Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 45
1.
Article En | MEDLINE | ID: mdl-38715433

Ultrastructure expansion microscopy (U-ExM) involves the physical magnification of specimens embedded in hydrogels, which allows for super-resolution imaging of subcellular structures using a conventional diffraction-limited microscope. Methods for expansion microscopy exist for several organisms, organs, and cell types, and used to analyze cellular organelles and substructures in nanoscale resolution. Here, we describe a simple step-by-step U-ExM protocol for the expansion, immunostaining, imaging, and analysis of cytoskeletal and organellar structures in kidney tissue. We detail the critical modified steps to optimize isotropic kidney tissue expansion, and preservation of the renal cell structures of interest. We demonstrate the utility of the approach using several markers of renal cell types, centrioles, cilia, the extracellular matrix, and other cytoskeletal elements. Finally, we show that the approach works well on mouse and human kidney samples that were preserved using different fixation and embedding conditions. Overall, this protocol provides a simple and cost-effective approach to analyze both preclinical and clinical renal samples in high detail, using conventional lab supplies and standard widefield or confocal microscopy.

2.
bioRxiv ; 2024 Feb 17.
Article En | MEDLINE | ID: mdl-38405695

Ultrastructure expansion microscopy (U-ExM) involves the physical magnification of specimens embedded in hydrogels, which allows for super-resolution imaging of subcellular structures using a conventional diffraction-limited microscope. Methods for expansion microscopy exist for several organisms, organs, and cell types, and used to analyze cellular organelles and substructures in nanoscale resolution. Here, we describe a simple step-by-step U-ExM protocol for the expansion, immunostaining, imaging, and analysis of cytoskeletal and organellar structures in kidney tissue. We detail the critical modified steps to optimize isotropic kidney tissue expansion, and preservation of the renal cell structures of interest. We demonstrate the utility of the approach using several markers of renal cell types, centrioles, cilia, the extracellular matrix, and other cytoskeletal elements. Finally, we show that the approach works well on mouse and human kidney samples that were preserved using different fixation and storage conditions. Overall, this protocol provides a simple and cost-effective approach to analyze both pre-clinical and clinical renal samples in high detail, using conventional lab supplies and standard widefield or confocal microscopy.

3.
JCI Insight ; 9(4)2024 Feb 22.
Article En | MEDLINE | ID: mdl-38385746

Autosomal dominant polycystic kidney disease (ADPKD) is a monogenic disorder accounting for approximately 5% of patients with renal failure, yet therapeutics for the treatment of ADPKD remain limited. ADPKD tissues display abnormalities in the biogenesis of the centrosome, a defect that can cause genome instability, aberrant ciliary signaling, and secretion of pro-inflammatory factors. Cystic cells form excess centrosomes via a process termed centrosome amplification (CA), which causes abnormal multipolar spindle configurations, mitotic catastrophe, and reduced cell viability. However, cells with CA can suppress multipolarity via "centrosome clustering," a key mechanism by which cells circumvent apoptosis. Here, we demonstrate that inhibiting centrosome clustering can counteract the proliferation of renal cystic cells with high incidences of CA. Using ADPKD human cells and mouse models, we show that preventing centrosome clustering with 2 inhibitors, CCB02 and PJ34, blocks cyst initiation and growth in vitro and in vivo. Inhibiting centrosome clustering activates a p53-mediated surveillance mechanism leading to apoptosis, reduced cyst expansion, decreased interstitial fibrosis, and improved kidney function. Transcriptional analysis of kidneys from treated mice identified pro-inflammatory signaling pathways implicated in CA-mediated cystogenesis and fibrosis. Our results demonstrate that centrosome clustering is a cyst-selective target for the improvement of renal morphology and function in ADPKD.


Cysts , Polycystic Kidney, Autosomal Dominant , Humans , Mice , Animals , Polycystic Kidney, Autosomal Dominant/pathology , Cell Proliferation , Kidney/pathology , Centrosome/metabolism , Fibrosis , Cysts/metabolism , Cysts/pathology
4.
EMBO Rep ; 25(1): 428-454, 2024 Jan.
Article En | MEDLINE | ID: mdl-38177914

Mutations in genes that disrupt centrosome structure or function can cause congenital kidney developmental defects and lead to fibrocystic pathologies. Yet, it is unclear how defective centrosome biogenesis impacts renal progenitor cell physiology. Here, we examined the consequences of impaired centrosome duplication on kidney stromal progenitor cell growth, differentiation, and fate. Conditional deletion of the ciliopathy gene Cep120, which is essential for centrosome duplication, in the stromal mesenchyme resulted in reduced abundance of interstitial lineages including pericytes, fibroblasts and mesangial cells. These phenotypes were caused by a combination of delayed mitosis, activation of the mitotic surveillance pathway leading to apoptosis, and changes in both Wnt and Hedgehog signaling that are key for differentiation of stromal cells. Cep120 ablation resulted in small hypoplastic kidneys with medullary atrophy and delayed nephron maturation. Finally, Cep120 and centrosome loss in the interstitium sensitized kidneys of adult mice, causing rapid fibrosis after renal injury via enhanced TGF-ß/Smad3-Gli2 signaling. Our study defines the cellular and developmental defects caused by loss of Cep120 and aberrant centrosome biogenesis in the embryonic kidney stroma.


Hedgehog Proteins , Kidney , Mice , Animals , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Kidney/pathology , Cell Differentiation/genetics , Stromal Cells , Stem Cells , Cell Cycle Proteins/metabolism
5.
Cells ; 12(23)2023 11 21.
Article En | MEDLINE | ID: mdl-38067099

BACKGROUND: Gliomas are the most malignant tumors of the central nervous system. One of the factors in their high drug resistance is avoiding programmed death (PCD) induction. This is related to the overexpression of intracellular survival pathways: PI3K-Akt/PKB-mTOR and Ras-Raf-MEK-ERK. Apoptosis and autophagy are co-existing processes due to the interactions between Bcl-2 and beclin-1 proteins. Their complex may be a molecular "toggle-switch" between PCD types. The aim of this research was to investigate the role of Bcl-2:beclin-1 complex in glioma cell elimination through the combined action of LY294002 and sorafenib. METHODS: Drug cytotoxicity was estimated with an MTT test. The type of cell death was evaluated using variant microscopy techniques (fluorochrome staining, immunocytochemistry, and transmission electron microscopy), as well as the Bcl-2:beclin-1 complex formation and protein localization. Molecular analysis of PCD indicators was conducted through immunoblotting, immunoprecipitation, and ELISA testing. SiRNA was used to block Bcl-2 and beclin-1 expression. RESULTS: The results showed the inhibitors used in simultaneous application resulted in Bcl-2:beclin-1 complex formation and apoptosis becoming dominant. This was accompanied by changes in the location of the tested proteins. CONCLUSIONS: "Switching" between apoptosis and autophagy using PI3K and Raf inhibitors with Bcl-2:beclin-1 complex formation opens new therapeutic perspectives against gliomas.


Glioma , Phosphatidylinositol 3-Kinases , Sorafenib , Humans , Apoptosis , Autophagy , Beclin-1 , Glioma/drug therapy , Glioma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Sorafenib/pharmacology , Sorafenib/therapeutic use
6.
bioRxiv ; 2023 Apr 05.
Article En | MEDLINE | ID: mdl-37066241

Defective centrosome function can disrupt embryonic kidney development, by causing changes to the renal interstitium that leads to fibrocystic disease pathologies. Yet, it remains unknown how mutations in centrosome genes impact kidney interstitial cells. Here, we examined the consequences of defective centrosome biogenesis on stromal progenitor cell growth, differentiation and fate. Conditional deletion of Cep120 , a ciliopathy gene essential for centrosome duplication, in the stromal mesenchyme resulted in reduced abundance of pericytes, interstitial fibroblasts and mesangial cells. This was due to delayed mitosis, increased apoptosis, and changes in Wnt and Hedgehog signaling essential for differentiation of stromal lineages. Cep120 ablation resulted in hypoplastic kidneys with medullary atrophy and delayed nephron maturation. Finally, centrosome loss in the interstitium sensitized kidneys of adult mice, causing rapid fibrosis via enhanced TGF-ß/Smad3-Gli2 signaling after renal injury. Our study defines the cellular and developmental defects caused by centrosome dysfunction in embryonic kidney stroma. Highlights: Defective centrosome biogenesis in kidney stroma causes:Reduced abundance of stromal progenitors, interstitial and mesangial cell populationsDefects in cell-autonomous and paracrine signalingAbnormal/delayed nephrogenesis and tubular dilationsAccelerates injury-induced fibrosis via defective TGF-ß/Smad3-Gli2 signaling axis.

7.
Int J Mol Sci ; 22(10)2021 May 13.
Article En | MEDLINE | ID: mdl-34068110

The aim of the study was to investigate the anticancer potential of LY294002 (PI3K inhibitor) and temozolomide using glioblastoma multiforme (T98G) and anaplastic astrocytoma (MOGGCCM) cells. Apoptosis, autophagy, necrosis, and granules in the cytoplasm were identified microscopically (fluorescence and electron microscopes). The mitochondrial membrane potential was studied by flow cytometry. The activity of caspases 3, 8, and 9 and Akt was evaluated fluorometrically, while the expression of Beclin 1, PI3K, Akt, mTOR, caspase 12, and Hsp27 was determined by immunoblotting. SiRNA was used to block Hsp27 and PI3K expression. Cell migration and localization of Hsp27 were tested with the wound healing assay and immunocytochemistry, respectively. LY294002 effectively diminished the migratory potential and increased programmed death of T98G and MOGGCCM. Autophagy was dominant in MOGGCCM, while apoptosis was dominant in T98G. LY294002 with temozolomide did not potentiate cell death but redirected autophagy toward apoptosis, which was correlated with ER stress. A similar effect was observed after blocking PI3K expression with siRNA. Transfection with Hsp27 siRNA significantly increased apoptosis related to ER stress. Our results indicate that inhibition of the PI3K/Akt/mTOR pathway sensitizes glioma cells to apoptosis upon temozolomide treatment, which was correlated with ER stress. Hsp27 increases the resistance of glioma cells to cell death upon temozolomide treatment.


Biomarkers, Tumor/metabolism , Chromones/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Temozolomide/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Enzyme Inhibitors/pharmacology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Membrane Potential, Mitochondrial , Necrosis , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Tumor Cells, Cultured
8.
Molecules ; 25(21)2020 Nov 08.
Article En | MEDLINE | ID: mdl-33171577

Coumarins, which occur naturally in the plant kingdom, are diverse class of secondary metabolites. With their antiproliferative, chemopreventive and antiangiogenetic properties, they can be used in the treatment of cancer. Their therapeutic potential depends on the type and location of the attachment of substituents to the ring. Therefore, the aim of our study was to investigate the effect of simple coumarins (osthole, umbelliferone, esculin, and 4-hydroxycoumarin) combined with sorafenib (specific inhibitor of Raf (Rapidly Accelerated Fibrosarcoma) kinase) in programmed death induction in human glioblastoma multiforme (T98G) and anaplastic astrocytoma (MOGGCCM) cells lines. Osthole and umbelliferone were isolated from fruits: Mutellina purpurea L. and Heracleum leskowii L., respectively, while esculin and 4-hydroxycoumarin were purchased from Sigma Aldrich (St. Louis, MO, USA). Apoptosis, autophagy and necrosis were identified microscopically after straining with specific fluorochromes. The level of caspase 3, Beclin 1, PI3K (Phosphoinositide 3-kinase), and Raf kinases were estimated by immunoblotting. Transfection with specific siRNA (small interfering RNA) was used to block Bcl-2 (B-cell lymphoma 2), Raf, and PI3K expression. Cell migration was tested with the wound healing assay. The present study has shown that all the coumarins eliminated the MOGGCCM and T98G tumor cells mainly via apoptosis and, to a lesser extent, via autophagy. Osthole, which has an isoprenyl moiety, was shown to be the most effective compound. Sorafenib did not change the proapoptotic activity of this coumarin; however, it reduced the level of autophagy. At the molecular level, the induction of apoptosis was associated with a decrease in the expression of PI3K and Raf kinases, whereas an increase in the level of Beclin 1 was observed in the case of autophagy. Inhibition of the expression of this protein by specific siRNA eliminated autophagy. Moreover, the blocking of the expression of Bcl-2 and PI3K significantly increased the level of apoptosis. Osthole and sorafenib successfully inhibited the migration of the MOGGCCM and T98G cells.


Antineoplastic Agents, Phytogenic/pharmacology , Coumarins/pharmacology , Glioblastoma/drug therapy , Magnoliopsida/chemistry , Plant Extracts/pharmacology , Sorafenib/pharmacology , 4-Hydroxycoumarins/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Autophagy/drug effects , Beclin-1/genetics , Beclin-1/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Esculin/pharmacology , Gene Expression Regulation/drug effects , Humans , Necrosis/drug therapy , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/metabolism , Umbelliferones/pharmacology , raf Kinases/metabolism
9.
Int J Mol Sci ; 21(21)2020 Oct 26.
Article En | MEDLINE | ID: mdl-33114713

Tryptophan metabolites: kynurenine (KYN), kynurenic acid (KYNA) and 6-formylindolo[3,2-b]carbazole (FICZ) are considered aryl hydrocarbon receptor (AhR) ligands. AhR is mainly expressed in barrier tissues, including skin, and is involved in various physiological and pathological processes in skin. We studied the effect of KYN, KYNA and FICZ on melanocyte and melanoma A375 and RPMI7951 cell toxicity, proliferation and cell death. KYN and FICZ inhibited DNA synthesis in both melanoma cell lines, but RPMI7951 cells were more resistant to pharmacological treatment. Tested compounds were toxic to melanoma cells but not to normal human adult melanocytes. Changes in the protein level of cyclin D1, CDK4 and retinoblastoma tumor suppressor protein (Rb) phosphorylation revealed different mechanisms of action of individual AhR ligands. Importantly, all tryptophan metabolites induced necrosis, but only KYNA and FICZ promoted apoptosis in melanoma A375 cells. This effect was not observed in RPMI7951 cells. KYN, KYNA and FICZ in higher concentrations inhibited the protein level of AhR but did not affect the gene expression. To conclude, despite belonging to the group of AhR ligands, KYN, KYNA and FICZ exerted different effects on proliferation, toxicity and induction of cell death in melanoma cells in vitro.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Carbazoles/pharmacology , Kynurenic Acid/pharmacology , Kynurenine/pharmacology , Melanoma/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanocytes/drug effects , Melanocytes/metabolism , Melanoma/drug therapy , Phosphorylation/drug effects , Retinoblastoma Protein/metabolism
10.
Eur J Pharmacol ; 881: 173207, 2020 Aug 15.
Article En | MEDLINE | ID: mdl-32446712

In the recent years, coumarin bioactive compounds have been identified to posess anticancer properties. Therefore, the aim of the present study was to investigate for the first time the efficacy of osthole, umbelliferone, esculin, and 4-hydroxycoumarin, alone and in combination with Temozolomide, in the elimination of deadly brain tumors, anaplastic astrocytoma (AA) and glioblastoma multiforme (GBM) cells via programmed death. Our results indicated that osthole, umbelliferone, esculin, and 4-hydroxycoumarin initiated mainly apoptosis in the T98G and MOGGCCM cells. Osthole was the most effective. It also initiated autophagy in a small percentage of the cell population. The co-incubation with Temozolomide did not increase the pro-apoptotic potential of natural compounds but decreased the level of autophagy in the T98G cells. Apoptosis was associated with reduced mitochondrial membrane potential, activation of caspase 3, inhibition of Bcl-2 expression and the presence of a Bcl-2/Beclin 1. Blocking of Bcl-2 expression resulted in promotion of apoptosis, but not autophagy, in the MOGGCCM and T98G lines. It also sensitized astrocytoma cells, but not GBM, to the combined osthole and TMZ treatment, which was correlated with a reduced level of Beclin 1 and increased expression of caspase 3. Osthole and TMZ, alone and in combination, inhibited the migratory phenotype of the GBM and AA cells. In summary, our results indicated that osthole effectively eliminated glioma cells via apoptosis, what was correlated with Bcl-2/Beclin 1 complex formation. Considering the anti-migratory effect, osthole and Temozolomide display antiglioma potential but it needs further extensive studies.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Coumarins/pharmacology , Glioma/drug therapy , Temozolomide/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Beclin-1/genetics , Beclin-1/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , Membrane Potential, Mitochondrial/drug effects , Necrosis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction
11.
Molecules ; 25(7)2020 Apr 03.
Article En | MEDLINE | ID: mdl-32260268

8-Hydroxyquinaldic acid, the end-metabolite of tryptophan, is well-known metal chelator; however, its role in humans, especially in cancer promotion and progression, has not been fully revealed. Importantly, 8-hydroxyquinaldic acid is the analog of kynurenic acid with evidenced antiproliferative activity towards various cancer cells. In this study, we revealed that 8-hydroxyquinaldic acid inhibited not only proliferation and mitochondrial activity in colon cancer HT-29 and LS-180 cells, but it also decreased DNA synthesis up to 90.9% for HT-29 cells and 76.1% for LS-180 cells. 8-Hydroxyquinaldic acid induced changes in protein expression of cell cycle regulators (CDK4, CDK6, cyclin D1, cyclin E) and CDKs inhibitors (p21 Waf1/Cip1, p27 Kip1), but the effect was dependent on the tested cell line. Moreover, 8-hydroxyquinaldic acid inhibited migration of colon cancer HT-29 and LS-180 cells and increased the expression of ß-catenin and E-cadherin. Importantly, antiproliferative and anti-migratory concentrations of 8-hydroxyquinaldic acid were non-toxic in vitro and in vivo. We reported for the first time antiproliferative and anti-migratory activity of 8-hydroxyquinaldic acid against colon cancer HT-29 and LS-180 cells.


Cell Cycle Proteins/metabolism , Colorectal Neoplasms/metabolism , Kynurenic Acid/analogs & derivatives , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Kynurenic Acid/pharmacology , Mitochondria/metabolism , Tryptophan/chemistry , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
12.
J Food Biochem ; 43(4): e12802, 2019 04.
Article En | MEDLINE | ID: mdl-31353575

Lycopene, sulforaphane, quercetin, and curcumin, ingredients of daily diet, show significant anticancer and chemopreventive potential; however, no data are available showing thorough evaluation of jointly used phytochemicals on cancer cell proliferation. Here, we compare anticancer potential of mentioned substances applied separately or in combination (as MIX) by measuring mitochondrial activity (MTT test), DNA synthesis (BrdU test) and lactate dehydrogenase release (LDH test) in colon epithelial (CCD841 CoTr), and colon cancer (HT-29, LS174T) cells. Additive inhibitory effect of simultaneously used phytochemicals on cancer cells proliferation has been shown. In epithelial cells, tested combination effectively inhibited mitochondrial activity, but not DNA synthesis. LDH test revealed cytotoxicity of tested mixture against cancer cells without negative effect on normal cells. Furthermore, we demonstrated that MIX enhances antiproliferative effect of common cytostatics: 5-fluorouracil and cisplatin. Presented data suggest chemopreventive potential of the proposed combination of natural substances and their usefulness as adjuvant strategy during chemotherapy. PRACTICAL APPLICATIONS: Colorectal cancer is one of the most common causes of cancer death worldwide. Since its development and progression is strongly correlated with dietary habits, healthy diet as well as supplementation with proved anticancer agents seems to be reasonable strategy of colon cancer prevention and treatment. In the present study, we have focused on four natural compounds abundantly found in daily diet i.e., lycopene, sulforaphane, quercetin, and curcumin, with well established anticancer potential. Their individual and collective impact both on normal colon epithelium cells and colon cancer cells viability, growth, and proliferation was examined. Furthermore, activity of the substances combined as MIX to influence antiproliferative potential of commonly used in colon cancer treatment cytostatics, 5-fluorouracil, and cisplatin was verified. Proposed in the study combination of phytochemicals with experimentally proven antiproliferative activity may propose an effective strategy for prevention and treatment of colon cancer.


Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/physiopathology , Curcumin/pharmacology , Isothiocyanates/pharmacology , Lycopene/pharmacology , Quercetin/pharmacology , Cell Survival/drug effects , Colonic Neoplasms/genetics , Drug Synergism , HT29 Cells , Humans , Mitochondria/drug effects , Mitochondria/genetics , Sulfoxides
13.
Pharmacol Rep ; 71(2): 189-193, 2019 Apr.
Article En | MEDLINE | ID: mdl-30780127

BACKGROUND: Origin, synthesis and activity of quinaldic acid (QA), proposed derivative of kynurenic acid, have been poorly studied to date. Previously, we have demonstrated the antiproliferative effect of QA in a colon cancer model in vitro. The goal of present study was to verify QA activity to modify the expression of p53 tumor suppressor in colon cancer cells, and to relate it to its cancer cell growth inhibiting activity in vitro. METHODS: LS180 colon cancer cells possessing the wild type form of p53 were used in the study. Real-time PCR and immunobloting techniques were used to test the expression of p53 at gene and protein level, respectively. Next, immunocytochemistry was used to visualize the localization of p53 protein within the cells. Furthermore, the antiproliferative activity of QA was retested in cells with siRNA silenced P53 gene. RESULTS: The activity of QA to modify both the expression and phosphorylation of p53 protein as well as the level of P53 gene is shown. Concomitantly, the nuclear and cytoplasmic localization of phospho-p53 protein upon QA treatment is also presented. Moreover, reduced activity of QA in colon cancer cells with silenced p53 expression is observed. CONCLUSION: QA affects the expression of p53 tumor suppressor, both at gene and protein level. The prominent contribution of p53 to the antiproliferative effect of QA in LS180 colon cancer cells can be suggested.


Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Quinolines/pharmacology , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , RNA, Small Interfering/administration & dosage , Real-Time Polymerase Chain Reaction
14.
Eur J Pharm Sci ; 129: 42-57, 2019 Mar 01.
Article En | MEDLINE | ID: mdl-30594731

The treatment of epilepsy remains difficult mostly since almost 30% of patients suffer from pharmacoresistant forms of the disease. Therefore, there is an urgent need to search for new antiepileptic drug candidates. Previously, it has been shown that 4-alkyl-5-substituted-1,2,4-triazole-3-thione derivativatives possessed strong anticonvulsant activity in a maximal electroshock-induced seizure model of epilepsy. In this work, we examined the effect of the chemical structure of the 1,2,4-triazole-3-thione-based molecules on the anticonvulsant activity and the binding to voltage-gated sodium channels (VGSCs) and GABAA receptors. Docking simulations allowed us to determine the mode of interactions between the investigated compounds and binding cavity of the human VGSC. Selected compounds were also investigated in a panel of ADME-Tox assays, including parallel artificial membrane permeability assay (PAMPA), single cell gel electrophoresis (SCGE) and cytotoxicity evaluation in HepG2 cells. The obtained results indicated that unbranched alkyl chains, from butyl to hexyl, attached to 1,2,4-triazole core are essential both for good anticonvulsant activity and strong interactions with VGSCs. The combined in-vivo, in-vitro and in-silico studies emphasize 4-alkyl-5-substituted-1,2,4-triazole-3-thiones as promising agents in the development of new anticonvulsants.


Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Triazoles/chemistry , Triazoles/pharmacology , Voltage-Gated Sodium Channels/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Computer Simulation , Electroshock/methods , HEK293 Cells , Hep G2 Cells , Humans , Mice , Molecular Docking Simulation/methods , Receptors, GABA-A/metabolism , Seizures/drug therapy
15.
Pharmacol Rep ; 70(2): 277-283, 2018 Apr.
Article En | MEDLINE | ID: mdl-29477035

BACKGROUND: Previously, we have demonstrated that kynurenic acid (KYNA), an endogenous metabolite of tryptophan formed along kynurenine pathway, is present in synovial fluid of rheumatoid arthritis (RA) and osteoarthritis (OA) patients. In this study, the goal was to investigate the presence of quinaldic acid (QUDA), a putative metabolite of KYNA, in synovial fluid of RA and OA patients. METHODS: The effect of QUDA on proliferation and motility of synovial fibroblasts and its interaction with KYNA were determined in vitro. The study was conducted on synovial fluid obtained from 38 patients with RA and 15 patients with OA. QUDA was identified and quantified using the gas chromatography-mass spectrometry (GC-MS) method. In vitro experiments were conducted on rabbit synoviocyte cell line HIG-82. RESULTS: Presence of QUDA was detected in all 53 samples of synovial fluid. The concentration of QUDA in synovial fluid obtained from patients with RA was 28.6 ±â€¯14.9 pmol/ml, which was lower in comparison with OA 42.3 ±â€¯10.0 pmol/ml. QUDA content positively correlated with the number of tender joints and negatively with the total cell counts determined in synovial fluid of RA patients. It did not correlate with KYNA content. QUDA reduced both proliferation and motility of synoviocytes in a dose-dependent manner. The enhancement of antiproliferative action of QUDA by KYNA was evidenced. CONCLUSIONS: Data show a local deficit of QUDA in RA patients and suggest its potential role as an endogenous substance controlling synoviocyte viability.


Arthritis, Rheumatoid/metabolism , Osteoarthritis/metabolism , Quinolines/metabolism , Synovial Fluid/metabolism , Synoviocytes/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Proliferation/physiology , Cells, Cultured , Female , Fibroblasts/metabolism , Humans , Kynurenic Acid/metabolism , Male , Middle Aged , Rabbits , Synovial Membrane/metabolism , Young Adult
16.
Pharmacol Rep ; 69(4): 779-787, 2017 Aug.
Article En | MEDLINE | ID: mdl-28587939

BACKGROUND: Gliomas are aggressive brain tumors with very high resistance to chemotherapy. Therefore, the aim of the present study was to investigate the effectiveness of sorafenib and Temozolomide in elimination of human glioma cells through apoptosis and autophagy. METHODS: MOGGCCM (anaplastic astrocytoma) and T98G (glioblastoma multiforme) cell lines incubated with sorafenib and/or Temozolomide were used in the experiments. Cell morphology (ER stress, apoptosis, autophagy, and necrosis) was analyzed microscopically while apoptosis and mitochondrial membrane potential were assessed with flow cytometry. Beclin1, LC3, p62, Hsp27, and Hsp72 levels were analyzed by immunoblotting. The activity of caspase 3, 8, and 9 was evaluated fluorometrically. Expression of Hsps was blocked by transfection with specific siRNA. RESULTS: In MOGGCCM cells, Temozolomide most frequently induced autophagy, which was accompanied by decreased p62 and increased beclin1 and LC3II levels. Sorafenib initiated mainly apoptosis. Additional incubation with Temozolomide, synergistically potentiated the pro-apoptotic properties of sorafenib, but it was mediated in a caspase-independent way. In T98G cells, the effect of the analyzed drugs on programmed cell death induction was different from that in MOGGCCM cells. Sorafenib induced autophagy, while Temozolomide initiated mainly apoptosis. After simultaneous drug application, apoptosis dominated, suggesting synergistic action of both drugs. Inhibition of Hsp27 and Hsp72 expression increased the sensitivity of both cell lines to ER stress and, to a lesser extent, to induction of apoptosis, but not autophagy. CONCLUSIONS: Sorafenib and Temozolomide applied in combination are potent apoptosis inducers in T98G and MOGGCCM cells. ER stress precedes the elimination. Blocking of Hsp expression has a greater impact on ER stress rather than apoptosis induction.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Astrocytoma/drug therapy , Dacarbazine/analogs & derivatives , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Cell Line, Tumor , Dacarbazine/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Niacinamide/pharmacology , RNA Interference , Sorafenib , Temozolomide
17.
Neurochem Res ; 42(3): 838-845, 2017 Mar.
Article En | MEDLINE | ID: mdl-27444613

Kynurenic acid (KYNA) is an end stage product of tryptophan metabolism with a variety of functions in the human body, both in the central nervous system (CNS) and in other organs. Although its activity in the human brain has been widely studied and effects on neural cells were emphasized, the effect of KYNA on oligodendroglial cells remains unknown. Present study aims at describing the activity of high concentration of KYNA in OLN-93 cells. The inhibition of OLN-93 oligodendrocytes viability by KYNA in a medium with reduced serum concentration has been demonstrated. Although decreased metabolic activity of KYNA treated OLN-93 cells was shown, the cells proliferation was not altered. KYNA treatment did not alter morphology as well as expression level of cell cycle and proliferation regulating proteins. Furthermore, glutamate receptor antagonists and agonists did not alter the inhibitory effect of KYNA on viability of OLN-93 oligodendrocytes. This study contributes to the elucidation of effects of KYNA on oligodendrocytes in vitro, yet further analyses are necessary to explain the mechanisms behind the damage and loss of myelin sheaths.


Glutamates/metabolism , Kynurenic Acid/metabolism , Oligodendroglia/metabolism , Animals , Cell Line , Cell Proliferation , Cell Survival , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Kynurenic Acid/pharmacology , Oligodendroglia/cytology , Oligodendroglia/drug effects , Rats
18.
Bioorg Med Chem ; 24(6): 1356-61, 2016 Mar 15.
Article En | MEDLINE | ID: mdl-26897091

2-(2,4-Dihydroxyphenyl)thieno-1,3-thiazin-4-ones are a group of new compounds with potential anticancer activity. This type of derivatives was poorly investigated in the area of synthesis and biological activities. In the present study the antiproliferative action of the most active derivative BChTT was described. The aim of biological evaluation was to investigate the ability of the compound to inhibit cancer cell proliferation and identify mechanism involved in its action on the molecular level. BChTT inhibited the proliferation of lung cancer A549, colon cancer HT-29 and glioma C6 cells in the concentration-dependent manner. It was not toxic to normal cells including skin fibroblasts, hepatocytes and oligodendrocytes in the antiproliferative concentrations. BChTT decreased the DNA synthesis in the treated cancer cells and induced cell cycle arrest in the G0/G1 phase. Moreover, the ability of the compound to activate p38 kinase and decrease cyclin D1 expression was estimated. Participation of p38 kinase in the antiproliferative action of the compound was confirmed by the analysis of BChTT activity in the cells with the p38 silenced gene. The obtained results may suggest the ability of the tested derivative to inhibit cancer cells proliferation by induction of p38-mediated cyclin D1 downregulation.


Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Neoplasms/enzymology , Neoplasms/pathology , Thiazines/pharmacology , Thiophenes/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HT29 Cells , Humans , Molecular Structure , Rats , Structure-Activity Relationship , Thiazines/chemical synthesis , Thiazines/chemistry , Thiophenes/chemical synthesis , Thiophenes/chemistry
19.
Monatsh Chem ; 146(8): 1315-1327, 2015.
Article En | MEDLINE | ID: mdl-26190864

ABSTRACT: We reported the synthesis and characterization of a series of azolo- and azino[1,3]thiazinones containing the 2,4-dihydroxyphenyl substituent. The compounds were prepared by a new one-step reaction of aryl-modified sulfinylbis[(2,4-dihydroxyphenyl)methanethione]s and the corresponding aminoazolo(azino)carboxamides. Their chemical structures were confirmed by IR, NMR: 1H, 13C, HSQC, and EI-MS spectral data. The compounds inhibited proliferation and viability of lung cancer A549, colon cancer HT-29, and glioma C6 cells in a structure- and concentration-dependent manner. The activity of some analogues was below 10 µmol dm-3 (IC50). Glioma C6 cells were the most sensitive to tested compounds. Generally, the derivatives were not toxic for the skin fibroblast HSF culture. Moreover, some of them exerted a protective effect on the treated normal cells. Evaluation of compound properties in silico showed that they possess significant drug-like characteristics and most of them display a low toxicity.

20.
Mol Divers ; 19(4): 725-36, 2015 Nov.
Article En | MEDLINE | ID: mdl-25920594

A new one-step synthesis of novel biologically active 2-substituted 2,4-dihydroxyphenyl-4[Formula: see text]-thieno[3,2-[Formula: see text]][1,3]thiazin-4-ones and 4[Formula: see text]-thieno[2,3-[Formula: see text]][1,3]thiazin-4-ones has been elaborated and described. The compounds were prepared by the reaction of aryl-modified sulfinylbis [(2,4-dihydroxyphenyl)methanethione]s and the corresponding aminothiophenecarboxamides. The derivatives showed anticancer activity in vitro. These compounds inhibited the proliferation and viability of lung cancer A549, colon cancer HT-29 and glioma C6 cells in a concentration-dependent manner. Some of the derivatives had no influence on normal skin fibroblasts culture viability. Moreover, one compound (1b) showed the ability to inhibit DNA synthesis in cancer cells, especially in C6 cells, and was not toxic for normal oligodendrocytes and hepatocytes. Using reversed phase RP 18 HPLC and immobilised artificial membrane (IAM) chromatography the phase affinity of the compounds was determined. The influence of lipophilicity on the activity of compounds has been discussed.


Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Thiazines/chemical synthesis , Thiazines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography , Drug Screening Assays, Antitumor , HT29 Cells , Humans , Molecular Structure
...