Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Metabolites ; 11(1)2021 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-33430006

RESUMO

The primacy of lipids as essential components of cellular membranes is conserved across taxonomic domains. In addition to this crucial role as a semi-permeable barrier, lipids are also increasingly recognized as important signaling molecules with diverse functional mechanisms ranging from cell surface receptor binding to the intracellular regulation of enzymatic cascades. In this review, we focus on ether lipids, an ancient family of lipids having ether-linked structures that chemically differ from their more prevalent acyl relatives. In particular, we examine ether lipid biosynthesis in the peroxisome of mammalian cells, the roles of selected glycerolipids and glycerophospholipids in signal transduction in both prokaryotes and eukaryotes, and finally, the potential therapeutic contributions of synthetic ether lipids to the treatment of cancer.

2.
Sci Rep ; 9(1): 16891, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31729453

RESUMO

We introduce machine learning (ML) to perform classification and quantitation of images of nuclei from human blood neutrophils. Here we assessed the use of convolutional neural networks (CNNs) using free, open source software to accurately quantitate neutrophil NETosis, a recently discovered process involved in multiple human diseases. CNNs achieved >94% in performance accuracy in differentiating NETotic from non-NETotic cells and vastly facilitated dose-response analysis and screening of the NETotic response in neutrophils from patients. Using only features learned from nuclear morphology, CNNs can distinguish between NETosis and necrosis and between distinct NETosis signaling pathways, making them a precise tool for NETosis detection. Furthermore, by using CNNs and tools to determine object dispersion, we uncovered differences in NETotic nuclei clustering between major NETosis pathways that is useful in understanding NETosis signaling events. Our study also shows that neutrophils from patients with sickle cell disease were unresponsive to one of two major NETosis pathways. Thus, we demonstrate the design, performance, and implementation of ML tools for rapid quantitative and qualitative cell analysis in basic science.


Assuntos
Diagnóstico por Imagem/métodos , Armadilhas Extracelulares/metabolismo , Processamento de Imagem Assistida por Computador/métodos , Aprendizado de Máquina , Neutrófilos/patologia , Morte Celular/fisiologia , Humanos , Necrose/metabolismo , Necrose/patologia , Redes Neurais de Computação , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia
3.
Blood Adv ; 3(22): 3818-3828, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31770438

RESUMO

We previously reported the discovery of a novel lipid deacetylase in platelets, arylacetamide deacetylase-like 1 (AADACL1/NCEH1), and that its inhibition impairs agonist-induced platelet aggregation, Rap1 GTP loading, protein kinase C (PKC) activation, and ex vivo thrombus growth. However, precise mechanisms by which AADACL1 impacts platelet signaling and function in vivo are currently unknown. Here, we demonstrate that AADACL1 regulates the accumulation of ether lipids that impact PKC signaling networks crucial for platelet activation in vitro and in vivo. Human platelets treated with the AADACL1 inhibitor JW480 or the AADACL1 substrate 1-O-hexadecyl-2-acetyl-sn-glycerol (HAG) exhibited decreased platelet aggregation, granule secretion, Ca2+ flux, and PKC phosphorylation. Decreased aggregation and secretion were rescued by exogenous adenosine 5'-diphosphate, indicating that AADACL1 likely functions to induce dense granule secretion. Experiments with P2Y12-/- and CalDAG GEFI-/- mice revealed that the P2Y12 pathway is the predominate target of HAG-mediated inhibition of platelet aggregation. HAG itself displayed weak agonist properties and likely mediates its inhibitory effects via conversion to a phosphorylated metabolite, HAGP, which directly interacted with the C1a domains of 2 distinct PKC isoforms and blocked PKC kinase activity in vitro. Finally, AADACL1 inhibition in rats reduced platelet aggregation, protected against FeCl3-induced arterial thrombosis, and delayed tail bleeding time. In summary, our data support a model whereby AADACL1 inhibition shifts the platelet ether lipidome to an inhibitory axis of HAGP accumulation that impairs PKC activation, granule secretion, and recruitment of platelets to sites of vascular damage.


Assuntos
Plaquetas/metabolismo , Metabolismo dos Lipídeos , Esterol Esterase/metabolismo , Trombose/etiologia , Trombose/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Modelos Biológicos , Fosforilação , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Testes de Função Plaquetária , Ligação Proteica , Proteína Quinase C/metabolismo , Ratos , Receptores Purinérgicos P2Y12/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esterol Esterase/antagonistas & inibidores , Especificidade por Substrato , Trombose/tratamento farmacológico
4.
Physiol Rep ; 5(5)2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28275112

RESUMO

We generated and studied CLIC1 null (C1KO) mice to investigate the physiological role of this protein. C1KO and matched wild-type (WT) mice were studied in two models of acute toxic tissue injury. CLIC1 expression is upregulated following acute injury of WT kidney and pancreas and is absent in C1KOs. Acute tissue injury is attenuated in the C1KOs and this correlates with an absence of the rise in tissue reactive oxygen species (ROS) that is seen in WT mice. Infiltration of injured tissue by inflammatory cells was comparable between WT and C1KOs. Absence of CLIC1 increased PMA-induced superoxide production by isolated peritoneal neutrophils but dramatically decreased PMA-induced superoxide production by peritoneal macrophages. CLIC1 is expressed in both neutrophils and macrophages in a peripheral pattern consistent with either plasma membrane or the cortical cytoskeleton in resting cells and redistributes away from the periphery following PMA stimulation in both cell types. Absence of CLIC1 had no effect on redistribution or dephosphorylation of Ezrin/ERM cytoskeleton in macrophages. Plasma membrane chloride conductance is altered in the absence of CLIC1, but not in a way that would be expected to block superoxide production. NADPH oxidase redistributes from an intracellular compartment to the plasma membrane when WT macrophages are stimulated to produce superoxide and this redistribution fails to occur in C1KO macrophages. We conclude that the role of CLIC1 in macrophage superoxide production is to support redistribution of NADPH oxidase to the plasma membrane, and not through major effects on ERM cytoskeleton or by acting as a plasma membrane chloride channel.


Assuntos
Injúria Renal Aguda/metabolismo , Canais de Cloreto/metabolismo , Macrófagos/metabolismo , Superóxidos/metabolismo , Injúria Renal Aguda/genética , Animais , Membrana Celular/metabolismo , Canais de Cloreto/genética , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Camundongos , Camundongos Knockout , NADPH Oxidases/metabolismo , Fosforilação , Espécies Reativas de Oxigênio/metabolismo
5.
Blood ; 128(23): 2597-2598, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27932328
6.
Angew Chem Int Ed Engl ; 55(3): 950-4, 2016 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-26636264

RESUMO

Although peptide-based therapeutics are finding increasing application in the clinic, extensive structural modification is typically required to prevent their rapid degradation by proteases in the blood. We have evaluated the ability of erythrocytes to serve as reservoirs, protective shields (against proteases), and light-triggered launch pads for peptides. We designed lipidated peptides that are anchored to the surface of red blood cells, which furnishes a protease-resistant environment. A photocleavable moiety is inserted between the lipid anchor and the peptide backbone, thereby enabling light-triggered peptide release from erythrocytes. We have shown that a cell-permeable peptide, a hormone (melanocyte stimulating hormone), and a blood-clotting agent can be anchored to erythrocytes, protected from proteases, and photolytically released to create the desired biological effect.


Assuntos
Membrana Celular/efeitos dos fármacos , Proteínas de Membrana/química , Peptídeos/uso terapêutico , Sequência de Aminoácidos , Membrana Celular/metabolismo , Células HEK293 , Células HeLa , Humanos , Peptídeos/química
7.
Chem Biol ; 20(9): 1125-34, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-23993462

RESUMO

A comprehensive knowledge of the platelet proteome is necessary for understanding thrombosis and for envisioning antiplatelet therapies. To discover other biochemical pathways in human platelets, we screened platelets with a carbamate library designed to interrogate the serine hydrolase subproteome and used competitive activity-based protein profiling to map the targets of active carbamates. We identified an inhibitor that targets arylacetamide deacetylase-like 1 (AADACL1), a lipid deacetylase originally identified in invasive cancers. Using this compound, along with highly selective second-generation inhibitors of AADACL1, metabolomics, and RNA interference, we show that AADACL1 regulates platelet aggregation, thrombus growth, RAP1 and PKC activation, lipid metabolism, and fibrinogen binding to platelets and megakaryocytes. These data provide evidence that AADACL1 regulates platelet and megakaryocyte activation and highlight the value of this chemoproteomic strategy for target discovery in platelets.


Assuntos
Plaquetas/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Carbamatos/química , Carbamatos/metabolismo , Carbamatos/farmacologia , Hidrolases de Éster Carboxílico/antagonistas & inibidores , Hidrolases de Éster Carboxílico/genética , Linhagem Celular , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Fibrinogênio/metabolismo , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Metabolômica , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Ligação Proteica , Proteína Quinase C/metabolismo , Proteômica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Esterol Esterase , Proteínas rap1 de Ligação ao GTP/metabolismo
8.
Cell Rep ; 3(3): 678-88, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23434512

RESUMO

MyD88, the intracellular adaptor of most TLRs, mediates either proinflammatory or immunosuppressive signaling that contributes to chronic inflammation-associated diseases. Although gene-specific chromatin modifications regulate inflammation, the role of MyD88 signaling in establishing such epigenetic landscapes under different inflammatory states remains elusive. Using quantitative proteomics to enumerate the inflammation-phenotypic constituents of the MyD88 interactome, we found that in endotoxin-tolerant macrophages, protein phosphatase 2A catalytic subunit α (PP2Ac) enhances its association with MyD88 and is constitutively activated. Knockdown of PP2Ac prevents suppression of proinflammatory genes and resistance to apoptosis. Through site-specific dephosphorylation, constitutively active PP2Ac disrupts the signal-promoting TLR4-MyD88 complex and broadly suppresses the activities of multiple proinflammatory/proapoptotic pathways as well, shifting proinflammatory MyD88 signaling to a prosurvival mode. Constitutively active PP2Ac translocated with MyD88 into the nuclei of tolerant macrophages establishes the immunosuppressive pattern of chromatin modifications and represses chromatin remodeling to selectively silence proinflammatory genes, coordinating the MyD88-dependent inflammation control at both signaling and epigenetic levels under endotoxin-tolerant conditions.


Assuntos
Epigênese Genética/imunologia , Tolerância Imunológica/genética , Lipopolissacarídeos/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Proteína Fosfatase 2/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Apoptose , Núcleo Celular/metabolismo , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Células HEK293 , Humanos , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/genética , Fenótipo , Fosforilação , Ligação Proteica , Proteína Fosfatase 2/genética , Proteoma/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
9.
Circ Res ; 111(5): e111-22, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22740088

RESUMO

RATIONALE: Mating type switching/sucrose non-fermenting (SWI/SNF) chromatin-remodeling complexes utilize either BRG1 or BRM as a catalytic subunit to alter nucleosome position and regulate gene expression. BRG1 is required for vascular endothelial cell (VEC) development and embryonic survival, whereas BRM is dispensable. OBJECTIVE: To circumvent embryonic lethality and study Brg1 function in adult tissues, we used conditional gene targeting. To evaluate possible Brg1-Brm redundancy, we analyzed Brg1 mutant mice on wild-type and Brm-deficient backgrounds. METHODS AND RESULTS: The inducible Mx1-Cre driver was used to mutate Brg1 in adult mice. These conditional-null mutants exhibited a tissue-specific phenotype and unanticipated functional compensation between Brg1 and Brm. Brg1 single mutants were healthy and had a normal lifespan, whereas Brg1/Brm double mutants exhibited cardiovascular defects and died within 1 month. BRG1 and BRM were required for the viability of VECs but not other cell types where both genes were also knocked out. The VEC phenotype was most evident in the heart, particularly in the microvasculature of the outer myocardium, and was recapitulated in primary cells ex vivo. VEC death resulted in vascular leakage, cardiac hemorrhage, secondary death of cardiomyocytes due to ischemia, and ventricular dissections. CONCLUSIONS: BRG1-catalyzed SWI/SNF complexes are particularly important in cardiovascular tissues. However, in contrast to embryonic development, in which Brm does not compensate, Brg1 is required in adult VECs only when Brm is also mutated. These results demonstrate for the first time that Brm functionally compensates for Brg1 in vivo and that there are significant changes in the relative importance of BRG1- and BRM-catalyzed SWI/SNF complexes during the development of an essential cell lineage.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , DNA Helicases/metabolismo , Células Endoteliais/metabolismo , Cardiopatias Congênitas/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Fatores Etários , Animais , Catálise , Morte Celular/fisiologia , Linhagem da Célula/fisiologia , Sobrevivência Celular/fisiologia , Proteínas Cromossômicas não Histona/genética , Vasos Coronários/embriologia , Vasos Coronários/metabolismo , Vasos Coronários/patologia , DNA Helicases/genética , Ecocardiografia , Células Endoteliais/patologia , Coração/embriologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Homeostase/fisiologia , Camundongos , Camundongos Transgênicos , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Proteínas Nucleares/genética , Derrame Pleural/genética , Derrame Pleural/metabolismo , Derrame Pleural/patologia , Fatores de Transcrição/genética
10.
Curr Drug Targets ; 12(12): 1859-70, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21718236

RESUMO

Platelets are dynamic blood cells that form life-threatening thrombi in response to a variety of pathological conditions such as atherosclerosis, diabetes, metastatic cancer, sickle cell disease and obesity. These thrombi can lead directly to myocardial infarction (MI), stroke and other thrombotic events that contribute to over a million deaths every year in the United States. Even though multiple, effective drugs have been developed to combat these pathologies by antagonizing platelet receptors and their ligands, clinical use of these drugs can result in serious bleeding consequences. With the advent of increasingly powerful and accessible systems biology approaches, however, new opportunities are available to identify novel platelet targets and elucidate potentially safer antiplatelet strategies. Here we provide an overview of some of these exciting systems approaches ranging from genomics to aptamer discovery and emphasize the importance of identifying and exploiting novel platelet targets for therapeutic benefit.


Assuntos
Plaquetas/fisiologia , Inibidores da Agregação Plaquetária/farmacologia , Biologia de Sistemas/métodos , Trombose/prevenção & controle , Animais , Plaquetas/efeitos dos fármacos , Descoberta de Drogas/tendências , Monitoramento de Medicamentos , Humanos , Terapia de Alvo Molecular/tendências , Farmacogenética/tendências , Inibidores da Agregação Plaquetária/efeitos adversos , Inibidores da Agregação Plaquetária/uso terapêutico , Proteômica/tendências , Medição de Risco/tendências , Transdução de Sinais/efeitos dos fármacos , Trombose/tratamento farmacológico , Trombose/etiologia , Pesquisa Translacional Biomédica
11.
Curr Proteomics ; 8(3): 216-228, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22308104

RESUMO

Human platelets are thought to express approximately 2000-3000 proteins, but post-translational modifications, alternatively spliced variants and a rich diversity of vertebrate domain architectures likely make this a conservative estimate. Even though rapidly advancing proteomic techniques have facilitated the identification of roughly one third of the platelet proteome, a combination of abundance-based and activity-based proteomics methodologies is needed for elucidation of platelet functional characteristics including the definition of a "core proteome" and recognition of diverse enzyme activity profiles associated with various physiological states. In this review, we describe the latest mass spectrometry-based techniques capable of providing some of these physiological details required for more comprehensive evaluation of the human platelet repertoire.

12.
Mol Biol Cell ; 16(5): 2458-69, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15772154

RESUMO

The Ras family GTPase, R-Ras, elicits important integrin-dependent cellular behaviors such as adhesion, spreading and migration. While oncogenic Ras GTPases and R-Ras share extensive sequence homology, R-Ras induces a distinct set of cellular behaviors. To explore the structural basis for these differences, we asked whether the unique N-terminal 26 amino acid extension of R-Ras was responsible for R-Ras-specific signaling events. Using a 32D mouse myeloid cell line, we show that full-length R-Ras activates Rac and induces Rac-dependent cell spreading. In contrast, truncated R-Ras lacking its first 26 amino acids fails to activate Rac, resulting in reduced cell spreading. Truncated R-Ras also stimulates more beta3 integrin-dependent cell migration than full-length R-Ras, suggesting that the N-terminus may negatively regulate cell movement. However, neither the subcellular localization of R-Ras nor its effects on cell adhesion are affected by the presence or absence of the N-terminus. These results indicate that the N-terminus of R-Ras positively regulates specific R-Ras functions such as Rac activation and cell spreading but negatively regulates R-Ras-mediated cell migration.


Assuntos
Movimento Celular/fisiologia , GTP Fosfo-Hidrolases/química , GTP Fosfo-Hidrolases/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas ras/química , Proteínas ras/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Adesão Celular , Linhagem Celular , Membrana Celular/metabolismo , DNA/genética , GTP Fosfo-Hidrolases/genética , Camundongos , Dados de Sequência Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Deleção de Sequência , Proteínas ras/genética , Proteínas rho de Ligação ao GTP/metabolismo
13.
J Biol Chem ; 277(13): 10813-23, 2002 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-11799108

RESUMO

Cytosolic GTP-bound Ras has been shown to act as a dominant negative (DN) inhibitor of Ras by sequestering Raf in non-productive cytosolic complexes. Nevertheless, this distinct class of DN mutants has been neither well characterized nor extensively used to analyze Ras signaling. In contrast, DN Ras17N, which functions by blocking Ras guanine nucleotide exchange factors, has been well characterized and is widely used. Cytosolic GTP-bound Ras mutants could be used to inhibit particular Ras effectors by introducing additional mutations (T35S, E37G or Y40C) that permit them to associate selectively with and inhibit Raf, RalGDS, or phosphoinositide 3-kinase, respectively. When the wild-type Ras effector binding region is used, cytosolic Ras should associate with all Ras effectors, even those that are not yet identified, making these DN Ras mutants effective inhibitors of multiple Ras functions. We generated cytosolic GTP-bound H-, N-, and K-Ras, and we assessed their ability to inhibit Ras-induced phenotypes. In fibroblasts, cytosolic H-, N-, and K-Ras inhibited Ras-induced Elk-1 activation and focus formation, induced a flattened cell morphology, and increased adhesion to fibronectin through modulation of a beta(1)-subunit-containing integrin, thereby demonstrating that DN activity is not limited to a subset of Ras isoforms. We also generated cytosolic GTP-bound Ras effector domain mutants (EDMs), each of which reduced the ability of cytosolic GTP-bound Ras proteins to inhibit Elk-1 activation and to induce cell flattening, implicating multiple pathways in these phenotypes. In contrast, Ras-induced focus formation, platelet-derived growth factor (PDGF)-, or Ras-induced phospho-Akt levels and cell adhesion to fibronectin were affected by T35S and Y40C EDMs, whereas PDGF- or Ras-induced phospho-Erk levels were affected only by the T35S EDM, implying that a more limited set of Ras-mediated pathways participate in these phenotypes. These data constitute the first extensive characterization of this functionally distinct class of DN Ras inhibitor proteins.


Assuntos
Adesão Celular , Citosol/metabolismo , Proteínas de Ligação a DNA , Genes Dominantes , Guanosina Trifosfato/metabolismo , Mutação , Transdução de Sinais , Fatores de Transcrição , Proteínas ras/metabolismo , Células 3T3 , Animais , Fibronectinas/metabolismo , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Elk-1 do Domínio ets , Proteínas ras/antagonistas & inibidores , Proteínas ras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA