and blockage of the HMGB/TLR4 axis represents a potentially effective therapeutic strategy aimed at ameliorating diabetes-induced inflammation and ensuing liver injury.This is a post-peer-review, pre-copyedit version of an article published in Journal of Physiology and Biochemistry. The final authenticated version is available online at: [http://dx.doi.org/10.1007/s13105-018-0626-0

    New anti-glioblastoma strategy with natural compounds sclareol and doxorubicin

    Get PDF
    Background: Doxorubicin (DOX) has been very effective against glioblastoma invitro. Its application in vivo is hampered because it cannot pass the blood–brainbarrier (BBB). Significant research efforts are invested to overcome this limitation.Sclareol (SC) is an aromatic compound naturally found in clary sage. Thecombination of SC and DOX showed promising effects in different tumor types invitro and in vivo. Therefore, we tested their combination and innovative hybridmolecules (SC:DOX) on glioblastoma cells with the expression of P-glycoprotein, amajor component of BBB and cancer multidrug resistance marker. Methods:Cytotoxicity and selectivity towards glioblastoma cells of SC, DOX, theircombination, and SC:DOX were examined by MTT assay. The effect of SC on DOXaccumulation was determined by flow cytometry. We also studied SC:DOXaccumulation, cellular uptake, localization imaging, and DNA damage induction.Results: The effects of simultaneous SC and DOX treatments demonstrated theconsiderable potential of SC to reverse DOX resistance in glioblastoma cells andincrease DOX accumulation. SC:DOX hybrids, named CON1 and CON2 were lesscytotoxic than DOX, but with reduced resistance and increased selectivity towardsglioblastoma cells. Cellular uptake of CON1 and CON2 was increased in glioblastomacells compared to DOX. Perinuclear localization of CON1 and CON2 vs. nuclearlocalization of DOX as well as no DNA damaging effects suggest a differentmechanism of action for SC:DOX. Conclusion: The combination of SC and DOX, andtheir innovative hybrids, could be considered a promising strategy that can overcomethe limitations of DOX application in glioblastoma.Kanazir S, SaviΔ‡ D, editors. Book of abstracts: 8th Congress of Serbian neuroscience society with international participation; 2023 May 31 - Jun 2; Belgrade, Serbia. Belgrade : Serbian Neuroscience Society; 2023. p. 71

    Unveiling Anticancer Potential of COX-2 and 5-LOX Inhibitors: Cytotoxicity, Radiosensitization Potential and Antimigratory Activity against Colorectal and Pancreatic Carcinoma

    Get PDF
    Apart from cytotoxicity, inhibitors of the COX-2 enzyme have demonstrated additional effects important for cancer treatment (such as radiosensitization of tumor cells and cell antimigratory effects); however, the relationship between the inhibition of other inflammation-related enzyme 5-LOX inhibitors and anticancer activity is still not well understood. In our study, the cytotoxicity of thirteen COX-2 and 5-LOX inhibitors previously presented by our group (1-13) was tested on three cancer cell lines (HCT 116, HT-29 and BxPC-3) and one healthy cell line (MRC-5). Compounds 3, 5, 6 and 7 showed moderate cytotoxicity, but good selectivity towards cancer cell lines. IC50 values were in the range of 22.99-51.66 Β΅M (HCT 116 cell line), 8.63-41.20 Β΅M (BxPC-3 cell line) and 24.78-81.60 Β΅M (HT-29 cell line; compound 7 > 100 Β΅M). In comparison to tested, commercially available COX-2 and 5-LOX inhibitors, both cytotoxicity and selectivity were increased. The addition of compounds 6 and 7 to irradiation treatment showed the most significant decrease in cell proliferation of the HT-29 cell line (p < 0.001). The antimigratory potential of the best dual COX-2 and 5-LOX inhibitors (compounds 1, 2, 3 and 5) was tested by a wound-healing assay using the SW620 cell line. Compounds 1 and 3 were singled out as compounds with the most potent effect (relative wound closure was 3.20% (24 h), 5,08% (48 h) for compound 1 and 3.86% (24 h), 7.68% (48 h) for compound 3). Considering all these results, compound 3 stood out as the compound with the most optimal biological activity, with the best dual COX-2 and 5-LOX inhibitory activity, good selectivity towards tested cancer cell lines, significant cell antimigratory potential and a lack of toxic effects at therapeutic doses
    corecore