Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Article in English | MEDLINE | ID: mdl-38916832

ABSTRACT

Chemotherapy resistance is a major obstacle in cancer therapy, and identifying novel druggable targets to reverse this phenomenon is essential. The exosome-mediated transmittance of drug resistance has been shown in various cancer models including ovarian and prostate cancer models. In this study, we aimed to investigate the role of exosomal miRNA transfer in chronic myeloid leukemia drug resistance. For this purpose, firstly exosomes were isolated from imatinib sensitive (K562S) and resistant (K562R) chronic myeloid leukemia (CML) cells and named as Sexo and Rexo, respectively. Then, miRNA microarray was used to compare miRNA profiles of K562S, K562R, Sexo, Rexo, and Rexo-treated K562S cells. According to our results, miR-125b-5p and miR-99a-5p exhibited increased expression in resistant cells, their exosomes, and Rexo-treated sensitive cells compared to their sensitive counterparts. On the other hand, miR-210-3p and miR-193b-3p were determined to be the two miRNAs which exhibited decreased expression profile in resistant cells and their exosomes compared to their sensitive counterparts. Gene targets, signaling pathways, and enrichment analysis were performed for these miRNAs by TargetScan, KEGG, and DAVID. Potential interactions between gene candidates at the protein level were analyzed via STRING and Cytoscape software. Our findings revealed CCR5, GRK2, EDN1, ARRB1, P2RY2, LAMC2, PAK3, PAK4, and GIT2 as novel gene targets that may play roles in exosomal imatinib resistance transfer as well as mTOR, STAT3, MCL1, LAMC1, and KRAS which are already linked to imatinib resistance. MDR1 mRNA exhibited higher expression in Rexo compared to Sexo as well as in K562S cells treated with Rexo compared to K562S cells which may suggest exosomal transfer of MDR1 mRNA.

2.
Toxicol In Vitro ; 95: 105754, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38061604

ABSTRACT

In this study, we aimed to analyze the effects of first and second-generation Bcr-Abl tyrosine kinase inhibitors, imatinib and nilotinib on LPS/IFN gamma activated RAW 264.7 macrophages. Our data revealed that imatinib was less effective on nitrite levels and more toxic on macrophages compared to nilotinib. Therefore, we further analysed the effect of nilotinib on various inflammatory markers including iNOS, COX-2, NFkB, IL-6, p-ERK, p-p38 and p-JNK in LPS/IFN gamma activated RAW264.7 macrophages. Spectrophotometric viability test and Griess assay,western blot, RT-PCR and luciferase reporter assays were used to analyze the biological activity of nilotinib. Our findings revealed that nilotinib decreases nitrite levels, iNOS mRNA, iNOS and p-p38 protein expressions significantly whereas induces IL-6 mRNA and p-JNK protein expressions at particular doses. We did not find significant effect of nilotinib on COX-2, p-ERK and nuclear p65 proteins and NFkB transcriptional activity. In addition, the binding mode of nilotinib to iNOS protein was predicted by molecular docking. According to the docking analyses, nilotinib exhibited hydrophobic interactions between MET349, ALA191, VAL346, PHE363, TYR367, MET368, CYS194, TRP366 residues at the binding pocket and the molecule as well as van der Waals interactions at specific residues. In conclusion, our results reveal that, in addition to its anticancer activity, nilotinib can exhibit immune modulatory effects on macrophages through its effects on iNOS, IL-6, p-p38 and p-JNK.


Subject(s)
Lipopolysaccharides , Nitrites , Imatinib Mesylate/pharmacology , Lipopolysaccharides/pharmacology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Nitrites/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Molecular Docking Simulation , Macrophages , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Pyrimidines/toxicity , RNA, Messenger/metabolism
3.
Adv Med Sci ; 68(2): 238-248, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37421850

ABSTRACT

PURPOSE: Chronic myeloid leukemia (CML) is a hematological malignancy characterized by the presence of BCR-ABL protein. Imatinib (IMA) is considered as the first line therapy in management of CML which particularly targets the BCR-ABL tyrosine kinase protein. However, emergence of resistance to IMA hinders its clinical efficiency. Hence, identifying novel targets for therapeutic approaches in CML treatment is of great importance. Here, we characterize a new subpopulation of highly adherent IMA-resistant CML cells that express stemness and adhesion markers compared to naive counterparts. MATERIALS AND METHODS: We performed several experimental assays including FISH, flow cytometry, and gene expression assays. Additionally, bioinformatics analysis was performed by normalized web-available microarray data (GSE120932) to revalidate and introduce probable biomarkers. Protein-protein interactions (PPI) network was analyzed by the STRING database employing Cytoscape v3.8.2. RESULTS: Our findings demonstrated that constant exposure to 5 â€‹µM IMA led to development of the adherent phenotype (K562R-adh). FISH and BCR-ABL expression analysis indicated that K562R-adh cells were derived from the original cells (K562R). In order to determine the role of various genes involved in epithelial-mesenchymal transition (EMT) and stem cell characterization, up/down-regulation of various genes including cancer stem cell (CSC), adhesion and cell surface markers and integrins were observed which was similar to the findings of the GSE120932 dataset. CONCLUSION: Treating CML patients with tyrosine kinase inhibitors (TKIs) as well as targeting adhesion molecules deemed to be effective approaches in prevention of IMA resistance emergence which in turn may provide promising effects in the clinical management of CML patients.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Protein Kinase Inhibitors , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Drug Resistance, Neoplasm/genetics , K562 Cells , Apoptosis , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Fusion Proteins, bcr-abl/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Phenotype
4.
Cytoskeleton (Hoboken) ; 80(9-10): 367-381, 2023.
Article in English | MEDLINE | ID: mdl-36961307

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most common forms of brain tumor. As an excessively invasive tumor type, GBM cannot be fully cured due to its invasion ability into healthy brain tissues. Therefore, molecular mechanisms behind GBM migration and invasion need to be deeply investigated for the development of effective GBM treatments. Cellular motility and invasion are strictly associated with the cytoskeleton, especially with actins and tubulins. Palladin, an actin-binding protein, tightly bundles actins during initial invadopodia and contraction fiber formations, which are essential for cellular motility. Spastin, a microtubule-binding protein, cuts microtubules into small pieces and acts on invadopodia elongation and cellular trafficking of invadopodia-associated proteins. Regulation of proteins such as spastin and palladin involved in dynamic reorganization of the cytoskeleton, are rapidly carried out by microRNAs at the posttranscriptional level. Therefore, determining possible regulatory miRNAs of spastin and palladin is critical to elucidate GBM motility. miR96 and miR182 down-regulate SPAST and PALLD at both transcript and protein levels. Over-expression of miR96 and miR182 resulted in inhibition of the motility. However, over-expression of spastin and palladin induced the motility. Spastin and palladin rescue of miR96- or miR182-transfected U251 MG cells resulted in diminished effects of the miRNAs and rescued the motility. Our results demonstrate that miR96 and miR182 over-expressions inhibit GBM motility by regulating cytoskeleton through spastin and palladin. These findings suggest that miR96 and miR182 should be investigated in more detail for their potential use in GBM therapy.

5.
Cells ; 12(3)2023 01 27.
Article in English | MEDLINE | ID: mdl-36766769

ABSTRACT

Microtubule-severing protein Spastin has been shown to co-localize with actin in migratory glioblastoma cells and is linked to glioblastomas' migration and invasion capacity. However, the effectiveness of Spastin in glioblastoma migration and the molecular mechanism underpinning the orientation of Spastin towards actin filaments remain unknown. Here, we demonstrated that Spastin plays an active role in glioblastoma migration by showing a reduced migratory potential of T98G glioblastoma cells using real-time cell analysis (RTCA) in Spastin-depleted cells. Pull-down assays revealed that a cis-trans isomerase Pin1 interacts with Spastin through binding to the phosphorylated Pin1 recognition motifs in the microtubule-binding domain (MBD), and immunocytochemistry analysis showed that interaction with Pin1 directs Spastin to actin filaments in extended cell regions. Consequently, by utilizing RTCA, we proved that the migration and invasion capacity of T98G glioblastoma cells significantly increased with the overexpression of Spastin, of which the Pin1 recognition motifs in MBD are constitutively phosphorylated, while the overexpression of phospho-mutant form did not have a significant effect on migration and invasion of T98G glioblastoma cells. These findings demonstrate that Pin1 is a novel interaction partner of Spastin, and their interaction drives Spastin to actin filaments, allowing Spastin to contribute to the glioblastomas' migration and invasion abilities.


Subject(s)
Glioblastoma , Humans , Glioblastoma/metabolism , Microtubules/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Protein Binding , Spastin/metabolism
6.
Fundam Clin Pharmacol ; 37(3): 557-565, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36690337

ABSTRACT

Chemotherapy with targeted drugs is the first line therapy option for acute and chronic myeloid leukemia. However, hematopoietic stem cell transplantation may be used in high-risk patients or patients with failed responses to chemo drugs. Discovery and development of more effective new agents with lower side effects is the main aim of leukemia treatment. In this study, a novel retinoid compound with tetrahydronaphthalene ring was synthesized and evaluated for anticancer activity in human chronic and acute myeloid leukemia cell lines K562 and HL-60. Novel N-(1H-indol-1-yl)-5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalene-2-carboxamide was synthesized based on molecular hybridization of the two different bioactive structures retinoid head and indole. The effects of the synthesized carboxamide compound, which was referred to as compound 5, were determined in K562 chronic myeloid leukemia and HL-60 acute myeloid leukemia cell lines and L929 fibroblast cell line, which served as a control. Colorimetric MTT and caspase3 activity tests, flow cytometry, western blot, and microscopic examinations were used to evaluate biological activity. Compound 5 more effectively induced cell death in HL60 cells in comparison to K562 cells and L929 fibroblast cells. Therefore, further mechanism of cell death was investigated in HL60 cell line. It was found that compound 5 induced remarkable cytotoxicity, caspase3 activation, and PARP fragmentation in HL60 cells. Flow cytometric staining showed that the percentage of cells arrested in G0/G1 was also increased with compound 5 treatment. Important modulator proteins of cell proliferation p-ERK, p-AKT, and p-m-TOR were also found to be inhibited with compound 5 treatment. Collectively, our results reveal compound 5, which is a novel indole retinoid compound as a potential active agent for the treatment of acute promyelocytic leukemia.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukemia, Myeloid, Acute , Humans , HL-60 Cells , Proto-Oncogene Proteins c-akt , Retinoids/pharmacology , Apoptosis , Cell Cycle Checkpoints , Cell Proliferation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Indoles/pharmacology , Tetrahydronaphthalenes/pharmacology
7.
Mol Genet Genomics ; 297(4): 1141-1150, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35704118

ABSTRACT

In this study, we aimed to determine the genetic basis of a Turkish family related to hereditary spastic paraplegia (HSP) by exome sequencing. HSP is a progressive neurodegenerative disorder and displays genetic and clinical heterogeneity. The major symptoms are muscle weakness and spasticity, especially in the lower extremities. We studied seven affected and seven unaffected family members, as well as a clinically undetermined member, to identify the disease-causing gene. Exome sequencing was performed for four affected and two unaffected individuals. The variants were firstly filtered for HSP-associated genes, and we found a common variant in the ZFYVE27 gene, which has been previously implied for association with HSP. Due to the incompletely penetrant segregation pattern of the ZFYVE27 variant, revealed by Sanger sequencing, with the disease in this family, filtering was re-performed according to the mode of inheritance and allelic frequencies. The resulting 14 rare variants were further evaluated in terms of their cellular functions, and three candidate variants in ATAD3C, VPS16, and MYO1H genes were selected as possible causative variants, which were analyzed for their familial segregation. ATAD3C and VPS16 variants were eliminated due to incomplete penetrance. Eventually, the MYO1H variant NM_001101421.3:c.2972_2974del (p.Glu992del, rs372231088) was found as the possible disease-causing deletion for HSP in this family. This is the first study reporting the possible role of a MYO1H variant in HSP pathogenesis. Further studies on the cellular roles of Myo1h protein are needed to validate the causality of MYO1H gene at the onset of HSP.


Subject(s)
Myosin Type I , Spastic Paraplegia, Hereditary , Humans , Inheritance Patterns , Mutation , Myosin Type I/genetics , Pedigree , Proteins/genetics , Spastic Paraplegia, Hereditary/diagnosis , Spastic Paraplegia, Hereditary/genetics , Vesicular Transport Proteins/genetics , Exome Sequencing
8.
Hum Mol Genet ; 31(11): 1844-1859, 2022 06 04.
Article in English | MEDLINE | ID: mdl-34935948

ABSTRACT

Hereditary spastic paraplegia (HSP) is a disease in which dieback degeneration of corticospinal tracts, accompanied by axonal swellings, leads to gait deficiencies. SPG4-HSP, the most common form of the disease, results from mutations of human spastin gene (SPAST), which is the gene that encodes spastin, a microtubule-severing protein. The lack of a vertebrate model that recapitulates both the etiology and symptoms of SPG4-HSP has stymied the development of effective therapies for the disease. hSPAST-C448Y mice, which express human mutant spastin at the ROSA26 locus, display corticospinal dieback and gait deficiencies but not axonal swellings. On the other hand, mouse spastin gene (Spast)-knockout (KO) mice display axonal swellings but not corticospinal dieback or gait deficiencies. One possibility is that reduced spastin function, resulting in axonal swellings, is not the cause of the disease but exacerbates the toxic effects of the mutant protein. To explore this idea, Spast-KO and hSPAST-C448Y mice were crossbred, and the offspring were compared with the parental lines via histological and behavioral analyses. The crossbred animals displayed axonal swellings as well as earlier onset, worsened gait deficiencies and corticospinal dieback compared with the hSPAST-C448Y mouse. These results, together with observations on changes in histone deacetylases 6 and tubulin modifications in the axon, indicate that each of these three transgenic mouse lines is valuable for investigating a different component of the disease pathology. Moreover, the crossbred mice are the best vertebrate model to date for testing potential therapies for SPG4-HSP.


Subject(s)
Spastic Paraplegia, Hereditary , Spastin , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Gain of Function Mutation , Humans , Loss of Function Mutation , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Spastin/genetics
9.
Leuk Res ; 102: 106523, 2021 03.
Article in English | MEDLINE | ID: mdl-33607534

ABSTRACT

INTRODUCTION: Chronic Myeloid Leukemia (CML) is a hematological disease which is characterized by the presence of BCR-ABL fusion protein. Imatinib (IMA), a tyrosine kinase inhibitor of BCR-ABL, is used as a frontline treatment.Although IMA aids in killing a majority of leukemia cells, it may not kill CML stem cells which are the primary roots of disease and therapy resistance. Recently, antimicrobial drugs have been gaining attention because of their selective targeting of cancer cells. Therefore, we now ask if combinational therapy of IMA with a targeted antimicrobial drug Azithromycin (AZT) can enhance the treatment efficiency in IMA resistant CML. METHODS: K562S (IMA sensitive) and K562R (IMA resistant) cells were treated with increasing concentrations of AZT to determine its effects on cell proliferation and apoptosis. Cell viability, apoptosis, caspase3/7 activity and P-glycoprotein (Pgp) function were investigated with spectrophotometric MTT assay and flow cytometric Annexin V staining, caspase 3/7 activity, and Rhodamine123 staining assays respectively. The expression levels of pro-apoptotic (BAX, BAD and BIM), anti- apoptotic (BCL-XL and BCL-2) and drug transporter (MDR-1 and MRP-1) genes were assessed with qRT-PCR. RESULTS: AZT treatment alone inhibited cell viability, induced apoptosis and enhanced caspase 3/7 activity in both K562S and high MDR-1 (Pgp) expressing K562R cells. Moreover, combination of AZT/IMA suppressed cell viability, induced apoptosis and caspase3/7 activity more effectively and significantly compared to K562R cells treated with only IMA or AZT. Furthermore, AZT and AZT/IMA combination decreased Pgp function in K562R cells in comparison with their controls. Based on qRT-PCR data, single AZT and combined AZT/IMA treatment also induced BAX/BCL-2 ratio significantly in both K562S and K562R cells. CONCLUSION: Single AZT and AZT/IMA combinational treatment can be proposed as a promising and effective treatment strategy for CML. One of the mechanisms underlying the potent anticancer effect of combined AZT/IMA could be its ability to inhibit Pgp function and increase intracellular accumulation of IMA which leads to the induction of apoptosis in K562R cells.


Subject(s)
Antineoplastic Agents/pharmacology , Azithromycin/pharmacology , Drug Resistance, Neoplasm/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Imatinib Mesylate/pharmacology
10.
Turk J Pharm Sci ; 18(1): 75-79, 2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33634671

ABSTRACT

OBJECTIVES: Probucol is a bisphenol antioxidant with antiinflammatory, antilipidemic and antidiabetic effect. Development and progression of cancer is closely related to chronic inflammation and oxidative stress. Agents that target these processes have been shown to modulate cancer cell proliferation. In this regard, the effect of probucol on proliferation of different cancer cell lines was investigated. MATERIALS AND METHODS: Different concentrations of probucol solutions were prepared and applied to the following cancer cell lines: K562S (imatinib sensitive) and K562R (imatinib resistant) chronic myeloid leukemia (CML) cells; U937 histiocytic lymphoma cells; HL60 acute myeloid leukemia cells; U266, H929, and RPMI8226 multiple myeloma cells; and L929 fibroblast cells. Cell viability was conducted by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. RESULTS: Significant toxicity was not exhibited due to probucol treatment (0.1-10 µM) in K562S and K562R CML cells, U937 histiocytic lymphoma cells, HL60 acute myeloid leukemia cells, U266 multiple myeloma cells, and L929 fibroblast cells. However, probucol treatment significantly inhibited the viability of H929 and RPMI8226 multiple myeloma cells at the concentration of 0.5-10 µM and 5-10 µM, respectively. CONCLUSION: Probucol treatment slightly inhibited the viability of other cancer cell lines, but significantly inhibited the viability of H929 and RPMI8226 multiple myeloma cells. However, its effect was not potent, since a 50% reduction in cell viability could not be achieved at the concentrations of probucol treatment administered.

11.
Brain Res ; 1750: 147167, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33091395

ABSTRACT

BACKGROUND: Kufor-Rakeb Syndrome (KRS) is an autosomal recessive disease characterized by Parkinsonism, pyramidal signs, dementia, and supranuclear gaze palsy. KRS is caused by mutations in ATP13A2producing a transmembrane protein responsible for the regulation of intracellular inorganic cations. OBJECTIVE: Two siblings born to a Turkish family of consanguineous marriage had mixed neurological presentations with the presence of hypointense images on T2-weighted MRI and were pre-diagnosed as having autosomal recessive spastic paraparesis or ataxia.We aimed to identify the disease-causing mutation by whole-exome sequencing and elucidate the underlying molecular mechanism of the causative mutation. METHODS: Prussian blue staining was conducted for the detection of cellular iron accumulation. Disease-causing mutation inATP13A2was detected by whole-exome sequencing. Expression levels of ATP13A2 mRNA and protein were assessed by qRT-PCR and Western Blot. RESULTS: Iron deposits in the patients' fibroblasts were detected by Prussian blue staining. Novel homozygous mutation c.1422_1423del:p.P474fs was detected intheATP13A2. As this mutation caused a premature termination codon (PTC), the expression of mutant ATP13A2 mRNA through qRT-PCR analysis was found to be degraded by nonsense-mediated decay and this prevented the expression of ATP13A2 protein in the patients' fibroblasts. CONCLUSIONS: Novel frameshift mutation causing a PTC in ATP13A2 lead to degradation of ATP13A2 mRNA by NMD. Iron accumulation due to the absence of ATP13A2 protein in the patient's fibroblasts and hypointense areas on T2-weighted images may expand the spectrum of KRS to consider it as neurodegeneration with brain iron accumulation disorders.


Subject(s)
Iron/metabolism , Parkinsonian Disorders/genetics , Proton-Translocating ATPases/genetics , Adult , Brain/metabolism , Female , Humans , Iron/toxicity , Magnetic Resonance Imaging , Male , Mutation , Nonsense Mediated mRNA Decay/genetics , Parkinsonian Disorders/metabolism , Primary Cell Culture , Proton-Translocating ATPases/metabolism , Siblings , Exome Sequencing/methods
12.
Postepy Kardiol Interwencyjnej ; 16(2): 162-169, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32636900

ABSTRACT

INTRODUCTION: Periprocedural myocardial necrosis, which can range from a low level elevation of cardiac biomarkers to a large myocardial infarction (MI), is a common complication after percutaneous coronary intervention (PCI). AIM: We hypothesized that insulin-like growth factor-1 (IGF-1) levels may play a protective role in myocardial injury after coronary stent placement and aimed to investigate the relationship between IGF-1 levels and plaque characteristics assessed by optical coherence tomography (OCT). MATERIAL AND METHODS: Between May 2015 and December 2015 we prospectively enrolled 74 patients with stable angina pectoris in whom single de novo coronary artery stenosis was present. PCI was performed according to standard methods. OCT was applied to all patients. TnT was analyzed at admission, before PCI and at 6, 12, 24 and 48 h after PCI. Serum IGF-1 was measured prior to PCI. RESULTS: A total of 25 (33.7%) patients had periprocedural myocardial injury or type 4a myocardial infarction, and 49 (66.2%) patients had no events. IGF-1 level and reference intimal thickness, medial thickness, and plaque fibrous cap thickness in OCT had strong correlations (r = 0.88, 0.80 and 0.88 respectively, p < 0.001). IGF-1 was an independent predictor of periprocedural myocardial injury or type 4a MI in univariate (OR = 0.929, 95% CI: 0.895-0.964, p < 0.001) and multivariate regression analysis (OR = 0.757, 95% CI: 0.575-0.998, p = 0.04). Based on ROC analysis, the best cut-off value of IGF-1 for predicting periprocedural myocardial injury or type 4a myocardial infarction was 144.5 ng/ml, with a maximum sensitivity of 88% and specificity of 77.6% (AUC = 0.80, 95% CI: 0.69-0.88, p < 0.0001). CONCLUSIONS: The results from this study indicate that low IGF-1 levels are associated with plaque instability assessed by OCT. Low IGF-1 levels may identify patients who are at increased risk for periprocedural myocardial injury/infarction.

13.
Anticancer Agents Med Chem ; 20(14): 1728-1738, 2020.
Article in English | MEDLINE | ID: mdl-32357823

ABSTRACT

BACKGROUND: Poly (ADP-ribosyl) polymerase-1 (PARP-1) inhibitors are compounds that are used to treat cancers, which are defective in DNA-repair and DNA Damage-Response (DDR) pathways. OBJECTIVE: In this study, a series of potential PARP-1 inhibitor substituted (piperazine-1-carbonyl)phenyl)-1Hbenzo[ d]imidazole-4-carboxamide compounds were synthesised and tested for their PARP-1 inhibitory and anticancer activities. METHODS: Compounds were tested by cell-free colorimetric PARP-1 activity and MTT assay in MDA-MB-231, MDA-MB-436, MDA-MB-468 breast cancer, and L929 fibroblast cell lines. RESULTS: Our results showed that compound 6a inhibited viability in MDA-MB-231 and MDA-MB-468 cells whereas 8a inhibited viability in MDA-MB-468 cells. Compound 6b significantly inhibited cell viability in tested cancer cells. However, 6b exhibited toxicity in L929 cells, whereas 6a and 8a were found to be non-toxic for L929 cells. Compounds 6a, 6b and 8a exhibited significant inhibition of PARP-1 activity. CONCLUSION: These three compounds exhibited PARP-1 inhibitory activities and anticancer effects on breast cancer cells, and further research will enlighten the underlying mechanisms of their effects.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Molecular Docking Simulation , Molecular Structure , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Structure-Activity Relationship
14.
Gene ; 727: 144241, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31715301

ABSTRACT

Tumor suppressor protein p53, which functions in the cell cycle, apoptosis and neuronal differentiation via transcriptional regulations of target genes or interactions with several proteins, has been associated with neurite outgrowth through microtubule re-organization. We previously demonstrated in neurons that upon p53 induction, the level of microtubule severing protein Katanin-p60 increases, indicating that p53 might be a transcriptional regulator of the KATNA1 gene encoding Katanin-p60. In this context, we firstly elucidated the activity of KATNA1 regulatory regions and endogenous KATNA1 mRNA levels in the presence or absence of p53 using HCT 116 WT and HCT 116 p53 (-/-) cells. Next, we demonstrated the binding of p53 to the KATNA1 promoter and then investigated the role of p53 on KATNA1 gene expression by ascertaining KATNA1 mRNA and Katanin-p60 protein levels upon p53 overexpression and activation in both cells. Moreover, we showed changes in microtubule network upon increased Katanin-p60 level due to p53 overexpression. Also, the changes in KATNA1 mRNA and Katanin-p60 protein levels upon p53 knockdown were investigated. Our results indicate that p53 is an activator of KATNA1 gene expression and we show that both p53 and Katanin-p60 expression have strict regulations and are maintained at balanced levels as they are vital proteins to orchestrate either survival and apoptosis or differentiation.


Subject(s)
Katanin/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Gene Expression/genetics , Gene Expression Regulation/genetics , HCT116 Cells , Humans , Katanin/metabolism , Microtubules/metabolism , Neurons/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Transcription, Genetic/genetics
15.
Coron Artery Dis ; 30(4): 270-276, 2019 06.
Article in English | MEDLINE | ID: mdl-31026233

ABSTRACT

AIM: The aim of this study is to identify the predictors of angiographic no-reflow development in patients who underwent primary percutaneous coronary intervention and to investigate the long-term (median follow-up time=59 months) clinical endpoints. PATIENTS AND METHODS: We retrospectively evaluated 3205 patients (824 females, mean age: 58.6 years) with acute myocardial infarction (ST-segment elevation myocardial infarction) admitted within the first 12 h of chest pain and treated with primary percutaneous coronary intervention between January 2006 and January 2010. The patients were divided into angiographic no-reflow [final Thrombolysis In Myocardial Infarction (TIMI)<3 flow] (n=324) and reflow (final TIMI 3) (n=2881) groups. RESULTS: On multivariate logistic regression analysis age [odds ratio (OR)=1.02, 95% confidence interval (CI): 1.00-1.04, P=0.003], Killip class≥2 (OR=1.99, 95% CI: 1.30-3.04, P=0.002), pain-to-balloon time more than 4 h (OR=3.98, 95% CI: 2.50-6.32, P<0.001), baseline TIMI≤1 flow (OR=2.55, 95% CI: 1.05-6.22, P=0.038), lesion length of at least 15 mm (OR=4.31, 95% CI: 2.89-6.41, P<0.001), reference vessel diameter of at least 3.5 mm (OR=2.83, 95% CI: 1.87-4.27, P<0.001), cutoff occlusion pattern (OR=1.93, 95% CI: 1.03-3.62, P=0.04), and SYNTAX score of at least 19 (OR=1.76, 95% CI: 1.1.23-3.07, P<0.001)] were found as significant predictors for the development of no-reflow phenomenon. In no-reflow patients, in-hospital mortality (10.8 vs. 2.9%), heart failure (32.1 vs. 8.7%), and severe arrhythmias (23.1 vs. 9.3%) were significantly more common (P<0.001), for all. In the long-term follow-up, death (33.3 vs. 13.4%, P<0.001), advanced heart failure (12.5 vs. 5.4%, P<0.001), and stroke (3.5 vs. 1.7%, P=0.035) rates were significantly higher in the no-reflow group. CONCLUSION: The no-reflow predictors that were identified in this study might be useful in the determination of the patients who could benefit from aggressive pharmaco-invasive therapy. Development of no-reflow is associated with both in-hospital and long-term very high morbidity and mortality rates.


Subject(s)
No-Reflow Phenomenon/etiology , Percutaneous Coronary Intervention/adverse effects , ST Elevation Myocardial Infarction/therapy , Coronary Angiography , Coronary Circulation , Female , Humans , Male , Middle Aged , No-Reflow Phenomenon/diagnostic imaging , No-Reflow Phenomenon/mortality , No-Reflow Phenomenon/physiopathology , Percutaneous Coronary Intervention/mortality , Retrospective Studies , Risk Assessment , Risk Factors , ST Elevation Myocardial Infarction/diagnostic imaging , ST Elevation Myocardial Infarction/mortality , ST Elevation Myocardial Infarction/physiopathology , Time Factors , Treatment Outcome
16.
J Ultrasound Med ; 38(11): 2981-2988, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30927311

ABSTRACT

OBJECTIVES: We aimed to determine the effects of normal pregnancy on left atrial (LA) mechanics using 2-dimensional speckle-tracking echocardiography. METHODS: A total of 47 healthy women with singleton pregnancies were prospectively enrolled in this study. A total of 4 visits, including each trimester and postpartum at 6 months, were planned. Echocardiographic studies were performed with a Vivid 7 device equipped with a 2.5-MHz transducer (GE Vingmed Ultrasound AS, Horten, Norway). RESULTS: Although the LA reservoir phase strain showed a gradual decrease from the first trimester to the third trimester during pregnancy, the measurements in the postpartum period were found to return to initial levels (mean ± SD: first trimester, 40.3% ± 11.7%; second trimester, 37.5% ± 12.9%; third trimester, 33.5% ± 9.0%; postpartum, 42.1% ± 11.1%; P < .001). The LA pump function strain was also parallel to the LA reservoir strain and gradually decreased from the first trimester to the third trimester during pregnancy, and it was observed that rose to the initial level in the postpartum period (first trimester, 16.7% ± 7.4%; second trimester, 14.8% ± 5.5%; third trimester, 12.7% ± 4.3%; postpartum, 15.8% ± 5.5%; P < .001). CONCLUSIONS: We prospectively determined normal reference values for LA deformation parameters using speckle-tracking echocardiography in each trimester and the postpartum period in healthy pregnancy. These reference values may help identify subclinical LA dysfunction in several cardiovascular or systemic conditions. According to this study, these parameters decreased toward the third trimester during pregnancy and recovered in the postpartum period.


Subject(s)
Atrial Function, Left/physiology , Echocardiography/methods , Adult , Female , Follow-Up Studies , Heart Atria/diagnostic imaging , Humans , Postpartum Period , Pregnancy , Pregnancy Trimesters , Prospective Studies , Reference Values
17.
PLoS One ; 14(2): e0212518, 2019.
Article in English | MEDLINE | ID: mdl-30789974

ABSTRACT

Microtubule severing, which is highly critical for the survival of both mitotic and post-mitotic cells, has to be precisely adjusted by regulating the expression levels of severing proteins, katanin and spastin. Even though severing mechanism is relatively well-studied, there are limited studies for the transcriptional regulation of microtubule severing proteins. In this study, we identified the main regulatory region of KATNA1 gene encoding katanin-p60 as 5' UTR, which has a key role for its expression, and showed Elk1 binding to KATNA1. Furthermore, we identified that Elk1 decreased katanin-p60 and spastin protein expressions, while mRNA levels were increased upon Elk1 overexpression. In addition, SUMOylation is a known post-translational modification regulating Elk1 activity. A previous study suggested that K230, K249, K254 amino acids in the R domain are the main SUMOylation sites; however, we identified that these amino acids are neither essential nor substantial for Elk1 SUMOylation. Also, we determined that KATNA1 methylation results in the reduction of Elk1 binding whereas SPG4 methylation does not. Together, our findings emphasizing the impacts of both transcriptional and post-transcriptional regulations of katanin-p60 and spastin suggest that Elk1 has a key role for differential expression patterns of microtubule severing proteins, thereby regulating cellular functions through alterations of microtubule organization.


Subject(s)
Katanin/metabolism , Spastin/metabolism , ets-Domain Protein Elk-1/metabolism , 5' Untranslated Regions , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Cell Line , DNA Methylation , Humans , Katanin/chemistry , Katanin/genetics , Microtubules/genetics , Microtubules/metabolism , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spastin/chemistry , Spastin/genetics , Sumoylation , Transcription, Genetic , ets-Domain Protein Elk-1/chemistry , ets-Domain Protein Elk-1/genetics
18.
Hematology ; 23(10): 765-770, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29945498

ABSTRACT

OBJECTIVE: Chronic myleoid leukemia (CML) is a myeloproliferative disorder characterized with the constitutive activation of Bcr-Abl tyrosine kinase which is a target for imatinib, the first line treatment option for CML. Constitutive activation of NFκB and ß-catenin signaling promotes cellular proliferation and survival and resistance to Imatinib therapy in CML. Akirin-2 is a nuclear protein which is required for NFκB dependent gene expression as a cofactor and has been linked to Wnt/beta-catenin pathway. The purpose of this study is to examine Akirin-2, NFκB and ß-catenin in imatinib resistance of CML and to test if any direct physical protein-protein interaction exists between NFkB and both ß-catenin and Akirin-2. METHODS: RT-PCR and western blot were performed to determine Akirin-2, NFκB-p65 and ß-catenin gene and protein expressions, Co-immunoprecipitation and chromatin immunoprecipitation analysis were carried out to detect the direct physical interactions and binding of NFκB-p65 and ß-catenin proteins to MDR1 promoter region, respectively. RESULTS: ß-catenin and NFκB-p65 proteins bound to DNA promoter regions of MDR1 in imatinib-sensitive and resistant CML cells, whereas any direct protein-protein interaction could not be found between NFκB-p65 and Akirin-2 or ß-catenin proteins. Nuclear ß-catenin and NFκB-p65 levels increased in imatinib resistance. Moreover, increased Akirin-2 protein accumulation in the nucleus was shown for the first time in imatinib resistant CML cells. DISCUSSION: We show for the first time that Akirin-2 can be a novel biomarker in imatinib resistance. Targeting Akirin-2, NFκB and ß-catenin genes may provide an opportunity to overcome imatinib resistance in CML.


Subject(s)
Biomarkers, Tumor , DNA-Binding Proteins , Drug Resistance, Neoplasm , Gene Expression Regulation, Leukemic/drug effects , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Neoplasm Proteins , Transcription Factor RelA , Transcription Factors , beta Catenin , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression Profiling , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Transcription Factor RelA/biosynthesis , Transcription Factor RelA/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , beta Catenin/biosynthesis , beta Catenin/genetics
19.
J Chem Neuroanat ; 92: 41-47, 2018 10.
Article in English | MEDLINE | ID: mdl-29860071

ABSTRACT

Hyperphosphorylation of tau leading to neurofibrillary tangles (NFT) is one of the key pathological hallmarks in neurodegenerative disorders such as Alzheimer disease (AD). Peptidyl-prolyl cis-trans isomerase (Pin1) regulates the phosphorylation of Ser/Thr sites of tau protein, and promotes microtubule assembly. In this study, we aimed to determine the effect of tau hyperphosphorylation on Pin1 expression in primary cortical neurons in order to investigate the results of the pathological process on Pin1, an important enzyme involved in various cellular mechanisms. Primary cortical neurons were prepared from embryonic day 16 -Sprague Dawley rat embryos. The cultures were treated with 25 nM okadaic acid (OKA) on day 7 in order to promote tau hyperphosphorylation. The cytotoxicity was determined with LDH release and measured by ELISA. Tau phosphorylation was confirmed by western blot using anti-tau antibodies Thr231 and Tau-1. Pin1 mRNA expression level was determined by qRT-PCR at 8 and 24 h. Pin1 protein expression was analyzed with immunofluorescent labeling at 8 and 24 h. Tau phosphorylation on Thr231 was increased and non-phosphorylated Tau-1 was decreased in OKA treated group compared with the untreated control at 8 h of treatment. While Pin1 mRNA expression levels at 8 h post-OKA treatment were lower than that of control groups, there were no differences between OKA-treated group and control groups in Pin1 protein expression. Whereas no significant differences for Pin1 mRNA expression, protein expression levels were decreased OKA-treated group compared to control groups at 24 h of treatment. The LDH release of OKA-treated group was significantly increased at 24 h. Our study indicates that although OKA treatment suppressed Pin1 mRNA expression and induced tau phosphorylation at 8 h of treatment, its influence on Pin1 protein expression has 16 h phase delay. Given the important role of Pin1 in many cellular mechanisms these results might indicate that tau hyperphosphorylation involved in many neurodegenerative disorders may cause some alterations in brain microenvironment via Pin1.This is the first demonstration of the alteration of the Pin1 mRNA and protein expression in OKA induced model in primary cortical neurons.


Subject(s)
Cerebral Cortex/drug effects , Down-Regulation/drug effects , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neurons/drug effects , Okadaic Acid/pharmacology , tau Proteins/metabolism , Animals , Cerebral Cortex/metabolism , Neurons/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
20.
Turk J Med Sci ; 48(3): 620-627, 2018 Jun 14.
Article in English | MEDLINE | ID: mdl-29916222

ABSTRACT

Background/aim: MODY3 associated with HNF1A is the most common form of MODY and is clinically misdiagnosed as type 1 diabetes due to similar clinical symptoms. This study aimed to analyze the role of HNF1A-regulated miRNAs as a biomarker in the diagnosis of MODY3. Materials and methods: MIN6 cells were transfected with the HNF1A cDNA expression vector for overexpression or with siRNA specific to HNF1A to silence its expression. The HNF1A-regulated miRNAs were determined by RNA-Seq of the total RNA extract. Expressions of the candidate miRNAs in blood samples of MODY3, type 1 diabetes, and type 2 diabetes patients and in healthy subjects were compared statistically by Mann-Whitney U tests. Results: This study revealed the presence of 238 known HNF1A-regulated miRNAs in MIN6 cells. miR-129-1-3p, miR-200b-3p, and miR-378a-5p were selected as candidate miRNAs. The expression level of miR-378a-5p significantly decreased in type 2 diabetes and MODY3 patients, while miR-200b-3p expression was significantly decreased only in MODY3 patients. Conclusion: Although further studies with larger numbers of patients are required, this study demonstrated that the expression levels of miR-200b-3p and miR-378a-5p decrease in MODY3 patients and suggests that miR-200b-3p is an especially strong candidate to use clinically for selecting suspected MODY3 patients.

SELECTION OF CITATIONS
SEARCH DETAIL
...