Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Regul Toxicol Pharmacol ; 94: 293-298, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29454888

RESUMEN

Although parenteral iron products have been established to medicinal use decades before, their structure and pharmacokinetic properties are not fully characterized yet. With its' second reflection paper on intravenous iron-based nano-colloidal products (EMA/CHMP/SWP/620008/2012) the European Medicine Agency provided an extensive catalogue of methods for quality, non-clinical and pharmacokinetic studies for the comparison of nano-sized iron products to an originator (EMA, 2015). For iron distribution studies, the reflection paper assumed the use of rodents. In our tests, we used a turkey fetus model to investigate time dependent tissue concentrations in pharmacological and toxicological relevant tissues liver, heart and kidney. We found turkey embryos to be a suitable alternative to rodents with high discriminatory sensitivity. Clear differences were found between equimolar doses of iron products with hydroxyethyl amylopectin, sucrose, dextrane and carboxymaltose shell. A linear dose dependency for the tissue accumulation was also demonstrated.


Asunto(s)
Amilopectina/análogos & derivados , Amilopectina/farmacocinética , Embrión no Mamífero/metabolismo , Compuestos Férricos/farmacocinética , Ácido Glucárico/farmacocinética , Complejo Hierro-Dextran/farmacocinética , Maltosa/análogos & derivados , Nanopartículas , Amilopectina/administración & dosificación , Animales , Compuestos Férricos/administración & dosificación , Sacarato de Óxido Férrico , Ácido Glucárico/administración & dosificación , Complejo Hierro-Dextran/administración & dosificación , Riñón/metabolismo , Hígado/metabolismo , Maltosa/administración & dosificación , Maltosa/farmacocinética , Modelos Animales , Miocardio/metabolismo , Nanopartículas/administración & dosificación , Equivalencia Terapéutica , Turquía
2.
Toxicol Res (Camb) ; 6(1): 54-62, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-30090476

RESUMEN

Foci of altered hepatocytes (FAH) were induced in fetal turkey liver (FTL) by diethyl nitrosamine. FAH in FTL were resistant to iron overload similar to FAH in humans and rodents. The mitotic index was significantly higher in FAH (6.2 mitosis in 1000 hepatocytes) than in extrafocal liver tissue (1.8 mitosis in 1000 hepatocytes). The calculation of the net growth rate based on both cell proliferation (mitosis) and cell death (TUNEL positive) revealed a threefold growth advantage of the FAH over the surrounding liver. Two well established anti-tumor substances from different chemical classes, different modes of action and with different clinical use in the treatment of hepatocellular carcinoma (HCC) were used to study their effect on FAH. Sorafenib is the only approved drug for systemic pharmacological treatment of HCC; cisplatin has been used for many years in hepatic arterial infusion. Cisplatin had no clear effect on number of size of FAH, cell proliferation (mitosis) or cell loss (TUNEL positive). Sorafenib enhanced the development of FAH. Morphometric quantification revealed a sorafenib-induced 2-3-fold increase in number (FAH per cm2 and FAH per cm3), size and volume fraction of FAH. This unexpected finding was confirmed in two experiments. The effect was driven by an increased cell proliferation in the FAH, resulting in an increased, 5.4-fold growth advantage of FAH versus the surrounding liver in sorafenib-treated FTL. In this model, sorafenib has a promoting effect on preneoplastic FAH. This might be of relevance for the treatment of patients with long term survival perspective, e.g. in an adjuvant setting.

3.
Regul Toxicol Pharmacol ; 73(1): 65-72, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26111606

RESUMEN

Nanomedicines are more complex than most pharmacologically active substances or medicines and have been considered as non-biological complex drugs. For nanomedicines pivotal pharmacokinetic properties cannot be assessed by plasma concentration data from standard bioequivalence studies. Using intravenous iron complexes (IICs) as model we show that fetal avian tissues can be used to study time dependent tissue concentrations in heart and liver. Clear differences were found between equimolar doses of sucrose, gluconate or carboxymaltose coated iron particles. The range in tissue iron concentrations observed with these clinically widely used IICs provides an orientation as to what should be acceptable for any new IICs. Moreover, sensitivity of the experimental model was high enough to detect a 20% difference in tissue iron concentration. For the authorization of generic products under Article 10 (1) of Directive 2001/83/EC a plasma concentration of an active substance in the range of 80%-125% versus the reference product is usually considered acceptable. Based on its high discriminatory sensitivity this method was used to support a positive marketing authorization decision for a generic nanomedicine product.


Asunto(s)
Pollos/metabolismo , Corazón/fisiología , Compuestos de Hierro/metabolismo , Hierro/metabolismo , Hígado/metabolismo , Distribución Tisular/fisiología , Administración Intravenosa , Animales , Medicamentos Genéricos/administración & dosificación , Medicamentos Genéricos/metabolismo , Hierro/administración & dosificación , Compuestos de Hierro/administración & dosificación , Nanomedicina/métodos , Equivalencia Terapéutica
4.
Proc Natl Acad Sci U S A ; 109(52): 21366-71, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23236180

RESUMEN

129/SvEv mice with a loss-of-function mutation in the heterotrimeric G protein α-subunit gene Gnai3 have fusions of ribs and lumbar vertebrae, indicating a requirement for Gα(i) (the "inhibitory" class of α-subunits) in somite derivatives. Mice with mutations of Gnai1 or Gnai2 have neither defect, but loss of both Gnai3 and one of the other two genes increases the number and severity of rib fusions without affecting the lumbar fusions. No myotome defects are observed in Gnai3/Gnai1 double-mutant embryos, and crosses with a conditional allele of Gnai2 indicate that Gα(i) is specifically required in cartilage precursors. Penetrance and expressivity of the rib fusion phenotype is altered in mice with a mixed C57BL/6 × 129/SvEv genetic background. These phenotypes reveal a previously unknown role for G protein-coupled signaling pathways in development of the axial skeleton.


Asunto(s)
Desarrollo Óseo , Huesos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Mamíferos/crecimiento & desarrollo , Transducción de Señal , Alelos , Animales , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Vértebras Lumbares/anomalías , Vértebras Lumbares/crecimiento & desarrollo , Ratones , Ratones de la Cepa 129 , Ratones Mutantes , Mutación/genética , Fenotipo , Costillas/anomalías , Costillas/crecimiento & desarrollo , Fusión Vertebral , Esternón/anomalías , Esternón/crecimiento & desarrollo
5.
Transl Res ; 151(1): 36-44, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18061126

RESUMEN

In this study the toxicity and intracellular availability of iron from iron dextran (FeD), iron sucrose (FeS), and iron gluconate (FeG) was compared in organs of avian (turkey) embryos and in isolated cells (HepG2) in cell culture. Iron uptake was more pronounced in embryonic liver than in renal tissue. Cellular iron uptake in liver and kidney was more or less similar for the different compounds. Only some experiments showed slightly greater iron concentrations in liver and kidney with FeG compared with FeD and FeS. Significant differences were found in the survival ratios of the eggs and the embryo weights depending on the type of iron complex administered. The rank order of toxicities was FeG>FeS>FeD. Iron accumulation in HepG2-cells was extremely high with FeS and FeG, whereas FeD did not lead to a relevant iron uptake by HepG2 cells. The excessively high iron content of the cells is an in vitro phenomenon found neither in the in ovo model with the turkey embryos nor in the clinical use of the compounds. The rank order of toxicities in HepG2 cells was FeS>FeG>FeD. Iron uptake in cell culture does not reflect the in vivo situation. The in ovo model is more suitable to assess the cellular iron uptake and iron toxicity in cells and tissues than the in vitro model. In both in ovo and in vitro experiments, FeD seemed to be superior in terms of toxicity.


Asunto(s)
Embrión no Mamífero/efectos de los fármacos , Compuestos Férricos/toxicidad , Hematínicos/toxicidad , Complejo Hierro-Dextran/toxicidad , Hígado/citología , Animales , Peso Corporal/efectos de los fármacos , Carcinoma Hepatocelular , Línea Celular Tumoral , Embrión de Pollo , Compuestos Férricos/farmacocinética , Sacarato de Óxido Férrico , Ácido Glucárico , Gluconatos/farmacocinética , Gluconatos/toxicidad , Hematínicos/farmacocinética , Humanos , Complejo Hierro-Dextran/farmacocinética , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas , Nanopartículas , Tasa de Supervivencia , Pavos
6.
Immunology ; 122(1): 116-23, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17484771

RESUMEN

Heterotrimeric G(i) proteins play a role in signalling activated by lipopolysaccharide (LPS), Staphylococcus aureus (SA) and group B streptococci (GBS), leading to production of inflammatory mediators. We hypothesized that genetic deletion of G(i) proteins would alter cytokine and chemokine production induced by LPS, SA and GBS stimulation. LPS-induced, heat-killed SA-induced and heat-killed GBS-induced cytokine and chemokine production in peritoneal macrophages from wild-type (WT), Galpha(i2) (-/-) or Galpha(i1/3) (-/-) mice were investigated. LPS induced production of tumour necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), IL-10 and interferon-gamma-inducible protein-10 (IP-10); SA induced TNF-alpha, and IL-1beta production; and GBS induced TNF-alpha, IL-6, IL-1beta, macrophage inflammatory protein-1alpha (MIP-1alpha) and keratinocyte chemoattract (KC) production were all decreased (P < 0.05) in Galpha(i2) (-/-) or Galpha(i1/3) (-/-) mice compared with WT mice. In contrast to the role of G(i) proteins as a positive regulator of mediators, LPS-induced production of MIP-1alpha and granulocyte-macrophage colony-stimulating factor (GM-CSF) were increased in macrophages from Galpha(i1/3) (-/-) mice, and SA-induced MIP-1alpha production was increased in both groups of Galpha(i) protein-depleted mice. LPS-induced production of KC and IL-1beta, SA-induced production of GM-CSF, KC and IP-10, and GBS-induced production of IL-10, GM-CSF and IP-10 were unchanged in macrophages from Galpha(i2) (-/-) or Galpha(i1/3) (-/-) mice compared with WT mice. These data suggest that G(i2) and G(i1/3) proteins are both involved and differentially regulate murine inflammatory cytokine and chemokine production in response to both LPS and Gram-positive microbial stimuli.


Asunto(s)
Quimiocinas/biosíntesis , Lipopolisacáridos/inmunología , Macrófagos Peritoneales/inmunología , Staphylococcus aureus/inmunología , Streptococcus agalactiae/inmunología , Animales , Células Cultivadas , Citocinas/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP/inmunología , Genotipo , Ratones , Ratones Noqueados
7.
Proc Natl Acad Sci U S A ; 104(11): 4371-6, 2007 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-17360531

RESUMEN

The trafficking of leukocytes from the blood to sites of inflammation is the cumulative result of receptor-ligand-mediated signaling events associated with the leukocytes themselves as well as with the underlying vascular endothelium. Our data show that Galpha(i) signaling pathways in the vascular endothelium regulate a critical step required for leukocyte diapedesis. In vivo studies using knockout mice demonstrated that a signaling event in a non-lymphohematopoietic compartment of the lung prevented the recruitment of proinflammatory leukocytes. Intravital microscopy showed that blockade was at the capillary endothelial surface and ex vivo studies of leukocyte trafficking demonstrated that a Galpha(i)-signaling event in endothelial cells was required for transmigration. Collectively, these data suggest that specific Galpha(i2)-mediated signaling between endothelial cells and leukocytes is required for the extravasation of leukocytes and for tissue-specific accumulation.


Asunto(s)
Endotelio Vascular/metabolismo , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Leucocitos/metabolismo , Transducción de Señal , Alérgenos/metabolismo , Animales , Endotelio Vascular/citología , Endotoxinas/metabolismo , Eosinófilos/metabolismo , Inflamación , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Toxina del Pertussis/farmacología
8.
Proc Natl Acad Sci U S A ; 104(8): 3003-8, 2007 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-17296938

RESUMEN

Heterotrimeric G proteins of the G(i) class have been implicated in signaling pathways regulating growth and metabolism under physiological and pathophysiological conditions. Knockout mice carrying inactivating mutations in both of the widely expressed Galpha(i) class genes, Galpha(i2) and Galpha(i3), demonstrate shared as well as gene-specific functions. The presence of a single active allele of Galpha(i3) is sufficient for embryonic development, whereas at least one allele of Galpha(i2) is required for extrauterine life. Mice lacking both Galpha(i2) and Galpha(i3) are massively growth-retarded and die in utero. We have used biochemical and cell biological methods together with in situ liver perfusion experiments to study Galpha(i) isoform-specific functions in Galpha(i2)- and Galpha(i3)-deficient mice. The subcellular localization of Galpha(i3) in isolated mouse hepatocytes depends on the cellular metabolic status. Galpha(i3) localizes to autophagosomes upon starvation-induced autophagy and distributes to the plasma membrane upon insulin stimulation. Analysis of autophagic proteolysis in perfused mouse livers showed that mice lacking Galpha(i3) are deficient in the inhibitory action of insulin. These data indicate that Galpha(i3) is crucial for the antiautophagic action of insulin and suggest an as-yet-unrecognized function for Galpha(i3) on autophagosomal membranes.


Asunto(s)
Autofagia/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Insulina/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Animales , Anticuerpos/farmacología , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/inmunología , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Masculino , Ratones , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Fracciones Subcelulares/efectos de los fármacos
9.
Biochim Biophys Acta ; 1763(10): 1051-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16962188

RESUMEN

Heterotrimeric Gi proteins play a role in lipopolysaccharide (LPS) and Staphylococcus aureus (SA) activated signaling leading to inflammatory mediator production. We hypothesized that genetic deletion of Gi proteins would alter cytokine and chemokine production induced by LPS and SA. LPS- and heat killed SA-induced cytokine and chemokine production in splenocytes from wild type (WT), Galpha(i2) (-/-) or Galpha(i1/3) (-/-) mice were investigated. LPS- or SA-induced production of TNFalpha, IL-6, IFNgamma, IL-12, IL-17, GM-CSF, MIP-1alpha, MCP-1, MIG and IP-10 were significantly increased (1.2 to 33 fold, p<0.05) in splenocytes harvested from Galpha(i2)(-/-) mice compared with WT mice. The effect of Galpha(i) protein depletion was remarkably isoform specific. In splenocytes from Galpha(i1/3) (-/-) mice relative to WT mice, SA-induced IL-6, IFNgamma, GM-CSF, and IP-10 levels were decreased (59% to 86%, p<0.05), whereas other LPS- or SA-stimulated cytokines and chemokines were not different relative to WT mice. LPS- and SA-induced production of KC were unchanged in both groups of the genetic deficient mice. Splenocytes from both Galpha(i2) (-/-) and Galpha(i1/3) (-/-) mice did not exhibit changes in TLR2 and TLR4 expression. Also analysis of splenic cellular composition by flow cytometry demonstrated an increase in splenic macrophages and reduced CD4 T cells in both Galpha(i2) (-/-) and Galpha(i1/3) (-/-) mice relative to WT mice. The disparate response of splenocytes from the Galpha(i2) (-/-) relative to Galpha(i1/3) (-/-) mice therefore cannot be attributed to major differences in spleen cellular composition. These data demonstrate that G(i2) and G(i1/3) proteins are both involved and differentially regulate splenocyte inflammatory cytokine and chemokine production in a highly Gi isoform specific manner in response to LPS and Gram-positive microbial stimuli.


Asunto(s)
Quimiocinas/biosíntesis , Citocinas/biosíntesis , Subunidad alfa de la Proteína de Unión al GTP Gi2/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Lipopolisacáridos/farmacología , Bazo/metabolismo , Staphylococcus aureus/fisiología , Animales , Células Cultivadas , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Isoformas de Proteínas , Sensibilidad y Especificidad , Bazo/citología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
10.
Am J Physiol Cell Physiol ; 289(2): C293-301, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15788486

RESUMEN

Heterotrimeric G(i) proteins may play a role in lipopolysaccharide (LPS)-activated signaling through Toll-like receptor 4 (TLR4), leading to inflammatory mediator production. Although LPS is a TLR4 ligand, the gram-positive bacterium Staphylococcus aureus (SA) is a TLR2 ligand, and group B streptococci (GBS) are neither TLR2 nor TLR4 ligands but are MyD88 dependent. We hypothesized that genetic deletion of G(i) proteins would alter mediator production induced by LPS and gram-positive bacterial stimulation. We examined genetic deletion of Galpha(i2) or Galpha(i1/3) protein in Galpha(i2)-knockout (Galpha(i2)-/-) or Galpha(i1/3)-knockout (Galpha(i1/3)-/-) mice. LPS-, heat-killed SA-, or GBS-induced mediator production in splenocytes or peritoneal macrophages (MPhi) was investigated. There were significant increases in LPS-, SA-, and GBS-induced production of TNF-alpha and IFN-gamma in splenocytes from Galpha(i2)-/- mice compared with wild-type (WT) mice. Also, LPS-induced TNF-alpha was increased in splenocytes from Galpha(i1/3)-/- mice. In contrast to splenocytes, LPS-, SA-, and GBS-induced TNF-alpha, IL-10, and thromboxane B(2) (TxB(2)) production was decreased in MPhi harvested from Galpha(i2)-/- mice. Also, LPS-induced production of IL-10 and TxB(2) was decreased in MPhi from Galpha(i1/3)-/- mice. In subsequent in vivo studies, TNF-alpha levels after LPS challenge were significantly greater in Galpha(i2)-/- mice than in WT mice. Also, myeloperoxidase activity, a marker of tissue neutrophil infiltration, was significantly increased in the gut and lung of LPS-treated Galpha(i2)-/- mice compared with WT mice. These data suggest that G(i) proteins differentially regulate murine TLR-mediated inflammatory cytokine production in a cell-specific manner in response to both LPS and gram-positive microbial stimuli.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/inmunología , Bacterias Grampositivas/inmunología , Inflamación/inmunología , Lipopolisacáridos/inmunología , Animales , Western Blotting , Citocinas/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Inflamación/microbiología , Linfocitos/inmunología , Linfocitos/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like
11.
J Immunol ; 174(5): 3041-50, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15728518

RESUMEN

Complement and FcgammaR effector pathways are central triggers of immune inflammation; however, the exact mechanisms for their cooperation with effector cells and their nature remain elusive. In this study we show that in the lung Arthus reaction, the initial contact between immune complexes and alveolar macrophages (AM) results in plasma complement-independent C5a production that causes decreased levels of inhibitory FcgammaRIIB, increased levels of activating FcgammaRIII, and highly induced FcgammaR-mediated TNF-alpha and CXCR2 ligand production. Blockade of C5aR completely reversed such changes. Strikingly, studies of pertussis toxin inhibition show the essential role of G(i)-type G protein signaling in C5aR-mediated control of the regulatory FcgammaR system in vitro, and analysis of the various C5aR-, FcgammaR-, and G(i)-deficient mice verifies the importance of Galpha(i2)-associated C5aR and the FcgammaRIII-FcgammaRIIB receptor pair in lung inflammation in vivo. Moreover, adoptive transfer experiments of C5aR- and FcgammaRIII-positive cells into C5aR- and FcgammaRIII-deficient mice establish AM as responsible effector cells. AM lacking either C5aR or FcgammaRIII do not possess any such inducibility of immune complex disease, whereas reconstitution with FcgammaRIIB-negative AM results in an enhanced pathology. These data suggest that AM function as a cellular link of C5a production and C5aR activation that uses a Galpha(i2)-dependent signal for modulating the two opposing FcgammaR, FcgammaRIIB and FcgammaRIII, in the initiation of the inflammatory cascade in the lung Arthus reaction.


Asunto(s)
Reacción de Arthus/inmunología , Reacción de Arthus/patología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Mediadores de Inflamación/metabolismo , Pulmón/patología , Macrófagos Alveolares/inmunología , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Complemento/metabolismo , Receptores Fc/metabolismo , Animales , Reacción de Arthus/metabolismo , Línea Celular , Complemento C5a/fisiología , Subunidad alfa de la Proteína de Unión al GTP Gi2 , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Calor , Inmunoglobulina G/farmacología , Mediadores de Inflamación/fisiología , Pulmón/inmunología , Activación de Macrófagos/inmunología , Macrófagos Alveolares/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/fisiología , Receptor Cross-Talk/inmunología , Receptor de Anafilatoxina C5a , Receptores de Complemento/biosíntesis , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Receptores de IgG/deficiencia , Receptores de IgG/genética , Receptores de IgG/metabolismo , Receptores de IgG/fisiología
12.
Int Immunol ; 15(11): 1359-67, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14565934

RESUMEN

Heterotrimeric Gi signaling regulates immune homeostasis, since autoimmunity occurs upon disruption of this pathway. However, the role of the lymphocyte-expressed Galphai subunits (Galphai2 and 3) on T cell activation and cytokine production is poorly understood. To examine this role, we studied T lymphocytes from mice deficient in the Galphai2 or Galphai3 subunits. Galphai2(-/-) but not Galphai3(-/-) splenocytes were hyper-responsive for IFN-gamma and IL-4 production following activation through the TCR. Galphai2(-/-) T cells had a relaxed costimulatory requirement for IL-2 secretion and proliferation compared to wild-type cells. Purified naïve Galphai2(-/-) T cells produced more IL-2 than naïve wild-type T cells following TCR activation, indicating that the hyper-responsive cytokine profile was not due to the expanded Galphai2(-/-) memory T cells, but involved an intrinsic T cell alteration. Cytokine hyper-responsiveness was not seen when purified Galphai2(-/-) T cells were stimulated with phorbol myristic acetate/ionomycin, localizing the alteration to a proximal TCR-specific signaling pathway. Galphai2(-/-) CD4(+) T cells were distinguished from wild-type or Galphai3(-/-) T cells by a globally augmented TCR-induced calcium response. These findings indicate that Galphai2(-/-) mice have an intrinsic CD4(+) T cell abnormality in TCR signaling which may be one cause of augmented T cell effector function and Galphai2(-/-) autoimmune susceptibility.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Proteínas Proto-Oncogénicas/fisiología , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Diferenciación Celular , Citocinas/inmunología , Subunidad alfa de la Proteína de Unión al GTP Gi2 , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Interleucina-2/inmunología , Activación de Linfocitos , Ratones , Proteínas Proto-Oncogénicas/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal
13.
Mol Imaging Biol ; 5(4): 271-7, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14499142

RESUMEN

Treatment of inflammatory bowel disease (IBD) generally relies on long-term use of anti-inflammatory and immunosuppressive agents. The adverse effects of those drugs make it important to prescribe the minimal regimen that is effective. An objective method for noninvasively quantifying severity of bowel inflammation would thus be valuable in guiding inflammatory bowel disease therapy. Using positron emission tomography (PET), we show that white blood cells (WBCs) labeled with 2-deoxy-2-[18F]fluoro-D-glucose (FDG) can serve as a quantitative marker for identifying the presence and severity of intestinal inflammation. In both murine and human subjects, PET images of FDG-labeled WBCs demonstrated little tracer uptake in healthy gastrointestinal and urinary tracts, where physiologic distribution of FDG images of glucose metabolism often compromises abdominopelvic PET imaging of intestinal pathology. Intestinal foci of FDG-labeled WBCs were confirmed to represent inflamed bowel through histopathologic or colonoscopic analysis, and intensity of foci measured in PET images correlated well with histopathologic measures of degree of inflammation. FDG-labeled WBC's, in conjunction with PET, can be used to provide quantitative assessment of bowel inflammation noninvasively, accurately, and rapidly.


Asunto(s)
Colon/diagnóstico por imagen , Enfermedades Inflamatorias del Intestino/diagnóstico por imagen , Leucocitos/metabolismo , Tomografía Computarizada de Emisión , Animales , Colon/patología , Enfermedad de Crohn/diagnóstico por imagen , Enfermedad de Crohn/patología , Cinética , Ratones , Radiografía , Factores de Tiempo
14.
Mol Cell Neurosci ; 23(3): 398-413, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12837624

RESUMEN

Changes in the vesicular transmitter content modulate synaptic strength and may contribute to synaptic plasticity. Several transporters mediating transmitter uptake into small synaptic vesicles (SSVs) have been identified but their regulation is largely unknown. Here we show by quantitative immunoelectron microscopy that the heterotrimeric G-protein subunits Galphao(2), Galpha(q/11), Gbeta(2), and Ggamma(7) are associated with vesicle-containing areas in terminals of cerebellar parallel fibers. These terminals also contain the vesicular glutamate transporter 1 (VGLUT1). In contrast, SSVs of climbing fiber terminals that contain VGLUT2 express one of the Gbeta-subunits Gbeta(1), Gbeta(3), or Gbeta(4), Ggamma(7), and one Galpha-subunit, probably Galphao(2). Glutamate uptake into cerebellar SSVs was inhibited by more than 50% by GMppNp, an activator of G proteins. Thus, vesicle populations with different subtypes of vesicular glutamate transporters contain functional G proteins with distinct subunit profiles. Heterotrimeric G proteins may play an important role in the control of vesicular filling.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Ácido Glutámico/metabolismo , Proteínas de Transporte de Membrana , Neuronas/química , Transportadores de Anión Orgánico , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Proteínas de Transporte Vesicular , Animales , Especificidad de Anticuerpos , Proteínas Portadoras/análisis , Proteínas Portadoras/inmunología , Cerebelo/citología , Proteínas Transportadoras de GABA en la Membrana Plasmática , Proteínas de Unión al GTP/inmunología , Inmunohistoquímica , Proteínas de la Membrana/análisis , Proteínas de la Membrana/inmunología , Microscopía Inmunoelectrónica , Neuronas/ultraestructura , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Ratas , Proteína 1 de Transporte Vesicular de Glutamato , Proteína 2 de Transporte Vesicular de Glutamato , Ácido gamma-Aminobutírico/fisiología
15.
J Immunol ; 170(4): 1707-15, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12574334

RESUMEN

Null mutation of the Galphai2 trimeric G protein results in a discrete and profound mucosal disorder, including inflammatory bowel disease (IBD), attenuation of IL-10 expression, and immune function polarized to Th1 activity. Genetic and adoptive transfer experiments have established a role for B cells and IL-10 in mucosal immunologic homeostasis and IBD resistance. In this study, we addressed the hypothesis that Galphai2 is required for the development of IL-10-producing B cells. Galphai2(-/-) mice were reduced in the relative abundance of marginal zone (MZ), transitional type 2 (T2), and B-1a B cells and significantly increased in follicular mature and B-1b B cells. Reconstitution of RAG2(-/-) mice with Galphai2(-/-) bone marrow induced an IBD-like colitis and a deficiency in absolute numbers of MZ, T2, and B-1 B cells. Thus, the Galphai2(-/-) genotype in colitis susceptibility and B cell development involved a cis effect within the hemopoietic compartment. In vitro, the B cell population of Galphai2(-/-) mice was functionally deficient in LPS-induced proliferation and IL-10 production, consistent with the exclusive capacity of T2 and MZ cell subpopulations for LPS responsiveness. In vivo, Galphai2(-/-) mice were selectively impaired for the IgM response to T-independent type II, consistent with the relative depletion of MZ and peritoneal B-1 subpopulations. Collectively, these results reveal a selective role for Galphai2 in MZ and B-1 B cell development. Disorders of this Galphai2-dependent process in B cell development may represent a mechanism for IBD susceptibility.


Asunto(s)
Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Genes/fisiología , Proteínas Proto-Oncogénicas/genética , Animales , Antígenos T-Independientes/administración & dosificación , Antígenos T-Independientes/inmunología , Subgrupos de Linfocitos B/patología , Trasplante de Médula Ósea , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Colitis/genética , Colitis/inmunología , Colitis/patología , Subunidad alfa de la Proteína de Unión al GTP Gi2 , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Genes/inmunología , Predisposición Genética a la Enfermedad , Inmunofenotipificación , Inyecciones Intraperitoneales , Recuento de Linfocitos , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cavidad Peritoneal/patología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/fisiología , Receptores de Antígenos de Linfocitos B/fisiología , Bazo/inmunología , Bazo/patología
16.
J Cell Biol ; 160(1): 89-99, 2003 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-12507995

RESUMEN

Receptor-regulated class I phosphoinositide 3-kinases (PI3K) phosphorylate the membrane lipid phosphatidylinositol (PtdIns)-4,5-P2 to PtdIns-3,4,5-P3. This, in turn, recruits and activates cytosolic effectors with PtdIns-3,4,5-P3-binding pleckstrin homology (PH) domains, thereby controlling important cellular functions such as proliferation, survival, or chemotaxis. The class IB p110 gamma/p101 PI3K gamma is activated by G beta gamma on stimulation of G protein-coupled receptors. It is currently unknown whether in living cells G beta gamma acts as a membrane anchor or an allosteric activator of PI3K gamma, and which role its noncatalytic p101 subunit plays in its activation by G beta gamma. Using GFP-tagged PI3K gamma subunits expressed in HEK cells, we show that G beta gamma recruits the enzyme from the cytosol to the membrane by interaction with its p101 subunit. Accordingly, p101 was found to be required for G protein-mediated activation of PI3K gamma in living cells, as assessed by use of GFP-tagged PtdIns-3,4,5-P3-binding PH domains. Furthermore, membrane-targeted p110 gamma displayed basal enzymatic activity, but was further stimulated by G beta gamma, even in the absence of p101. Therefore, we conclude that in vivo, G beta gamma activates PI3K gamma by a mechanism assigning specific roles for both PI3K gamma subunits, i.e., membrane recruitment is mediated via the noncatalytic p101 subunit, and direct stimulation of G beta gamma with p110 gamma contributes to activation of PI3K gamma.


Asunto(s)
Membrana Celular/metabolismo , Subunidades beta de la Proteína de Unión al GTP , Subunidades gamma de la Proteína de Unión al GTP , Proteínas de Unión al GTP Heterotriméricas/fisiología , Proteínas de la Membrana , Fosfatidilinositol 3-Quinasas/metabolismo , Sitio Alostérico , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Línea Celular , Cromatografía en Gel , Dimerización , Relación Dosis-Respuesta a Droga , Transferencia de Energía , Proteínas Fluorescentes Verdes , Humanos , Immunoblotting , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Modelos Biológicos , Fosforilación , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Espectrometría de Fluorescencia , Transfección
17.
J Biol Chem ; 278(13): 11536-45, 2003 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-12502714

RESUMEN

Class I phosphoinositide 3-kinases (PI3Ks) are bifunctional enzymes possessing lipid kinase activity and the capacity to phosphorylate their catalytic and/or regulatory subunits. In this study, in vitro autophosphorylation of the G protein-sensitive p85-coupled class I(A) PI3K beta and p101-coupled class I(B) PI3K gamma was examined. Autophosphorylation sites of both PI3K isoforms were mapped to C-terminal serine residues of the catalytic p110 subunit (i.e. serine 1070 of p110 beta and serine 1101 of p110 gamma). Like other class I(A) PI3K isoforms, autophosphorylation of p110 beta resulted in down-regulated PI3K beta lipid kinase activity. However, no inhibitory effect of p110 gamma autophosphorylation on PI3K gamma lipid kinase activity was observed. Moreover, PI3K beta and PI3K gamma differed in the regulation of their autophosphorylation. Whereas p110 beta autophosphorylation was stimulated neither by G beta gamma complexes nor by a phosphotyrosyl peptide derived from the platelet-derived growth factor receptor, autophosphorylation of p110 gamma was significantly enhanced by G beta gamma in a time- and concentration-dependent manner. In summary, we show that autophosphorylation of both PI3K beta and PI3K gamma occurs in a C-terminal region of the catalytic p110 subunit but differs in its regulation and possible functional consequences, suggesting distinct roles of autophosphorylation of PI3K beta and PI3K gamma.


Asunto(s)
Isoenzimas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Catálisis , Línea Celular , Cartilla de ADN , Humanos , Isoenzimas/química , Datos de Secuencia Molecular , Fosfatidilinositol 3-Quinasas/química , Fosforilación , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Serina/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...