Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncoimmunology ; 10(1): 1873607, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33537176

RESUMEN

PD1 blockade to reinvigorate T cells has become part of standard of care for patients with NSCLC across disease stages. However, the majority of patients still do not respond. One potential mechanism of resistance is increased expression of other checkpoint inhibitory molecules on T cells leading to their suppression; however, this phenomenon has not been well studied in tumor-reactive, human T cells. The purpose of this study was to evaluate this compensatory mechanism in a novel model using human effector T cells infiltrating and reactive against human lung cancer. Immunodeficient mice with flank tumors established from a human lung cancer cell line expressing the NYESO1 antigen were treated with activated human T cells expressing a TCR reactive to NYESO1 (Ly95) with or without anti-PD1 alone and with combinations of anti-PD1 plus anti-TIM3 or anti-TIGIT. A month later, the effect on tumor growth and the phenotype and ex vivo function of the TILs were analyzed. Anti-PD1 and Ly95 T cells led to greater tumor control than Ly95 T cells alone; however, tumors continued to grow. The ex-vivo function of PD1-blocked Ly95 TILs was suppressed and was associated with increased T cell expression of TIM3/TIGIT. Administering combinatorial blockade of PD1+ TIM3 or PD1+ TIGIT with Ly95 T cells led to greater tumor control than blocking PD1 alone. In our model, PD1 blockade was suboptimally therapeutic alone. The effect of TIM3 and TIGIT was upregulated on T cells in response to PD1 blockade and anti-tumor activity could be enhanced when these inhibitory receptors were also blocked with antibodies in combination with anti-PD1 therapy.


Asunto(s)
Linfocitos Infiltrantes de Tumor , Linfocitos T , Traslado Adoptivo , Animales , Humanos , Ratones , Receptor de Muerte Celular Programada 1 , Receptores Inmunológicos
2.
Am J Respir Cell Mol Biol ; 59(6): 723-732, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30095976

RESUMEN

Pulmonary lymphangioleiomyomatosis (LAM) is a slow-progressing metastatic disease that is driven by mutations in the tumor suppressor tuberous sclerosis complex 1/2 (TSC1/2). Rapamycin inhibits LAM cell proliferation and is the only approved treatment, but it cannot cause the regression of existing lesions and can only stabilize the disease. However, in other cancers, immunotherapies such as checkpoint blockade against PD-1 and its ligand PD-L1 have shown promise in causing tumor regression and even curing some patients. Thus, we asked whether PD-L1 has a role in LAM progression. In vitro, PD-L1 expression in murine Tsc2-null cells is unaffected by mTOR inhibition with torin but can be upregulated by IFN-γ. Using immunohistochemistry and single-cell flow cytometry, we found increased PD-L1 expression both in human lung tissue from patients with LAM and in Tsc2-null lesions in a murine model of LAM. In this model, PD-L1 is highly expressed in the lung by antigen-presenting and stromal cells, and activated T cells expressing PD-1 infiltrate the affected lung. In vivo treatment with anti-PD-1 antibody significantly prolongs mouse survival in the model of LAM. Together, these data demonstrate that PD-1/PD-L1-mediated immunosuppression may occur in LAM, and suggest new opportunities for therapeutic targeting that may provide benefits beyond those of rapamycin.


Asunto(s)
Antígeno B7-H1/metabolismo , Neoplasias Pulmonares/metabolismo , Pulmón/metabolismo , Linfangioleiomiomatosis/metabolismo , Esclerosis Tuberosa/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Antígeno B7-H1/inmunología , Estudios de Casos y Controles , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Linfangioleiomiomatosis/tratamiento farmacológico , Linfangioleiomiomatosis/inmunología , Linfangioleiomiomatosis/patología , Ratones , Ratones Endogámicos C57BL , Esclerosis Tuberosa/tratamiento farmacológico , Esclerosis Tuberosa/inmunología , Esclerosis Tuberosa/patología , Regulación hacia Arriba
3.
PLoS One ; 13(5): e0197105, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29758070

RESUMEN

Lymphangioleiomyomatosis (LAM) is a rare, almost exclusively female lung disease linked to inactivating mutations in tuberous sclerosis complex 2 (TSC2), a tumor suppressor gene that controls cell metabolic state and growth via regulation of the mechanistic target of rapamycin (mTORC1) signaling. mTORC1 is frequently activated in human cancers and, although the mTORC1 inhibitor rapamycin has a cytostatic effect, it is, in general, unable to elicit a robust curative effect or tumor regression. Using RNA-Seq, we identified (1) Insulin-like Growth Factor (IGF2) as one of the genes with the highest fold-change difference between human TSC2-null and TSC2-expressing angiomyolipoma cells from a patient with LAM, and (2) the mouse IGF2 homolog Igf2, as a top-ranking gene according to fold change between Tsc2-/- and Tsc2+/+ mouse embryo fibroblasts (MEFs). We extended transcript-level findings to protein level, observing increased Igf2 protein expression and Igf2 secretion by Tsc2-/- MEFs. Increased Igf2 expression was not due to epigenetic imprinting, but was partially mediated through the Stat3 pathway and was completely insensitive to rapamycin treatment. An siRNA-mediated decrease of Igf2 resulted in decreased Stat3 phosphorylation, suggesting presence of an autocrine Igf2/Stat3 amplification cycle in Tsc2-/- MEFs. In human pulmonary LAM lesions and metastatic cell clusters, high levels of IGF2 were associated with mTORC1 activation. In addition, treatment of three primary IGF2-expressing LAM lung cell lines with rapamycin did not result in IGF2 level changes. Thus, targeting of IGF2 signaling may be of therapeutic value to LAM patients, particularly those who are unresponsive to rapamycin.


Asunto(s)
Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor II del Crecimiento Similar a la Insulina/biosíntesis , Neoplasias Pulmonares/metabolismo , Transducción de Señal , Sirolimus/farmacología , Proteínas Supresoras de Tumor/deficiencia , Animales , Línea Celular Tumoral , Embrión de Mamíferos/patología , Fibroblastos/patología , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Linfangioleiomiomatosis , Ratones , Ratones Noqueados , Proteína 2 del Complejo de la Esclerosis Tuberosa
4.
Nat Commun ; 8(1): 1216, 2017 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-29084966

RESUMEN

Platelets are increasingly recognized for their contributions to tumor metastasis. Here, we show that the phosphoinositide signaling modulated by phosphatidylinositol transfer protein type α (PITPα), a protein which shuttles phosphatidylinositol between organelles, is essential for platelet-mediated tumor metastasis. PITPα-deficient platelets have reduced intracellular pools of phosphoinositides and an 80% reduction in IP3 generation upon platelet activation. Unexpectedly, mice lacking platelet PITPα form thrombi normally at sites of intravascular injuries. However, following intravenous injection of tumor cells, mice lacking PITPα develop fewer lung metastases due to a reduction of fibrin formation surrounding the tumor cells, rendering the metastases susceptible to mucosal immunity. These findings demonstrate that platelet PITPα-mediated phosphoinositide signaling is inconsequential for in vivo hemostasis, yet is critical for in vivo dissemination. Moreover, this demonstrates that signaling pathways within platelets may be segregated into pathways that are essential for thrombosis formation and pathways that are important for non-hemostatic functions.


Asunto(s)
Plaquetas/metabolismo , Neoplasias Pulmonares/secundario , Proteínas de Transferencia de Fosfolípidos/metabolismo , Trombosis/metabolismo , Animales , Anticoagulantes/farmacología , Plaquetas/efectos de los fármacos , Plaquetas/patología , Fibrina/metabolismo , Eliminación de Gen , Hemostasis/efectos de los fármacos , Hiperplasia , Inmunidad Mucosa/efectos de los fármacos , Inositol 1,4,5-Trifosfato/metabolismo , Integrasas/metabolismo , Tejido Linfoide/patología , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Trombina/metabolismo , Trombosis/patología
5.
Nat Commun ; 5: 4691, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25178411

RESUMEN

PIKfyve is essential for the synthesis of phosphatidylinositol-3,5-bisphosphate [PtdIns(3,5)P2] and for the regulation of endolysosomal membrane dynamics in mammals. PtdIns(3,5)P2 deficiency causes neurodegeneration in mice and humans, but the role of PtdIns(3,5)P2 in non-neural tissues is poorly understood. Here we show that platelet-specific ablation of PIKfyve in mice leads to accelerated arterial thrombosis, and, unexpectedly, also to inappropriate inflammatory responses characterized by macrophage accumulation in multiple tissues. These multiorgan defects are attenuated by platelet depletion in vivo, confirming that they reflect a platelet-specific process. PIKfyve ablation in platelets induces defective maturation and excessive storage of lysosomal enzymes that are released upon platelet activation. Impairing lysosome secretion from PIKfyve-null platelets in vivo markedly attenuates the multiorgan defects, suggesting that platelet lysosome secretion contributes to pathogenesis. Our findings identify PIKfyve as an essential regulator for platelet lysosome homeostasis, and demonstrate the contributions of platelet lysosomes to inflammation, arterial thrombosis and macrophage biology.


Asunto(s)
Plaquetas/patología , Endosomas/patología , Enfermedades por Almacenamiento Lisosomal/patología , Lisosomas/patología , Fosfatidilinositol 3-Quinasas/deficiencia , Trombosis/patología , Animales , Plaquetas/enzimología , Peso Corporal , Gránulos Citoplasmáticos/enzimología , Gránulos Citoplasmáticos/patología , Endosomas/enzimología , Regulación de la Expresión Génica , Infertilidad/genética , Inflamación/complicaciones , Inflamación/enzimología , Inflamación/patología , Longevidad/genética , Enfermedades por Almacenamiento Lisosomal/complicaciones , Enfermedades por Almacenamiento Lisosomal/enzimología , Lisosomas/enzimología , Macrófagos/enzimología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosfatos de Fosfatidilinositol/metabolismo , Recuento de Plaquetas , Transducción de Señal , Trombosis/complicaciones , Trombosis/enzimología
6.
Haematologica ; 99(3): 554-60, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24293517

RESUMEN

Protein arginylation by arginyl-transfer RNA protein transferase (ATE1) is emerging as a regulator protein function that is reminiscent of phosphorylation. For example, arginylation of ß-actin has been found to regulate lamellipodial formation at the leading edge in fibroblasts. This finding suggests that similar functions of ß-actin in other cell types may also require arginylation. Here, we have tested the hypothesis that ATE1 regulates the cytoskeletal dynamics essential for in vivo platelet adhesion and thrombus formation. To test this hypothesis, we generated conditional knockout mice specifically lacking ATE1 in their platelets and in their megakaryocytes and analyzed the role of arginylation during platelet activation. Surprisingly, rather than finding an impairment of the actin cytoskeleton structure and its rearrangement during platelet activation, we observed that the platelet-specific ATE1 knockout led to enhanced clot retraction and in vivo thrombus formation. This effect might be regulated by myosin II contractility since it was accompanied by enhanced phosphorylation of the myosin regulatory light chain on Ser19, which is an event that activates myosin in vivo. Furthermore, ATE1 and myosin co-immunoprecipitate from platelet lysates. This finding suggests that these proteins directly interact within platelets. These results provide the first evidence that arginylation is involved in phosphorylation-dependent protein regulation, and that arginylation affects myosin function in platelets during clot retraction.


Asunto(s)
Aminoaciltransferasas/metabolismo , Plaquetas/metabolismo , Retracción del Coagulo , Miosinas/metabolismo , Trombosis/metabolismo , Actinas/metabolismo , Aminoaciltransferasas/química , Aminoaciltransferasas/deficiencia , Aminoaciltransferasas/genética , Animales , Retracción del Coagulo/genética , Modelos Animales de Enfermedad , Expresión Génica , Ratones , Ratones Noqueados , Modelos Moleculares , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Conformación Proteica , Trombosis/genética
7.
PLoS One ; 8(7): e69315, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935982

RESUMEN

RhoA plays a multifaceted role in platelet biology. During platelet development, RhoA has been proposed to regulate endomitosis, proplatelet formation, and platelet release, in addition to having a role in platelet activation. These processes were previously studied using pharmacological inhibitors in vitro, which have potential drawbacks, such as non-specific inhibition or incomplete disruption of the intended target proteins. Therefore, we developed a conditional knockout mouse model utilizing the CRE-LOX strategy to ablate RhoA, specifically in megakaryocytes and in platelets to determine its role in platelet development. We demonstrated that deleting RhoA in megakaryocytes in vivo resulted in significant macrothrombocytopenia. RhoA-null megakaryocytes were larger, had higher mean ploidy, and exhibited stiff membranes with micropipette aspiration. However, in contrast to the results observed in experiments relying upon pharmacologic inhibitors, we did not observe any defects in proplatelet formation in megakaryocytes lacking RhoA. Infused RhoA-null megakaryocytes rapidly released platelets, but platelet levels rapidly plummeted within several hours. Our evidence supports the hypothesis that changes in membrane rheology caused infused RhoA-null megakaryocytes to prematurely release aberrant platelets that were unstable. These platelets were cleared quickly from circulation, which led to the macrothrombocytopenia. These observations demonstrate that RhoA is critical for maintaining normal megakaryocyte development and the production of normal platelets.


Asunto(s)
Plaquetas/enzimología , Megacariocitos/enzimología , Ploidias , Trombopoyesis , Proteína de Unión al GTP rhoA/metabolismo , Animales , Membrana Celular/metabolismo , Tamaño de la Célula , Citoesqueleto/metabolismo , Eliminación de Gen , Marcación de Gen , Ratones , Mutación/genética , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Reología , Trombocitopenia/patología , Proteína de Unión al GTP rhoA/deficiencia , Proteína de Unión al GTP rhoA/genética
8.
Blood ; 121(14): 2743-52, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23372168

RESUMEN

Three isoforms of phosphatidylinositol-4-phosphate 5-kinase (PIP5KIα, PIP5KIß, and PIP5KIγ) can each catalyze the final step in the synthesis of phosphatidylinositol-4,5-bisphosphate (PIP2), which in turn can be either converted to second messengers or bind directly to and thereby regulate proteins such as talin. A widely quoted model speculates that only p90, a longer splice form of platelet-specific PIP5KIγ, but not the shorter p87 PIP5KIγ, regulates the ligand-binding activity of integrins via talin. However, when we used mice genetically engineered to lack only p90 PIP5KIγ, we found that p90 PIP5KIγ is not critical for integrin activation or platelet adhesion on collagen. However, p90 PIP5KIγ-null platelets do have impaired anchoring of their integrins to the underlying cytoskeleton. Platelets lacking both the p90 and p87 PIP5KIγ isoforms had normal integrin activation and actin dynamics, but impaired anchoring of their integrins to the cytoskeleton. Most importantly, they formed weak shear-resistant adhesions ex vivo and unstable vascular occlusions in vivo. Together, our studies demonstrate that, although PIP5KIγ is essential for normal platelet function, individual isoforms of PIP5KIγ fulfill unique roles for the integrin-dependent integrity of the membrane cytoskeleton and for the stabilization of platelet adhesion.


Asunto(s)
Plaquetas/citología , Plaquetas/enzimología , Integrinas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Adhesividad Plaquetaria/fisiología , Trombosis/enzimología , Citoesqueleto de Actina/fisiología , Empalme Alternativo/genética , Animales , Citoesqueleto/fisiología , Exones/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Isomerismo , Megacariocitos/citología , Megacariocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pinzas Ópticas , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Embarazo , Talina/metabolismo , Trombosis/genética
9.
Blood ; 113(15): 3577-84, 2009 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-19190246

RESUMEN

Pleckstrin, the platelet and leukocyte C kinase substrate, is a prominent substrate of PKC in platelets, monocytes, macrophages, lymphocytes, and granulocytes. Pleckstrin accounts for 1% of the total protein in these cells, but it is best known for containing the 2 prototypic Pleckstrin homology, or PH, domains. Overexpressed pleckstrin can affect polyphosphoinositide second messenger-based signaling events; however, its true in vivo role has been unknown. Here, we describe mice containing a null mutation within the pleckstrin gene. Platelets lacking pleckstrin exhibit a marked defect in exocytosis of delta and alpha granules, alphaIIbbeta3 activation, actin assembly, and aggregation after exposure to the PKC stimulant, PMA. Pleckstrin-null platelets aggregate normally in response to thrombin, but they fail to aggregate in response to thrombin in the presence of PI3K inhibitors, suggesting that a PI3K-dependent signaling pathway compensates for the loss of pleckstrin. Although pleckstrin-null platelets merged their granules in response to stimulation of PKC, they failed to empty their contents into the open canalicular system. This might be attributable to impaired actin assembly present in cells lacking pleckstrin. These data show that pleckstrin regulates the fusion of granules to the cell membrane and is an essential component of PKC-mediated exocytosis.


Asunto(s)
Plaquetas/metabolismo , Proteínas Sanguíneas/metabolismo , Exocitosis/fisiología , Fosfoproteínas/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal/fisiología , Actinas/metabolismo , Animales , Plaquetas/citología , Proteínas Sanguíneas/genética , Gránulos Citoplasmáticos/metabolismo , Fusión de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/genética , Activación Plaquetaria/fisiología
10.
Proc Natl Acad Sci U S A ; 105(37): 14064-9, 2008 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-18772378

RESUMEN

The three isoforms of PIP5KI (alpha, beta, and gamma) synthesize PI4,5P(2) (PIP(2)) by phosphorylating PI4P. Therefore, it is not clear why platelets, like all eukaryotic cells, have more than one isoform. To test the hypothesis that PIP5KI isoforms have nonoverlapping functions, we generated a murine line containing a null mutation of PIP5KIbeta and analyzed the effect on platelet signaling. PIP5KIbeta-null mice had normal platelet counts. In contrast to platelets lacking PIP5KIalpha, platelets lacking PIP5KIbeta exhibited impaired aggregation accompanied by disaggregation. Although platelets lacking PIP5KIbeta had only a moderate deficiency of PIP(2) under basal conditions, they had a striking deficiency in PIP(2) synthesis and IP(3) formation after thrombin stimulation. We have also observed that platelets lacking both PIP5KIalpha and PIP5KIbeta have a complete loss of thrombin-induced IP(3) synthesis even though they still contain PIP5KIgamma, the predominant PIP5KI isoform in platelets. These results demonstrate that PIP5KIbeta, like PIP5KIalpha, contributes to the rapid synthesis of a pool of PIP(2) that is required for second-messenger formation, whereas the pool of PIP(2) synthesized by PIP5KIgamma does not contribute to this process. Additionally, we found that PIP5KIbeta-null platelets failed to form arterial thrombi properly in vivo. Together, these data demonstrate that PIP5KIbeta is required for rapid PIP(2) synthesis, second-messenger production, and stable platelet adhesion under shear in vivo. These results also demonstrate that after stimulation of a G protein-coupled receptor, IP(3) is completely derived from a rapidly synthesized discrete pool of PIP(2) synthesized by PIP5KIalpha and PIP5KIbeta.


Asunto(s)
Inositol 1,4,5-Trifosfato/metabolismo , Fosfatidilinositol 4,5-Difosfato/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Plaquetas/citología , Plaquetas/enzimología , Línea Celular , Regulación Enzimológica de la Expresión Génica , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones Noqueados , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Agregación Plaquetaria , Trombosis/enzimología , Trombosis/genética
11.
J Exp Med ; 205(8): 1775-88, 2008 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-18663126

RESUMEN

Src homology 2 domain-containing leukocyte phosphoprotein of 76 kD (SLP76), an adaptor that plays a critical role in platelet activation in vitro, contains three N-terminal tyrosine residues that are essential for its function. We demonstrate that mice containing complementary tyrosine to phenylalanine mutations in Y145 (Y145F) and Y112 and Y128 (Y112/128F) differentially regulate integrin and collagen receptor signaling. We show that mutation of Y145 leads to severe impairment of glycoprotein VI (GPVI)-mediated responses while preserving outside-in integrin signaling. Platelets from Y112/128F mice, although having mild defects in GPVI signaling, exhibit defective actin reorganization after GPVI or alpha IIb beta 3 engagement. The in vivo consequences of these signaling defects correlate with the mild protection from thrombosis seen in Y112/128F mice and the near complete protection observed in Y145F mice. Using genetic complementation, we further demonstrate that all three phosphorylatable tyrosines are required within the same SLP76 molecule to support platelet activation by GPVI.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/fisiología , Plaquetas/fisiología , Integrinas/fisiología , Fosfoproteínas/química , Fosfoproteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Agammaglobulinemia Tirosina Quinasa , Sustitución de Aminoácidos , Animales , Antígenos CD36/fisiología , Femenino , Prueba de Complementación Genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Mutagénesis Sitio-Dirigida , Fosfolipasa C gamma/sangre , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Fosforilación , Agregación Plaquetaria/genética , Agregación Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/fisiología , Proteínas Tirosina Quinasas/sangre , Transducción de Señal , Trombosis/sangre , Trombosis/etiología , Trombosis/genética , Tirosina/química
12.
J Clin Invest ; 118(2): 812-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18188447

RESUMEN

Phosphatidylinositol-4,5-bisphosphate (PIP(2)) is an abundant phospholipid that contributes to second messenger formation and has also been shown to contribute to the regulation of cytoskeletal dynamics in all eukaryotic cells. Although the alpha, beta, and gamma isoforms of phosphatidylinositol-4-phosphate-5-kinase I (PIP5KI) all synthesize PIP2, mammalian cells usually contain more than one PIP5KI isoform. This raises the question of whether different isoforms of PIP5KI fulfill different functions. Given the speculated role of PIP(2) in platelet and megakaryocyte actin dynamics, we analyzed murine megakaryocytes lacking individual PIP5KI isoforms. PIP5KIgamma(-/-) megakaryocytes exhibited plasma membrane blebbing accompanied by a decreased association of the membrane with the cytoskeleton. This membrane defect was rescued by adding back wild-type PIP5KIgamma, but not by adding a catalytically inactive mutant or a splice variant lacking the talin-binding motif. Notably, both PIP5KIbeta- and PIP5KIgamma(-/-) cells had impaired PIP(2) synthesis. However, PIP5KIbeta-null cells lacked the membrane-cytoskeleton defect. Furthermore, overexpressing PIP5KIbeta in PIP5KIgamma(-/-) cells failed to revert this defect. Megakaryocytes lacking the PIP5KIgamma-binding partner, talin1, mimicked the membrane-cytoskeleton defect phenotype seen in PIP5KIgamma(-/-) cells. These findings demonstrate a unique role for PIP5KIgamma in the anchoring of the cell membrane to the cytoskeleton in megakaryocytes, probably through a pathway involving talin. These observations further demonstrate that individual PIP5KI isoforms fulfill distinct functions within cells.


Asunto(s)
Membrana Celular/ultraestructura , Citoesqueleto/ultraestructura , Megacariocitos/ultraestructura , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Talina/metabolismo , Animales , Citoesqueleto/enzimología , Masculino , Megacariocitos/enzimología , Ratones , Ratones Mutantes , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
13.
J Immunol ; 179(4): 2223-7, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17675482

RESUMEN

Chemokines acting through G protein-coupled receptors play an essential role in the immune response. PI3K and phospholipase C (PLC) are distinct signaling molecules that have been proposed in the regulation of chemokine-mediated cell migration. Studies with knockout mice have demonstrated a critical role for PI3K in G(alphai) protein-coupled receptor-mediated neutrophil and lymphocyte chemotaxis. Although PLCbeta is not essential for the chemotactic response of neutrophils, its role in lymphocyte migration has not been clearly defined. We compared the chemotactic response of peripheral T cells derived from wild-type mice with mice containing loss-of-function mutations in both of the two predominant lymphocyte PLCbeta isoforms (PLCbeta2 and PLCbeta3), and demonstrate that loss of PLCbeta2 and PLCbeta3 significantly impaired T cell migration. Because second messengers generated by PLCbeta lead to a rise in intracellular calcium and activation of PKC, we analyzed which of these responses was critical for the PLCbeta-mediated chemotaxis. Intracellular calcium chelation decreased the chemotactic response of wild-type lymphocytes, but pharmacologic inhibition of several PKC isoforms had no effect. Furthermore, calcium efflux induced by stromal cell-derived factor-1alpha was undetectable in PLCbeta2beta3-null lymphocytes, suggesting that the migration defect is due to the impaired ability to increase intracellular calcium. This study demonstrates that, in contrast to neutrophils, phospholipid second messengers generated by PLCbeta play a critical role in T lymphocyte chemotaxis.


Asunto(s)
Señalización del Calcio/inmunología , Quimiotaxis/inmunología , Isoenzimas/inmunología , Linfocitos T/inmunología , Fosfolipasas de Tipo C/inmunología , Animales , Señalización del Calcio/genética , Quimiocina CXCL12 , Quimiocinas/genética , Quimiocinas/inmunología , Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Quimiotaxis/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/inmunología , Isoenzimas/deficiencia , Isoenzimas/genética , Ratones , Ratones Noqueados , Mutación , Neutrófilos/enzimología , Neutrófilos/inmunología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Fosfolipasa C beta , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Linfocitos T/enzimología , Fosfolipasas de Tipo C/deficiencia
14.
Proc Natl Acad Sci U S A ; 104(28): 11748-53, 2007 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-17609388

RESUMEN

All eukaryotic cells contain the phospholipid phosphatidylinositol 4, 5-bisphosphate (PIP2) that serves multiple roles in signal transduction cascades. Type I phosphatidylinositol-4-phosphate 5-kinase (PIP5KI) catalyzes the synthesis of PIP2 by phosphorylating phosphatidylinositol 4 phosphate. Although the classical isoforms of PIP5KI (designated as alpha, beta, and gamma) all generate the same phospholipid product, they have significantly dissimilar primary structures and expression levels in different tissues, and they appear to localize within different compartments within the cell. Therefore, it appears likely that PIP5KI isoforms have overlapping, but not identical, functions. Here we show that targeted disruption of PIP5KIgamma causes widespread developmental and cellular defects. PIP5KIgamma-null embryos have myocardial developmental defects associated with impaired intracellular junctions that lead to heart failure and extensive prenatal lethality at embryonic day 11.5 of development. Loss of PIP5KIgamma also results in neural tube closure defects that were associated with impaired PIP2 production, adhesion junction formation, and neuronal cell migration. These data, along with those of other PIP5KI isoforms, indicate that individual PIP5KI isoenzymes fulfill specific roles in embryonic development.


Asunto(s)
Sistema Cardiovascular/citología , Sistema Cardiovascular/embriología , Diferenciación Celular/fisiología , Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Inositol 1,4,5-Trifosfato/fisiología , Neuronas/citología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Animales , Sistema Cardiovascular/enzimología , Sistema Cardiovascular/inervación , Diferenciación Celular/genética , Movimiento Celular/genética , Movimiento Celular/fisiología , Células Cultivadas , Sistema Nervioso Central/enzimología , Genes Letales , Inositol 1,4,5-Trifosfato/biosíntesis , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuronas/enzimología , Neuronas/patología , Fosfatidilinositol 4,5-Difosfato/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética
15.
Radiology ; 234(3): 749-55, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15734931

RESUMEN

PURPOSE: To measure the transverse relaxation rate (1/T2) and magnetic susceptibility of the heart in conditions of iron overload by using magnetic resonance (MR) imaging and to correlate these with the tissue iron concentration in a gerbil model. MATERIALS AND METHODS: With prior approval by the institutional animal care and use committee, iron overload was induced with one to 15 weekly subcutaneous injections of iron dextran. Nine gerbils had one to five injections, 10 had six to 10, and eight had 13-15. T2 of the whole heart was measured ex vivo (n=27), and the magnetic susceptibility of the tissue was estimated through measurement of the tissue lysate (n=25). The iron level was measured (in milligrams of iron per gram of wet tissue) with chemical analysis after MR imaging. While 1/T2 and magnetic susceptibility are not equivalent measures of the chemically determined tissue iron level, correlations were expected and were identified by using linear regression models. RESULTS: Iron concentration range was 0.28-1.95 mg/g wet tissue. Iron concentration was strongly correlated with 1/T2 (r=0.92, P <.001, and the root of the mean squares error of the linear prediction, epsilonRMS, was 0.17 mg Fe/g wet tissue with a repetition time of 700 msec). Iron concentration also was strongly correlated with magnetic susceptibility (r=0.90, P <.001, epsilonRMS=0.19 mg Fe/g wet tissue). Multiple regression analysis with combined 1/T2 (with repetition time of 700 msec) and magnetic susceptibility data led to a slight increase in r and decrease in epsilon(RMS) (r=0.93, P <.001, epsilonRMS=0.16 mg Fe/g wet tissue). CONCLUSION: The results of this animal model study demonstrate that 1/T2 and magnetic susceptibility values can be used for estimation of the iron level in the heart.


Asunto(s)
Sobrecarga de Hierro/diagnóstico , Hierro/metabolismo , Imagen por Resonancia Magnética/métodos , Miocardio/metabolismo , Animales , Modelos Animales de Enfermedad , Gerbillinae , Modelos Lineales
16.
Blood ; 106(1): 110-7, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15705797

RESUMEN

Stimulation of platelet G protein-coupled receptors results in the cleavage of phosphatidylinositol 4,5-trisphosphate (PIP(2)) into inositol 1,4,5-trisphosphate and 1,2-diacylglycerol by phospholipase C (PLCbeta). It also results in the phosphorylation of PIP2 by the gamma isoform of phosphatidylinositol 3-kinase (PI3Kgamma) to synthesize phosphatidylinositol 3,4,5-trisphosphate. To understand the role of PIP2 in platelet signaling, we evaluated knock-out mice lacking 2 isoforms of PLCbeta (PLCbeta2 and PLCbeta3) or lacking the G(betagamma)-activated isoform of PI3K (PI3Kgamma). Both knock-out mice were unable to form stable thrombi in a carotid injury model. To provide a functional explanation, knock-out platelets were studied ex vivo. PLCbeta2/beta3-/- platelets failed to assemble filamentous actin, had defects in both secretion and mobilization of intracellular calcium, and were unable to form stable aggregates following low doses of agonists. Platelets lacking PI3Kgamma disaggregated following low-dose adenosine diphosphate (ADP) and had a mildly impaired ability to mobilize intracellular calcium. Yet, they exhibited essentially normal actin assembly and secretion. Remarkably, both PLCbeta2/beta3-/- and PI3Kgamma-/- platelets spread more slowly upon fibrinogen. These results suggest substantial redundancy in platelet signaling pathways. Nonetheless, the diminished ability of knock-out platelets to normally spread after adhesion and to form stable thrombi in vivo suggests that both PLCbeta2/beta3 and PI3Kgamma play vital roles in platelet cytoskeletal dynamics.


Asunto(s)
Isoenzimas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Activación Plaquetaria/fisiología , Fosfolipasas de Tipo C/metabolismo , Actinas/metabolismo , Animales , Plaquetas/enzimología , Plaquetas/metabolismo , Calcio/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ib , Isoenzimas/genética , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosfolipasa C beta , Agregación Plaquetaria/fisiología , Sistemas de Mensajero Secundario/fisiología , Fosfolipasas de Tipo C/genética
17.
Br J Haematol ; 125(6): 788-95, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15180869

RESUMEN

Vanillin, a food additive, covalently binds with sickle haemoglobin (Hb S), inhibits cell sickling and shifts the oxygen equilibrium curve towards the left. These effects would potentially benefit patients with sickle cell disease (SCD). However, vanillin has no therapeutic effect if given orally because orally administered vanillin is rapidly decomposed in the upper digestive tract. To overcome this problem, a vanillin prodrug, MX-1520, which is biotransformed to vanillin in vivo, was synthesized. Studies using transgenic sickle mice, which nearly exclusively develop pulmonary sequestration upon exposure to hypoxia, showed that oral administration of MX-1520 prior to hypoxia exposure significantly reduced the percentage of sickled cells in the blood. The survival time under severe hypoxic conditions was prolonged from 6.6 +/- 0.8 min in untreated animals to 28.8 +/- 12 min (P < 0.05) and 31 +/- 7.5 min (P < 0.05) for doses of 137.5 and 275 mg/kg respectively. Intraperitoneal injection of MX-1520 to bypass possible degradation in the digestive tract showed that doses as low as 7 mg/kg prolonged the survival time and reduced the percentage of sickled cells during hypoxia exposure. These results demonstrate the potential for MX-1520 to be a new and safe anti-sickling agent for patients with SCD.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Benzaldehídos/farmacocinética , Aditivos Alimentarios/farmacocinética , Profármacos/farmacocinética , Anemia de Células Falciformes/etiología , Anemia de Células Falciformes/patología , Animales , Disponibilidad Biológica , Hipoxia/complicaciones , Hipoxia/patología , Inyecciones Intraperitoneales , Pulmón/patología , Ratones , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...