Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(12)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38927997

ABSTRACT

Oxidative stress represents a critical facet of the array of abiotic stresses affecting crop growth and yield. In this paper, we investigated the potential differences in the functions of two highly homologous Arabidopsis DSS1 proteins in terms of maintaining genome integrity and response to oxidative stress. In the context of homologous recombination (HR), it was shown that overexpressing AtDSS1(I) using a functional complementation test increases the resistance of the Δdss1 mutant of Ustilago maydis to genotoxic agents. This indicates its conserved role in DNA repair via HR. To investigate the global transcriptome changes occurring in dss1 plant mutant lines, gene expression analysis was conducted using Illumina RNA sequencing technology. Individual RNA libraries were constructed from three total RNA samples isolated from dss1(I), dss1(V), and wild-type (WT) plants under hydrogen peroxide-induced stress. RNA-Seq data analysis and real-time PCR identification revealed major changes in gene expression between mutant lines and WT, while the dss1(I) and dss1(V) mutant lines exhibited analogous transcription profiles. The Kyoto Encyclopedia of Genes and Genomes enrichment analysis revealed significantly enriched metabolic pathways. Notably, genes associated with HR were upregulated in dss1 mutants compared to the WT. Otherwise, genes of the metabolic pathway responsible for the synthesis of secondary metabolites were downregulated in both dss1 mutant lines. These findings highlight the importance of understanding the molecular mechanisms of plant responses to oxidative stress.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Gene Expression Regulation, Plant , Oxidative Stress , Seedlings , Transcriptome , Oxidative Stress/genetics , Arabidopsis/genetics , Arabidopsis/metabolism , Seedlings/genetics , Seedlings/metabolism , Seedlings/growth & development , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Gene Knockout Techniques , Gene Expression Profiling , Mutation , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism
2.
Int J Mol Sci ; 24(3)2023 Jan 26.
Article in English | MEDLINE | ID: mdl-36768765

ABSTRACT

Global climate change has a detrimental effect on plant growth and health, causing serious losses in agriculture. Investigation of the molecular mechanisms of plant responses to various environmental pressures and the generation of plants tolerant to abiotic stress are imperative to modern plant science. In this paper, we focus on the application of the well-established technology CRISPR/Cas9 genome editing to better understand the functioning of the intrinsically disordered protein DSS1 in plant response to oxidative stress. The Arabidopsis genome contains two highly homologous DSS1 genes, AtDSS1(I) and AtDSS1(V). This study was designed to identify the functional differences between AtDSS1s, focusing on their potential roles in oxidative stress. We generated single dss1(I) and dss1(V) mutant lines of both Arabidopsis DSS1 genes using CRISPR/Cas9 technology. The homozygous mutant lines with large indels (dss1(I)del25 and dss1(V)ins18) were phenotypically characterized during plant development and their sensitivity to oxidative stress was analyzed. The characterization of mutant lines revealed differences in root and stem lengths, and rosette area size. Plants with a disrupted AtDSS1(V) gene exhibited lower survival rates and increased levels of oxidized proteins in comparison to WT plants exposed to oxidative stress induced by hydrogen peroxide. In this work, the dss1 double mutant was not obtained due to embryonic lethality. These results suggest that the DSS1(V) protein could be an important molecular component in plant abiotic stress response.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , CRISPR-Cas Systems , Gene Editing/methods , Oxidative Stress/genetics , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism
3.
Protoplasma ; 258(4): 779-792, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33404921

ABSTRACT

DSS1 is a small protein, highly conserved across different species. As a member of the intrinsically disordered protein family, DSS1 interacts with different protein partners, thus forming complexes involved in diverse biological mechanisms: DNA repair, regulation of protein homeostasis, mRNA export, etc. Additionally, DSS1 has a novel intriguing role in the post-translational protein modification named DSSylation. Oxidatively damaged proteins are targeted for removal with DSS1 and then degraded by proteasome. Yet, DSS1 involvement in the maintenance of genome integrity through homologous recombination is the only function well studied in Arabidopsis research. The fact that animal DSS1 shows wide multifunctionality imposes a need to investigate the additional roles of two Arabidopsis thaliana DSS1 homologs. Having in mind the universality of various biological processes, we considered the possibility of plant DSS1 involvement in cellular homeostasis maintenance during stress exposure. Using real-time PCR and immunoblot analysis, we investigated the profiles of DSS1 gene and protein expression under oxidative stress. We grew and selected the homozygous Arabidopsis mutant line, carrying the T-DNA intron insertion in the DSS1(V) gene. The mutant line was phenotypically described during plant development, and its sensitivity to oxidative stress was characterized. This is the first report which indicates that plant DSS1 gene expression has an altered profile under the influence of oxidative stress. dss1(V)-/- plants showed an increased sensitivity to oxidative stress, germinated faster than WT, but generally showed developmental delay in further stages. Our results indicate that the DSS1 protein could be a crucial player in the molecular mechanisms underlying plant abiotic stress responses.


Subject(s)
Arabidopsis , Intrinsically Disordered Proteins , Animals , Arabidopsis/genetics , Arabidopsis/metabolism , Genes, Plant , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Oxidative Stress/genetics , Stress, Physiological
4.
Immunobiology ; 224(3): 470-476, 2019 05.
Article in English | MEDLINE | ID: mdl-30765133

ABSTRACT

Dendritic cells (DC) are responsible for the initiation and shaping of the adaptive immune response and are in the focus of autoimmunity research. We were interested in comparison of DC obtained from autoimmunity-prone Dark Agouti (DA) rats and autoimmunity-resistant Albino Oxford (AO) rats. DC were generated from bone marrow precursors and matured (mDC) by lipopolysaccharide. Tolerogenic DC (tolDC) obtained by vitamin D3 treatment were studied in parallel. Profile of cytokine production was different in AO and DA mDC and tolDC. Expression of MHC class II molecules and CD86 were higher in DA DC, while vitamin D3 reduced their expression in dendritic cells of both strains. Allogeneic proliferation of CD4+ T cells was reduced by AO tolDC, but not with DA tolDC in comparison to respective mDC. Finally, expression of various genes identified as differentially expressed in human mDC and tolDC was also analyzed in AO and DA DC. Again, AO and DA DC differed in the expression of the analyzed genes. To conclude, AO and DA DC differ in production of cytokines, expression of antigen presentation-related molecules and in regulation of CD4+ T proliferation. The difference is valuable for understanding the divergence of the strains in their susceptibility to autoimmunity.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Animals , Antigen Presentation , Autoimmunity , Cell Differentiation , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Resistance , Disease Susceptibility , Female , Genetic Background , Immune Tolerance , Lipopolysaccharides/immunology , Rats , Rats, Inbred Strains , Transcriptome
5.
Ann Bot ; 118(2): 271-80, 2016 08.
Article in English | MEDLINE | ID: mdl-27371693

ABSTRACT

BACKGROUND AND AIMS: Retranslocation of iron (Fe) from source tissues enhances plant tolerance to Fe deficiency. Previous work has shown that silicon (Si) can alleviate Fe deficiency by enhancing acquisition and root to shoot translocation of Fe. Here the role of Si in Fe mobilization in older leaves and the subsequent retranslocation of Fe to young leaves of cucumber (Cucumis sativus) plants growing under Fe-limiting conditions was investigated. METHODS: Iron ((57)Fe or naturally occurring isotopes) was measured in leaves at different positions on plants hydroponically growing with or without Si supply. In parallel, the concentration of the Fe chelator nicotianamine (NA) along with the expression of nicotianamine synthase (NAS) involved in its biosynthesis and the expression of yellow stripe-like (YSL) transcripts mediating Fe-NA transport were also determined. KEY RESULTS: In plants not receiving Si, approximately half of the total Fe content remained in the oldest leaf. In contrast, Si-treated plants showed an almost even Fe distribution among leaves with four different developmental stages, thus providing evidence of enhanced Fe remobilization from source leaves. This Si-stimulated Fe export was paralleled by an increased NA accumulation and expression of the YSL1 transporter for phloem loading/unloading of the Fe-NA complex. CONCLUSIONS: The results suggest that Si enhances remobilization of Fe from older to younger leaves by a more efficient NA-mediated Fe transport via the phloem. In addition, from this and previous work, a model is proposed of how Si acts to improve Fe homeostasis under Fe deficiency in cucumber.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Cucumis sativus/drug effects , Iron Deficiencies , Silicon/pharmacology , Alkyl and Aryl Transferases/genetics , Azetidinecarboxylic Acid/analogs & derivatives , Azetidinecarboxylic Acid/metabolism , Biological Transport , Cucumis sativus/metabolism , Homeostasis , Hydroponics , Models, Biological , Phloem/drug effects , Phloem/metabolism , Plant Leaves/drug effects , Plant Leaves/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism
6.
J Agric Food Chem ; 64(24): 4900-7, 2016 Jun 22.
Article in English | MEDLINE | ID: mdl-27225664

ABSTRACT

Cucurbitacin E (CucE) is a highly oxidized steroid consisting of a tetracyclic triterpene. It is a member of a Cucurbitacin family of biomolecules that are predominantly found in Cucurbitaceae plants. CucE has already been identified as a potent anti-inflammatory compound. Here, its effects on CD4(+) T helper (Th) cells and macrophages, as the major encephalitogenic cells in the autoimmunity of the central nervous system, were investigated. Production of major pathogenic Th cell cytokines: interferon-gamma and interleukin-17 were inhibited under the influence of CucE. The effects of CucE on CD4(+) T cells were mediated through the modulation of aryl hydrocarbon receptor, STAT3, NFκB, p38 MAPK, and miR-146 signaling. Further, production of nitric oxide and reactive oxygen species, as well as phagocytic ability, were inhibited in macrophages treated with CucE. These results imply that CucE possesses powerful antiencephalitogenic activity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Triterpenes/administration & dosage , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Signal Transduction/drug effects , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Zebrafish
7.
Biomed Pharmacother ; 74: 206-10, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26349986

ABSTRACT

MicroRNAs (miR) are small non-coding RNAs involved in the immune response regulation. miR-155 has been attributed a major pro-inflammatory role in the pathogenesis of multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). Here, a role of miR-155 in re-activation of encephalitogenic CD4(+) T cells was investigated. Dark Agouti rats were immunized with myelin basic protein (MBP) emulsified in complete Freund's adjuvant. CD4(+) T cells were purified from draining lymph node cells (DLNC) obtained in the inductive phase and from spinal cord immune cells (SCIC) isolated at the peak of EAE. CD4(+) T cells obtained from SCIC (i.e., in vivo re-activated cells) had markedly higher expression of miR-155 in comparison to those purified from DLNC (not re-activated). Likewise, in vitro re-activation of DLNC with MBP led to increase in miR-155 expression. Further, DLNC and DLNC CD4(+) T cells were transfected with an inhibitor of miR-155 during in vitro re-activation. As a result, expression of important CD4(+) T cell effector cytokines IFN-γ and IL-17, but not of regulatory cytokines IL-10 and TGF-ß, was reduced. These results imply that miR-155 supports re-activation of encephalitogenic CD4(+) T cells. Our results contribute to a view that miR-155 might be a valuable target in multiple sclerosis therapy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , MicroRNAs/genetics , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Expression Regulation , Multiple Sclerosis/genetics , Multiple Sclerosis/physiopathology , Myelin Basic Protein/immunology , Rats , Spinal Cord/immunology
8.
J Neuroimmunol ; 262(1-2): 72-8, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23890777

ABSTRACT

Transferrin (Tf) has a major role in T cell activation and proliferation. Here, we investigated whether Tf exerts immunomodulatory effects on T cells and in development of T-cell driven experimental autoimmune encephalomyelitis (EAE). While treatment of concanavalin A-stimulated splenocytes with apotransferrin (ApoTf) did not affect release of IL-1ß, TNF, IFN-γ, IL-17, IL-4, and IL-10, it markedly and dose-dependently down-regulated synthesis of IL-2 in these cells. ApoTf also inhibited IL-2 generation in purified CD3+ T cells and the effect was accompanied with down-regulation of MAPK p44/42 and NFκB signaling. Despite impeded IL-2 release, proliferation of splenocytes was not inhibited by ApoTf. Importantly, ApoTf ameliorated EAE in mice and significantly reduced ex vivo IL-2 production in proteolipid protein-specific lymphocytes. Thus ApoTf may be a promising beneficial agent for multiple sclerosis.


Subject(s)
Apoproteins/physiology , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , Transferrin/physiology , Animals , Apoproteins/administration & dosage , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunosuppressive Agents/administration & dosage , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred NOD , NF-kappa B/antagonists & inhibitors , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transferrin/administration & dosage
9.
Brain Res ; 1519: 120-6, 2013 Jun 26.
Article in English | MEDLINE | ID: mdl-23651977

ABSTRACT

CXCL12 plays a protective role in CNS autoimmunity. Expression of CXCL12-γ, which has distinct structural and functional properties than the other isoforms of CXCL12, was determined in spinal cords of rats immunized to develop experimental autoimmune encephalomyelitis (EAE). CNS expression of CXCL12-γ was markedly lower in EAE-prone Dark Agouti rats than in EAE-resistant Albino Oxford rats, both in spinal cord homogenates and micro-blood vessels isolated from spinal cords. Inhibition of nitric oxide (NO) synthesis in DA rats upregulated, while donation of NO in AO rats downregulated CNS expression of CXCL12-γ. NO inhibited CXCL12-γ expression in astrocytes in vitro. A splice variant of CXCL12-γ which migrates into nucleolus was not detected in spinal cord or astrocytes. Thus, CXCL12-γ is expressed in the CNS after EAE induction, but its expression is markedly suppressed in spinal cord affected with full blown inflammation. NO is an important regulator of CXCL12-γ expression in neuroinflammation.


Subject(s)
Chemokine CXCL12/metabolism , Encephalomyelitis, Autoimmune, Experimental/complications , Inflammation/etiology , Inflammation/pathology , Spinal Cord/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Chemokine CXCL12/genetics , Cytokines/pharmacology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Freund's Adjuvant/toxicity , Male , Nitric Oxide Synthase/metabolism , Rats , Rats, Inbred Strains , Species Specificity , Spinal Cord/drug effects , Time Factors , Up-Regulation
10.
J Neuroimmunol ; 259(1-2): 55-65, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23602714

ABSTRACT

NO-hybridization of the HIV protease inhibitor Saquinavir generates a new chemical entity named Saq-NO, that retains the anti-viral activity and exerts lower toxicity. We show that Saq-NO inhibited the generation of various cytokines in ConA-stimulated unfractionated murine spleen cells and rat lymph nodes stimulated with ConA as well as in purified CD4(+) T cells in vitro and reduced the circulating levels of cytokines in mice challenged with anti-CD3 antibody. Furthermore, Saq-NO reduced IL-17 and IFN-γ production in myelin basic protein (MBP)-specific cells isolated from rats immunized with MBP. These findings translated well into the in vivo setting as Saq-NO ameliorated the course of the disease in two preclinical models of multiple sclerosis. Our results demonstrate that Saq-NO exerts immunomodulatory effects that warrant studies on its application in autoimmune diseases.


Subject(s)
Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Saquinavir/analogs & derivatives , Animals , Benzofurans , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lymph Nodes/cytology , Mice , Mice, Inbred BALB C , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/immunology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Quinolines , Rats , Rats, Inbred Strains , Ribosomal Protein S6 Kinases/metabolism , Saquinavir/pharmacology , Spleen/cytology
11.
New Phytol ; 198(4): 1096-1107, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23496257

ABSTRACT

· Root responses to lack of iron (Fe) have mainly been studied in nutrient solution experiments devoid of silicon (Si). Here we investigated how Si ameliorates Fe deficiency in cucumber (Cucumis sativus) with focus on the storage and utilization of Fe in the root apoplast. · A combined approach was performed including analyses of apoplastic Fe, reduction-based Fe acquisition and Fe-mobilizing compounds in roots along with the expression of related genes. · Si-treated plants accumulated higher concentrations of root apoplastic Fe, which rapidly decreased when Fe was withheld from the nutrient solution. Under Fe-deficient conditions, Si also increased the accumulation of Fe-mobilizing compounds in roots. Si supply stimulated root activity of Fe acquisition at the early stage of Fe deficiency stress through regulation of gene expression levels of proteins involved in Fe acquisition. However, when the period of Fe deprivation was extended, these reactions further decreased as a consequence of Si-induced enhancement of the Fe status of the plants. · This work provides new evidence for the beneficial role of Si in plant nutrition and clearly indicates that Si-mediated alleviation of Fe deficiency includes an increase of the apoplastic Fe pool in roots and an enhancement of Fe acquisition.


Subject(s)
Cucumis sativus/metabolism , Extracellular Space/metabolism , Iron Deficiencies , Iron/metabolism , Plant Roots/metabolism , Silicon/pharmacology , Citrates/metabolism , Cucumis sativus/drug effects , Cucumis sativus/genetics , Cucumis sativus/growth & development , Extracellular Space/drug effects , Gene Expression Regulation, Plant/drug effects , Genes, Plant/genetics , Malates/metabolism , Plant Roots/drug effects , Plant Roots/genetics , Xylem/drug effects , Xylem/metabolism
12.
Growth Factors ; 30(6): 385-93, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23137174

ABSTRACT

Although several reports suggest a potentially deleterious role of macrophage migration inhibitory factor (MIF) in type 2 diabetes (T2D) pathology, it is still unclear how this pro-inflammatory cytokine acts on pancreatic beta cells. The aim of the present study was to evaluate MIF effects on murine beta cells in the in vitro settings mimicking T2D-associated conditions. Results indicate that recombinant MIF further increased apoptosis of pancreatic islets or MIN6 cells upon exposure to palmitic acid or glucose. This was accompanied by upregulation of several pro-apoptotic molecules. Furthermore, MIF potentiated nutrient-induced islet cell dysfunction, as revealed by lower glucose oxidation rate, ATP content, and depolarized mitochondrial membrane. The final outcome was potentiation of mitochondrial apoptotic pathway. The observed upregulation of nutrient-induced islet cell dysfunction and apoptosis by MIF implicates that silencing MIF may be beneficial for maintaining integrity of endocrine pancreas in obesity-associated T2D.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/cytology , Macrophage Migration-Inhibitory Factors/metabolism , Palmitic Acid/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Cell Line , Cytokines/metabolism , DNA Fragmentation , Diabetes Mellitus, Type 2/metabolism , Gene Expression Profiling , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Tetrazolium Salts/chemistry , Thiazoles/chemistry
13.
Int J Biochem Cell Biol ; 44(6): 838-41, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22484430

ABSTRACT

CXCL12, also known as SDF-1 (stromal cell derived factor-1) is a small protein that belongs to the chemokine family, whose members have a crucial role in directing cell migration. CXCL12 has an essential role in neural and vascular development, hematopoiesis and in immunity. It acts through two receptors, CXCR4 and CXCR7. While the former is a classic G protein-coupled transmembrane chemokine receptor, the latter primarily function as a scavenger of CXCL12. CXCL12 has been considered as a standard pro-inflammatory molecule for a long time, as it attracts leukocytes to inflammatory sites and contributes to their activation. However, recent findings indicate that this chemokine has the opposite role in neuroinflammation. In this review, basic data about molecular and functional properties of CXCL12 are presented, while its role in CNS autoimmunity is addressed in details.


Subject(s)
Chemokine CXCL12/physiology , Inflammation/physiopathology , Nervous System/physiopathology , Animals , Humans
14.
Food Chem Toxicol ; 50(5): 1499-507, 2012 May.
Article in English | MEDLINE | ID: mdl-22342526

ABSTRACT

Warfarin affects mainly vitamin K dependent (VKD) processes, but the effects on some non-VKD-related activities such as tumor growth inhibition and mononuclear cell-mediated immune reactions were shown as well. In this study, the effect of subchronic (30 days) oral warfarin (0.35 mg/l and 3.5mg/l) intake on peripheral blood granulocytes in rats was investigated. Increase in prothrombin and partial thromboplastin time at high warfarin dose reflected its basic activity. Priming effect for respiratory burst was noted at both warfarin doses, while only high warfarin dose resulted in priming for adhesion, the rise in intracellular myeloperoxidase content/release and stimulation of nitric oxide production. Differential effects of high warfarin dose were noted on granulocyte cytokines IL-6 (lack of the effect), TNF-α (decreased release and mRNA expression) and IL-12 (increase in mRNA for IL-12 subunits p35 and p40). Changes in granulocytes seems not to rely on mitogen activated kinases p38 and ERK. Warfarin intake was associated with an increase in circulating IL-6, fibrinogen and haptoglobin and with changes in the activity of erythrocyte antioxidant enzymes superoxide dismutase and catalase. The effects of oral warfarin intake on peripheral blood granulocytes demonstrated in this study might be relevant for oral anticoagulant therapy strategies in humans.


Subject(s)
Anticoagulants/administration & dosage , Granulocytes/drug effects , Warfarin/administration & dosage , Administration, Oral , Animals , Anticoagulants/pharmacology , Base Sequence , Catalase/metabolism , Cell Adhesion , DNA Primers , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Granulocytes/cytology , Interleukin-6/blood , Male , Protein Kinases/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/blood , Warfarin/pharmacology
15.
Immunol Cell Biol ; 90(7): 688-98, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22064706

ABSTRACT

As a result of chronic exposure to high levels of free fatty acids, glucose and inflammatory mediators ß-cell apoptosis occurs at the end stage of obesity-associated type 2 diabetes (T2D). One potentially deleterious molecule for ß-cell function associated with T2D and obesity in humans is macrophage migration inhibitory factor (MIF). Therefore, the aim of this study was to explore MIF expression in vivo during development of obesity and insulin resistance in high-fat diet (HFD)-fed C57BL/6 mice and whether MIF inhibition could affect ß-cell apoptosis and dysfunction induced by palmitic acid (PA) in vitro. Indeed, increase in systemic and locally produced MIF correlated well with the weight gain, triglyceride upregulation, glucose intolerance and insulin resistance, which developed in HFD-fed mice. In in vitro settings PA dose-dependently induced MIF secretion before apoptosis development in islets. Further, mif gene deletion, mRNA silencing or protein inhibition rescued ß-cells from PA-induced apoptosis as measured by MTT assay and histone-DNA enzyme linked immuno sorbent assay. Protection from induced apoptosis was mediated by altered activation of caspase pathway and correlated with changes in the level of Bcl-2 family members. Further, MIF inhibition conveyed a significant resistance to PA-induced downregulation of insulin and PDX-1 expression and ATP content. However, ß-cell function was not entirely preserved in the absence of MIF judging by low glucose oxidation and depolarized mitochondrial membrane. In conclusion, the observed considerable preservation of ß-cells from nutrient-induced apoptosis might implicate MIF as a potential therapeutic target in the later stage of obesity-associated T2D.


Subject(s)
Apoptosis/drug effects , Islets of Langerhans/metabolism , Macrophage Migration-Inhibitory Factors/deficiency , Palmitic Acid/pharmacology , Animals , Blood Glucose/metabolism , Caspase 9/genetics , Cell Line, Tumor , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Diet, High-Fat/adverse effects , Enzyme-Linked Immunosorbent Assay , Female , Humans , Insulin/blood , Islets of Langerhans/pathology , Isoxazoles/pharmacology , Macrophage Migration-Inhibitory Factors/blood , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood , Up-Regulation/drug effects , bcl-2-Associated X Protein/genetics
16.
J Cell Physiol ; 226(7): 1803-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21506111

ABSTRACT

We have recently shown that covalent attachment of the NO moiety to the HIV protease inhibitor Saquinavir (Saq) produced a qualitatively new chemical entity, named Saquinavir-NO (Saq-NO), with enhanced anticancer properties and reduced toxicity. In this study we evaluated the impact of Saq-NO on the growth of A375 human melanoma cells, as a prototype of NO-dependent cancer model. The novel compound strongly affected the in vitro and in vivo progression of A375 melanoma cell growth. The mechanism of antimelanoma action comprised dual drug activity-induction of apoptotic cell death and acquisition of melanoma cell responsiveness to TRAIL. Saq-NO-triggered apoptosis was dependent on transient AKT up-regulation and reduced pERK and iNOS expression that were observed within the first 12 h of exposure to the drug. Thereafter, however, Saq-NO up-regulated both iNOS transcription and NO endogenous synthesis and sensitized A375 cells to TRAIL. Furthermore, reduced YY1 expression was observed after 24 h of Saq-NO exposure, which correlated with increased expression of DR5. The biological relevance of this complex and powerful action of Saq-NO was consistent with the marked drug-induced inhibition of the growth of A375 xenotransplants in nude mice.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Melanoma/drug therapy , Nitric Oxide Synthase Type II/metabolism , Saquinavir/analogs & derivatives , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Melanoma/enzymology , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Nude , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Saquinavir/pharmacology , Time Factors , Up-Regulation , Xenograft Model Antitumor Assays , YY1 Transcription Factor/metabolism
17.
FEBS Lett ; 585(23): 3781-8, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21443873

ABSTRACT

Astrocytes are the most abundant cell population within the CNS of mammals. Their glial role is perfectly performed in the healthy CNS as they support functions of neurons. The omnipresence of astrocytes throughout the white and grey matter and their intimate relation with blood vessels of the CNS, as well as numerous immunity-related actions that these cells are capable of, imply that astrocytes should have a prominent role in neuroinflammatory disorders, such as multiple sclerosis (MS). The role of astrocytes in MS is rather ambiguous, as they have the capacity to both stimulate and restrain neuroinflammation and tissue destruction. In this paper we present some of the proved and the proposed functions of astrocytes in neuroinflammation and discuss the effect of MS therapeutics on astrocytes.


Subject(s)
Astrocytes/pathology , Multiple Sclerosis/pathology , Animals , Humans , Models, Immunological , Molecular Targeted Therapy
18.
Cell Cycle ; 10(3): 492-9, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21270522

ABSTRACT

The NO-derivative of the HIV protease inhibitor saquinavir (Saq-NO) is a nontoxic variant of the parental drug with enhanced anticancer activity on several cell lines. However, it is still unclear whether the p53 status of the target cell might influence the sensitivity to Saq-NO. In this study we evaluated the in vitro and in vivo activity of Saq-NO on the p53-deficient hormone resistant prostate cancer PC-3 cells. We demonstrate that the absence of functional p53 is not essential for the capacity of Saq-NO to reduce prostate cancer cell growth. In contrast to its previously described cytostatic action in B16 and C6 cell lines, Saq-NO exerted cytotoxic effects in PC-3 cells leading to dominant induction of apoptosis and enhanced production of proapoptotic Bim. In addition, differently from saquinavir, Saq-NO restored TRAIL sensitivity that was correlated with increased expression of DR5 independent from ROS/RNS production and YY1 repression. NF-κB activation may be responsible of the Saq-NO induced DR5 expression. Moreover, Saq-NO but not saquinavir, exerted synergistic activity with conventional cytostatic therapy. In agreement with these in vitro studies, Saq-NO inhibited the in vivo growth of PC-3 cells xenotransplants to a greater extent than the parental compound. Taken together, these data indicate that Saq-NO possesses powerful and suitable in vitro and in vivo chemotherapeutic potential to be further studied as a novel drug for the treatment of prostate cancer in the clinical setting.


Subject(s)
Antineoplastic Agents/pharmacology , Protease Inhibitors/pharmacology , Saquinavir/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Humans , Male , Mice , Mice, Nude , Paclitaxel/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Protease Inhibitors/therapeutic use , Saquinavir/pharmacology , Saquinavir/therapeutic use , Transplantation, Heterologous , Tumor Suppressor Protein p53/genetics
19.
Int J Cancer ; 128(8): 1955-65, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-20568104

ABSTRACT

Various constituents of the olive tree (Olea europaea) have been traditionally used in the treatment of infection, inflammation, prevention of chronic diseases, cardiovascular disorders and cancer. The anticancer potential of dry olive leaf extract (DOLE) represents the net effect of multilevel interactions between different biologically active compounds from the extract, cancer cells and conventional therapy. In this context, it was of primary interest to evaluate the influence of DOLE on progression of the highly malignant, immuno- and chemoresistant type of skin cancer-melanoma. DOLE significantly inhibited proliferation and subsequently restricted clonogenicity of the B16 mouse melanoma cell line in vitro. Moreover, late phase tumor treatment with DOLE significantly reduced tumor volume in a syngeneic strain of mice. DOLE-treated B16 cells were blocked in the G(0) /G(1) phase of the cell cycle, underwent early apoptosis and died by late necrosis. At the molecular level, the dying process started as caspase dependent, but finalized as caspase independent. In concordance, overexpression of antiapoptotic members of the Bcl-2 family, Bcl-2 and Bcl-XL, and diminished expression of their natural antagonists, Bim and p53, were observed. Despite molecular suppression of the proapoptotic process, DOLE successfully promoted cell death mainly through disruption of cell membrane integrity and late caspase-independent fragmentation of genetic material. Taken together, the results of this study indicate that DOLE possesses strong antimelanoma potential. When DOLE was applied in combination with different chemotherapeutics, various outcomes, including synergy and antagonism, were observed. This requires caution in the use of the extract as a supplementary antitumor therapeutic.


Subject(s)
Melanoma, Experimental/prevention & control , Olea/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry , Skin Neoplasms/prevention & control , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Blotting, Western , Caspases/genetics , Caspases/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Female , Male , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/pathology , Tumor Cells, Cultured
20.
J Plant Physiol ; 167(16): 1407-11, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20637525

ABSTRACT

Metallothionein type 3 (MT3) expression has previously been detected in leaves, fruits, and developing somatic embryos in different plant species. However, specific tissular and cellular localization of MT3 transcripts have remained unidentified. In this study, in situ RNA-RNA analysis revealed buckwheat metallothionein type 3 (FeMT3) transcript localization in vascular elements, mesophyll and guard cells of leaves, vascular tissue of roots and throughout the whole embryo. Changes in FeMT3 mRNA levels in response to drought and oxidative stress, as well as ROS scavenging abilities of the FeMT3 protein in yeast were also detected, indicating possible involvement of FeMT3 in stress defense and ROS related cellular processes.


Subject(s)
Fagopyrum/metabolism , Metallothionein/metabolism , Plant Proteins/metabolism , Droughts , Oxidative Stress/genetics , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...