Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L159-L173, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33949204

RESUMO

Chronic obstructive pulmonary disease (COPD) is composed of chronic airway inflammation and emphysema. Recent studies show that Class IA phosphatidylinositol 3-kinases (PI3Ks) play an important role in the regulation of inflammation and emphysema. However, there are few studies on their regulatory subunits. p55PIK is a regulatory subunit of Class IA PI3Ks, and its unique NH2-terminal gives it special functions. p55PIK expression in the lungs of nonsmokers, smokers, and patients with COPD was examined. We established a fusion protein TAT-N15 from the NH2-terminal effector sequence of p55PIK and TAT (the transduction domain of HIV transactivator protein) and investigated the effects of silencing p55PIK or adding TAT-N15 on cigarette smoke exposure at the cellular and animal level. p55PIK expression was increased in patients with COPD. p55PIK deficiency and TAT-N15 significantly inhibited the cigarette smoke extract-induced IL-6, IL-8, and activation of the Akt and the NF-κB pathway in BEAS-2B. p55PIK deficiency and TAT-N15 intranasal administration prevented emphysema and the lung function decline in mice exposed to smoke for 6 mo. p55PIK deficiency and TAT-N15 significantly inhibited lung inflammatory infiltration, reduced levels of IL-6 and KC in mice lung homogenate, and inhibited activation of the Akt and the NF-κB signaling in COPD mice lungs. Our studies indicate that p55PIK is involved in the pathogenesis of COPD, and its NH2-terminal derivative TAT-N15 could be an effective drug in the treatment of COPD by inhibiting the activation of the Akt and the NF-κB pathway.


Assuntos
Inflamação/prevenção & controle , Fosfatidilinositol 3-Quinases/deficiência , Doença Pulmonar Obstrutiva Crônica/complicações , Enfisema Pulmonar/prevenção & controle , Fumaça/efeitos adversos , Adulto , Idoso , Animais , Estudos de Casos e Controles , Feminino , Humanos , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Domínios Proteicos , Enfisema Pulmonar/etiologia , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patologia
2.
EMBO J ; 36(12): 1707-1718, 2017 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-28533230

RESUMO

Alveolar macrophages (AMs) are specialized tissue-resident macrophages that orchestrate the immune responses to inhaled pathogens and maintain organ homeostasis of the lung. Dysregulation of AMs is associated with allergic inflammation and asthma. Here, we examined the role of a phosphoinositide kinase PIKfyve in AM development and function. Mice with conditionally deleted PIKfyve in macrophages have altered AM populations. PIKfyve deficiency results in a loss of AKT activation in response to GM-CSF, a cytokine critical for AM development. Upon exposure to house dust mite extract, mutant mice display severe lung inflammation and allergic asthma accompanied by infiltration of eosinophils and lymphoid cells. Moreover, they have defects in production of retinoic acid and fail to support incorporation of Foxp3+ Treg cells in the lung, resulting in exacerbation of lung inflammation. Thus, PIKfyve plays a role in preventing excessive lung inflammation through regulating AM function.


Assuntos
Asma/patologia , Hipersensibilidade/patologia , Inflamação/patologia , Macrófagos Alveolares/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Alérgenos/administração & dosagem , Animais , Técnicas de Inativação de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Pulmão/patologia , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/deficiência , Deleção de Sequência , Transdução de Sinais , Linfócitos T Reguladores/imunologia
3.
Mol Cell ; 50(1): 29-42, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23434372

RESUMO

Autophagy is an evolutionarily conserved membrane trafficking process. Induction of autophagy in response to nutrient limitation or cellular stress occurs by similar mechanisms in organisms from yeast to mammals. Unlike yeast, metazoan cells rely more on growth factor signaling for a wide variety of cellular activities including nutrient uptake. How growth factor availability regulates autophagy is poorly understood. Here we show that, upon growth factor limitation, the p110ß catalytic subunit of the class IA phosphoinositide 3-kinases (PI3Ks) dissociates from growth factor receptor complexes and increases its interaction with the small GTPase Rab5. This p110ß-Rab5 association maintains Rab5 in its guanosine triphosphate (GTP)-bound state and enhances the Rab5-Vps34 interaction that promotes autophagy. p110ß mutants that fail to interact with Rab5 are defective in autophagy promotion. Hence, in mammalian cells, p110ß acts as a molecular sensor for growth factor availability and induces autophagy by activating a Rab5-mediated signaling cascade.


Assuntos
Autofagia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Classe I de Fosfatidilinositol 3-Quinases/deficiência , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Mutação , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais , Transfecção
4.
Mod Rheumatol ; 28(3): 530-541, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28880680

RESUMO

OBJECTIVE: Neutrophil extracellular traps (NETs) are peculiar structures composed of the externalized chromatin with intracellular proteins and formed by activated neutrophils in a reactive oxygen species (ROS)-dependent manner. Aberrant NETs are considered to be autoantigens for anti-neutrophil cytoplasmic antibodies (ANCAs) underling the development of microscopic polyangiitis (MPA). However, little is known regarding the therapeutic efficacy of in vivo inhibition of NET formation (NETosis) on MPA pathogenesis. This study determines whether reducing NETosis prevents ANCA production and improves characteristic involvement. METHODS: A mouse model of MPA induced by administering a novel extract from Candida albicans was devised. By applying this method to mice lacking phosphoinositide 3-kinase gamma (PI3K-gamma), which is indispensable for ROS production in neutrophils, we investigated the levels of in vivo NETs, ANCA titers and histological damage. RESULTS: Our model exhibited accumulation of NETs in vivo, elevation of ANCA titers and characteristic pathologies mimicking human MPA, including small-vessel vasculitis and crescentic glomerulonephritis. Strikingly, these abnormalities were reduced by genetically and/or pharmacologically blocking PI3K-gamma. Moreover, a pharmacological PI3K-gamma blockade decreased the levels of human NETs. CONCLUSION: Our results suggest that in vivo inhibition of NETosis by blocking PI3K-gamma could be a promising therapeutic strategy for the pathogenesis of MPA.


Assuntos
Anticorpos Anticitoplasma de Neutrófilos/metabolismo , Armadilhas Extracelulares/metabolismo , Poliangiite Microscópica/metabolismo , Fosfatidilinositol 3-Quinases/deficiência , Animais , Produtos Biológicos/toxicidade , Candida/química , Armadilhas Extracelulares/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Poliangiite Microscópica/etiologia , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia
5.
Cell Commun Signal ; 15(1): 28, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28724384

RESUMO

BACKGROUND: Phosphoinositide 3-kinase γ (PI3Kγ) and PI3Kδ are second messenger-generating enzymes with key roles in proliferation, differentiation, survival, and function of leukocytes. Deficiency of the catalytic subunits p110γ and p110δ of PI3Kγ and PI3Kδ in p110γ/δ-/- mice leads to defective B- and T-cell homeostasis. Here we examined the role of p110γ and p110δ in the homeostasis of neutrophils by analyzing p110γ-/-, p110δ-/- and p110γ/δ-/- mice. METHODS: Neutrophils and T cells in leukocyte suspensions from the bone marrow (BM), blood, spleen and lung were analyzed by flow cytometry. Serum concentrations of IL-17, of the neutrophilic growth factor G-CSF, and of the neutrophil mobilizing CXC chemokines CXCL1/KC and CXCL2/MIP-2 were measured by Bio-Plex assay. Production of G-CSF and CXCL1/KC by IL-17-stimulated primary lung tissue cells were determined by ELISA, whereas IL-17-dependent signaling in lung tissue cells was analyzed by measuring Akt phosphorylation using immunoblot. RESULTS: We found that in contrast to single knock-out mice, p110γ/δ-/- mice exhibited significantly elevated neutrophil counts in blood, spleen, and lung. Increased granulocytic differentiation stages in the bone marrow of p110γ/δ-/- mice were paralleled by increased serum concentrations of G-CSF, CXCL1/KC, and CXCL2/MIP-2. As IL-17 induces neutrophilia via the induction of G-CSF and CXC chemokines, we measured IL-17 and IL-17-producing T cells. IL-17 serum concentrations and frequencies of IL-17+ splenic T cells were significantly increased in p110γ/δ-/- mice. Moreover, IFN-γ+, IL-4+, and IL-5+ T cell subsets were drastically increased in p110γ/δ-/- mice, suggesting that IL-17+ T cells were up-regulated in the context of a general percentage increase of other cytokine producing T cell subsets. CONCLUSIONS: We found that p110γ/δ deficiency in mice induces complex immunological changes, which might in concert contribute to neutrophilia. These findings emphasize a crucial but indirect role of both p110γ and p110δ in the regulation of neutrophil homeostasis.


Assuntos
Transtornos Leucocíticos/genética , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinases/deficiência , Animais , Células Cultivadas , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Homeostase , Interleucina-17/metabolismo , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , Transtornos Leucocíticos/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Baço/metabolismo , Linfócitos T/metabolismo
6.
Am J Med Genet A ; 170(3): 622-33, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26663319

RESUMO

The cause of posterior urethral valves (PUV) is unknown, but genetic factors are suspected given their familial occurrence. We examined cases of isolated PUV to identify novel copy number variants (CNVs). We identified 56 cases of isolated PUV from all live-births in New York State (1998-2005). Samples were genotyped using Illumina HumanOmni2.5 microarrays. Autosomal and sex-linked CNVs were identified using PennCNV and cnvPartition software. CNVs were prioritized for follow-up if they were absent from in-house controls, contained ≥ 10 consecutive probes, were ≥ 20 Kb in size, had ≤ 20% overlap with variants detected in other birth defect phenotypes screened in our lab, and were rare in population reference controls. We identified 47 rare candidate PUV-associated CNVs in 32 cases; one case had a 3.9 Mb deletion encompassing BMP7. Mutations in BMP7 have been associated with severe anomalies in the mouse urethra. Other interesting CNVs, each detected in a single PUV case included: a deletion of PIK3R3 and TSPAN1, duplication/triplication in FGF12, duplication of FAT1--a gene essential for normal growth and development, a large deletion (>2 Mb) on chromosome 17q that involves TBX2 and TBX4, and large duplications (>1 Mb) on chromosomes 3q and 6q. Our finding of previously unreported novel CNVs in PUV suggests that genetic factors may play a larger role than previously understood. Our data show a potential role of CNVs in up to 57% of cases examined. Investigation of genes in these CNVs may provide further insights into genetic variants that contribute to PUV.


Assuntos
Proteína Morfogenética Óssea 7/genética , Caderinas/genética , Variações do Número de Cópias de DNA , Fatores de Crescimento de Fibroblastos/genética , Fosfatidilinositol 3-Quinases/genética , Deleção de Sequência , Tetraspaninas/genética , Estreitamento Uretral/genética , Sequência de Bases , Proteína Morfogenética Óssea 7/deficiência , Caderinas/deficiência , Estudos de Casos e Controles , Pré-Escolar , Cromossomos Humanos Par 17 , Cromossomos Humanos Par 3 , Cromossomos Humanos Par 6 , Hibridização Genômica Comparativa , Fatores de Crescimento de Fibroblastos/deficiência , Expressão Gênica , Genótipo , Humanos , Lactente , Masculino , Dados de Sequência Molecular , New York/epidemiologia , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Fosfatidilinositol 3-Quinases/deficiência , Polimorfismo de Nucleotídeo Único , Tetraspaninas/deficiência , Uretra/metabolismo , Uretra/patologia , Estreitamento Uretral/diagnóstico , Estreitamento Uretral/epidemiologia , Estreitamento Uretral/patologia
7.
Arterioscler Thromb Vasc Biol ; 35(2): 368-77, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25503990

RESUMO

OBJECTIVE: An aneurysm is an inflammatory vascular condition. Phosphatidylinositol 3-kinases δ is highly expressed in leukocytes, and play a key role in innate immunity. However, the link between phosphatidylinositol 3-kinases δ and aneurysm development has not yet been elucidated. APPROACH AND RESULTS: Carotid ligation unexpectedly induced characteristic aneurysm formation beneath the ligation point in p110δ(D910A/D910A) mice (n=25; P<0.001 versus wild-type). Besides, p110δ inactivation exacerbated CaCl2-induced abdominal aortic aneurysms development. A reverse transcription polymerase chain reaction microarray revealed significant extracellular matrix components degradation and matrix metalloproteinases (MMPs) upregulation in the abdominal aorta of p110δ(D910A/D910A) mice. Similarly, the expression of both collagen I and IV was significantly decreased (n=10; P<0.05 versus wild-type) in carotid artery. Western blot assay confirmed that MMP-12 was significantly upregulated in arteries of p110δ(D910A/D910A) mice (n=10; P<0.01 versus wild-type). In vitro, p110δ inactivation marked increase peritoneal macrophages recruitment and synergistically enhance tumor necrosis factor-α-induced recruitment. A specific phosphatidylinositol 3-kinases δ inhibitor (IC87114) or genetic p110δ inactivation upregulated MMP-12 expression and c-Jun phosphorylation (n=6; P<0.05 versus wild-type macrophages). IC87114 also increased activator protein-1 DNA-binding activity (n=6; P<0.001 versus control) and enhanced the effect of tumor necrosis factor-α on activator protein-1-binding activity (n=5; P<0.01 versus tumor necrosis factor-α treatment groups). Knockdown of c-Jun suppressed the effect of the IC87114 and tumor necrosis factor-α on MMP-12 mRNA expression (n=5 in each group; P<0.01 versus scrRNA treatment groups). CONCLUSIONS: Our findings demonstrate that p110δ inactivation leads to extracellular matrix degradation in vessels and promotes aneurysm development by inducing macrophages migration and upregulating the activator protein-1/MMP-12 pathway in macrophages.


Assuntos
Aorta Abdominal/enzimologia , Aneurisma da Aorta Abdominal/enzimologia , Lesões das Artérias Carótidas/enzimologia , Artéria Carótida Primitiva/enzimologia , Macrófagos Peritoneais/enzimologia , Metaloproteinase 12 da Matriz/metabolismo , Fosfatidilinositol 3-Quinases/deficiência , Fator de Transcrição AP-1/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Aorta Abdominal/efeitos dos fármacos , Aorta Abdominal/patologia , Aorta Abdominal/cirurgia , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/patologia , Cloreto de Cálcio , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Artéria Carótida Primitiva/efeitos dos fármacos , Artéria Carótida Primitiva/patologia , Artéria Carótida Primitiva/cirurgia , Linhagem Celular , Classe I de Fosfatidilinositol 3-Quinases , Modelos Animais de Doenças , Ativação Enzimática , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Regulação Enzimológica da Expressão Gênica , Ligadura , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Interferência de RNA , Transdução de Sinais , Fator de Transcrição AP-1/genética , Transfecção , Fator de Necrose Tumoral alfa/farmacologia
8.
Biochemistry (Mosc) ; 81(7): 691-9, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27449615

RESUMO

The intracellular PI3K-AKT-mTOR pathway is involved in regulation of numerous important cell processes including cell growth, differentiation, and metabolism. The PI3Kα isoform has received particular attention as a novel molecular target in gene therapy, since this isoform plays critical roles in tumor progression and tumor blood flow and angiogenesis. However, the role of PI3Kα and other class I isoforms, i.e. PI3Kß, γ, δ, in the regulation of vascular tone and regional blood flow are largely unknown. We used novel isoform-specific PI3K inhibitors and mice deficient in both PI3Kγ and PI3Kδ (Pik3cg(-/-)/Pik3cd(-/-)) to define the putative contribution of PI3K isoform(s) to arterial vasoconstriction. Wire myography was used to measure isometric contractions of isolated murine mesenteric arterial rings. Phenylephrine-dependent contractions were inhibited by the pan PI3K inhibitors wortmannin (100 nM) and LY294002 (10 µM). These vasoconstrictions were also inhibited by the PI3Kα isoform inhibitors A66 (10 µM) and PI-103 (1 µM), but not by the PI3Kß isoform inhibitor TGX 221 (100 nM). Pik3cg(-/-)/Pik3cd(-/-)-arteries showed normal vasoconstriction. We conclude that PI3Kα is an important downstream element in vasoconstrictor GPCR signaling, which contributes to arterial vasocontraction via α1-adrenergic receptors. Our results highlight a regulatory role of PI3Kα in the cardiovascular system, which widens the spectrum of gene therapy approaches targeting PI3Kα in cancer cells and tumor angiogenesis and regional blood flow.


Assuntos
Fosfatidilinositol 3-Quinases/metabolismo , Androstadienos/farmacologia , Animais , Cromonas/farmacologia , Classe I de Fosfatidilinositol 3-Quinases , Classe Ib de Fosfatidilinositol 3-Quinase/deficiência , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Furanos/farmacologia , Artérias Mesentéricas/fisiologia , Camundongos , Camundongos Knockout , Morfolinas/farmacologia , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neoplasias/terapia , Neovascularização Patológica , Fenilefrina/farmacologia , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Piridinas/farmacologia , Pirimidinas/farmacologia , Pirimidinonas/farmacologia , Receptores Adrenérgicos alfa 1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Wortmanina
9.
Eur J Immunol ; 44(11): 3353-67, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25124254

RESUMO

Alternatively activated or M2 macrophages have been reported to protect mice from intestinal inflammation, but the mechanism of protection has not been elucidated. In this study, we demonstrate that mice deficient in the p110δ catalytic subunit activity of class I phosphatidylinositol 3-kinase (PI3Kp110δ) have increased clinical disease activity and histological damage during dextran sodium sulfate (DSS) induced colitis. Increased disease severity in PI3Kp110δ-deficient mice is dependent on professional phagocytes and correlates with reduced numbers of arginase I+ M2 macrophages in the colon and increased production of inflammatory nitric oxide. We further demonstrate that PI3Kp110δ-deficient macrophages are defective in their ability to induce arginase I when skewed to an M2 phenotype with IL-4. Importantly, adoptive transfer of IL-4-treated macrophages derived from WT mice, but not those from PI3Kp110δ-deficient mice, protects mice during DSS-induced colitis. Moreover, M2 macrophages mediated protection is lost when mice are cotreated with inhibitors that block arginase activity or during adoptive transfer of arginase I deficient M2 macrophages. Taken together, our data demonstrate that arginase I activity is required for M2 macrophages mediated protection during DSS-induced colitis in PI3Kp110δ-deficient mice.


Assuntos
Arginase/biossíntese , Colite/patologia , Macrófagos/enzimologia , Macrófagos/imunologia , Fosfatidilinositol 3-Quinases/genética , Transferência Adotiva , Animais , Arginase/antagonistas & inibidores , Classe I de Fosfatidilinositol 3-Quinases , Colite/induzido quimicamente , Colite/imunologia , Colo/imunologia , Colo/patologia , Sulfato de Dextrana , Inflamação/imunologia , Inflamação/patologia , Interleucina-4/farmacologia , Ativação de Macrófagos/imunologia , Macrófagos/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Óxido Nítrico/biossíntese , Fosfatidilinositol 3-Quinases/deficiência
10.
Arterioscler Thromb Vasc Biol ; 34(8): 1681-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24903091

RESUMO

OBJECTIVE: Platelet hyperactivity is a contributing factor in the pathogenesis of cardiovascular disease and can be induced by elevated levels of circulating growth factors, such as insulin-like growth factor-1 (IGF-1). IGF-1 is a primer that cannot stimulate platelet activation by itself, but in combination with physiological stimuli can potentiate platelet functional responses via a phosphoinositide 3-kinase-dependent mechanism. In this study, we explored the role of the phosphoinositide 3-kinase p110α isoform in IGF-1-mediated enhancement of platelet function. APPROACH AND RESULTS: Using a platelet-specific p110α knockout murine model, we demonstrate that genetic deletion, similar to pharmacological inactivation of p110α, did not affect proteinase-activated receptor 4 signaling to Akt/protein kinase B but significantly reduced IGF-1-mediated Akt phosphorylation. The p110ß inhibitor TGX-221 abolished IGF-1-induced Akt phosphorylation in p110α-deficient platelets, demonstrating that both p110α and p110ß contribute to IGF-1-mediated Akt phosphorylation. Genetic deletion of p110α had no effect on IGF-1-mediated increases in thrombus formation on collagen and enhancement of proteinase-activated receptor 4-mediated integrin activation and α-granule secretion. In contrast, pharmacological inhibition of p110α blocked IGF-1-mediated potentiation of integrin activation and α-granule secretion. Functional enhancement by IGF-1 in p110α knockout samples was lost after TGX-221 treatment, suggesting that p110ß drives priming in the absence of the p110α isoform. CONCLUSIONS: Together, these results demonstrate that both p110α and p110ß are involved in Akt signaling by IGF-1, but that it is the p110α isoform that is responsible for IGF-1-mediated potentiation of platelet function.


Assuntos
Plaquetas/enzimologia , Fator de Crescimento Insulin-Like I/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ativação Plaquetária , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases , Integrina alfa2/metabolismo , Integrina beta3/metabolismo , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Ativação Plaquetária/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Receptores de Trombina/metabolismo , Vesículas Secretórias/metabolismo , Transdução de Sinais , Trombose/sangue , Trombose/enzimologia , Fatores de Tempo
11.
J Immunol ; 191(1): 395-406, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23733875

RESUMO

Cyclooxygenase (Cox)-2 dependent PGs modulate several functions in many pathophysiological processes, including migration of immune cells. In this study, we addressed the role of Cox-2 in macrophage migration by using in vivo and in vitro models. Upon thioglycolate challenge, CD11b(+) F4/80(+) macrophages showed a diminished ability to migrate to the peritoneal cavity in cox-2(-/-) mice. In vivo migration of cox-2(-/-) macrophages from the peritoneal cavity to lymph nodes, as well as cell adhesion to the mesothelium, was reduced in response to LPS. In vitro migration of cox-2(-/-) macrophages toward MCP-1, RANTES, MIP-1α, or MIP-1ß, as well as cell adhesion to ICAM-1 or fibronectin, was impaired. Defects in cell migration were not due to changes in chemokine receptor expression. Remarkably, cox-2(-/-) macrophages showed a deficiency in focal adhesion formation, with reduced phosphorylation of paxillin (Tyr(188)). Interestingly, expression of the p110γ catalytic subunit of PI3K was severely reduced in the absence of Cox-2, leading to defective Akt phosphorylation, as well as cdc42 and Rac-1 activation. Our results indicate that the paxillin/p110γ-PI3K/Cdc42/Rac1 axis is defective in cox-2(-/-) macrophages, which results in impaired cell adhesion and migration.


Assuntos
Inibição de Migração Celular/imunologia , Classe Ib de Fosfatidilinositol 3-Quinase/deficiência , Ciclo-Oxigenase 2/deficiência , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/imunologia , Fosfatidilinositol 3-Quinases/deficiência , Transdução de Sinais/imunologia , Animais , Adesão Celular/genética , Adesão Celular/imunologia , Inibição de Migração Celular/genética , Células Cultivadas , Classe Ib de Fosfatidilinositol 3-Quinase/fisiologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/fisiologia , Macrófagos Peritoneais/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Imunológicos , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/genética , Proteína cdc42 de Ligação ao GTP/deficiência , Proteínas rac1 de Ligação ao GTP/deficiência
12.
Nature ; 454(7205): 776-9, 2008 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-18594509

RESUMO

On activation by receptors, the ubiquitously expressed class IA isoforms (p110alpha and p110beta) of phosphatidylinositol-3-OH kinase (PI(3)K) generate lipid second messengers, which initiate multiple signal transduction cascades. Recent studies have demonstrated specific functions for p110alpha in growth factor and insulin signalling. To probe for distinct functions of p110beta, we constructed conditional knockout mice. Here we show that ablation of p110beta in the livers of the resulting mice leads to impaired insulin sensitivity and glucose homeostasis, while having little effect on phosphorylation of Akt, suggesting the involvement of a kinase-independent role of p110beta in insulin metabolic action. Using established mouse embryonic fibroblasts, we found that removal of p110beta also had little effect on Akt phosphorylation in response to stimulation by insulin and epidermal growth factor, but resulted in retarded cell proliferation. Reconstitution of p110beta-null cells with a wild-type or kinase-dead allele of p110beta demonstrated that p110beta possesses kinase-independent functions in regulating cell proliferation and trafficking. However, the kinase activity of p110beta was required for G-protein-coupled receptor signalling triggered by lysophosphatidic acid and had a function in oncogenic transformation. Most strikingly, in an animal model of prostate tumour formation induced by Pten loss, ablation of p110beta (also known as Pik3cb), but not that of p110alpha (also known as Pik3ca), impeded tumorigenesis with a concomitant diminution of Akt phosphorylation. Taken together, our findings demonstrate both kinase-dependent and kinase-independent functions for p110beta, and strongly indicate the kinase-dependent functions of p110beta as a promising target in cancer therapy.


Assuntos
Proliferação de Células , Transformação Celular Neoplásica , Glucose/metabolismo , Insulina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Fibroblastos/citologia , Intolerância à Glucose/enzimologia , Intolerância à Glucose/genética , Homeostase , Humanos , Insulina/farmacologia , Resistência à Insulina/genética , Fígado/enzimologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
13.
J Exp Med ; 204(3): 497-510, 2007 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-17325199

RESUMO

Recent observations using multiphoton intravital microscopy (MP-IVM) have uncovered an unexpectedly high lymphocyte motility within peripheral lymph nodes (PLNs). Lymphocyte-expressed intracellular signaling molecules governing interstitial movement remain largely unknown. Here, we used MP-IVM of murine PLNs to examine interstitial motility of lymphocytes lacking the Rac guanine exchange factor DOCK2 and phosphoinositide-3-kinase (PI3K)gamma, signaling molecules that act downstream of G protein-coupled receptors, including chemokine receptors (CKRs). T and B cells lacking DOCK2 alone or DOCK2 and PI3Kgamma displayed markedly reduced motility inside T cell area and B cell follicle, respectively. Lack of PI3Kgamma alone had no effect on migration velocity but resulted in increased turning angles of T cells. As lymphocyte egress from PLNs requires the sphingosine-1-phosphate (S1P) receptor 1, a G(alphai) protein-coupled receptor similar to CKR, we further analyzed whether DOCK2 and PI3Kgamma contributed to S1P-triggered signaling events. S1P-induced cell migration was significantly reduced in T and B cells lacking DOCK2, whereas T cell-expressed PI3Kgamma contributed to F-actin polymerization and protein kinase B phosphorylation but not migration. These findings correlated with delayed lymphocyte egress from PLNs in the absence of DOCK2 but not PI3Kgamma, and a markedly reduced cell motility of DOCK2-deficient T cells in close proximity to efferent lymphatic vessels. In summary, our data support a central role for DOCK2, and to a lesser extent T cell-expressed PI3Kgamma, for signal transduction during interstitial lymphocyte migration and S1P-mediated egress.


Assuntos
Movimento Celular/imunologia , Proteínas Ativadoras de GTPase/fisiologia , Linfócitos/citologia , Linfócitos/metabolismo , Lisofosfolipídeos/fisiologia , Esfingosina/análogos & derivados , Animais , Comunicação Celular/genética , Comunicação Celular/imunologia , Classe Ib de Fosfatidilinositol 3-Quinase , Proteínas Ativadoras de GTPase/deficiência , Proteínas Ativadoras de GTPase/genética , Fatores de Troca do Nucleotídeo Guanina , Isoenzimas/deficiência , Isoenzimas/genética , Linfonodos/citologia , Linfonodos/imunologia , Linfonodos/metabolismo , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Esfingosina/fisiologia
14.
Proc Natl Acad Sci U S A ; 107(12): 5471-6, 2010 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-20212113

RESUMO

The phosphatidylinositol 3-kinase (PI3K) signaling pathway is deregulated in many human diseases including cancer, diabetes, obesity, and autoimmunity. PI3K consists of a p110 catalytic protein and a p85alpha regulatory protein, required for the stabilization and localization of p110-PI3K activity. The p110-PI3K enzyme generates the key signaling lipid phosphatidylinositol 3,4,5-trisphosphate, which is dephosphorylated by the PI3-phosphatase PTEN. Here we show another function for the p85alpha regulatory protein: it binds directly to and enhances PTEN lipid phosphatase activity. We demonstrate that ectopically expressed FLAG-tagged p85 coimmunoprecipitates endogenous PTEN in an epidermal growth factor dependent manner. We also show epidermal growth factor dependent coimmunoprecipitation of endogenous p85 and PTEN proteins in HeLa cells. Thus p85 regulates both p110-PI3K and PTEN-phosphatase enzymes through direct interaction. This finding underscores the need for caution in analyzing PI3K activity because anti-p85 immunoprecipitations may contain both p85:p110-PI3K and p85:PTEN-phosphatase enzymes and thus measure net PI3K activity. We identify the N-terminal SH3-BH region of p85alpha, absent in the smaller p55alpha and p50alpha isoforms, as the region that mediates PTEN binding and regulation. Cellular expression of p85DeltaSH3-BH results in substantially increased magnitude and duration of pAkt levels in response to growth factor stimulation. The ability of p85 to bind and directly regulate both p110-PI3K and PTEN-PI3-phosphatase allows us to explain the paradoxical insulin signaling phenotypes observed in mice with reduced PI3K or PTEN proteins. This discovery will impact ongoing studies using therapeutics targeting the PI3K/PTEN/Akt pathway.


Assuntos
PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Células COS , Chlorocebus aethiops , Fator de Crescimento Epidérmico/metabolismo , Células HeLa , Humanos , Insulina/metabolismo , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/química , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fenótipo , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Transdução de Sinais
15.
Proc Natl Acad Sci U S A ; 107(29): 12901-6, 2010 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-20616072

RESUMO

G protein-coupled receptor-regulated PI3Kgamma is abundantly expressed in myeloid cells and has been implicated as a promising drug target to treat various inflammatory diseases. However, its role in bone homeostasis has not been investigated, despite the fact that osteoclasts are derived from myeloid lineage. We therefore carried out thorough bone phenotypic characterization of a PI3Kgamma-deficient mouse line and found that PI3Kgamma-deficient mice had high bone mass. Our analyses further revealed that PI3Kgamma deficiency did not affect bone formation because no significant changes in osteoblast number and bone formation rate were observed. Instead, the lack of PI3Kgamma was associated with decreased bone resorption, as evidenced by decreased osteoclast number in vivo and impaired osteoclast formation in vitro. The decreased osteoclast formation was accompanied by down-regulated expression of osteoclastogenic genes, compromised chemokine receptor signaling, and an increase in apoptosis during osteoclast differentiation. Together, these data suggest that PI3Kgamma regulates bone homeostasis by modulating osteoclastogenesis. Our study also suggests that inhibition of PI3Kgamma, which is being considered as a potential therapeutic strategy for treating chronic inflammatory disorders, may result in an increase in bone mass.


Assuntos
Osso e Ossos/enzimologia , Homeostase , Osteoclastos/enzimologia , Osteogênese , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/citologia , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Contagem de Células , Quimiocinas/farmacologia , Classe Ib de Fosfatidilinositol 3-Quinase , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Deleção de Genes , Homeostase/efeitos dos fármacos , Isoenzimas/deficiência , Isoenzimas/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/deficiência , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo
16.
Nat Genet ; 26(3): 379-82, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11062485

RESUMO

Phosphoinositide 3-kinases produce 3'-phosphorylated phosphoinositides that act as second messengers to recruit other signalling proteins to the membrane. Pi3ks are activated by many extracellular stimuli and have been implicated in a variety of cellular responses. The Pi3k gene family is complex and the physiological roles of different classes and isoforms are not clear. The gene Pik3r1 encodes three proteins (p85 alpha, p55 alpha and p50 alpha) that serve as regulatory subunits of class IA Pi3ks (ref. 2). Mice lacking only the p85 alpha isoform are viable but display hypoglycaemia and increased insulin sensitivity correlating with upregulation of the p55 alpha and p50 alpha variants. Here we report that loss of all protein products of Pik3r1 results in perinatal lethality. We observed, among other abnormalities, extensive hepatocyte necrosis and chylous ascites. We also noted enlarged skeletal muscle fibres, brown fat necrosis and calcification of cardiac tissue. In liver and muscle, loss of the major regulatory isoform caused a great decrease in expression and activity of class IA Pi3k catalytic subunits; nevertheless, homozygous mice still displayed hypoglycaemia, lower insulin levels and increased glucose tolerance. Our findings reveal that p55 alpha and/or p50 alpha are required for survival, but not for development of hypoglycaemia, in mice lacking p85 alpha.


Assuntos
Anormalidades Múltiplas/genética , Ascite Quilosa/genética , Genes Letais , Hipoglicemia/genética , Fígado/patologia , Fosfatidilinositol 3-Quinases/deficiência , Isoformas de Proteínas/deficiência , Tecido Adiposo Marrom/patologia , Animais , Animais não Endogâmicos , Calcinose/genética , Cardiomiopatias/genética , Catálise , Cruzamentos Genéticos , Dimerização , Indução Enzimática , Feminino , Genes , Genótipo , Vida Livre de Germes , Glucose/metabolismo , Glucose/farmacologia , Hipertrofia , Insulina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Fibras Musculares Esqueléticas/patologia , Necrose , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Processamento de Proteína Pós-Traducional/genética , Subunidades Proteicas , Sistemas do Segundo Mensageiro/genética
17.
Nat Genet ; 21(2): 230-5, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9988280

RESUMO

The hallmark of type 2 diabetes, the most common metabolic disorder, is a defect in insulin-stimulated glucose transport in peripheral tissues. Although a role for phosphoinositide-3-kinase (PI3K) activity in insulin-stimulated glucose transport and glucose transporter isoform 4 (Glut4) translocation has been suggested in vitro, its role in vivo and the molecular link between activation of PI3K and translocation has not yet been elucidated. To determine the role of PI3K in glucose homeostasis, we generated mice with a targeted disruption of the gene encoding the p85alpha regulatory subunit of PI3K (Pik3r1; refs 3-5). Pik3r1-/- mice showed increased insulin sensitivity and hypoglycaemia due to increased glucose transport in skeletal muscle and adipocytes. Insulin-stimulated PI3K activity associated with insulin receptor substrates (IRSs) was mediated via full-length p85 alpha in wild-type mice, but via the p50 alpha alternative splicing isoform of the same gene in Pik3r1-/- mice. This isoform switch was associated with an increase in insulin-induced generation of phosphatidylinositol(3,4,5)triphosphate (PtdIns(3,4,5)P3) in Pik3r1-/- adipocytes and facilitation of Glut4 translocation from the low-density microsome (LDM) fraction to the plasma membrane (PM). This mechanism seems to be responsible for the phenotype of Pik3r1-/- mice, namely increased glucose transport and hypoglycaemia. Our work provides the first direct evidence that PI3K and its regulatory subunit have a role in glucose homeostasis in vivo.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/deficiência , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Hipoglicemia/genética , Insulina/farmacologia , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Animais , Transporte Biológico/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Cruzamentos Genéticos , Desoxiglucose/metabolismo , Ativação Enzimática/genética , Glucose/metabolismo , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Camundongos , Camundongos Knockout , Músculo Esquelético/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Frações Subcelulares/enzimologia
18.
J Immunol ; 184(3): 1492-8, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20028656

RESUMO

High mobility group box 1 (HMGB1) is an alarmin actively secreted by immune cells and passively released by necrotic nonimmune cells. HMGB1 has been implicated in both cardiac contractile dysfunction and the lethality associated with sepsis/endotoxemia. The aim of the current study was to assess whether viable cardiomyocytes could produce HMGB1 and whether HMGB1 can affect myocardial contractility. LPS was used as a model of sepsis/endotoxemia in mice and isolated cardiac myocytes. LPS increased myocardial expression of HMGB1 in vivo (immunohistochemistry) and production and secretion of HMGB1 by viable cardiac myocytes in vitro (Western). LPS increased the phosphorylation status of PI3Kgamma in cardiac myocytes, an effect not observed in TLR4(-/-) myocytes. Genetic (PI3Kgamma(-/-)) or pharmacologic (AS605240) blockade of PI3Kgamma ameliorated the LPS-induced 1) cardiomyocyte production and secretion of HMGB1 in vitro and 2) HMGB1 expression in the myocardium in vivo. The LPS-induced depression of myocardial contractility was prevented by the HMGB1 antagonist, A-box. Genetic (PI3Kgamma(-/-)) or pharmacologic (AS605240) blockade of PI3Kgamma ameliorated the LPS-induced decrease in myocardial contractility. No evidence of inflammatory infiltrate was noted in any of the in vivo studies. The findings of the current study indicate that 1) LPS can induce HMGB1 secretion by viable cardiac myocytes through a TLR4/PI3Kgamma signaling pathway, and 2) HMGB1 plays a role in the LPS-induced myocardial contractile dysfunction. The results of the current study also have broader implications (i.e., that viable parenchymal cells, such as cardiac myocytes, participate in the alarmin response).


Assuntos
Cardiomiopatias/imunologia , Citocinas/biossíntese , Proteína HMGB1/biossíntese , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/fisiologia , Animais , Animais Recém-Nascidos , Cardiomiopatias/enzimologia , Cardiomiopatias/fisiopatologia , Sobrevivência Celular/imunologia , Células Cultivadas , Classe Ib de Fosfatidilinositol 3-Quinase , Citocinas/metabolismo , Citocinas/fisiologia , Modelos Animais de Doenças , Endotoxemia/enzimologia , Endotoxemia/imunologia , Endotoxemia/fisiopatologia , Proteína HMGB1/metabolismo , Proteína HMGB1/fisiologia , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/fisiologia , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica/genética , Contração Miocárdica/imunologia , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética
19.
J Immunol ; 184(6): 3098-105, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20154209

RESUMO

We showed previously that mice with an inactivating knockin mutation in the p110delta isoform of PI3K (referred to as p110delta(D910A) mice) displayed enhanced primary resistance to Leishmania major despite mounting paradoxically impaired T cell responses. In this study, we show that p110delta(D910A) mice are impaired in their secondary (memory) anti-Leishmania responses in vitro and in vivo. Following secondary L. major challenge, p110delta(D910A) mice exhibited reduced delayed-type hypersensitivity response and weaker parasite control compared to wild-type mice. Using adoptive transfer experiments, we show that immune T cells from healed p110delta(D910A) mice were impaired in their proliferation and effector cytokine (IFN-gamma) responses upon L. major challenge. Interestingly, Leishmania-reactive T cells from healed p110delta(D910A) mice contain severalfold lower numbers of CD62L(lo) and CD62(hi) T cells than those from healed wild-type mice. The reduction in numbers of CD62L(lo) T cells in p110delta(D910A) mice is due to failure of their CD62L(hi) T cells to downregulate CD62L expression in response to L. major. Furthermore, although CD62L(lo) cells from p110delta(D910A) mice could home efficiently to lymphoid organs, their ability to exit these tissues and emigrate to cutaneous sites of infection was greatly impaired. Collectively, our data identify PI3K signaling as important events that control memory T cell subset differentiation, generation, effector function, and recruitment to cutaneous tissues and suggest that manipulating this pathway could provide means of enhancing desired memory T cell subset, response during vaccination, or both.


Assuntos
Diferenciação Celular/imunologia , Imunização Secundária/métodos , Memória Imunológica , Leishmania major/imunologia , Leishmaniose Cutânea/enzimologia , Leishmaniose Cutânea/imunologia , Fosfatidilinositol 3-Quinases/fisiologia , Subpopulações de Linfócitos T/enzimologia , Subpopulações de Linfócitos T/imunologia , Animais , Diferenciação Celular/genética , Classe I de Fosfatidilinositol 3-Quinases , Citocinas/biossíntese , Citocinas/deficiência , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Técnicas de Introdução de Genes , Inativação Gênica/imunologia , Hipersensibilidade Tardia/enzimologia , Hipersensibilidade Tardia/imunologia , Hipersensibilidade Tardia/parasitologia , Memória Imunológica/genética , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/fisiologia , Leishmaniose Cutânea/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Vacinas Protozoárias/administração & dosagem , Vacinas Protozoárias/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/parasitologia
20.
Nature ; 441(7091): 366-70, 2006 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-16625210

RESUMO

The eight catalytic subunits of the mammalian phosphoinositide-3-OH kinase (PI(3)K) family form the backbone of an evolutionarily conserved signalling pathway; however, the roles of most PI(3)K isoforms in organismal physiology and disease are unknown. To delineate the role of p110alpha, a ubiquitously expressed PI(3)K involved in tyrosine kinase and Ras signalling, here we generated mice carrying a knockin mutation (D933A) that abrogates p110alpha kinase activity. Homozygosity for this kinase-dead p110alpha led to embryonic lethality. Mice heterozygous for this mutation were viable and fertile, but displayed severely blunted signalling via insulin-receptor substrate (IRS) proteins, key mediators of insulin, insulin-like growth factor-1 and leptin action. Defective responsiveness to these hormones led to reduced somatic growth, hyperinsulinaemia, glucose intolerance, hyperphagia and increased adiposity in mice heterozygous for the D933A mutation. This signalling function of p110alpha derives from its highly selective recruitment and activation to IRS signalling complexes compared to p110beta, the other broadly expressed PI(3)K isoform, which did not contribute to IRS-associated PI(3)K activity. p110alpha was the principal IRS-associated PI(3)K in cancer cell lines. These findings demonstrate a critical role for p110alpha in growth factor and metabolic signalling and also suggest an explanation for selective mutation or overexpression of p110alpha in a variety of cancers.


Assuntos
Crescimento/fisiologia , Insulina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Adiposidade , Animais , Peso Corporal , Domínio Catalítico , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Ingestão de Alimentos , Perda do Embrião/enzimologia , Perda do Embrião/genética , Perda do Embrião/metabolismo , Ativação Enzimática , Glucose/metabolismo , Heterozigoto , Homozigoto , Hiperinsulinismo/metabolismo , Proteínas Substratos do Receptor de Insulina , Leptina/metabolismo , Camundongos , Mutação/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Fosfoproteínas/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA