Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Ther ; 25(3): 593-605, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28190779

RESUMEN

Recently, an engineered Homeobox-nucleoporin fusion gene, NUP98-HOXA10HD or NA10HD, was reported to expand and maintain murine hematopoietic stem cells (HSCs). We postulated that NA10HD would increase the number of human γ-globin-expressing cells to therapeutic levels. We developed a double gene lentiviral vector encoding both human γ-globin and NA10HD, which was used to transduce human peripheral blood CD34+ cells and increased engraftment 2- to 2.5-fold at 15 weeks post-transplantation in immunodeficient mice. In ß-thalassemic mice transplanted with ß-thalassemic HSCs transduced with the γ-globin/NA10HD vector, the number of fetal hemoglobin (HbF)-expressing cells was significantly increased after 3 months, leading to resolution of the anemia. Furthermore, the increases in HbF were maintained at 6 months and persisted after secondary transplantation. In addition, NA10HD enrichment of transduced HSCs led to HbF increases without affecting homeostasis of the white blood cell lineages. Our results suggest that NA10HD increases the number of γ-globin-transduced HSCs that engraft, leading to an elevated number of fetal hemoglobin-containing red cells. These effects of NA10HD provide an improved platform for testing of the therapeutic efficacy of novel globin vectors and provide further impetus to develop safe and effective methods for selective expansion of genetically modified cells.


Asunto(s)
Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Lentivirus/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Talasemia beta/genética , gamma-Globinas/genética , Animales , Modelos Animales de Enfermedad , Eritrocitos/citología , Eritrocitos/metabolismo , Hemoglobina Fetal/metabolismo , Orden Génico , Técnicas de Transferencia de Gen , Sitios Genéticos , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Proteínas Homeobox A10 , Humanos , Ratones , Transducción Genética , Trasplante Heterólogo , Talasemia beta/metabolismo , Talasemia beta/terapia
2.
Blood ; 126(20): 2307-19, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26405223

RESUMEN

Controlling the activation of platelets is a key strategy to mitigate cardiovascular disease. Previous studies have suggested that the ATP-binding cassette (ABC) transporter, ABCC4, functions in platelet-dense granules. Using plasma membrane biotinylation and super-resolution microscopy, we demonstrate that ABCC4 is primarily expressed on the plasma membrane of both mouse and human platelets. Platelets lacking ABCC4 have unchanged dense-granule function, number, and volume, but harbor a selective impairment in collagen-induced aggregation. Accordingly, Abcc4 knockout (KO) platelet attachment to a collagen substratum was also faulty and associated with elevated intracellular cyclic AMP (cAMP) and reduced plasma membrane localization of the major collagen receptor, GPVI. In the ferric-chloride vasculature injury model, Abcc4 KO mice exhibited markedly impaired thrombus formation. The attenuation of platelet aggregation by the phosphodiesterase inhibitor EHNA (a non-ABCC4 substrate), when combined with Abcc4 deficiency, illustrated a crucial functional interaction between phosphodiesterases and ABCC4. This was extended in vivo where EHNA dramatically prolonged the bleeding time, but only in Abcc4 KO mice. Further, we demonstrated in human platelets that ABCC4 inhibition, when coupled with phosphodiesterase inhibition, strongly impaired platelet aggregation. These findings have important clinical implications because they directly highlight an important relationship between ABCC4 transporter function and phosphodiesterases in accounting for the cAMP-directed activity of antithrombotic agents.


Asunto(s)
Plaquetas/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Agregación Plaquetaria , Adenina/análogos & derivados , Adenina/farmacología , Animales , Plaquetas/patología , AMP Cíclico/genética , AMP Cíclico/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Trombosis/genética , Trombosis/metabolismo , Trombosis/patología
3.
Blood ; 119(8): 1915-21, 2012 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-22130804

RESUMEN

Sickle cell anemia is characterized by chronic hemolysis coupled with extensive vascular inflammation. This inflammatory state also mechanistically promotes a high risk of lethal, invasive pneumococcal infection. Current treatments to reduce vaso-occlusive complications include chronic hydroxyurea therapy to induce fetal hemoglobin. Because hydroxyurea also reduces leukocytosis, an understanding of the impact of this treatment on pneumococcal pathogenesis is needed. Using a sickle cell mouse model of pneumococcal pneumonia and sepsis, administration of hydroxyurea was found to significantly improve survival. Hydroxyurea treatment decreased neutrophil extravasation into the infected lung coincident with significantly reduced levels of E-selectin in serum and on pulmonary epithelia. The protective effect of hydroxyurea was abrogated in mice deficient in E-selectin. The decrease in E-selectin levels was also evident in human sickle cell patients receiving hydroxyurea therapy. These data indicate that in addition to induction of fetal hemoglobin, hydroxyurea attenuates leukocyte-endothelial interactions in sickle cell anemia, resulting in protection against lethal pneumococcal sepsis.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Selectina E/metabolismo , Hidroxiurea/uso terapéutico , Neumonía Neumocócica/prevención & control , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/metabolismo , Animales , Antidrepanocíticos/uso terapéutico , Niño , Modelos Animales de Enfermedad , Selectina E/sangre , Selectina E/genética , Femenino , Humanos , Inmunohistoquímica , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Neumonía Neumocócica/complicaciones , Análisis de Supervivencia
4.
Cancer Cell ; 2(4): 279-88, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12398892

RESUMEN

We show here that a zinc finger transcriptional repressor, Slug, which is aberrantly upregulated by the E2A-HLF oncoprotein in pro-B cell acute leukemia, functions as an antiapoptotic factor in normal hematopoietic progenitor cells. Slug(-/-) mice were much more radiosensitive than wild-type mice, dying earlier and showing accentuated decreases in peripheral blood cell counts, as well as abundant microhemorrhages and widely disseminated bacterial microabscesses throughout the body. Slug expression was detected in diverse subsets of hematopoietic progenitors, but not in more differentiated B and T lymphoid cells, and there was a significant increase in apoptotic (TUNEL-positive) bone marrow progenitor cells in irradiated Slug(-/-) mice compared to wild-type controls. These results implicate Slug in a novel survival pathway that protects hematopoietic progenitors from apoptosis after DNA damage.


Asunto(s)
Apoptosis/efectos de la radiación , Células Madre Hematopoyéticas/citología , Factores de Transcripción/fisiología , Dedos de Zinc/fisiología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Recuento de Células Sanguíneas , Plaquetas/metabolismo , Médula Ósea/metabolismo , Linaje de la Célula , Transformación Celular Neoplásica , Citoprotección , Daño del ADN , Cartilla de ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Rayos gamma , Regulación Neoplásica de la Expresión Génica , Hematopoyesis/fisiología , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/efectos de la radiación , Hemoglobinas/metabolismo , Homocigoto , Etiquetado Corte-Fin in Situ , Leucemia de Células B/genética , Leucemia de Células B/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Reacción en Cadena de la Polimerasa , Recombinación Genética , Factores de Transcripción de la Familia Snail , Bazo/metabolismo , Tasa de Supervivencia , Timo/efectos de la radiación , Proteína p53 Supresora de Tumor/metabolismo , Irradiación Corporal Total
5.
Blood ; 113(23): 5747-56, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19365082

RESUMEN

Correction of murine models of beta-thalassemia has been achieved through high-level globin lentiviral vector gene transfer into mouse hematopoietic stem cells (HSCs). However, transduction of human HSCs is less robust and may be inadequate to achieve therapeutic levels of genetically modified erythroid cells. We therefore developed a double gene lentiviral vector encoding both human gamma-globin under the transcriptional control of erythroid regulatory elements and methylguanine methyltransferase (MGMT), driven by a constitutive cellular promoter. MGMT expression provides cellular resistance to alkylator drugs, which can be administered to kill residual untransduced, diseased HSCs, whereas transduced cells are protected. Mice transplanted with beta-thalassemic HSCs transduced with a gamma-globin/MGMT vector initially had subtherapeutic levels of red cells expressing gamma-globin. To enrich gamma-globin-expressing cells, transplanted mice were treated with the alkylator agent 1,3-bis-chloroethyl-1-nitrosourea. This resulted in significant increases in the number of gamma-globin-expressing red cells and the amount of fetal hemoglobin, leading to resolution of anemia. Selection of transduced HSCs was also obtained when cells were drug-treated before transplantation. Mice that received these cells demonstrated reconstitution with therapeutic levels of gamma-globin-expressing cells. These data suggest that MGMT-based drug selection holds promise as a modality to improve gene therapy for beta-thalassemia.


Asunto(s)
Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Lentivirus/genética , Proteínas Supresoras de Tumor/metabolismo , Talasemia beta/metabolismo , gamma-Globinas/metabolismo , Animales , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Resistencia a Medicamentos , Eritrocitos/metabolismo , Femenino , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Masculino , Ratones , Proteínas Supresoras de Tumor/genética , Talasemia beta/genética , Talasemia beta/patología , Talasemia beta/terapia , gamma-Globinas/genética
6.
Haematologica ; 95(9): 1599-603, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20378564

RESUMEN

Hydroxyurea has proven clinical efficacy in patients with sickle cell disease. Potential mechanisms for the beneficial effects include fetal hemoglobin induction and the reduction of cell adhesive properties, inflammation and hypercoagulability. Using a murine model of sickle cell disease in which fetal hemoglobin induction does not occur, we evaluated whether hydroxyurea administration would still yield improvements in hematologic parameters and reduce end-organ damage. Animals given a maximally tolerated dose of hydroxyurea that resulted in significant reductions in the neutrophil and platelet counts showed no improvement in hemolytic anemia and end-organ damage compared to control mice. In contrast, animals having high levels of fetal hemoglobin due to gene transfer with a gamma-globin lentiviral vector showed correction of anemia and organ damage. These data suggest that induction of fetal hemoglobin by hydroxyurea is an essential mechanism for its clinical benefits.


Asunto(s)
Anemia de Células Falciformes/terapia , Hemoglobina Fetal/administración & dosificación , Terapia Genética/métodos , Hidroxiurea/uso terapéutico , Animales , Recuento de Células Sanguíneas , Modelos Animales de Enfermedad , Hemoglobina Fetal/genética , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento
7.
Mol Ther ; 17(2): 245-52, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19050697

RESUMEN

Increased levels of red cell fetal hemogloblin, whether due to hereditary persistence of expression or from induction with hydroxyurea therapy, effectively ameliorate sickle cell disease (SCD). Therefore, we developed erythroid-specific, gamma-globin lentiviral vectors for hematopoietic stem cell (HSC)-targeted gene therapy with the goal of permanently increasing fetal hemoglobin (HbF) production in sickle red cells. We evaluated two different gamma-globin lentiviral vectors for therapeutic efficacy in the BERK sickle cell mouse model. The first vector, V5, contained the gamma-globin gene driven by 3.1 kb of beta-globin regulatory sequences and a 130-bp beta-globin promoter. The second vector, V5m3, was identical except that the gamma-globin 3'-untranslated region (3'-UTR) was replaced with the beta-globin 3'-UTR. Adult erythroid cells have beta-globin mRNA 3'-UTR-binding proteins that enhance beta-globin mRNA stability and we postulated this design might enhance gamma-globin expression. Stem cell gene transfer was efficient and nearly all red cells in transplanted mice expressed human gamma-globin. Both vectors demonstrated efficacy in disease correction, with the V5m3 vector producing a higher level of gamma-globin mRNA which was associated with high-level correction of anemia and secondary organ pathology. These data support the rationale for a gene therapy approach to SCD by permanently enhancing HbF using a gamma-globin lentiviral vector.


Asunto(s)
Anemia de Células Falciformes/terapia , Hemoglobina Fetal/fisiología , Terapia Genética/métodos , Vectores Genéticos/genética , Lentivirus/genética , gamma-Globinas/genética , Animales , Southern Blotting , Células Cultivadas , Electroforesis , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Citometría de Flujo , Trasplante de Células Madre Hematopoyéticas , Ratones , Modelos Genéticos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/metabolismo , Bazo/patología
8.
Sci Rep ; 7: 43308, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28256526

RESUMEN

Sickle cell disease (SCD) is a major global health concern. Patients with SCD experience disproportionately greater morbidity and mortality in response to influenza infection than do others. Viral infection is one contributing factor for the development of Acute Chest Syndrome (ACS), a major cause of morbidity and mortality in SCD patients. We determined whether the heightened sensitivity to influenza infection could be reproduced in the two different SCD murine models to ascertain the underlying mechanisms of increased disease severity. In agreement with clinical observations, we found that both genetic and bone marrow-transplanted SCD mice had greater mortality in response to influenza infection than did wild-type animals. Despite similar initial viral titers and inflammatory responses between wild-type and SCD animals during infection, SCD mice continued to deteriorate and failed to resolve the infection, resulting in increased mortality. Histopathology of the lung tissues revealed extensive pulmonary edema and vascular damage following infection, a finding confirmed by heightened vascular permeability following virus challenge. These findings implicate the development of exacerbated pulmonary permeability following influenza challenge as the primary factor underlying heightened mortality. These studies highlight the need to focus on prevention and control strategies against influenza infection in the SCD population.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Permeabilidad Capilar , Susceptibilidad a Enfermedades , Gripe Humana/inmunología , Animales , Modelos Animales de Enfermedad , Histocitoquímica , Humanos , Pulmón/patología , Ratones , Análisis de Supervivencia
9.
Exp Hematol ; 31(12): 1198-205, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14662325

RESUMEN

OBJECTIVE: To determine the role of the c-terminal half of c-Mpl in Mpl-L-induced myeloprotection and the importance of Stat5 isoforms in the survival signaling pathways induced by Mpl ligand. MATERIALS AND METHODS: Delta60-Mpl knockin mice, Stat5a(-/-)/b(-/-), Stat5a(-/-), and Stat5b(-/-) mice and wild-type (WT) controls were given a lethal myelosuppressive regimen: 80 mg/kg carboplatin intravenously followed by 7.5 or 6.5 Gy 137Cs total-body irradiation. A single dose of PEG-rmMGDF (65 microg/kg) was intravenously injected immediately after myelosuppression. Mice survival and blood counts were monitored for 22 days posttreatment. RESULTS: Knockin Delta60-Mpl mice lacking the c-terminal half of the intracellular domain of c-Mpl show reduced ability of Mpl-L to prevent lethal myelosuppression and an impaired thrombopoietic response to exogenous c-Mpl ligand. The survival of Mpl-L-treated Stat5a(-/-)/b(-/-) mice exposed to the lethal myelosuppressive regimen was substantially compromised compared to that of WT mice. Reduced survival of Stat5a(-/-)/b(-/-) mice was due to more severe hematopoietic suppression. Deletion of Stat5a did not result in a defect in hematopoietic recovery. In contrast, Mpl-L-treated Stat5b-deficient mice demonstrated significantly delayed hematopoietic recovery compared to WT controls. CONCLUSIONS: Myeloprotective signaling transduced by the terminal 60 amino acids of the intracellular domain of c-Mpl is essential for complete protection from lethal myelosuppression provided by Mpl-L. Our studies differentiate the functions of Stat5 isoforms in hematopoietic stress and reveal a pivotal role of Stat5b in Mpl-L-induced hematopoietic recovery in this lethal myelosuppression model.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Hematopoyesis , Proteínas de la Leche , Agonistas Mieloablativos/farmacología , Isoformas de Proteínas/fisiología , Trombopoyetina/farmacología , Transactivadores/fisiología , Animales , Proteínas de Unión al ADN/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Agonistas Mieloablativos/administración & dosificación , Proteínas Oncogénicas/genética , Isoformas de Proteínas/genética , Receptores de Citocinas/genética , Receptores de Trombopoyetina , Factor de Transcripción STAT5 , Eliminación de Secuencia , Transducción de Señal , Tasa de Supervivencia , Trombopoyetina/administración & dosificación , Transactivadores/genética
10.
Exp Hematol ; 43(7): 565-77.e1-10, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25907033

RESUMEN

The transcription factor GATA2 is highly expressed in hematopoietic stem cells and is downregulated during lineage maturation. Gain of function mutations, loss of function mutations, and overexpression of GATA2 have been reported in acute myeloid leukemia. In previous studies, we and others showed that GATA2 overexpression at high levels, similar to that seen in hematopoietic stem cells, blocked differentiation of hematopoietic stem cells and progenitors. To better understand the effects of GATA2, we designed a Tamoxifen-inducible GATA2-estrogen receptor (ERT) vector. In the absence of Tamoxifen, small amounts of GATA2-ERT were still able to enter the nucleus in mouse bone marrow (BM) cells, providing us with a tool to test the effects of low-level GATA2 overexpression. We observed that this low-level GATA2 overexpression enhanced self-renewal of myeloid progenitors in vitro and resulted in immortalization of BM cells to myeloid cell lines. Continuous GATA2-ERT expression was required for the proliferation of these immortalized lines. Myeloid expansion and a block in T and B lineage differentiation were observed in mice transplanted with GATA2-ERT-expressing BM cells. Myeloid expansion occurred after the granulocyte monocyte progenitor stage, and lymphoid block was distal to the common lymphoid progenitor in transgenic mice. GATA2 appeared to induce growth via downstream activation of Nmyc and Hoxa9. Our results demonstrate that GATA2 overexpression at low level confers self-renewal capacity to myeloid progenitors and is relevant to myeloid leukemia development.


Asunto(s)
Células de la Médula Ósea/patología , Transformación Celular Neoplásica/genética , Factor de Transcripción GATA2/fisiología , Regulación Leucémica de la Expresión Génica , Linfopoyesis/genética , Células Mieloides/patología , Mielopoyesis/genética , Animales , Linfocitos B/patología , Células de la Médula Ósea/metabolismo , División Celular , Núcleo Celular/metabolismo , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Factor de Transcripción GATA2/genética , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Genes Sintéticos , Genes myc , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Células Mieloides/metabolismo , Receptores de Estrógenos/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/patología , Tamoxifeno/farmacología
11.
Mol Ther Methods Clin Dev ; 2: 15045, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26665131

RESUMEN

Patients with severe sickle cell disease (SCD) are candidates for gene therapy using autologous hematopoietic stem cells (HSCs), but concomitant multi-organ disease may contraindicate pretransplant conditioning with full myeloablation. We tested whether nonmyeloablative conditioning, a regimen used successfully for allogeneic bone marrow transplantation of adult SCD patients, allows engraftment of γ-globin gene-corrected cells to a therapeutic level in the Berkeley mouse model of SCD. Animals transplanted according to this regimen averaged 35% engraftment of transduced hematopoietic stem cells with an average vector copy < 2.0. Fetal hemoglobin (HbF) levels ranged from 20 to 44% of total hemoglobin and approximately two-thirds of circulating red blood cells expressed HbF detected by immunofluorescence (F-cells). Gene therapy treatment of SCD mice ameliorated anemia, reduced hyperleukocytosis, improved renal function, and reduced iron accumulation in liver, spleen, and kidneys. Thus, modest levels of chimerism with donor cells expressing high levels of HbF from an insulated γ-globin lentiviral vector can improve the pathology of SCD in mice, thereby illustrating a potentially safe and effective strategy for gene therapy in humans.

12.
Cell Host Microbe ; 15(5): 587-599, 2014 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-24832453

RESUMEN

Sickle cell disease (SCD) patients are at high risk of contracting pneumococcal infection. To address this risk, they receive pneumococcal vaccines, and antibiotic prophylaxis and treatment. To assess the impact of SCD and these interventions on pneumococcal genetic architecture, we examined the genomes of more than 300 pneumococcal isolates from SCD patients over 20 years. Modern SCD strains retained invasive capacity but shifted away from the serotypes used in vaccines. These strains had specific genetic changes related to antibiotic resistance, capsule biosynthesis, metabolism, and metal transport. A murine SCD model coupled with Tn-seq mutagenesis identified 60 noncapsular pneumococcal genes under differential selective pressure in SCD, which correlated with aspects of SCD pathophysiology. Further, virulence determinants in the SCD context were distinct from the general population, and protective capacity of potential antigens was lost over time in SCD. This highlights the importance of understanding bacterial pathogenesis in the context of high-risk individuals.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Proteínas Bacterianas/genética , Interacciones Huésped-Patógeno , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/genética , Adaptación Fisiológica , Adolescente , Niño , Preescolar , Femenino , Genómica , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Filogenia , Infecciones Neumocócicas/etiología , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/administración & dosificación , Estudios Prospectivos , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/aislamiento & purificación , Streptococcus pneumoniae/fisiología
13.
J Clin Invest ; 120(2): 627-35, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20093777

RESUMEN

Sickle cell disease (SCD) is characterized by intravascular hemolysis and inflammation coupled to a 400-fold greater incidence of invasive pneumococcal infection resulting in fulminant, lethal pneumococcal sepsis. Mechanistically, invasive infection is facilitated by a proinflammatory state that enhances receptor-mediated endocytosis of pneumococci into epithelial and endothelial cells. As statins reduce chronic inflammation, in addition to their serum cholesterol-lowering effects, we hypothesized that statin therapy might improve the outcome of pneumococcal infection in SCD. In this study, we tested this hypothesis in an experimental SCD mouse model and found that statin therapy prolonged survival following pneumococcal challenge. The protective effect resulted in part from decreased platelet-activating factor receptor expression on endothelia and epithelia, which led to reduced bacterial invasion. An additional protective effect resulted from inhibition of host cell lysis by pneumococcal cholesterol-dependent cytotoxins (CDCs), including pneumolysin. We conclude therefore that statins may be of prophylactic benefit against invasive pneumococcal disease in patients with SCD and, more broadly, in settings of bacterial pathogenesis driven by receptor-mediated endocytosis and the CDC class of toxins produced by Gram-positive invasive bacteria.


Asunto(s)
Anemia de Células Falciformes/microbiología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Infecciones Neumocócicas/prevención & control , Anemia de Células Falciformes/patología , Animales , Antiinflamatorios/uso terapéutico , Proteínas Bacterianas/uso terapéutico , Citotoxinas/antagonistas & inhibidores , Citotoxinas/toxicidad , Modelos Animales de Enfermedad , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Glicoproteínas de Membrana Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/fisiología , Infecciones Neumocócicas/patología , Infecciones Neumocócicas/fisiopatología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/fisiología , Estreptolisinas/uso terapéutico
14.
J Infect Dis ; 195(4): 581-4, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17230418

RESUMEN

Children with sickle cell disease have a 600-fold increased incidence of invasive pneumococcal disease. Platelet-activating factor receptor (PAFr) mediates pneumococcal invasion, and up-regulation of PAFr on chronically activated endothelia could contribute to increased bacterial invasion. Mice transplanted with sickle cell bone marrow developed more extensive infection, and 57% died, compared with 16% of wild-type mice. Histopathological analysis revealed that sickle cell mice expressed significantly more PAFr on endothelia and epithelia. Pharmacological blockade or genetic deletion of PAFr protected sickle cell mice from mortality. We conclude that PAFr plays an important role in hypersusceptibility to pneumococcal infection in sickle cell disease.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/metabolismo , Susceptibilidad a Enfermedades , Glicoproteínas de Membrana Plaquetaria/fisiología , Infecciones Neumocócicas/etiología , Infecciones Neumocócicas/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Animales , Modelos Animales de Enfermedad , Células Endoteliales/química , Endotelio Vascular/química , Células Epiteliales/química , Inmunohistoquímica , Pulmón/irrigación sanguínea , Pulmón/química , Ratones , Ratones Endogámicos C57BL , Glicoproteínas de Membrana Plaquetaria/análisis , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Glicoproteínas de Membrana Plaquetaria/biosíntesis , Neumonía Neumocócica/etiología , Neumonía Neumocócica/metabolismo , Receptores Acoplados a Proteínas G/análisis , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/biosíntesis , Regulación hacia Arriba
15.
Blood ; 106(8): 2750-6, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15985541

RESUMEN

Binding of von Willebrand factor (VWF) to the platelet membrane glycoprotein (GP) Ib-IX-V complex initiates a signaling cascade that causes alphaIIbbeta3 activation and platelet aggregation. Previous work demonstrated that botrocetin (bt)/VWF-mediated agglutination activates alphaIIbbeta3 and elicits adenosine triphosphate (ATP) secretion in a thromboxane A2 (TxA2)- and Ca2+-dependent manner. This agglutination-elicited TxA2 production occurs in the absence of ATP secretion. However, the signaling components and signaling network or pathway activated by GPIb-mediated agglutination to cause TxA2 production have not been identified. Therefore, the focus of this study was to elucidate at least part of the signal transduction network or pathway activated by GPIb-mediated agglutination to cause TxA2 production. The phosphatidylinositol 3-kinase (PI3K) selective inhibitor wortmannin, and mouse platelets deficient in Lyn, Src, Syk, Src homology 2 (SH2) domain-containing leukocyte protein 76 (SLP-76), phospholipase Cgamma2 (PLCgamma2), linker for activation of T cells (LAT), or Fc receptor gamma-chain (FcRgamma-chain) were used for these studies. LAT and FcRgamma-chain were found not to be required for agglutination-driven TxA2 production or activation of alphaIIbbeta3, but were required for granule secretion and aggregation. The results also clearly demonstrate that bt/VWF-mediated agglutination-induced TxA2 production is dependent on signaling apparently initiated by Lyn, enhanced by Src, and propagated through Syk, SLP-76, PI3K, PLCgamma2, and protein kinase C (PKC).


Asunto(s)
Antígenos CD36/metabolismo , Venenos de Crotálidos/farmacología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Transducción de Señal , Tromboxano A2/metabolismo , Factor de von Willebrand/farmacología , Adenosina Trifosfato/metabolismo , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Precursores Enzimáticos/deficiencia , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfolipasa C gamma , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/fisiología , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/deficiencia , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/deficiencia , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptores Fc/metabolismo , Transducción de Señal/efectos de los fármacos , Quinasa Syk , Fosfolipasas de Tipo C/deficiencia , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/metabolismo , Familia-src Quinasas/metabolismo
16.
Blood ; 99(7): 2442-7, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11895777

RESUMEN

Members of the Src family of kinases are abundant in platelets. Although their localization is known, their role(s) in platelet function are not well understood. Lyn is a Src-family kinase that participates in signal transduction pathways elicited by collagen-related peptide; it has also been implicated through biochemical studies in the regulation of von Willebrand factor signaling. Here, we provide evidence that Lyn plays a role in gamma-thrombin activation of platelets. Unlike the wild-type platelets, platelets from Lyn-deficient mice do not undergo irreversible aggregation, produce thromboxane A2, or secrete adenosine diphosphate in response to submaximal gamma-thrombin concentrations that cause secretion-dependent irreversible aggregation. Phosphorylation of Akt, a downstream effector of phosphatidylinositol 3-kinase, also requires a higher concentration of gamma-thrombin in Lyn-deficient platelets than in wild-type platelets. These findings demonstrate that Lyn signaling is required for thrombin induction of secretion-dependent platelet aggregation. Specifically, Lyn is required under these conditions to enable thrombin-induced TxA2 production and adenosine diphosphate secretion, necessary steps in secretion-dependent platelet aggregation.


Asunto(s)
Adenosina Difosfato/sangre , Plaquetas/fisiología , Agregación Plaquetaria/efectos de los fármacos , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/sangre , Trombina/farmacología , Tromboxano A2/sangre , Familia-src Quinasas/genética , Animales , Plaquetas/efectos de los fármacos , Gránulos Citoplasmáticos/fisiología , Técnicas In Vitro , Cinética , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/sangre , Proteínas Proto-Oncogénicas c-akt , Tromboxano A2/biosíntesis , Familia-src Quinasas/sangre , Familia-src Quinasas/deficiencia
17.
Biochem Biophys Res Commun ; 292(4): 916-21, 2002 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-11944902

RESUMEN

The work presented here demonstrates that platelets from mice lacking LAT (linker for the activation of T cells) show reversible aggregation in response to concentrations of collagen that cause TxA2/ADP-dependent irreversible aggregation of control platelets. The aggregation defect of the LAT-deficient platelets was shown to be the result of almost no TxA2 production and significantly diminished ADP secretion. In contrast, the LAT deficiency does not affect aggregation induced by high concentrations of collagen because that aggregation is not dependent on TxA2 and/or ADP. Even though ADP and TxA2 provide amplification signals for platelet activation in response to low concentrations of collagen, LAT-deficient platelets hyperaggregate to low levels of U46619, a TxA2 analog, or ADP. Though the mechanism(s) of costimulatory signals by collagen, ADP, and TxA2 remains unidentified, it is clear that LAT plays a positive role in collagen-induced, TxA2/ADP-dependent aggregation, and a negative role in TxA2 or ADP-induced platelet aggregation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Adenosina Difosfato/farmacología , Plaquetas/metabolismo , Proteínas Portadoras/metabolismo , Colágeno/farmacología , Proteínas de la Membrana , Fosfoproteínas/metabolismo , Transducción de Señal/fisiología , Tromboxano A2/farmacología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Adenosina Difosfato/metabolismo , Animales , Anticuerpos/farmacología , Ácido Araquidónico/metabolismo , Plaquetas/efectos de los fármacos , Proteínas Portadoras/genética , Relación Dosis-Respuesta a Droga , Ratones , Ratones Noqueados , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tromboxano A2/biosíntesis , Vasoconstrictores/farmacología
18.
Blood ; 101(7): 2646-51, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12446460

RESUMEN

Collagen-induced activation of platelets in suspension leads to alpha(IIb)beta(3)-mediated outside-in signaling, granule release, thromboxane A2 (TxA2) production, and aggregation. Although much is known about collagen-induced platelet signaling, the roles of TxA2 production, adenosine diphosphate (ADP) and dense-granule secretion, and alpha(IIb)beta(3)-mediated outside-in signaling in this process are unclear. Here, we demonstrate that TxA2 and ADP are required for collagen-induced platelet activation in response to a low, but not a high, level of collagen and that alpha(IIb)beta(3)-mediated outside-in signaling is required, at least in part, for this TxA2 production and ADP secretion. A high level of collagen can activate platelets deficient in PLC gamma 2, G alpha q, or TxA2 receptors, as well as platelets treated with a protein kinase C inhibitor, Ro31-8220. Thus, activation of alpha(IIb)beta(3) in response to a high level of collagen does not require these signaling proteins. Furthermore, a high level of collagen can cause weak TxA2 and ADP-independent aggregation, but maximal aggregation induced by a high level of collagen requires TxA2 or secretion.


Asunto(s)
Adenosina Difosfato/fisiología , Colágeno/farmacología , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/fisiología , Transducción de Señal , Tromboxano A2/fisiología , Adenosina Difosfato/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Proteínas de Unión al GTP Heterotriméricas , Cinética , Ratones , Fosfolipasa C gamma , Tromboxano A2/biosíntesis , Fosfolipasas de Tipo C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA