Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Res Pract Thromb Haemost ; 7(4): 100184, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37538496

RESUMEN

Background: CD34+ cells, megakaryocytes (MKs), and platelets express toll-like receptors (TLRs) that enable these cells to amplify the host innate immune response. However, the role of TLR7/TLR8 activation in megakaryopoiesis has not yet been investigated. Objectives: We evaluated the effect of coxsackievirus B3 (CVB3) and synthetic TLR7/TLR8 agonists on the development of human MKs and production of platelets. Methods: CD34+ cells from human umbilical cord were inoculated with CVB3 or stimulated with synthetic TLR7/TLR8 agonists and then cultured in the presence of thrombopoietin. Results: CD34+ cells, MK progenitor cells, and mature MKs expressed TLR7 and TLR8, and exposure to CVB3 resulted in productive infection, as determined by the presence of viral infectious particles in culture supernatants. Cell expansion, differentiation into MKs, MK maturation, and platelet biogenesis were significantly reduced in CD34+-infected cultures. The reduction in MK growth was not due to an alteration in cellular proliferation but was accompanied by an increase in cellular apoptosis and pyroptosis. Impairment of MK generation and maturation of viable cells were also associated with decreased expression of transcription factors involved in these processes. These effects were completely abrogated by TLR7 but not TLR8 antagonists and mimicked by TLR7 but not TLR8 agonists. CVB3 infection of CD34+ cells increased the immunophenotype of MKs characterized as CD148+/CD48+ or CD41+/CD53+ cells. Conclusion: These data suggest a novel role of TLR7 in megakaryo/thrombopoiesis that may contribute to a better understanding of the molecular basis underlying thrombocytopenia and the immunologic role of MKs in viral infection processes.

2.
J Thromb Haemost ; 17(12): 2196-2210, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31397069

RESUMEN

BACKGROUND: Platelet Toll-like receptor (TLR)2/4 are key players in amplifying the host immune response; however, their role in human megakaryo/thrombopoiesis has not yet been defined. OBJECTIVES: We evaluated whether Pam3CSK4 or lipopolysaccharide (LPS), TLR2/4 ligands respectively, modulate human megakaryocyte development and platelet production. METHODS: CD34+ cells from human umbilical cord were stimulated with LPS or Pam3CSK4 with or without thrombopoietin (TPO). RESULTS: CD34+ cells and megakaryocytes express TLR2 and TLR4 at both RNA and protein level; however, direct stimulation of CD34+ cells with LPS or Pam3CSK4 had no effect on cell growth. Interestingly, both TLR ligands markedly increased TPO-induced CD34+ cell proliferation, megakaryocyte number and maturity, proplatelet and platelet production when added at day 0. In contrast, this synergism was not observed when TLR agonists were added 7 days after TPO addition. Interleukin-6 (IL-6) release was observed upon CD34+ or megakaryocyte stimulation with LPS or Pam3CSK4 but not with TPO and this effect was potentiated in combination with TPO. The increased proliferation and IL-6 production induced by TPO + LPS or Pam3CSK4 were suppressed by TLR2/4 or IL-6 neutralizing antibodies, as well as by PI3K/AKT and nuclear factor-κB inhibitors. Additionally, increased proplatelet and platelet production were associated with enhanced nuclear translocation of nuclear factor-E2. Finally, the supernatants of CD34+ cells stimulated with TPO+LPS-induced CFU-M colonies. CONCLUSIONS: Our data suggest that the activation of TLR2 and TLR4 in CD34+ cells and megakaryocytes in the presence of TPO may contribute to warrant platelet provision during infection episodes by an autocrine IL-6 loop triggered by PI3K/NF-κB axes.


Asunto(s)
Antígenos CD34/metabolismo , Plaquetas/efectos de los fármacos , Lipopéptidos/farmacología , Lipopolisacáridos/farmacología , Megacariocitos/efectos de los fármacos , Trombopoyesis/efectos de los fármacos , Trombopoyetina/farmacología , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 4/agonistas , Plaquetas/inmunología , Plaquetas/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Interleucina-6/metabolismo , Megacariocitos/inmunología , Megacariocitos/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Transducción de Señal , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
3.
Clin Immunol ; 183: 254-262, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28863968

RESUMEN

NETosis is a host defense mechanism associated with inflammation and tissue damage. Experimental models show that platelets and von Willebrand factor (VWF) are key elements for intravascular NETosis. We determined NETosis in septic and burn patients at 1 and 4days post-admission (dpa). Nucleosomes were elevated in patients. In septics, they correlated with Human Neutrophil Elastase (HNE)-DNA complexes and SOFA score at 1dpa, and were associated with mortality. Patient's neutrophils had spontaneous NETosis and were unresponsive to stimulation. Although platelet P-selectin and TNF-α were increased in both groups, higher platelet TLR-4 expression, VWF levels and IL-6 were found in septics at 1dpa. Neither platelet activation markers nor cytokines correlated with nucleosomes or HNE-DNA. Nucleosomes could be indicators of organ damage and predictors of mortality in septic but not in burn patients. Platelet activation, VWF and cytokines do not appear to be key mediators of NETosis in these patient groups.


Asunto(s)
Quemaduras/metabolismo , Trampas Extracelulares/fisiología , Nucleosomas/metabolismo , Sepsis/metabolismo , Adulto , Anciano , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación , Modelos Lineales , Masculino , Persona de Mediana Edad , Activación Plaquetaria , Factor de von Willebrand
4.
Front Biosci (Landmark Ed) ; 22(11): 1867-1883, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28410150

RESUMEN

Besides their undiscussed role in hemostasis and thrombosis, platelets are also key effector cells capable of assisting and modulating inflammatory reactions and immune responses. Platelets play a sentinel role in immune surveillance by recognizing danger signals from pathogens and cell damage through the expression of toll-like receptors (TLRs) on its surface and internal compartments. Platelets express all 10 TLRs transcripts and its signalosome, including adaptor proteins and transcription factors. The activation of these receptors in platelets triggers hemostatic and inflammatory responses which participate in the host's response to bacterial and viral infections, linking thrombosis with infection and immunity. Among the responses elicited by the activation of platelet TLRs are platelet adhesion and aggregation, formation of mixed platelet-leukocyte aggregates, expression and secretion of cytokines and quemokines, and thrombin generation. TLRs also are expressed in megakaryocytes, and their activation regulates not only platelet biogenesis, but also pro-inflammatory and antiviral responses. This review will focus on work that has shown the role of platelet TLRs, mainly in thromboinflammatory responses elicited by platelets and megakaryocytes.


Asunto(s)
Plaquetas/inmunología , Hemostasis/inmunología , Inflamación/inmunología , Trombosis/inmunología , Receptores Toll-Like/inmunología , Plaquetas/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Inflamación/metabolismo , Megacariocitos/inmunología , Megacariocitos/metabolismo , Activación Plaquetaria/inmunología , Transducción de Señal/inmunología , Trombosis/metabolismo , Receptores Toll-Like/metabolismo
5.
J Leukoc Biol ; 99(1): 153-62, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26320263

RESUMEN

In addition to being key elements in hemostasis and thrombosis, platelets amplify neutrophil function. We aimed to gain further insight into the stimuli, mediators, molecular pathways, and regulation of neutrophil extracellular trap formation mediated by human platelets. Platelets stimulated by lipopolysaccharide, a wall component of gram-negative bacteria, Pam3-cysteine-serine-lysine 4, a mimetic of lipopeptide from gram-positive bacteria, Escherichia coli, Staphylococcus aureus, or physiologic platelet agonists promoting neutrophil extracellular trap formation and myeloperoxidase-associated DNA activity under static and flow conditions. Although P-selectin or glycoprotein IIb/IIIa were not involved, platelet glycoprotein Ib, neutrophil cluster of differentiation 18, and the release of von Willebrand factor and platelet factor 4 seemed to be critical for the formation of neutrophil extracellular traps. The secretion of these molecules depended on thromboxane A(2) production triggered by lipopolysaccharide or Pam3-cysteine-serine-lysine 4 but not on high concentrations of thrombin. Accordingly, aspirin selectively inhibited platelet-mediated neutrophil extracellular trap generation. Signaling through extracellular signal-regulated kinase, phosphatidylinositol 3-kinase, and Src kinases, but not p38 or reduced nicotinamide adenine dinucleotide phosphate oxidase, was involved in platelet-triggered neutrophil extracellular trap release. Platelet-mediated neutrophil extracellular trap formation was inhibited by prostacyclin. Our results support a role for stimulated platelets in promoting neutrophil extracellular trap formation, reveal that an endothelium-derived molecule contributes to limiting neutrophil extracellular trap formation, and highlight platelet inhibition as a potential target for controlling neutrophil extracellular trap cell death.


Asunto(s)
Plaquetas/metabolismo , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Activación Plaquetaria , Transducción de Señal , Células Endoteliales/metabolismo , Humanos , Lipopéptidos/inmunología , Lipopolisacáridos/inmunología , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/inmunología , Receptores de Superficie Celular/metabolismo
6.
Thromb Haemost ; 107(1): 99-110, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22159527

RESUMEN

Acidosis is one of the hallmarks of tissue injury such as trauma, infection, inflammation, and tumour growth. Although platelets participate in the pathophysiology of all these processes, the impact of acidosis on platelet biology has not been studied outside of the quality control of laboratory aggregation assays or platelet transfusion optimization. Herein, we evaluate the effect of physiologically relevant changes in extracellular acidosis on the biological function of platelets, placing particular emphasis on haemostatic and secretory functions. Platelet haemostatic responses such as adhesion, spreading, activation of αIIbß3 integrin, ATP release, aggregation, thromboxane B2 generation, clot retraction and procoagulant activity including phosphatidilserine exposure and microparticle formation, showed a statistically significant inhibition of thrombin-induced changes at pH of 7.0 and 6.5 compared to the physiological pH (7.4). The release of alpha granule content was differentially regulated by acidosis. At low pH, thrombin or collagen-induced secretion of vascular endothelial growth factor and endostatin were dramatically reduced. The release of von Willebrand factor and stromal derived factor-1α followed a similar, albeit less dramatic pattern. In contrast, the induction of CD40L was not changed by low pH, and P-selectin exposure was significantly increased. While the generation of mixed platelet-leukocyte aggregates and the increased chemotaxis of neutrophils mediated by platelets were further augmented under acidic conditions in a P-selectin dependent manner, the increased neutrophil survival was independent of P-selectin expression. In conclusion, our results indicate that extracellular acidosis downregulates most of the haemostatic platelet functions, and promotes those involved in amplifying the neutrophil-mediated inflammatory response.


Asunto(s)
Acidosis/metabolismo , Plaquetas/metabolismo , Neutrófilos/citología , Adenosina Trifosfato/metabolismo , Coagulación Sanguínea , Quimiocina CXCL12/metabolismo , Endostatinas/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Hemostasis , Humanos , Inflamación , Microscopía Fluorescente/métodos , Fosfatidilserinas/química , Fosforilación , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Transfusión de Plaquetas , Tromboxano B2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de von Willebrand/metabolismo
7.
Exp Hematol ; 39(7): 763-72, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21549176

RESUMEN

OBJECTIVE: Megakaryo/thrombopoiesis is a complex process regulated by multiple signals provided by the bone marrow microenvironment. Because macrophages are relevant components of the bone marrow stroma and their activation induces an upregulation of molecules that can regulate hematopoiesis, we analyzed the impact of these cells on the control of megakaryocyte development and platelet biogenesis. MATERIALS AND METHODS: The different stages of megakaryo/thrombopoiesis were analyzed by flow cytometry using an in vitro model of human cord blood CD34(+) cells stimulated with thrombopoietin in either a transwell system or conditioned media from monocyte-derived macrophages isolated from peripheral blood. Cytokines secreted from macrophages were characterized by protein array and enzyme-linked immunosorbent assay. RESULTS: Resting macrophages released soluble factors that promoted megakaryocyte growth, cell ploidy, a size increase, proplatelet production, and platelet release. Lipopolysaccharide stimulation triggered the secretion of cytokines that exerted opposite effects together with a dramatic switch of CD34(+) commitment to the megakaryocytic lineage toward the myeloid lineage. Neutralization of interleukin-8 released by stimulated macrophages partially reversed the inhibition of megakaryocyte growth. Activation of nuclear factor κB had a major role in the synthesis of molecules involved in the megakaryocyte inhibition mediated by lipopolysaccharide-stimulated macrophages. CONCLUSIONS: Our study extends our understanding about the role of the bone marrow microenvironment in the regulation of megakaryo/thrombopoiesis by showing that soluble factors derived from macrophages positively or negatively control megakaryocyte growth, differentiation, maturation, and their ability to produce platelets.


Asunto(s)
Citocinas/farmacología , Macrófagos/metabolismo , Comunicación Paracrina , Trombopoyesis/efectos de los fármacos , Antígenos CD34/metabolismo , Plaquetas/citología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Células Cultivadas , Quimiocinas/farmacología , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Ensayo de Inmunoadsorción Enzimática , Femenino , Sangre Fetal/citología , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Lipopolisacáridos/farmacología , Macrófagos/citología , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , FN-kappa B/metabolismo , Trombopoyetina/farmacología
8.
Transfusion ; 51(8): 1784-95, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21332728

RESUMEN

BACKGROUND: Transplanted hematopoietic progenitor cells (CD34+) have shown great promise in regenerative medicine. However, the therapeutic potential of transplanted cells is limited by their poor viability. It is well known that the microenvironment in which progenitors reside substantially affects their behavior. Because extracellular acidosis is a common feature of injured tissues or the tumor microenvironment and is a critical regulator of cell survival and activation, we evaluated the impact of acidosis on CD34+ cell biology. STUDY DESIGN AND METHODS: Apoptosis was evaluated by fluorescence microscopy and binding of annexin V, hypodiploid cells, Bcl-xL expression, active caspase-3, and mitochondrial membrane potential was determined by flow cytometry. Colony-forming units were studied by clonogenic assays, and cell cycle was evaluated by flow cytometry. RESULTS: Exposure of CD34+ cells to low pH (7.0-6.5) caused intracellular acidification, decreased cell proliferation, and triggered apoptosis via the mitochondrial pathway. Whereas exposure to thrombopoietin (TPO), stem cell factor (SCF), interleukin (IL)-3 or increases in cyclic adenosine monophosphate (cAMP) levels prevented CD34+ cell death induced by acidic conditions, granulocyte-macrophage-colony-stimulating factor, FMS-like tyrosine kinase 3-ligand, erythropoietin, and vascular endothelial growth factor had no effect. Despite their cytoprotective effect, CD34+ cell expansion triggered by TPO, SCF, or IL-3 was significantly impaired at low pH. However, a cocktail of these three cytokines synergistically supported proliferation, cell cycle progression, and colony formation. DISCUSSION: Our findings indicate that an acidic milieu is deleterious for CD34+ cells and that a combination of certain cytokines and cAMP donors may improve cell viability and function. These data may be useful to develop new therapeutic strategies or to optimize protocols for regenerative medicine.


Asunto(s)
Ácidos/farmacología , Antígenos CD34/metabolismo , AMP Cíclico/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Interleucina-3/farmacología , Factor de Células Madre/farmacología , Trombopoyetina/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Sinergismo Farmacológico , Sangre Fetal/citología , Sangre Fetal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Humanos , Concentración de Iones de Hidrógeno , Regulación hacia Arriba/efectos de los fármacos
9.
Exp Hematol ; 34(10): 1420-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16982335

RESUMEN

OBJECTIVE: Although cAMP is involved in a number of physiologic functions, its role in hematopoietic cell fate decision remains poorly understood. We have recently demonstrated that in CD34(+)-derived megakaryocytes, cAMP-related agents prevent apoptosis. In this study we addressed the question of whether cAMP also regulates survival of their precursors, CD34(+) cells. METHODS: Apoptosis was evaluated by fluorescence microscopy, and detection of hypodiploid or annexin V(+) cells by flow cytometry. Mitochondrial membrane potential and bcl-xL or caspase-3 expression were assessed by flow cytometry. Colony-forming units were studied by clonogenic assays in methylcellulose. RESULTS: We found that two different cAMP analogs such as Dibutiril-cAMP and sp-5,6-DCl-BIMPS (BIMPS) promoted survival of human umbilical cord-derived CD34(+) cells by suppressing apoptosis induced by either nitric oxide (NO) or serum deprivation. Involvement of PKA and PI3K pathway was demonstrated by the ability of their specific inhibitors Rp-cAMP and Wortmannin or LY294002 respectively to reverse the antiapoptotic effect of BIMPS. Treatment of CD34(+) cell with BIMPS not only restrained the bcl-xL downregulation but also suppressed the loss of mitochondrial membrane potential and caspase-3 activation induced by serum starvation. While thrombopoietin (TPO), granulocyte colony-stimulating factor (G-CSF) or stem cell factor (SCF) were not able to increase cAMP levels, the antiapoptotic activity exerted by these growth factors was blocked by inhibition of the adenylate cyclase and synergized by BIMPS. Cyclic AMP analogs suppressed the decreased colony formation in cells exposed to NO or serum deprivation. CONCLUSION: Altogether, our results strongly suggest that cAMP appears to be not only a key pathway controlling CD34(+) survival, but also a mediator of the TPO-, G-CSF- and SCF-mediated cytoprotection.


Asunto(s)
Antígenos CD34 , Apoptosis/efectos de los fármacos , Bucladesina/farmacología , Diclororribofuranosil Benzoimidazol/análogos & derivados , Células Madre Hematopoyéticas/metabolismo , Megacariocitos/metabolismo , Transducción de Señal/efectos de los fármacos , Tionucleótidos/farmacología , Bucladesina/metabolismo , Caspasa 3 , Caspasas/metabolismo , Células Cultivadas , Cromonas/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diclororribofuranosil Benzoimidazol/farmacología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Inhibidores Enzimáticos/farmacología , Sangre Fetal/citología , Sangre Fetal/metabolismo , Sustancias de Crecimiento/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Megacariocitos/citología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Mitocondrias/metabolismo , Morfolinas/farmacología , Óxido Nítrico/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Proteína bcl-X/biosíntesis
10.
J Pharmacol Exp Ther ; 319(2): 972-9, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16936242

RESUMEN

During inflammation, polymorphonuclear leukocyte (PMN) apoptosis can be delayed by different proinflammatory mediators. Classically, it has been accepted that the widely used anti-inflammatory drug acetyl salicylic acid (ASA) exerts its action through inhibition of cyclooxygenases and subsequent prostaglandin synthesis. We hypothesized that another anti-inflammatory action of ASA could be the shortening of PMN survival. We found that at therapeutic concentrations (1-3 mM), ASA and its metabolite salicylate (NaSal), but not indomethacin or ibuprofen, counteracted the prolonged PMN survival mediated by lipopolysaccharide (LPS) through inhibition of nuclear factor-kappaB (NF-kappaB) activation. Both salicylates also inhibited interleukin (IL)-1alpha or acidic conditions antiapoptotic activity. Higher concentrations of both drugs had a direct apoptotic effect. Salicylates were not effective when PMN apoptosis delay was induced by granulocyte macrophage-colony-stimulating factor (GM-CSF), a NF-kappaB-independent cytokine. Promotion of PMN survival by the combination of IL-1alpha and LPS was also reversed by salicylates, but higher concentrations were required. ASA concentrations that did not trigger PMN death increase the zymosan- or tumor necrosis factor-alpha-mediated proapoptotic effect. The LPS- and IL-1alpha- but not GM-CSF-mediated antiapoptotic effect was markedly reduced in PMNs from donors who had ingested ASA. Using a thioglycolate-induced peritonitis model, we showed that in ASA- or NaSal-treated mice there was not only a decrease in the number of cells recruited but also an increase in the percentage of apoptotic PMNs as well as an enhancement of phagocytosis compared with controls. Our findings demonstrate that acceleration of PMN apoptosis by turning off the NF-kappaB-mediated survival signals elicited by proinflammatory stimuli is another anti-inflammatory action of ASA and NaSal.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Aspirina/farmacología , Neutrófilos/efectos de los fármacos , Salicilato de Sodio/farmacología , Células Cultivadas , Ciclooxigenasa 2/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interleucina-1/farmacología , Lipopolisacáridos/farmacología , FN-kappa B/antagonistas & inhibidores
11.
Br J Pharmacol ; 145(3): 283-92, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15778737

RESUMEN

1 We have previously demonstrated that nitric oxide (NO) triggers CD34(+)-derived megakaryocyte apoptosis. We here show that prostacyclin (PGI(2)) inhibits PAPA/NO-induced megakaryocyte death detected by fluorescent microscopy and flow cytometry. 2 The cAMP-specific phosphodiesterase inhibitor, Ro 20-1724, and the permeable analog dibutyryl-cAMP also delayed apoptosis. PGI(2) effect was fully prevented when adenylyl cyclase activity was suppressed by SQ 22536, and partially reversed by the permeable protein kinase A inhibitor PKI 14-22 amide. ELISA showed that while both PGI(2) and NO alone or synergistically raised cAMP, only NO was able to increase intracellular cGMP levels. 3 Treatment of megakaryocytes with PGI(2) abolished both basal and NO-raised cGMP levels. Addition of 8-pCPT-cGMP or activation of soluble guanylyl cyclase by BAY 41-2272 induced cell death in a concentration-dependent manner, and ODQ, an inhibitor of guanylyl cyclase, prevented both PAPA/NO- or BAY 41-2272-induced apoptosis. Specific cGMP phosphodiesterase inhibition by Zaprinast or suppression of adenylyl cyclase by SQ 22536 enhanced the PAPA/NO proapoptotic effect. 4 PGI(2) completely inhibited NO-mediated generation and the increased activity of the cleaved form of caspase-3. 5 In conclusion, our results demonstrate that contrary to their well-known direct and synergistic inhibitory effects on platelets, PGI(2) and NO regulate opposite megakaryocyte survival responses through a delicate balance between intracellular cyclic nucleotide levels and caspase-3 activity control.


Asunto(s)
Apoptosis/efectos de los fármacos , Epoprostenol/farmacología , Megacariocitos/efectos de los fármacos , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/farmacología , Apoptosis/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Humanos , Megacariocitos/metabolismo
12.
Thromb Haemost ; 90(2): 326-33, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12888881

RESUMEN

Hematologic involvement is the main feature of Argentine hemorrhagic fever (AHF), an endemo-epidemic disease caused by Junin virus (JV). Since endothelial dysfunction could play a role in AHF-altered hemostasis, we studied human umbilical vein endothelial cell (HUVEC) infection with a virulent (JVv) and a non-virulent (JVa) JV strain. Cells were infected by the two JV variants with no detectable apoptosis or cytopathic effect. Both viral variants up-regulated ICAM-1 and VCAM-1 levels, while von Willebrand factor (VWF) production was decreased. Prostacyclin (PGI2) release and decay accelerating factor (DAF) expression were greater in JVv- than in JVa-infected or control cells. Furthermore, nitric oxide (NO) production and endothelial nitric oxide synthase (eNOS) expression was only raised in JVv-infected supernatants. Significant NO and PGI2 values were also detected in AHF patient sera. These data demonstrate that endothelial cell responses are triggered subsequently by JV infection, suggesting that such alterations play a major role in the pathogenesis of AHF and perhaps in other viral-induced hemorrhagic diseases.


Asunto(s)
Infecciones por Arenaviridae/complicaciones , Infecciones por Arenaviridae/fisiopatología , Endotelio Vascular/fisiopatología , Fiebres Hemorrágicas Virales/fisiopatología , Fiebres Hemorrágicas Virales/virología , Virus Junin , Apoptosis , Infecciones por Arenaviridae/sangre , Antígenos CD55/metabolismo , Células Cultivadas , Epoprostenol/metabolismo , Humanos , Óxido Nítrico/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de von Willebrand/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...