Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(5): 159496, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38649008

ABSTRACT

This work aims to understand better the mechanism of cellular processes accompanying the activation of human T cells and to develop a novel, fast, label-free approach to identify molecular biomarkers for this process. The standard methodology for confirming the activation state of T cells is based on flow cytometry and using antibodies recognizing activation markers. The method provide high specificity detection but may be susceptible to background staining or non-specific secondary antibody reactions. Here, we evaluated the potential of Raman-based molecular imaging in distinguishing non-activated and activated human T cells. Confocal Raman microscopy was performed on T cells followed by chemometrics to obtain comprehensive molecular information, while Stimulated Raman Scattering imaging was used to quickly provide high-resolution images of selected cellular components of activated and non-activated cells. For the first time, carotenoids, lipids, and proteins were shown to be important biomarkers of T-cell activation. We found that T-cell activation was accompanied by lipid accumulation and loss of carotenoid content. Our findings on the biochemical, morphological, and structural changes associated with activated mature T cells provide insights into the molecular changes that occur during therapeutic manipulation of the immune response. The methodology for identifying activated T cells is based on a novel imaging method and supervised and unsupervised chemometrics. It unambiguously identifies specific and unique molecular changes without the need for staining, fixation, or any other sample preparation.


Subject(s)
Biomarkers , Carotenoids , Lipid Metabolism , Lymphocyte Activation , Spectrum Analysis, Raman , T-Lymphocytes , Humans , Carotenoids/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Spectrum Analysis, Raman/methods , Biomarkers/metabolism , Proteins/metabolism
2.
Int J Mol Sci ; 25(6)2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38542349

ABSTRACT

Raman spectroscopy is a molecular spectroscopic technique able to provide detailed information about the chemical structure, phase, crystallinity, and molecular interactions of virtually any analyzed sample. Although its medical applications have been studied for several decades, only recent advances in microscopy, lasers, detectors, and better understanding of the principles of the Raman effect have successfully expanded its applicability to clinical settings. The promise of a rapid, label-free diagnostic method able to evaluate the metabolic status of a cell in vivo makes Raman spectroscopy particularly attractive for hematology and oncology. Here, we review widely studied hematological applications of Raman spectroscopy such as leukocyte activation status, evaluation of treatment response, and differentiation between cancer and non-malignant cells, as well as its use in still unexplored areas in hematology. We also discuss limitations and challenges faced by Raman spectroscopy-based diagnostics as well as recent advances and modifications of the method aimed to increase its applicability to clinical hematooncology.


Subject(s)
Hematology , Neoplasms , Humans , Spectrum Analysis, Raman/methods , Neoplasms/diagnosis , Neoplasms/pathology , Antigen Presentation , Head
3.
Spectrochim Acta A Mol Biomol Spectrosc ; 309: 123795, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38184880

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL), the most common non-Hodgkin's lymphoma in adults, is a genetically and metabolically heterogeneous group of aggressive malignancies. The complexity of their molecular composition and the variability in clinical presentation make clinical diagnosis and treatment selection a serious challenge. The challenge is therefore to quickly and correctly classify DLBCL cells. In this work, we show that Raman imaging is a tool with high diagnostic potential, providing unique information about the biochemical components of tumor cells and their metabolism. We present models of classification of lymphoma cells based on their Raman spectra. The models automatically and efficiently identify DLBCL cells and assign them to a given cell-of-origin (COO) subtype (activated B cell-like (ABC) or germinal center B cell-like (GCB)) or, respectively, to a comprehensive cluster classification (CCC) subtype (OxPhos/non-OxPhos). In addition, we describe each lymphoma subtype by its unique spectral profile, linking it to biochemical, genetic, or metabolic features.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Adult , Humans , Lymphoma, Large B-Cell, Diffuse/diagnosis , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Germinal Center/pathology
4.
Molecules ; 27(24)2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36557944

ABSTRACT

Endophytic fungi are a diverse group of microorganisms that colonize the inter- or intracellular spaces of plants and exhibit mutual benefits. Their interactions with the host plant and other microbiomes are multidimensional and play a crucial role in the production of secondary metabolites. We screened bioactive compounds present in the extracts of Aspergillus flavus, an endophytic fungus isolated from the roots of the medicinal grass Cynodon dactylon, for its anticancer potential. An in vitro analysis of the Ethyl acetate extract from A. flavus showed significant cytostatic effects (IC50: 16.25 µg/mL) against breast cancer cells (MCF-7). A morphological analysis of the cells and a flow cytometry of the cells with annexin V/Propidium Iodide suggested that the extract induced apoptosis in the MCF-7 cells. The extract of A. flavus increased reactive oxygen species (ROS) generation and caused a loss of mitochondrial membrane potential in MCF-7 cells. To identify the metabolites that might be responsible for the anticancer effect, the extract was subjected to a gas chromatography-mass spectrometry (GC-MS) analysis. Interestingly, nine phytochemicals that induced cytotoxicity in the breast cancer cell line were found in the extract. The in silico molecular docking and molecular dynamics simulation studies revealed that two compounds, 2,4,7-trinitrofluorenone and 3α, 5 α-cyclo-ergosta-7,9(11), 22t-triene-6beta-ol exhibited significant binding affinities (-9.20, and -9.50 Kcal/mol, respectively) against Bcl-2, along with binding stability and intermolecular interactions of its ligand-Bcl-2 complexes. Overall, the study found that the endophytic A. flavus from C. dactylon contains plant-like bioactive compounds that have a promising effect in breast cancer.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Humans , Female , Aspergillus flavus/metabolism , Cynodon/metabolism , Breast Neoplasms/drug therapy , Molecular Docking Simulation , Fungi/chemistry , Antineoplastic Agents/chemistry
5.
Biomolecules ; 12(7)2022 06 25.
Article in English | MEDLINE | ID: mdl-35883443

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that can cause acute and severe infections. Increasing resistance to antibiotics has given rise to the urgent need for an alternative antimicrobial agent. A promising strategy is the inhibition of iron sequestration in the bacteria. The current work aimed to screen for inhibitors of pyoverdine-mediated iron sequestration in P. aeruginosa. As a drug target, we choose l-ornithine-N5-monooxygenase (PvdA), an enzyme involved in the biosynthesis of pyoverdine that catalyzes the FAD-dependent hydroxylation of the side chain amine of ornithine. As drug repurposing is a fast and cost-efficient way of discovering new applications for known drugs, the approach may help to solve emerging clinical problems. In this study, we use data about molecules from drug banks for screening. A total of 15 drugs that are similar in structure to l-ornithine, the substrate of PvdA, and 30 drugs that are sub-structures of l-ornithine were virtually docked against PvdA. N-2-succinyl ornithine and cilazapril were found to be the top binders with a binding energy of -12.8 and -9.1 kcal mol-1, respectively. As the drug-likeness and ADME properties of the drugs were also found to be promising, molecular dynamics studies were performed to further confirm the stability of the complexes. The results of this in silico study indicate that N-2-succinyl ornithine could potentially be explored as a drug for the treatment of P. aeruginosa infections.


Subject(s)
Mixed Function Oxygenases , Pseudomonas Infections , Drug Repositioning , Humans , Iron/metabolism , Mixed Function Oxygenases/metabolism , Ornithine/metabolism , Pseudomonas aeruginosa/metabolism
6.
Int J Mol Sci ; 22(2)2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33467555

ABSTRACT

Acute leukemias, the most common cancers in children, are characterized by excessive proliferation of malignant progenitor cells. As a consequence of impaired blood cell production, leukemia patients are susceptible to infectious complications-a major cause of non-relapse mortality. Neutrophil extracellular traps (NETs) are involved in various pathologies, from autoimmunity to cancer. Although aberrant NETs formation may be partially responsible for immune defects observed in acute leukemia, still little is known on the NET release in the course of leukemia. Here, we present the first comprehensive evaluation of NETs formation by neutrophils isolated from children with acute leukemia in different stages of the disease and treatment stimulated in vitro with phorbol 12-myristate 13-acetate (PMA), N-formyl-methionyl-leucyl-phenylalanine (fMLP), and calcium ionophore (CI). NETs release was measured using quantitative fluorescent method and visualized microscopically. In this setting, NETs release was significantly impaired in leukemic children both at the diagnosis and during the treatment, and full restoration of neutrophil function was achieved only after successful completion of the leukemia treatment. We suggest that neutrophil function impairment may result from both disease- and treatment-related factors. In this context, deficient innate immune response observed in acute leukemia patients may be present regardless of neutrophil count and contribute to secondary immunodeficiency observed in this population.


Subject(s)
Extracellular Traps/immunology , Immunity, Innate/immunology , Leukemia/immunology , Neutrophils/immunology , Acute Disease , Adolescent , Calcium Ionophores/pharmacology , Cells, Cultured , Child , Child, Preschool , Humans , Immunity, Innate/drug effects , Infant , Leukemia/blood , Leukemia/drug therapy , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Tetradecanoylphorbol Acetate/pharmacology
7.
Eur J Pharm Sci ; 154: 105512, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32801003

ABSTRACT

Cancer is a heterogeneous disease, and its treatment requires the identification of new ways to thwart tumor cells. Amongst such emerging targets are glucose transporters (GLUTs, SLC2 family), which are overexpressed by almost all types of cancer cells; their inhibition provides a strategy to disrupt tumor metabolism selectively, leading to antitumor effects. Here, novel thiazolidinedione (TZD) derivatives were designed, synthesized, characterized, and evaluated for their GLUT1, GLUT4, and GLUT5 inhibitory potential, followed by in-vitro cytotoxicity determination in leukemic cell lines. Compounds G5, G16, and G17 inhibited GLUT1, with IC50 values of 5.4 ± 1.3, 26.6 ± 1.8, and 12.6 ± 1.2 µM, respectively. G17 was specific for GLUT1, G16 inhibited GLUT4 (IC50 = 21.6 ± 4.5 µM) comparably but did not affect GLUT5. The most active compound, G5, inhibited all three GLUT types, with GLUT4 IC50 = 9.5 ± 2.8 µM, and GLUT5 IC50 = 34.5 ± 2.4 µM. Docking G5, G16, and G17 to the inward- and outward-facing structural models of GLUT1 predicted ligand binding affinities consistent with the kinetic inhibition data and implicated E380 and W388 of GLUT1 vs. their substitutions in GLUT5 (A388 and A396, respectively) in inhibitor preference for GLUT1. G5 inhibited the proliferation of leukemia CEM cells at low micromolar range (IC50 = 13.4 µM) while being safer for normal blood cells. Investigation of CEM cell cycle progression after treatment with G5 showed that cells accumulated in the G2/M phase. Flow cytometric apoptosis studies revealed that compound G5 induced both early and late-stage apoptosis in CEM cells.


Subject(s)
Glucose Transport Proteins, Facilitative , Thiazolidinediones , Apoptosis , Cell Line , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Glucose Transporter Type 1/antagonists & inhibitors , Hypoglycemic Agents , Thiazolidinediones/pharmacology
8.
Eur J Med Chem ; 202: 112603, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32634629

ABSTRACT

Cancer cells increase their glucose uptake and glycolytic activity to meet the high energy requirements of proliferation. Glucose transporters (GLUTs), which facilitate the transport of glucose and related hexoses across the cell membrane, play a vital role in tumor cell survival and are overexpressed in various cancers. GLUT1, the most overexpressed GLUT in many cancers, is emerging as a promising anti-cancer target. To develop GLUT1 inhibitors, we rationally designed, synthesized, structurally characterized, and biologically evaluated in-vitro and in-vivo a novel series of furyl-2-methylene thiazolidinediones (TZDs). Among 25 TZDs tested, F18 and F19 inhibited GLUT1 most potently (IC50 11.4 and 14.7 µM, respectively). F18 was equally selective for GLUT4 (IC50 6.8 µM), while F19 was specific for GLUT1 (IC50 152 µM in GLUT4). In-silico ligand docking studies showed that F18 interacted with conserved residues in GLUT1 and GLUT4, while F19 had slightly different interactions with the transporters. In in-vitro antiproliferative screening of leukemic/lymphoid cells, F18 was most lethal to CEM cells (CC50 of 1.7 µM). Flow cytometry analysis indicated that F18 arrested cell cycle growth in the subG0-G1 phase and lead to cell death due to necrosis and apoptosis. Western blot analysis exhibited alterations in cell signaling proteins, consistent with cell growth arrest and death. In-vivo xenograft study in a CEM model showed that F18 impaired tumor growth significantly.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 4/antagonists & inhibitors , Thiazolidinediones/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/metabolism , Humans , Mice , Mice, SCID , Models, Molecular , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/chemistry
9.
Bioorg Chem ; 100: 103934, 2020 07.
Article in English | MEDLINE | ID: mdl-32446120

ABSTRACT

Epigenetics plays a fundamental role in cancer progression, and developing agents that regulate epigenetics is crucial for cancer management. Among Class I and Class II HDACs, HDAC8 is one of the essential epigenetic players in cancer progression. Therefore, we designed, synthesized, purified, and structurally characterized novel compounds containing N-substituted TZD (P1-P25). Cell viability assay of all compounds on leukemic cell lines (CEM, K562, and KCL22) showed the cytotoxic potential of P8, P9, P10, P12, P19, and P25. In-vitro screening of different HDACs isoforms revealed that P19 was the most potent and selective inhibitor for HDAC8 (IC50 - 9.3 µM). Thermal shift analysis (TSA) confirmed the binding of P19 to HDAC8. In-vitro screening of all compounds on the transport activity of GLUT1, GLUT4, and GLUT5 indicated that P19 inhibited GLUT1 (IC50 - 28.2 µM). P10 and P19 induced apoptotic cell death in CEM cells (55.19% and 60.97% respectively) and P19 was less cytotoxic on normal WBCs (CC50 - 104.2 µM) and human fibroblasts (HS27) (CC50 - 105.0 µM). Thus, among this novel series of TZD derivatives, compound P19 was most promising HDAC8 inhibitor and cytotoxic on leukemic cells. Thus, P19 could serve as a lead for further development of optimized molecules with enhanced selectivity and potency.


Subject(s)
Histone Deacetylase Inhibitors/metabolism , Repressor Proteins/antagonists & inhibitors , Thiazolidinediones/chemistry , Apoptosis/drug effects , Binding Sites , Cell Line , Cell Survival/drug effects , Drug Evaluation, Preclinical , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Molecular Docking Simulation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Repressor Proteins/metabolism , Structure-Activity Relationship , Thiazolidinediones/metabolism , Thiazolidinediones/pharmacology
10.
Mater Sci Eng C Mater Biol Appl ; 110: 110675, 2020 May.
Article in English | MEDLINE | ID: mdl-32204102

ABSTRACT

Integration of multiple features including shape memory, biodegradation, and sustained drug delivery in a single material offers the opportunity to significantly improve the abilities of implantable devices for cardiovascular system regeneration. Two types of shape memory polyurethanes (SMPUs): PU-PLGA and PU-PLLA/PEG differing in soft segments composition that comprising blends of various biodegradable polyols, i.e. D,l-lactide-co-glycolide diol (o-PLGA), poly(e-caprolactone) diols (o-PCL) with various molecular weights, poly-l-lactide diol (o-PLLA), polyethylene glycol (o-PEG) were synthesized and further utilized to electrospun nanofibrous - rapamycin (Rap) delivery system. Structure characterization by Fourier transform infrared spectroscopy (FTIR), differential scanning calorimetry (DCS) and hydrophilicity measurements were performed to gain more insights on the influence of the particular units of the softs segments on the transition temperature (Ttrans), shape recovery, degradation profile, and drug release kinetics. In vitro study in PBS solution revealed that incorporation of o-PLGA segments to SMPUs is favorable over o-PEG as increased shape memory performance was observed. Moreover, presence of PLGA in PU-PLGA gave more predictable degradation profile in comparison to PU-PLLA/PEG system. Human Cardiac Fibroblasts (HCF) viability tests in vitro confirmed that the amount of Rap released from evaluated PU-PLLA/PEG/Rap and PU-PLGA/Rap drug delivery systems was sufficient to inhibit cells growth on the surface of the tested materials.


Subject(s)
Drug Delivery Systems , Polyurethanes/chemistry , Smart Materials/chemistry , Tissue Engineering , Biocompatible Materials/chemistry , Calorimetry, Differential Scanning , Cell Survival , Crystallization , Drug Liberation , Fibroblasts/cytology , Humans , Kinetics , Lactates , Molecular Weight , Polyesters , Polyethylene Glycols , Spectroscopy, Fourier Transform Infrared , Temperature , Tensile Strength
11.
Bioorg Chem ; 95: 103522, 2020 01.
Article in English | MEDLINE | ID: mdl-31901516

ABSTRACT

Histone deacetylases (HDACs) are being explored as a therapeutic target for interventions in different types of cancer. HDAC8 is a class I HDAC that is implicated as a therapeutic target in various indication areas, including different types of cancer and particularly childhood neuroblastoma. Most previously described HDAC8-selective inhibitors contain a hydroxamate function as zinc binding group (ZBG) to confer potency. However, hydroxamate class HDAC inhibitors have raised increasing concerns about their mutagenic character. Therefore, non-hydroxamate based inhibitors could prove to be safer than hydroxamates. In the present work, a series of novel 5-naphthylidene-2,4-thiazolidinedione was designed and evaluated as potential antiproliferative agents targeting selectively HDAC8 enzyme. Eleven novel derivatives were synthesized, purified and characterized by spectroscopic techniques. Compounds 3k and 3h was found to be most potent selective inhibitors of HDAC8 with IC50 values of 2.7 µM and 6.3 µM respectively. 3a to 3i was found to be most cytotoxic in leukemic cell lines. 3a and 3 h both were found to induce apoptosis and cause cell cycle arrest in G2/M phase.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Repressor Proteins/antagonists & inhibitors , Thiazolidinediones/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Histone Deacetylases , Humans , Molecular Docking Simulation , Structure-Activity Relationship , Thiazolidinediones/chemistry
12.
Curr Cancer Drug Targets ; 20(3): 197-215, 2020.
Article in English | MEDLINE | ID: mdl-31814555

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPARγ) is a nuclear receptor acting as a transcription factor involved in the regulation of energy metabolism, cell cycle, cell differentiation, and apoptosis. These unique properties constitute a strong therapeutic potential that place PPARγ agonists as one of the most interesting and widely studied anticancer molecules. Although PPARγ agonists exert significant, antiproliferative and tumoricidal activity in vitro, their anticancer efficacy in animal models is ambiguous, and their effectiveness in clinical trials in monotherapy is unsatisfactory. However, due to pleiotropic effects of PPARγ activation in normal and tumor cells, PPARγ ligands interact with many antitumor treatment modalities and synergistically potentiate their effectiveness. The most spectacular example is a combination of PPARγ ligands with tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia (CML). In this setting, PPARγ activation sensitizes leukemic stem cells, resistant to any previous form of treatment, to targeted therapy. Thus, this combination is believed to be the first pharmacological therapy able to cure CML patients. Within the last decade, a significant body of data confirming the benefits of the addition of PPARγ ligands to various antitumor therapies, including chemotherapy, hormonotherapy, targeted therapy, and immunotherapy, has been published. Although the majority of these studies have been carried out in vitro or animal tumor models, a few successful attempts to introduce PPARγ ligands into anticancer therapy in humans have been recently made. In this review, we aim to summarize shines and shadows of targeting PPARγ in antitumor therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/drug effects , PPAR gamma/agonists , Animals , Drug Therapy, Combination , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
13.
Folia Histochem Cytobiol ; 52(4): 297-307, 2014.
Article in English | MEDLINE | ID: mdl-25401764

ABSTRACT

INTRODUCTION: The formation and maintenance of tissues is regulated by various signals triggered by biological, chemical, and physical factors. Data increasingly confirm that matrix or tissue elasticity plays an influential role in regulating numerous cell functions. The aim of the present study was to better understand the regulation of cellular differentiation by mechanical cues. We studied the influence of matrix stiffness on the osteodifferentiation of two cell lineages characterized by different responses: mesenchymal stromal/stem cells isolated from the Wharton's jelly of the umbilical cord (UC-MSCs) with strong stiffness-dependent responses; and bone-derived cells (BDCs), which are insensitive to changes in matrix rigidity. The study also aimed to delineate how matrix stiffness affects intracellular signaling through focal adhesion kinase (FAK) activity­one of the key components in integrin-mediated signaling pathways. MATERIAL AND METHODS: The effect of substrate stiffness on the expression of α2, α5, and ß1 integrin was studied using real time PCR and Western blot using cells cultured in an osteogenic medium on tunable polyacrylamide gels coated with type I collagen, with elasticities corresponding to Young's moduli of 1.46 kPa and 26.12 kPa. FAK activity was monitored using ELISA assays. RESULTS: We demonstrate for the first time the changes in the expression of α2, α5, and ß1 integrin subunits in perinatal stem cells and in adult osteoblast precursor cells during in vitro osteogenic differentiation on surfaces characterized by different stiffness. We found that matrix rigidity significantly affects the osteogenic differentiation of UC-MSCs through α2 integrin-mediated mechanotransduction events, though not through the α5 integrin subunit. In BDCs, there were no significant changes in the expression levels of the tested protein associated with varying stiffness. CONCLUSIONS: Our results provide evidence that matrix rigidity affects the osteogenic differentiation of UC-MSCs via mechanotransduction events mediated by α2 integrin subunits.


Subject(s)
Gene Expression Regulation, Developmental , Integrin alpha2beta1/metabolism , Osteogenesis/physiology , Signal Transduction , Stem Cells/cytology , Umbilical Cord/cytology , Wharton Jelly/cytology , Cell Differentiation , Cell Survival , Humans , Integrin alpha2beta1/genetics , Polymerase Chain Reaction
14.
Exp Hematol ; 42(6): 439-47, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24667683

ABSTRACT

Despite undisputed success of tyrosine kinase inhibitors in the therapy of chronic myeloid leukemia (CML), development of drug resistance and inability to cure the disease challenge clinicians and researchers. Additionally, recent reports regarding cardiovascular toxicities of second and third generation tyrosine kinase inhibitors prove that there is still a place for novel therapeutic combinations in CML. We have previously shown that statins are able to modulate activity of chemotherapeutics or antibodies used in oncology. Therefore, we decided to verify that statins are able to potentiate antileukemic activity of imatinib, still a frontline treatment of CML. Lovastatin, a cholesterol lowering drug, synergistically potentiates antileukemic activity of imatinib in cell lines and in primary CD34+ CML cells from patients in different phases of the disease, including patients resistant to imatinib with no detectable mutations. This effect is related to increased intracellular concentration of imatinib in CD34+ CML cells and cell lines measured using uptake of (14)C-labeled imatinib. Lovastatin does not influence influx but significantly inhibits efflux of imatinib mediated by ATP-binding cassette (ABC) transporters: ABCB1 and ABCG2. The addition of cholesterol completely reverses these effects. Statins do not affect expression of ABCB1 and ABCG2 genes. The effects are drug-class specific, as observed with other statins. Our results suggest that statins may offer a valuable addition to imatinib in a select group of CML patients.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP-Binding Cassette Transporters/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Leukemia, Myeloid, Chronic-Phase/physiopathology , Neoplasm Proteins/antagonists & inhibitors , Piperazines/pharmacology , Pyrimidines/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Antineoplastic Agents/therapeutic use , Blotting, Western , Cell Line , Cell Survival/drug effects , Drug Synergism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Imatinib Mesylate , K562 Cells , Leukemia, Myeloid, Chronic-Phase/drug therapy , Leukemia, Myeloid, Chronic-Phase/metabolism , Lovastatin/pharmacology , Mice
16.
Cell Biol Int ; 37(6): 608-16, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23447501

ABSTRACT

There is a profound dependence of cell behaviour on the stiffness of its microenvironment. To gain a better understanding of the regulation of cellular differentiation by mechanical cues, we investigated the influence of matrix stiffness (E = 1.46 kPa and E = 26.12 kPa) on differentiated osteogenic cell lineage of bone marrow stem cells (BM-MSCs) and bone-derived cells (BDCs) using flexible collagen-coated polyacrylamide substrates. Differentiation potential was determined by measuring alkaline phosphatase activity, expression of osteoblast-specific markers including alkaline phosphatase, osteocalcin, Runx2 and collagen type I, as well as assessment of mineralisation (Alizarin Red S staining). We found that osteogenic differentiation can be regulated by the rigidity of the substrate, which may depend on the commitment in multi- or uni-potent targeting cells. Osteogenic differentiation of BM-MSCs was enhanced on a stiff substrate compared to a soft one, whereas BDCs osteogenic differentiation did not vary depending on the substrate stiffness. The data help in understanding the role of the external mechanical determinants in stem cell differentiation, and can also be useful in translational approach in functional tissue engineering.


Subject(s)
Acrylic Resins/chemistry , Bone Marrow Cells/cytology , Bone and Bones/cytology , Cell Differentiation , Bone Marrow Cells/metabolism , Bone and Bones/metabolism , Cell Lineage , Gels , Humans , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Phenotype , Tissue Engineering
17.
Przegl Lek ; 69(3): 115-9, 2012.
Article in Polish | MEDLINE | ID: mdl-22764653

ABSTRACT

Although tissues may exist regardless of reduced oxygen pressure, this requires glycolytic ATP generation, which is very expensive from the energetic viewpoint. Hypoxia is defined as the condition in which oxygen pressure is reduced at the level of bodily tissues. There are many clinical situations during which decreased tissue oxygenation may occur. It may be transient or chronic, as well as systemic or local. An emergent need exists for monitoring and diagnosis with respect to numerous possible clinical circumstances leading to hypoxia and its life-threatening consequences. The assessment of global oxygen homeo-stasis relies on blood gas analysis and lactate concentration, but such an approach does not fully reflect the local oxygenation of tissues. Oxygen needle microelectrode measurements reveal great differences in tissue pO2 levels. Local pO2 levels depend on many factors, among which the most important are: the distance to the nearest capillary, the extracellular and intracellular fluid diffusion rates and intracellular measurements of the number and activity levels of mitochondria. Thus, nowadays, it is impossible to establish an accurate normal value ranges for local tissue pO2. Oxygen deficiency is an important gene regulator. A sequence-specific DNA-binding factor, the hypoxia induced factor (HIF), is the fundamental hypoxia response protein. 70 genes identified so far have been found to be HIF-dependent. They are responsible for increased oxygen delivery, i.e. by boosting angiogensis due to vascular endothelial growth factor (VEGF) release and the enhancement of red blood cell production by erythropoietin (EPO). VEGF-induced angiogenesis is one of several key hypoxia adaptations. An enhanced vascular bed in response to hypoxia affects almost every bodily tissue and organ. This was observed particularly in skeletal muscles as well as in the brain. The expression of a few hypoxia markers does not require HIF activation. An especially interesting member of this group is osteopontin (OPN), whose synthesis increases during hypoxia. OPN was originally linked to bone remodeling, but currently it seems to posses an important role in immunity, inflammation and tumor pathogenesis. Quantification of hypoxia is clinically essential both for therapy and prognosis. Taking account of the fact that the concept of oxygen pressure at the tissue level is not quantitative (norms do not exist, results are incomparable), biochemical markers are preferable. Particularly significant in this context are hypoxia-induced proteins such as HIF, EPO, VEGF or potentially OPN.


Subject(s)
Hypoxia/diagnosis , Hypoxia/metabolism , Adaptation, Physiological , Biomarkers/metabolism , Cell Hypoxia/physiology , Erythropoietin/metabolism , Humans , Hypoxia-Inducible Factor 1/metabolism , Osteopontin/metabolism , Oxygen/metabolism , Vascular Endothelial Growth Factor A/metabolism
18.
Cancer Res ; 69(10): 4235-43, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19435917

ABSTRACT

Photodynamic therapy (PDT) is an approved therapeutic procedure that exerts cytotoxic activity toward tumor cells by inducing production of reactive oxygen species such as singlet oxygen. PDT leads to oxidative damage of cellular macromolecules, including proteins that undergo multiple modifications such as fragmentation, cross-linking, and carbonylation that result in protein unfolding and aggregation. Because the major mechanism for elimination of carbonylated proteins is their degradation by proteasomes, we hypothesized that a combination of PDT with proteasome inhibitors might lead to accumulation of carbonylated proteins in endoplasmic reticulum (ER), aggravated ER stress, and potentiated cytotoxicity toward tumor cells. We observed that Photofrin-mediated PDT leads to robust carbonylation of cellular proteins and induction of unfolded protein response. Pretreatment of tumor cells with three different proteasome inhibitors, including bortezomib, MG132, and PSI, gave increased accumulation of carbonylated and ubiquitinated proteins in PDT-treated cells. Proteasome inhibitors effectively sensitized tumor cells of murine (EMT6 and C-26) as well as human (HeLa) origin to PDT-mediated cytotoxicity. Significant retardation of tumor growth with 60% to 100% complete responses was observed in vivo in two different murine tumor models (EMT6 and C-26) when PDT was combined with either bortezomib or PSI. Altogether, these observations indicate that combination of PDT with proteasome inhibitors leads to potentiated antitumor effects. The results of these studies are of immediate clinical application because bortezomib is a clinically approved drug that undergoes extensive clinical evaluations for the treatment of solid tumors.


Subject(s)
Dihematoporphyrin Ether/therapeutic use , Endoplasmic Reticulum/physiology , Photochemotherapy/methods , Proteasome Inhibitors , Protein Denaturation/drug effects , Adenocarcinoma , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colonic Neoplasms , HeLa Cells/drug effects , Humans , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/pathology , Porphyrins/therapeutic use , Reactive Oxygen Species/metabolism , Singlet Oxygen/metabolism , Ubiquitin/drug effects , Ubiquitin/metabolism , Verteporfin
19.
PLoS Med ; 5(3): e64, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18366248

ABSTRACT

BACKGROUND: Rituximab is used in the treatment of CD20+ B cell lymphomas and other B cell lymphoproliferative disorders. Its clinical efficacy might be further improved by combinations with other drugs such as statins that inhibit cholesterol synthesis and show promising antilymphoma effects. The objective of this study was to evaluate the influence of statins on rituximab-induced killing of B cell lymphomas. METHODS AND FINDINGS: Complement-dependent cytotoxicity (CDC) was assessed by MTT and Alamar blue assays as well as trypan blue staining, and antibody-dependent cellular cytotoxicity (ADCC) was assessed by a 51Cr release assay. Statins were found to significantly decrease rituximab-mediated CDC and ADCC of B cell lymphoma cells. Incubation of B cell lymphoma cells with statins decreased CD20 immunostaining in flow cytometry studies but did not affect total cellular levels of CD20 as measured with RT-PCR and Western blotting. Similar effects are exerted by other cholesterol-depleting agents (methyl-beta-cyclodextrin and berberine), but not filipin III, indicating that the presence of plasma membrane cholesterol and not lipid rafts is required for rituximab-mediated CDC. Immunofluorescence microscopy using double staining with monoclonal antibodies (mAbs) directed against a conformational epitope and a linear cytoplasmic epitope revealed that CD20 is present in the plasma membrane in comparable amounts in control and statin-treated cells. Atomic force microscopy and limited proteolysis indicated that statins, through cholesterol depletion, induce conformational changes in CD20 that result in impaired binding of anti-CD20 mAb. An in vivo reduction of cholesterol induced by short-term treatment of five patients with hypercholesterolemia with atorvastatin resulted in reduced anti-CD20 binding to freshly isolated B cells. CONCLUSIONS: Statins were shown to interfere with both detection of CD20 and antilymphoma activity of rituximab. These studies have significant clinical implications, as impaired binding of mAbs to conformational epitopes of CD20 elicited by statins could delay diagnosis, postpone effective treatment, or impair anti-lymphoma activity of rituximab.


Subject(s)
Antibodies, Monoclonal/drug effects , Antibodies, Monoclonal/therapeutic use , Antigens, CD20/drug effects , Antineoplastic Agents/antagonists & inhibitors , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Lymphoma, B-Cell/drug therapy , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD20/chemistry , B-Lymphocytes/metabolism , Cell Line, Tumor , Cholesterol/pharmacology , Cytotoxicity, Immunologic/drug effects , Humans , Hypercholesterolemia/blood , Hypercholesterolemia/drug therapy , Lovastatin/pharmacology , Membrane Microdomains/drug effects , Protein Conformation/drug effects , Rituximab
20.
Acta Biochim Pol ; 55(1): 75-84, 2008.
Article in English | MEDLINE | ID: mdl-18327303

ABSTRACT

Thiazolidinediones are oral antidiabetic agents that activate peroxisome proliferator-activated receptor-gamma (PPAR-gamma) and exert potent antioxidant and anti-inflammatory properties. It has also been shown that PPAR-gamma agonists induce G0/G1 arrest and apoptosis of malignant cells. Some of these effects have been suggested to result from inhibition of proteasome activity in target cells. The aim of our studies was to critically evaluate the cytostatic/cytotoxic effects of one of thiazolidinediones (pioglitazone) and its influence on proteasome activity. Pioglitazone exerted dose-dependent cytostatic/cytotoxic effects in MIA PaCa-2 cells. Incubation of tumor cells with pioglitazone resulted in increased levels of p53 and p27 and decreased levels of cyclin D1. Accumulation of polyubiquitinated proteins within cells incubated with pioglitazone suggested dysfunction of proteasome activity. However, we did not observe any influence of pioglitazone on the activity of isolated proteasome and on the proteolytic activity in lysates of pioglitazone-treated MIA PaCa-2 cells. Further, treatment with pioglitazone did not cause an accumulation of fluorescent proteasome substrates in transfected HeLa cells expressing unstable GFP variants. Our results indicate that pioglitazone does not act as a direct or indirect proteasome inhibitor.


Subject(s)
Neoplasms/drug therapy , PPAR gamma/metabolism , Proteasome Inhibitors , Thiazolidinediones/pharmacology , Cell Line, Tumor , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Cytostatic Agents/pharmacology , Dose-Response Relationship, Drug , HeLa Cells , Humans , Hypoglycemic Agents/pharmacology , Neoplasms/metabolism , Pioglitazone , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Tumor Suppressor Protein p53/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...