Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 14(2): 397-410, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26663311

ABSTRACT

UNLABELLED: ESSENTIALS: Extracellular histones are highly augmented in sites of neovessel formation, such as regeneration tissues. We studied histone effect on survival and angiogenic activity of mature and progenitor endothelial cells. Extracellular histones trigger apoptosis and pyroptosis and reduce angiogenesis in vivo and in vitro. Histone blockade can be useful as a therapeutic strategy to improve angiogenesis and tissue regeneration. BACKGROUND: Extracellular histones are highly augmented in sites of neovessel formation, like regeneration tissues. Their cytotoxic effect has been studied in endothelial cells, although the mechanism involved and their action on endothelial colony-forming cells (ECFCs) remain unknown. OBJECTIVE: To study the effect of histones on ECFC survival and angiogenic functions and compare it with mature endothelial cells. METHODS AND RESULTS: Nuclear morphology analysis showed that each human recombinant histone triggered both apoptotic-like and necrotic-like cell deaths in both mature and progenitor endothelial cells. While H1 and H2A exerted a weak toxicity, H2B, H3 and H4 were the most powerful. The percentage of apoptosis correlated with the percentage of ECFCs exhibiting caspase-3 activation and was zeroed by the pan-caspase inhibitor Z-VAD-FMK. Necrotic-like cell death was also suppressed by this compound and the caspase-1 inhibitor Ac-YVAD-CMK, indicating that histones triggered ECFC pyroptosis. All histones, at non-cytotoxic concentrations, reduced migration and H2B, H3 and H4 induced cell cycle arrest and impaired tubulogenesis via p38 activation. Neutrophil-derived histones exerted similar effects. In vivo blood vessel formation in the quail chorioallantoic membrane was also reduced by H2B, H3 and H4. Their cytotoxic and antiangiogenic effects were suppressed by unfractioned and low-molecular-weight heparins and the combination of TLR2 and TLR4 blocking antibodies. CONCLUSIONS: Histones trigger both apoptosis and pyroptosis of ECFCs and inhibit their angiogenic functions. Their cytotoxic and antiangiogenic effects are similar in mature endothelial cells and disappear after heparin addition or TLR2/TLR4 blockade, suggesting both as therapeutic strategies to improve tissue regeneration.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelial Progenitor Cells/drug effects , Histones/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Animals , Apoptosis/drug effects , Caspase 1/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Pyroptosis/drug effects , Quail , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Platelets ; 26(8): 799-804, 2015.
Article in English | MEDLINE | ID: mdl-26082997

ABSTRACT

BACKGROUND: Platelets mediate angiogenesis through the secretion of several factors, including the pro-angiogenic vascular endothelial growth factor (VEGF) and the anti-angiogenic endostatin. Although previous findings indicated that these molecules are packed into different alpha-granules and selectively released by specific stimulation of protease-activated receptor (PAR)-1 or PAR-4, recent evidences are against this hypothesis. OBJECTIVES: To elucidate the controversies about the VEGF and endostatin release and the overall angiogenic effect of PARs-stimulated platelets. METHODS: VEGF and endostatin were quantified by enzyme linked-immunosorbent assay (ELISA). Endothelial proliferation (pNPP assay), wound healing (scratch assay) and tubule formation (matrigel) of human microvascular endothelial cells (HMEC-1) and endothelial progenitor cells (EPC) were determined using supernatants from PAR-1- or PAR-4-stimulated platelets. RESULTS: Activation of washed platelets (WPs) by PAR-1- or PAR-4-activating peptide (AP) promoted the VEGF and endostatin secretion in a concentration-dependent manner, being PAR-1-AP more potent than PAR-4-AP. The release of both molecules was abrogated by pre-incubation of platelets with PAR antagonists. Activation of platelet-rich plasma (PRP) with either PAR-1-AP or PAR-4-AP induced a significant VEGF secretion. Quantification of platelet-endostatin secretion was not possible in PRP due to the high levels of plasmatic endostatin vs. platelet content. Releasates from PAR-1- or PAR-4-activated WPs promoted similar pattern of angiogenic responses of HMEC-1 or EPC. Moreover, proliferation of HMEC-1 mediated by PAR-stimulated PRP releasates was delayed and significantly lower compared with that induced by PAR-stimulated WPs. CONCLUSIONS: Our results are in contrast with the previously described differential release of VEGF and endostatin induced by the selective PAR-1 or PAR-4 stimulation, and support the notion that while circulating endostatin accounts for the maintenance of a systemic anti-angiogenic state, locally, the release of platelet alpha-granule content promotes angiogenesis.


Subject(s)
Blood Platelets/metabolism , Endostatins/metabolism , Receptor, PAR-1/agonists , Receptors, Thrombin/agonists , Vascular Endothelial Growth Factor A/metabolism , Blood Platelets/drug effects , Endothelial Cells/metabolism , Humans , Neovascularization, Physiologic , Oligopeptides/pharmacology , Platelet Activation/drug effects
3.
J Thromb Haemost ; 13(2): 271-82, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25393316

ABSTRACT

BACKGROUND: To further understand the role of platelets in the pathogenesis of viral infections we explored platelet interaction with Coxsackieviruses B (CVB) 1 and 3. CVB is a group of viruses that cause the majority of human enterovirus-related viral myocarditis; their receptor (CAR) is expressed on the platelet surface and there is a well-characterized CVB3-induced myocarditis murine model. METHODS: Human platelets were infected with CVB1 and 3 and viruses were detected in pellets and in supernatants. C57BL/6J mice with or without platelet depletion were inoculated with CVB3 and peripheral blood and heart samples collected at different times post-infection. RESULTS: CVB1 and 3 RNA and a capsid protein were detected in infected platelets. Despite the fact that titration assays in Vero cells showed increasing infectivity titers over time, supernatants and pellets from infected platelets showed similar levels, suggesting that platelets were not susceptible to a replicative infectivity cycle. CVB binding was CAR-independent and resulted in P-selectin and phosphatidylserine (PS) exposure. CVB3-infected mice showed a rapid thrombocytopenia that correlated with an increase in platelet PS exposure and platelet-leukocyte aggregates without modification of platelet P-selectin expression or von Willebrand factor levels. Mortality, viremia, heart viral titers and myocarditis were significantly higher in platelet-depleted than normal animals. Type I IFN levels were not changed but IgG levels were lower in infected and platelet-depleted mice. CONCLUSIONS: Our data reveal that platelets play a critical role in host survival and immune response against CVB3 infection.


Subject(s)
Blood Platelets/virology , Coxsackievirus Infections/blood , Coxsackievirus Infections/virology , Enterovirus B, Human/pathogenicity , Myocarditis/blood , Myocarditis/virology , Animals , Blood Platelets/immunology , Blood Platelets/metabolism , Capsid Proteins/blood , Capsid Proteins/genetics , Chlorocebus aethiops , Coxsackievirus Infections/immunology , Disease Models, Animal , Enterovirus B, Human/genetics , Enterovirus B, Human/immunology , Enterovirus B, Human/metabolism , Female , Host-Pathogen Interactions , Humans , Immunoglobulin G/blood , Male , Mice, Inbred C57BL , Myocarditis/immunology , P-Selectin/blood , Phosphatidylserines/blood , RNA, Viral/blood , Thrombocytopenia/blood , Thrombocytopenia/virology , Time Factors , Vero Cells , Virus Replication
4.
Cell Death Dis ; 5: e1507, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25375372

ABSTRACT

Solid tumors are infiltrated by immune cells where macrophages and senescent T cells are highly represented. Within the tumor microenvironment, a cross-talk between the infiltrating cells may occur conditioning the characteristic of the in situ immune response. Our previous work showed that tumors induce senescence of T cells, which are powerful suppressors of lympho-proliferation. In this study, we report that Tumor-Induced Senescent (TIS)-T cells may also modulate monocyte activation. To gain insight into this interaction, CD4+ or CD8+TIS-T or control-T cells were co-incubated with autologous monocytes under inflammatory conditions. After co-culture with CD4+ or CD8+TIS-T cells, CD14+ monocytes/macrophages (Mo/Ma) exhibit a higher expression of CD16+ cells and a reduced expression of CD206. These Mo/Ma produce nitric oxide and reactive oxygen species; however, TIS-T cells do not modify phagocyte capacity of Mo/Ma. TIS-T modulated-Mo/Ma show a higher production of pro-inflammatory cytokines (TNF, IL-1ß and IL-6) and angiogenic factors (MMP-9, VEGF-A and IL-8) and a lower IL-10 and IP-10 secretion than monocytes co-cultured with controls. The mediator(s) present in the supernatant of TIS-T cell/monocyte-macrophage co-cultures promote(s) tubulogenesis and tumor-cell survival. Monocyte-modulation induced by TIS-T cells requires cell-to-cell contact. Although CD4+ shows different behavior from CD8+TIS-T cells, blocking mAbs against T-cell immunoglobulin and mucin protein 3 and CD40 ligand reduce pro-inflammatory cytokines and angiogenic factors production, indicating that these molecules are involved in monocyte/macrophage modulation by TIS-T cells. Our results revealed a novel role for TIS-T cells in human monocyte/macrophage modulation, which may have deleterious consequences for tumor progression. This modulation should be considered to best tailor the immunotherapy against cancer.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD40 Ligand/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Membrane Proteins/metabolism , Monocytes/metabolism , CD4-Positive T-Lymphocytes/cytology , CD40 Ligand/genetics , CD8-Positive T-Lymphocytes/cytology , Cell Communication , Cell Survival , Cellular Senescence , Coculture Techniques , Cytokines/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation , HeLa Cells , Hepatitis A Virus Cellular Receptor 2 , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mannose Receptor , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/genetics , Monocytes/cytology , Nitric Oxide/metabolism , Primary Cell Culture , Reactive Oxygen Species/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
5.
Oncogenesis ; 2: e78, 2013 Nov 04.
Article in English | MEDLINE | ID: mdl-24189977

ABSTRACT

First-hits in the multi-hit process of leukemogenesis originate in germline or hematopoietic stem cells (HSCs), yet leukemia-initiating cells (LICs) usually have a lineage-committed phenotype. The molecular mechanisms underlying this compartment shift during leukemia evolution have not been a major focus of investigation and remain poorly understood. Here a mechanism underlying this shift was examined in the context of Runx1 deficiency, a frequent leukemia-initiating event. Lineage-negative cells isolated from the bone marrow of Runx1-haploinsufficient and wild-type control mice were cultured in granulocyte-colony-stimulating factor to force lineage commitment. Runx1-haploinsufficient cells demonstrated significantly greater and persistent exponential cell growth than wild-type controls. Not surprisingly, the Runx1-haploinsufficient cells were differentiation-impaired, by morphology and by flow-cytometric evaluation for granulocyte differentiation markers. Interestingly, however, this impaired differentiation was not because of decreased granulocyte lineage commitment, as RNA and protein upregulation of the master granulocyte lineage-commitment transcription factor Cebpa, and Hoxb4 repression, was similar in wild-type and Runx1-haploinsufficient cells. Instead, RNA and protein expression of Cebpe, a key driver of progressive maturation after lineage commitment, were significantly decreased in Runx1-haploinsufficient cells. Primary acute myeloid leukemia cells with normal cytogenetics and RUNX1 mutation also demonstrated this phenotype of very high CEBPA mRNA expression but paradoxically low expression of CEBPE, a CEBPA target gene. Chromatin-immunoprecipitation analyses suggested a molecular mechanism for this phenotype: in wild-type cells, Runx1 binding was substantially greater at the Cebpe than at the Cebpa enhancer. Furthermore, Runx1 deficiency substantially diminished high-level Runx1 binding at the Cebpe enhancer, but lower-level binding at the Cebpa enhancer was relatively preserved. Thus, Runx1-deficiency permits Cebpa upregulation and the exponential cell growth that accompanies lineage commitment, but by impairing activation of Cebpe, a key proliferation-terminating maturation gene, extends this exponential growth. These mechanisms facilitate germline cell or HSC of origin, yet evolution into LIC with lineage-committed phenotype.

6.
Thromb Haemost ; 110(5): 1035-45, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23965842

ABSTRACT

Histones are highly alkaline proteins found in cell nuclei and they can be released by either dying or inflammatory cells. The recent observations that histones are major components of neutrophil extracellular traps and promote platelet aggregation and platelet-dependent thrombin generation have shown that these proteins are potent prothrombotic molecules. Because the mechanism(s) of platelet activation by histones are not completely understood, we explored the ability of individual recombinant human histones H1, H2A, H2B, H3 and H4 to induce platelet activation as well as the possible molecular mechanisms involved. All histones were substrates for platelet adhesion and spreading and triggered fibrinogen binding, aggregation, von Willebrand factor release, P-selectin and phosphatidylserine (PS) exposure and the formation of platelet-leukocyte aggregates; however, H4 was the most potent. Histone-mediated fibrinogen binding, P-selectin and PS exposure and the formation of mixed aggregates were potentiated by thrombin. Histones induced the activation of ERK, Akt, p38 and NFκB. Accordingly, histone-induced platelet activation was significantly impaired by pretreatment of platelets with inhibitors of ERK (U 0126), PI3K/Akt (Ly 294002), p38 (SB 203580) and NFκB (BAY 11-7082 and Ro 106-9920). Preincubation of platelets with either aspirin or dexamethasone markedly decreased fibrinogen binding and the adhesion mediated by histones without affecting P-selectin exposure. Functional platelet responses induced by H3 and H4, but not H1, H2A and H2B, were partially mediated through interaction with Toll-like receptors -2 and -4. Our data identify histones as important triggers of haemostatic and proinflammatory platelet responses, and only haemostatic responses are partially inhibited by anti-inflammatory drugs.


Subject(s)
Blood Platelets/immunology , Histones/physiology , Thrombin/metabolism , Butadienes/pharmacology , Cell Adhesion/drug effects , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibrinogen/metabolism , Hemostasis/drug effects , Humans , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , P-Selectin , Platelet Activation/drug effects , Signal Transduction/drug effects , Sulfones/pharmacology
7.
Br J Pharmacol ; 170(2): 255-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23713888

ABSTRACT

BACKGROUND AND PURPOSE: Platelets are major players in every step of vessel development through the local delivery of angiogenesis-modulating factors, including the pro-angiogenic protein VEGF and the anti-angiogenic endostatin. Although thrombin is a potent agonist and is highly elevated in angiogenesis-related diseases, studies regarding its action on the release of platelet angiogenic factors are scarce and controversial. Herein, we have investigated the role of thrombin not only in VEGF and endostatin release but also in net platelet angiogenic activity. EXPERIMENTAL APPROACH: Human platelets were stimulated with thrombin in the presence of the various inhibitors of the signalling pathways involved in platelet activation. Supernatants/releasates were used to determine the levels of angiogenic molecules and to induce angiogenic responses. KEY RESULTS: We found that thrombin induced the secretion of both VEGF and endostatin; however, the overall effect of the releasates was pro-angiogenic as they promoted tubule-like formation and increased the proliferation of endothelial cells. Both responses were only slightly suppressed in the presence of a VEGF receptor-neutralizing antibody. Pharmacological studies revealed that while inhibitors of PKC, p38, ERK1/2, Src kinases or PI3K/Akt exerted only partial inhibitory effects, aspirin fully blocked the pro-angiogenic activity of the releasate. CONCLUSIONS AND IMPLICATIONS: In contrast to current belief, platelet pro-angiogenic responses are independent of VEGF and appear to be the result of the combined action of several molecules. Moreover, our data reinforce the notion that aspirin is a good candidate for a therapeutic agent to treat angiogenesis-related diseases.


Subject(s)
Blood Platelets/metabolism , Neovascularization, Physiologic/physiology , Thrombin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Aspirin/pharmacology , Cell Proliferation , Endostatins/metabolism , Endothelial Cells/metabolism , Humans , In Vitro Techniques , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/drug effects , Platelet Activation/physiology , Platelet Aggregation Inhibitors/pharmacology , Signal Transduction/physiology , Thrombin/administration & dosage
8.
Leukemia ; 26(2): 244-54, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21836612

ABSTRACT

The DNA hypomethylating drug decitabine maintains normal hematopoietic stem cell (HSC) self-renewal but induces terminal differentiation in acute myeloid leukemia (AML) cells. The basis for these contrasting cell fates, and for selective CpG hypomethylation by decitabine, is poorly understood. Promoter CpGs, with methylation measured by microarray, were classified by the direction of methylation change with normal myeloid maturation. In AML cells, the methylation pattern at maturation-responsive CpGs suggested at least partial maturation. Consistent with partial maturation, in gene expression analyses, AML cells expressed high levels of the key lineage-specifying factor CEBPA, but relatively low levels of the key late-differentiation driver CEBPE. In methylation analysis by mass spectrometry, CEBPE promoter CpGs that are usually hypomethylated during granulocyte maturation were significantly hypermethylated in AML cells. Decitabine-induced hypomethylation was greatest at these and other promoter CpGs that are usually hypomethylated with myeloid maturation, accompanied by cellular differentiation of AML cells. In contrast, decitabine-treated normal HSCs retained immature morphology, and methylation significantly decreased at CpGs that are less methylated in immature cells. High expression of lineage-specifying factor and aberrant epigenetic repression of some key late-differentiation driver genes distinguishes AML cells from normal HSCs, and could explain the contrasting differentiation and methylation responses to decitabine.


Subject(s)
Antineoplastic Agents/therapeutic use , Azacitidine/analogs & derivatives , CpG Islands , DNA Methylation , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/drug therapy , Azacitidine/therapeutic use , Base Sequence , Cell Lineage , DNA Primers , Decitabine , Humans , Leukemia, Myeloid, Acute/pathology , Polymerase Chain Reaction , Promoter Regions, Genetic
9.
J Thromb Haemost ; 9(12): 2477-85, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22136495

ABSTRACT

BACKGROUND: Type I interferons (IFN-I) negatively regulate megakaryo/thrombopoiesis. However, expression of the IFN-I receptor (IFNAR) in the megakaryocytic lineage is poorly characterized. OBJECTIVES: To study the expression and functionality of IFNAR in the megakaryocytic lineage. METHODS AND RESULTS: Although IFNAR mRNA was found in every cell type studied, its protein expression showed differences between them. According to flow cytometry and immunofluorescence, IFNAR1 was observed in Meg-01, Dami, CD34+ cells and megakaryocytes, but not in proplatelets or platelets. Immunoblotting assays showed that IFNAR1 and IFNAR2 were highly expressed in all cell types, except in platelets where it was barely detectable. Regarding IFNAR1, 130- and 90-kDa bands were detected in Meg-01 and Dami, whereas 130- and 60-kDa bands were found in CD34+ cells and megakaryocytes. Activation of megakaryocytic IFNAR by IFN-ß induced pSTAT1/2 and upregulated the antiviral genes IRF7 and MXA. The latter response was completely suppressed by IFNAR blockade. In contrast, the low levels of IFNAR in platelets were not functional as pSTAT1/2, aggregation and P-selectin expression were not induced by IFN-I. In addition, megakaryocytes increased IFN-I transcript levels and produced IFN-ß upon stimulation with PolyI:C, a synthetic dsRNA that mimics viral infection. CONCLUSIONS: Early progenitors and mature megakaryocytes, but not platelets, express functional IFNAR and synthetize/release IFN-ß, revealing not only that megakaryo/thrombopoiesis regulation by IFN-I is associated with a specific interaction with its receptor, but also that megakaryocytes may play a role in the antiviral defense by being both IFN producers and responders.


Subject(s)
Megakaryocytes/metabolism , Receptor, Interferon alpha-beta/physiology , Blotting, Western , Cell Line , Cell Lineage , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , Megakaryocytes/cytology , Real-Time Polymerase Chain Reaction
10.
J Thromb Haemost ; 9(8): 1562-71, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21649851

ABSTRACT

BACKGROUND: Hyperthermia is one of the main disturbances of homeostasis occurring during sepsis or hypermetabolic states such as cancer. Platelets are important mediators of the inflammation that accompanies these processes, but very little is known about the changes in platelet function that occur at different temperatures. OBJECTIVES: To explore the effect of higher temperatures on platelet physiology. METHODS: Platelet responses including adhesion, spreading (fluorescence microscopy), α(IIb)ß(3) activation (flow cytometry), aggregation (turbidimetry), ATP release (luminescence), thromboxane A(2) generation, alpha-granule protein secretion (ELISA) and protein phosphorylation from different signaling pathways (immunoblotting) were studied. RESULTS: Preincubation of platelets at temperatures higher than 37 °C (38.5-42 °C) inhibited thrombin-induced hemostasis, including platelet adhesion, aggregation, ATP release and thromboxane A(2) generation. The expression of P-selectin and CD63, as well as vascular endothelial growth factor (VEGF) release, was completely inhibited by hyperthermia, whereas von Willebrand factor (VWF) and endostatin levels remained substantially increased at high temperatures. This suggested that release of proteins from platelet granules is modulated not only by classical platelet agonists but also by microenvironmental factors. The observed gradation of response involved not only antiangiogenesis regulators, but also other cargo proteins. Some signaling pathways were more stable than others. While ERK1/2 and AKT phosphorylation were resistant to changes in temperature, Src, Syk, p38 phosphorylation and IkappaB degradation were decreased in a temperature-dependent fashion. CONCLUSIONS: Higher temperatures, such as those observed with fever or tissue invasion, inhibit the hemostatic functions of platelets and selectively regulate the release of alpha-granule proteins.


Subject(s)
Blood Platelets/metabolism , Fever/blood , Hemostasis , Platelet Activation , Secretory Vesicles/metabolism , Adenosine Triphosphate/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hot Temperature , Humans , Microscopy, Fluorescence , Nephelometry and Turbidimetry , Phosphorylation , Platelet Adhesiveness , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Kinases/metabolism , Signal Transduction , Thrombin/metabolism , Thromboxane A2/metabolism , Time Factors
11.
Leukemia ; 25(11): 1739-50, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21701495

ABSTRACT

Suppression of apoptosis by TP53 mutation contributes to resistance of acute myeloid leukemia (AML) to conventional cytotoxic treatment. Using differentiation to induce irreversible cell cycle exit in AML cells could be a p53-independent treatment alternative, however, this possibility requires evaluation. In vitro and in vivo regimens of the deoxycytidine analogue decitabine that deplete the chromatin-modifying enzyme DNA methyl-transferase 1 without phosphorylating p53 or inducing early apoptosis were determined. These decitabine regimens but not equimolar DNA-damaging cytarabine upregulated the key late differentiation factors CCAAT enhancer-binding protein ɛ and p27/cyclin dependent kinase inhibitor 1B (CDKN1B), induced cellular differentiation and terminated AML cell cycle, even in cytarabine-resistant p53- and p16/CDKN2A-null AML cells. Leukemia initiation by xenotransplanted AML cells was abrogated but normal hematopoietic stem cell engraftment was preserved. In vivo, the low toxicity allowed frequent drug administration to increase exposure, an important consideration for S phase specific decitabine therapy. In xenotransplant models of p53-null and relapsed/refractory AML, the non-cytotoxic regimen significantly extended survival compared with conventional cytotoxic cytarabine. Modifying in vivo dose and schedule to emphasize this pathway of decitabine action can bypass a mechanism of resistance to standard therapy.


Subject(s)
Epigenesis, Genetic , Genes, p53 , Leukemia, Myeloid, Acute/drug therapy , Transplantation, Heterologous , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Blotting, Western , Cell Differentiation , Cytarabine/therapeutic use , DNA Damage , Decitabine , Electrophoresis, Polyacrylamide Gel , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Phosphorylation
12.
J Thromb Haemost ; 7(8): 1333-43, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19566544

ABSTRACT

BACKGROUND: Although platelets are anucleated cells, they express several transcription factors that exert non-genomic functions, including the positive and negative regulation of platelet activation. NF-kappaB is a major transcriptional regulator of genes involved in survival, proliferation and inflammation. OBJECTIVE: Because platelets play a critical role not only in hemostasis, but also in inflammation and tumor progression, we evaluated the role of NF-kappaB in platelet physiology. RESULTS: Immunofluorescence, Western blotting and ELISA studies revealed that platelets express IkappaBalpha and NF-kappaB, and that stimulation with thrombin triggers IkappaBalpha phosphorylation and degradation and the binding of platelet NF-kappaB p65 subunit to synthetic oligonucleotides containing the consensus sequence for NF-kappaB. Two specific unrelated inhibitors of NF-kappaB activation, BAY 11-7082 and Ro 106-9920, reduced PAC-1 and fibrinogen binding to integrin alpha(IIb)beta3 and restricted platelet spreading on immobilized fibrinogen. Both inhibitors impaired aggregation mediated by ADP, epinephrine, collagen or thrombin, but not arachidonic acid. ATP release, TXB2 formation, P-selectin expression, ERK phosphorylation and cPLA2 activity stimulated by thrombin were reduced in BAY 11-7082- or Ro 106-9920-treated platelets. Although bleeding time was not affected, ADP-induced platelet aggregation was impaired in mice treated with BAY 11-7082. CONCLUSIONS: Our results suggest that NF-kappaB may be a novel mediator of platelet responses. The blockade of platelet function by NF-kappaB inhibitors might be relevant in those clinical situations where these drugs are being considered for anti-tumor and/or anti-inflammatory therapy.


Subject(s)
NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Platelet Activation , Animals , Cells, Cultured , Dual Specificity Phosphatase 2 , Fibrinogen/metabolism , Humans , I-kappa B Proteins/metabolism , Mice , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation , Platelet Adhesiveness , Protein Binding
13.
Curr Med Chem ; 16(4): 417-29, 2009.
Article in English | MEDLINE | ID: mdl-19199914

ABSTRACT

Nitric oxide (NO) is a diffusible, short-lived, diatomic free radical ubiquitously produced by mammalian cells. The generation of NO from L-arginine is enzymatically regulated by three different isoforms of NO synthases. The NO signaling pathway involves mainly the activation of soluble guanylyl cyclase to produce cyclic GMP (cGMP) as a second messenger and downstream mediator. In addition, the free radical activity of NO can cause cellular damage through a phenomenon known as nitrosative stress. NO is a pleiotropic biomodulator in several systems, including the cardiovascular, nervous and immune systems. In the hematopoietic system, NO is thought to be an autocrine or paracrine messenger but also an intracellular effector molecule. Megakaryopoiesis and subsequent thrombopoiesis occur through complex biologic steps that involve hematopoietic stem cell commitment to megakaryocytic lineage, megakaryocyte maturation and finally, platelet release. Here, we summarize the current knowledge regarding the role of exogenous and endogenous NO in hematopoietic stem cell biology, megakaryocyte development and platelet biogenesis as well as relevance of platelet-derived NO generation on platelet function. Dysregulation of NO synthesis has been observed in several diseases, and the evaluation of a series of pharmacological agents with the ability to modulate the NO/cGMP pathway in platelets will also be discussed.


Subject(s)
Blood Platelets/physiology , Hematopoietic Stem Cells/physiology , Nitric Oxide/physiology , Animals , Humans , Nitric Oxide/blood , Nitric Oxide Synthase/metabolism
15.
Biochem. biophys. res. commun ; 333(4): 1341-1347, 2005.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP, SESSP-IBACERVO | ID: biblio-1060801

ABSTRACT

The HlyX, a putative hemolysin identified from the Leptospira genomes, was cloned, expressed in Escherichia coli, purified, and its hemolytic activity was confirmed. Mouse polyclonal antiserum against the recombinant HlyX recognized HlyX-related antigens in a panel of Leptospira species extracts and it was also able to abolish the hemolytic activity of HlyX. A mixture of HlyX and LipL32, a known hemolysin from Leptospira, induced hemolysis in a synergistic way that was fully inhibited by antiserum against either protein. Moreover, sera from patients with leptospirosis also recognized the recombinant HlyX, showing that it is presented to the host immune system during Leptospira infection.


Subject(s)
Female , Humans , Animals , Mice , Escherichia coli/genetics , Escherichia coli/metabolism , Leptospira interrogans/classification , Leptospira interrogans/metabolism , Escherichia coli Proteins/metabolism , Hemolysis , Hemolysis/physiology , Hemolysin Proteins/pharmacology , Hemolysin Proteins/chemistry , Recombinant Proteins/analysis , Recombinant Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
17.
Thromb Haemost ; 90(2): p.326-33, 2003.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib10691

Subject(s)
Genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...