Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 21(3): 298-308, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32066953

RESUMEN

Depleting regulatory T cells (Treg cells) to counteract immunosuppressive features of the tumor microenvironment (TME) is an attractive strategy for cancer treatment; however, autoimmunity due to systemic impairment of their suppressive function limits its therapeutic potential. Elucidating approaches that specifically disrupt intratumoral Treg cells is direly needed for cancer immunotherapy. We found that CD36 was selectively upregulated in intrautumoral Treg cells as a central metabolic modulator. CD36 fine-tuned mitochondrial fitness via peroxisome proliferator-activated receptor-ß signaling, programming Treg cells to adapt to a lactic acid-enriched TME. Genetic ablation of Cd36 in Treg cells suppressed tumor growth accompanied by a decrease in intratumoral Treg cells and enhancement of antitumor activity in tumor-infiltrating lymphocytes without disrupting immune homeostasis. Furthermore, CD36 targeting elicited additive antitumor responses with anti-programmed cell death protein 1 therapy. Our findings uncover the unexplored metabolic adaptation that orchestrates the survival and functions of intratumoral Treg cells, and the therapeutic potential of targeting this pathway for reprogramming the TME.


Asunto(s)
Antígenos CD36/inmunología , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Animales , Apoptosis/inmunología , Antígenos CD36/deficiencia , Antígenos CD36/genética , Línea Celular Tumoral , Femenino , Homeostasis/inmunología , Humanos , Inmunoterapia , Metabolismo de los Lípidos/genética , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/metabolismo , Neoplasias/patología , PPAR-beta/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Microambiente Tumoral/inmunología
2.
Cell ; 165(1): 111-124, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26972052

RESUMEN

Normal platelet function is critical to blood hemostasis and maintenance of a closed circulatory system. Heightened platelet reactivity, however, is associated with cardiometabolic diseases and enhanced potential for thrombotic events. We now show gut microbes, through generation of trimethylamine N-oxide (TMAO), directly contribute to platelet hyperreactivity and enhanced thrombosis potential. Plasma TMAO levels in subjects (n > 4,000) independently predicted incident (3 years) thrombosis (heart attack, stroke) risk. Direct exposure of platelets to TMAO enhanced sub-maximal stimulus-dependent platelet activation from multiple agonists through augmented Ca(2+) release from intracellular stores. Animal model studies employing dietary choline or TMAO, germ-free mice, and microbial transplantation collectively confirm a role for gut microbiota and TMAO in modulating platelet hyperresponsiveness and thrombosis potential and identify microbial taxa associated with plasma TMAO and thrombosis potential. Collectively, the present results reveal a previously unrecognized mechanistic link between specific dietary nutrients, gut microbes, platelet function, and thrombosis risk.


Asunto(s)
Plaquetas/metabolismo , Microbioma Gastrointestinal , Metilaminas/metabolismo , Trombosis/metabolismo , Animales , Calcio/metabolismo , Traumatismos de las Arterias Carótidas/patología , Ciego/microbiología , Cloruros , Colina/metabolismo , Dieta , Femenino , Compuestos Férricos , Vida Libre de Gérmenes , Humanos , Metilaminas/sangre , Ratones , Ratones Endogámicos C57BL , Trombosis/patología
3.
Circ Res ; 125(12): 1087-1102, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31625810

RESUMEN

RATIONALE: A hallmark of chronic inflammatory disorders is persistence of proinflammatory macrophages in diseased tissues. In atherosclerosis, this is associated with dyslipidemia and oxidative stress, but mechanisms linking these phenomena to macrophage activation remain incompletely understood. OBJECTIVE: To investigate mechanisms linking dyslipidemia, oxidative stress, and macrophage activation through modulation of immunometabolism and to explore therapeutic potential targeting specific metabolic pathways. METHODS AND RESULTS: Using a combination of biochemical, immunologic, and ex vivo cell metabolic studies, we report that CD36 mediates a mitochondrial metabolic switch from oxidative phosphorylation to superoxide production in response to its ligand, oxidized LDL (low-density lipoprotein). Mitochondrial-specific inhibition of superoxide inhibited oxidized LDL-induced NF-κB (nuclear factor-κB) activation and inflammatory cytokine generation. RNA sequencing, flow cytometry, 3H-labeled palmitic acid uptake, lipidomic analysis, confocal and electron microscopy imaging, and functional energetics revealed that oxidized LDL upregulated effectors of long-chain fatty acid uptake and mitochondrial import, while downregulating fatty acid oxidation and inhibiting ATP5A (ATP synthase F1 subunit alpha)-an electron transport chain component. The combined effect is long-chain fatty acid accumulation, alteration of mitochondrial structure and function, repurposing of the electron transport chain to superoxide production, and NF-κB activation. Apoe null mice challenged with high-fat diet showed similar metabolic changes in circulating Ly6C+ monocytes and peritoneal macrophages, along with increased CD36 expression. Moreover, mitochondrial reactive oxygen species were positively correlated with CD36 expression in aortic lesional macrophages. CONCLUSIONS: These findings reveal that oxidized LDL/CD36 signaling in macrophages links dysregulated fatty acid metabolism to oxidative stress from the mitochondria, which drives chronic inflammation. Thus, targeting to CD36 and its downstream effectors may serve as potential new strategies against chronic inflammatory diseases such as atherosclerosis.


Asunto(s)
Antígenos CD36/deficiencia , Reprogramación Celular/fisiología , Macrófagos/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Animales , Antígenos CD36/genética , Células Cultivadas , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Masculino , Metabolismo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética
4.
Arterioscler Thromb Vasc Biol ; 39(2): 263-275, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30567481

RESUMEN

Objective- Dysregulated proliferation of vascular smooth muscle cells (VSMC) plays an essential role in neointimal hyperplasia. CD36 functions critically in atherogenesis and thrombosis. We hypothesize that CD36 regulates VSMC proliferation and contributes to the development of obstructive vascular diseases. Approach and Results- We found by immunofluorescent staining that CD36 was highly expressed in human vessels with obstructive diseases. Using guidewire-induced carotid artery injury and shear stress-induced intima thickening models, we compared neointimal hyperplasia in Apoe-/-, Cd36-/- /Apoe-/-, and CD36 specifically deleted in VSMC (VSMC cd36-/-) mice. CD36 deficiency, either global or VSMC-specific, dramatically reduced injury-induced neointimal thickening. Correspondingly, carotid artery blood flow was significantly increased in Cd36-/- /Apoe-/- compared with Apoe-/- mice. In cultured VSMCs from thoracic aorta of wild-type and Cd36-/- mice, we found that loss of CD36 significantly decreased serum-stimulated proliferation and increased cell populations in S phase, suggesting that CD36 is necessary for VSMC S/G2-M-phase transition. Treatment of VSMCs with a TSR (thrombospondin type 1 repeat) peptide significantly increased wild-type, but not Cd36-/- VSMC proliferation. TSR or serum treatment significantly increased cyclin A expression in wild-type, but not in Cd36-/- VSMCs. STAT3 (signal transducer and activator of transcription), which reportedly enhances both VSMC differentiation and maturation, was higher in Cd36-/- VSMCs. CD36 deficiency significantly decreased expression of Col1A1 (type 1 collagen A1 chain) and TGF-ß1 (transforming growth factor beta 1), and increased expression of contractile proteins, including calponin 1 and smooth muscle α actin, and dramatically increased cell contraction. Conclusions- CD36 promotes VSMC proliferation via upregulation of cyclin A expression that contributes to the development of neointimal hyperplasia, collagen deposition, and obstructive vascular diseases.


Asunto(s)
Antígenos CD36/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Neointima/patología , Animales , Antígenos CD36/análisis , Proliferación Celular , Ciclina A/análisis , Hiperplasia , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/fisiología
5.
Int J Mol Sci ; 21(16)2020 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-32824688

RESUMEN

(1) Background: Recently we have noted that adipocyte specific expression of the peptide, NaKtide, which was developed to attenuate the Na,K-ATPase oxidant amplification loop, could ameliorate the phenotypical features of uremic cardiomyopathy. We performed this study to better characterize the cellular transcriptomes that are involved in various biological pathways associated with adipocyte function occurring with renal failure. (2) Methods: RNAseq was performed on the visceral adipose tissue of animals subjected to partial nephrectomy. Specific expression of NaKtide in adipocytes was achieved using an adiponectin promoter. To better understand the cause of gene expression changes in vivo, 3T3L1 adipocytes were exposed to indoxyl sulfate (IS) or oxidized low density lipoprotein (oxLDL), with and without pNaKtide (the cell permeant form of NaKtide). RNAseq was also performed on these samples. (3) Results: We noted a large number of adipocyte genes were altered in experimental renal failure. Adipocyte specific NaKtide expression reversed most of these abnormalities. High correlation with some cardiac specific phenotypical features was noted amongst groups of these genes. In the murine adipocytes, both IS and oxLDL induced similar pathway changes as were noted in vivo, and pNaKtide appeared to reverse these changes. Network analysis demonstrated tremendous similarities between the network revealed by gene expression analysis with IS compared with oxLDL, and the combined in vitro dataset was noted to also have considerable similarity to that seen in vivo with experimental renal failure. (4) Conclusions: This study suggests that the myriad of phenotypical features seen with experimental renal failure may be fundamentally linked to oxidant stress within adipocytes.


Asunto(s)
Adipocitos/metabolismo , Estrés Oxidativo , Fragmentos de Péptidos/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Transcriptoma , Células 3T3 , Animales , Redes Reguladoras de Genes , Indicán/metabolismo , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/genética , ATPasa Intercambiadora de Sodio-Potasio/genética
6.
Curr Opin Hematol ; 26(5): 357-365, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31261174

RESUMEN

PURPOSE OF REVIEW: Metabolic diseases, including dyslipidemia, diabetes mellitus, and chronic inflammation are risk factors for clinically significant thrombotic events. Thrombosis in these settings is multifaceted with coordinated mechanisms between platelet activation and the hemostatic pathways. This review focuses on recent advances in platelet procoagulant and apoptotic signaling with emphasis on the pathophysiologic mechanisms induced by platelet CD36 in dyslipidemia, and the key unaddressed questions relating to the field. RECENT FINDINGS: CD36 promotes platelet activation and increases the risk for thrombosis through signaling events. These include generation of reactive oxygen species, activation of redox-sensitive MAP kinase ERK5, and promotion of a pro-thrombotic phenotype. CD36 promotes phosphatidylserine externalization leading to a procoagulant function downstream from MAP kinase ERK5 that is separate from a pro-aggregatory function. Phosphatidylserine externalization requires maladaptive caspase activation, promotes assembly of the factor tenase and prothrombinase complex, and promotes fibrin formation. It is distinct from the canonical pathways mediating platelet procoagulant function by strong physiologic stimuli or by the platelet apoptotic-like Bak/Bax-mediated pathway for cellular clearance. SUMMARY: Understanding CD36 signaling in the context of dyslipidemia, or other metabolic diseases will identify important and novel signaling hubs that could be potential therapeutic targets for intervention without impacting hemostasis.


Asunto(s)
Apoptosis , Coagulación Sanguínea , Antígenos CD36/metabolismo , Dislipidemias/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Activación Plaquetaria , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Antígenos CD36/genética , Humanos , Proteína Quinasa 7 Activada por Mitógenos/genética , Transducción de Señal
7.
Blood ; 129(21): 2917-2927, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28336528

RESUMEN

Atherothrombosis is a process mediated by dysregulated platelet activation that can cause life-threatening complications and is the leading cause of death by cardiovascular disease. Platelet reactivity in hyperlipidemic conditions is enhanced when platelet scavenger receptor CD36 recognizes oxidized lipids in oxidized low-density lipoprotein (oxLDL) particles, a process that induces an overt prothrombotic phenotype. The mechanisms by which CD36 promotes platelet activation and thrombosis remain incompletely defined. In this study, we identify a mechanism for CD36 to promote thrombosis by increasing activation of MAPK extracellular signal-regulated kinase 5 (ERK5), a protein kinase known to be exquisitely sensitive to redox stress, through a signaling pathway requiring Src kinases, NADPH oxidase, superoxide radical anion, and hydrogen peroxide. Pharmacologic inhibitors of ERK5 blunted platelet activation and aggregation in response to oxLDL and targeted genetic deletion of ERK5 in murine platelets prevented oxLDL-induced platelet deposition on immobilized collagen in response to arterial shear. Importantly, in vivo thrombosis experiments after bone marrow transplantation from platelet-specific ERK5 null mice into hyperlipidemic apolipoprotein E null mice showed decreased platelet accumulation and increased thrombosis times compared with mice transplanted with ERK5 expressing control bone marrows. These findings suggest that atherogenic conditions critically regulate platelet CD36 signaling by increasing superoxide radical anion and hydrogen peroxide through a mechanism that promotes activation of MAPK ERK5.


Asunto(s)
Plaquetas/inmunología , Antígenos CD36/inmunología , Hiperlipidemias/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Proteína Quinasa 7 Activada por Mitógenos/inmunología , Activación Plaquetaria/inmunología , Trombosis/inmunología , Aloinjertos , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/inmunología , Plaquetas/patología , Trasplante de Médula Ósea , Antígenos CD36/genética , Humanos , Hiperlipidemias/genética , Hiperlipidemias/patología , Lipoproteínas LDL/genética , Lipoproteínas LDL/inmunología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Mutantes , Proteína Quinasa 7 Activada por Mitógenos/genética , NADPH Oxidasas/genética , NADPH Oxidasas/inmunología , Activación Plaquetaria/genética , Trombosis/genética , Trombosis/patología
8.
Arterioscler Thromb Vasc Biol ; 37(8): 1462-1469, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28619997

RESUMEN

OBJECTIVE: Circulating levels of cardiotonic steroids (CTS) are elevated in various chronic inflammatory conditions, but the role of CTS in inflammation remains largely unknown. We have previously shown that the CTS ouabain stimulates proinflammatory responses in murine macrophages. In this study, we aim to explore the mechanism how CTS induce proinflammatory responses in primary murine and human macrophages. APPROACH AND RESULTS: Using both murine peritoneal macrophages and human monocyte-derived macrophages, we demonstrated that ouabain activated NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), leading to proinflammatory cytokine (eg, MCP-1 [monocyte chemotactic protein 1], TNF-α [tumor necrosis factor-α], IL-1ß [interleukin-1ß], and IL-6) production. By applying siRNA techniques and murine peritoneal macrophages isolated from genetically modified mice, we showed that macrophages partially deficient in Na/K-ATPase, the receptor for CTS, or fully deficient in the scavenger receptor CD36 or TLR4 (Toll-like receptor) were resistant to ouabain-induced NF-κB activation, suggesting an indispensable role of these 3 receptors in this pathway. Mechanistically, this effect of ouabain was independent of the ion transport function of the Na/K-ATPase. Instead, ouabain stimulated a signaling complex, including Na/K-ATPase, CD36, and TLR4. Subsequently, TLR4 recruited MyD88 adaptor protein for NF-κB activation. Furthermore, intraperitoneal injection of ouabain into mice specifically recruited Ly6C+CCR2+ monocyte subtypes to the peritoneal cavities, indicating that the CTS ouabain triggers inflammation in vivo. CONCLUSIONS: CTS activate NF-κB leading to proinflammatory cytokine production in primary macrophages through a signaling complex, including CD36, TLR4, and Na/K-ATPase. These findings warrant further studies on endogenous CTS in chronic inflammatory diseases, such as atherosclerosis.


Asunto(s)
Antígenos CD36/metabolismo , Cardiotónicos/toxicidad , Mediadores de Inflamación/metabolismo , Inflamación/inducido químicamente , Macrófagos Peritoneales/efectos de los fármacos , Ouabaína/toxicidad , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Antígenos CD36/deficiencia , Antígenos CD36/genética , Células Cultivadas , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Femenino , Inflamación/enzimología , Inflamación/genética , Macrófagos Peritoneales/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/deficiencia , ATPasa Intercambiadora de Sodio-Potasio/genética , Factores de Tiempo , Transfección
10.
Blood ; 125(17): 2591-3, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25907902

RESUMEN

In this issue of Blood, Magwenzi et al from the University of Hull report a novel mechanistic connection between oxidized low-density lipoprotein (oxLDL)-induced prothrombotic platelet signaling and the inhibition of endogenous platelet anti-activating signaling mediated by the nitric oxide (NO)/guanosine 3',5'-cyclic monophosphate (cGMP)/protein kinase G (PKG) pathway.


Asunto(s)
Plaquetas/citología , Antígenos CD36/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Lipoproteínas LDL/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Transducción de Señal , Animales , Humanos
11.
Arterioscler Thromb Vasc Biol ; 36(3): 534-44, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26821945

RESUMEN

OBJECTIVE: Literature on the effect of cell-derived extracellular vesicles (EV), ≤1 µm vesicles shed from various cell types during activation or apoptosis, on microvascular endothelial cell (MVEC) signaling is conflicting. Thrombospondin-1 and related proteins induce anti-angiogenic signals in MVEC via CD36. CD36 binds EV via phosphatidylserine exposed on their surface but the effects of this interaction on MVEC functions are not known. We hypothesized that EV would inhibit angiogenic MVEC functions via CD36. APPROACH AND RESULTS: EV generated in vitro from various cell types or isolated from plasma inhibited MVEC tube formation in in vitro matrigel assays and endothelial cell migration in Boyden chamber assays. Exosomes derived from the same cells did not have inhibitory activity. Inhibition of migration required endothelial cell expression of CD36. In mouse in vivo matrigel plug assays, EV inhibited cell migration into matrigel plugs in wild type but not in cd36 null animals. Annexin V, an anionic phospholipid binding protein, when incubated with EV partially reversed inhibition of migration, suggesting a phosphatidylserine-dependent effect. EV exposure induced reactive oxygen species generation in MVEC in a NADPH oxidase and Src family kinase-dependent manner, and their inhibition by apocynin and PP2, respectively, partially reversed the EV-mediated inhibition of migration. Annexin V partially reversed EV-induced reactive oxygen species generation in murine CD36 cDNA-transfected HVUEC but not in CD36-negative human umbilical vein endothelial cell. CONCLUSIONS: These studies establish a general inhibitory effect of EV on endothelial cell proangiogenic responses and identify a CD36-mediated mechanistic pathway through which EV inhibit MVEC migration and tube formation.


Asunto(s)
Antígenos CD36/metabolismo , Movimiento Celular , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Microvasos/metabolismo , Neovascularización Fisiológica , Transducción de Señal , Animales , Anexina A5/metabolismo , Antígenos CD36/deficiencia , Antígenos CD36/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/efectos de los fármacos , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Fosfatidilserinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 36(6): 1197-208, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27013613

RESUMEN

OBJECTIVE: CD36 is a scavenger and antiangiogenic receptor that is important in atherothrombotic diseases, diabetes mellitus, cancer, and obesity. Lysophosphatidic acid, a phospholipid signaling mediator, abolishes endothelial cell responses to antiangiogenic proteins containing thrombospondin type 1 homology domains by downregulating endothelial CD36 transcription via protein kinase D1 (PKD-1) signaling. We aimed to understand mechanisms by which lysophosphatidic acid-mediated angiogenic signaling is integrated to regulate CD36 transcription and endothelial cell function via a nuclear transcriptional complex. APPROACH AND RESULTS: Microvascular endothelial cells expressing CD36 were used for studying angiogenic signaling and CD36 transcription. Gene transfection and transduction, RT-qPCR, avidin-biotin-conjugated DNA-binding assay, chromatin immunoprecipitation assay, co-immunoprecipitation, proximal ligation assay, and immunofluorescence microscopy showed that lysophosphatidic acid-mediated CD36 transcriptional repression involved PKD-1 signaling mediated formation of forkhead box protein O1-histone deacetylase 7 complex in the nucleus. Unexpectedly, turning off CD36 transcription initiated reprogramming microvascular endothelial cells to express ephrin B2, a critical molecular signature involved in angiogenesis and arteriogenesis. Spheroid-based angiogenesis and in vivo Matrigel angiogenesis assays indicated that angiogenic branching morphogenesis and in vivo angiogenesis were dependent on PKD-1 signaling. A mouse tumor angiogenesis model revealed enhanced PKD-1 signaling and expression of ephrin B2 and smooth muscle actin in neovessels of Lewis Lung Carcinomas, along with low-CD36 expression or CD36 deficiency. CONCLUSIONS: Lysophosphatidic acid/PKD-1 signaling leads to nuclear accumulation of histone deacetylase 7, where it interacts with forkhead box protein O1 to suppress endothelial CD36 transcription and mediates silencing of antiangiogenic switch, resulting in proangiogenic and proarteriogenic reprogramming. Targeting this signaling cascade could be a novel approach for ischemic cardiovascular disease and cancer.


Asunto(s)
Antígenos CD36/metabolismo , Reprogramación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Lisofosfolípidos/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Proteína Quinasa C/metabolismo , Transcripción Genética/efectos de los fármacos , Animales , Antígenos CD36/genética , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Núcleo Celular/enzimología , Células Cultivadas , Regulación hacia Abajo , Células Endoteliales/enzimología , Efrina-B2/metabolismo , Proteína Forkhead Box O1/genética , Histona Desacetilasas/metabolismo , Humanos , Lisofosfolípidos/metabolismo , Ratones , Neovascularización Patológica , Proteína Quinasa C/genética , Interferencia de ARN , Transfección
13.
J Infect Dis ; 214(12): 1865-1875, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27683817

RESUMEN

Klebsiella pneumoniae remains an important cause of intrapulmonary infection and invasive disease worldwide. K. pneumoniae can evade serum killing and phagocytosis primarily through the expression of a polysaccharide capsule, but its pathogenicity is also influenced by host factors. We examined whether CD36, a scavenger receptor that recognizes pathogen and modified self ligands, is a host determinant of K. pneumoniae pathogenicity. Despite differences in serum sensitivity and virulence of 3 distinct K. pneumoniae (hypermucoviscous K1, research K2, and carbapenemase-producing ST258) strains, the absence of CD36 significantly increased host susceptibility to acute intrapulmonary infection by K. pneumoniae, regardless of strain. We demonstrate that CD36 enhances LPS responsiveness to K. pneumoniae to increase downstream cytokine production and macrophage phagocytosis that is independent of polysaccharide capsular antigen. Our study provides new insights into host determinants of K. pneumoniae pathogenicity and raises the possibility that functional mutations in CD36 may predispose individuals to K. pneumoniae syndromes.


Asunto(s)
Antígenos CD36/metabolismo , Interacciones Huésped-Patógeno , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/inmunología , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Fagocitosis , Animales , Femenino , Macrófagos/microbiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Bacteriana/inmunología
14.
Circ Res ; 115(12): 997-1006, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25287063

RESUMEN

RATIONALE: Platelets contain abundant thymidine phosphorylase (TYMP), which is highly expressed in diseases with high risk of thrombosis, such as atherosclerosis and type II diabetes mellitus. OBJECTIVE: To test the hypothesis that TYMP participates in platelet signaling and promotes thrombosis. METHODS AND RESULTS: By using a ferric chloride (FeCl3)-induced carotid artery injury thrombosis model, we found time to blood flow cessation was significantly prolonged in Tymp(-/-) and Tymp(+/-) mice compared with wild-type mice. Bone marrow transplantation and platelet transfusion studies demonstrated that platelet TYMP was responsible for the antithrombotic phenomenon in the TYMP-deficient mice. Collagen-, collagen-related peptide-, adenosine diphosphate-, or thrombin-induced platelet aggregation were significantly attenuated in Tymp(+/-) and Tymp(-/-) platelets, and in wild type or human platelets pretreated with TYMP inhibitor KIN59. Tymp deficiency also significantly decreased agonist-induced P-selectin expression. TYMP contains an N-terminal SH3 domain-binding proline-rich motif and forms a complex with the tyrosine kinases Lyn, Fyn, and Yes in platelets. TYMP-associated Lyn was inactive in resting platelets, and TYMP trapped and diminished active Lyn after collagen stimulation. Tymp/Lyn double haploinsufficiency diminished the antithrombotic phenotype of Tymp(+/-) mice. TYMP deletion or inhibition of TYMP with KIN59 dramatically increased platelet-endothelial cell adhesion molecule 1 tyrosine phosphorylation and diminished collagen-related peptide- or collagen-induced AKT phosphorylation. In vivo administration of KIN59 significantly inhibited FeCl3-induced carotid artery thrombosis without affecting hemostasis. CONCLUSIONS: TYMP participates in multiple platelet signaling pathways and regulates platelet activation and thrombosis. Targeting TYMP might be a novel antiplatelet and antithrombosis therapy.


Asunto(s)
Plaquetas/enzimología , Transducción de Señal , Trombosis/enzimología , Timidina Fosforilasa/metabolismo , Secuencia de Aminoácidos , Animales , Plaquetas/efectos de los fármacos , Trasplante de Médula Ósea , Cloruros , Inhibidores Enzimáticos/farmacología , Compuestos Férricos , Haploinsuficiencia , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Fenotipo , Fosforilación , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/farmacología , Transfusión de Plaquetas , Proteínas Proto-Oncogénicas c-akt/sangre , Proteínas Proto-Oncogénicas c-fyn/sangre , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-yes/sangre , Selenoproteína P/sangre , Transducción de Señal/efectos de los fármacos , Trombosis/sangre , Trombosis/inducido químicamente , Trombosis/prevención & control , Timidina Fosforilasa/antagonistas & inhibidores , Timidina Fosforilasa/sangre , Timidina Fosforilasa/deficiencia , Timidina Fosforilasa/genética , Factores de Tiempo , Familia-src Quinasas/sangre , Familia-src Quinasas/genética
16.
Blood ; 122(10): 1822-32, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23896411

RESUMEN

Thrombospondin-1 (TSP-1) inhibits growth factor signaling at the receptor level in microvascular endothelial cells (MVEC), and CD36 has been suggested to be involved in this inhibition, but the mechanisms are not known. We hypothesized that CD36-TSP-1 interaction recruits Src homology 2 domain-containing protein tyrosine phosphatase (SHP)-1 to the vascular endothelial growth factor receptor 2 (VEGFR2) signaling complex and attenuates vascular endothelial growth factor (VEGF) signaling. Western blots of anti-CD36 and anti-VEGFR2 immunoprecipitates from VEGF-treated MVEC showed that exposure of the cells to a recombinant protein containing the CD36 binding domain of thrombospondin-1 (known as the TSR domain) induced association of SHP-1 with the VEGFR2/CD36 signaling complex and thereby suppressed VEGFR2 phosphorylation. SHP-1 phosphatase activity was increased in immunoprecipitated VEGFR2 complexes from TSR-treated cells. Silencing CD36 expression in MVEC by small interfering RNA (siRNA) or genetic deletion of cd36 in mice showed that TSR-induced SHP-1/VEGFR2 complex formation required CD36 in vitro and in vivo. Silencing SHP-1 expression in MVEC by siRNA abrogated TSR-mediated inhibition of VEGFR2 phosphorylation as well as TSR-mediated inhibition of VEGF-induced endothelial cell migration and tube formation. These studies reveal a SHP-1-mediated antiangiogenic pathway induced by CD36-TSP-1 interaction that inhibits VEGFR2 signaling and they provide a novel target to modulate angiogenesis therapeutically.


Asunto(s)
Antígenos CD36/metabolismo , Células Endoteliales/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Transducción de Señal , Trombospondina 1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Antígenos CD36/deficiencia , Movimiento Celular , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Microvasos/citología , Modelos Biológicos , Neovascularización Fisiológica , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína
17.
Stem Cells ; 32(7): 1746-58, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24737733

RESUMEN

Glioblastoma (GBM) contains a self-renewing, tumorigenic cancer stem cell (CSC) population which contributes to tumor propagation and therapeutic resistance. While the tumor microenvironment is essential to CSC self-renewal, the mechanisms by which CSCs sense and respond to microenvironmental conditions are poorly understood. Scavenger receptors are a broad class of membrane receptors well characterized on immune cells and instrumental in sensing apoptotic cellular debris and modified lipids. Here, we provide evidence that CSCs selectively use the scavenger receptor CD36 to promote their maintenance using patient-derived CSCs and in vivo xenograft models. CD36 expression was observed in GBM cells in addition to previously described cell types including endothelial cells, macrophages, and microglia. CD36 was enriched in CSCs and was able to functionally distinguish self-renewing cells. CD36 was coexpressed with integrin alpha 6 and CD133, previously described CSC markers, and CD36 reduction resulted in concomitant loss of integrin alpha 6 expression, self-renewal, and tumor initiation capacity. We confirmed oxidized phospholipids, ligands of CD36, were present in GBM and found that the proliferation of CSCs, but not non-CSCs, increased with exposure to oxidized low-density lipoprotein. CD36 was an informative biomarker of malignancy and negatively correlated to patient prognosis. These results provide a paradigm for CSCs to thrive by the selective enhanced expression of scavenger receptors, providing survival, and metabolic advantages.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Antígenos CD36/metabolismo , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Antígenos CD36/genética , Proliferación Celular , Progresión de la Enfermedad , Femenino , Expresión Génica , Glioblastoma/mortalidad , Glioblastoma/patología , Estimación de Kaplan-Meier , Lipoproteínas LDL/fisiología , Ratones Desnudos , Trasplante de Neoplasias , Células Tumorales Cultivadas
18.
Arterioscler Thromb Vasc Biol ; 34(1): 160-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24177323

RESUMEN

OBJECTIVE: Proteasome inhibitors used in the treatment of hematologic cancers also reduce thrombosis. Whether the proteasome participates in platelet activation or function is unclear because little is known of the proteasome in these terminally differentiated cells. APPROACH AND RESULTS: Platelets displayed all 3 primary proteasome protease activities, which MG132 and bortezomib (Velcade) inhibited. Proteasome substrates are marked by ubiquitin, and platelets contained a functional ubiquitination system that modified the proteome by monoubiquitination and polyubiquitination. Systemic MG132 strongly suppressed the formation of occlusive, platelet-rich thrombi in FeCl3-damaged carotid arteries. Transfusion of platelets treated ex vivo with MG132 and washed before transfusion into thrombocytopenic mice also reduced carotid artery thrombosis. Proteasome inhibition reduced platelet aggregation by low thrombin concentrations and ristocetin-stimulated agglutination through the glycoprotein Ib-IX-V complex. This receptor was not appropriately internalized after proteasome inhibition in stimulated platelets, and spreading and clot retraction after MG132 exposure also were decreased. The effects of proteasome inhibitors were not confined to a single receptor as MG132 suppressed thrombin-stimulated, ADP-stimulated, and lipopolysaccharide-stimulated microparticle shedding. Proteasome inhibition increased ubiquitin decoration of cytoplasmic proteins, including the cytoskeletal proteins Filamin A and Talin-1. Mass spectrometry revealed a single MG132-sensitive tryptic cleavage after R1745 in an extended Filamin A loop, which would separate its actin-binding domain from its carboxy terminal glycoprotein Ibα-binding domain. CONCLUSIONS: Platelets contain a ubiquitin/proteasome system that marks cytoskeletal proteins for proteolytic modification to promote productive platelet-platelet and platelet-wall interactions.


Asunto(s)
Plaquetas/enzimología , Proteínas del Citoesqueleto/sangre , Activación Plaquetaria , Complejo de la Endopetidasa Proteasomal/sangre , Trombosis/enzimología , Adenosina Difosfato/farmacología , Animales , Plaquetas/efectos de los fármacos , Micropartículas Derivadas de Células/metabolismo , Cloruros , Modelos Animales de Enfermedad , Compuestos Férricos , Fibrinolíticos/farmacología , Filaminas/sangre , Humanos , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Activación Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria , Agregación Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Transfusión de Plaquetas , Inhibidores de Proteasoma/farmacología , Proteolisis , Talina/sangre , Trombina/farmacología , Trombosis/sangre , Trombosis/inducido químicamente , Trombosis/prevención & control , Factores de Tiempo , Ubiquitinación
19.
J Immunol ; 191(10): 5196-203, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24081990

RESUMEN

LPS activates platelets through TLR4, aiding productive sepsis, with stimulated splicing and translation of stored heteronuclear pro-IL-1ß RNA. Although the IL-1R type 1 (IL-1R1) receptor for IL-1 shares downstream components with the TLR4 receptor, platelets are not known to express IL-1R1, nor are they known to respond to this cytokine. We show by flow cytometry and Western blotting that platelets express IL-1R1, and that IL-1ß and IL-1α stimulate heteronuclear I-1ß splicing and translation of the newly made mRNA in platelets. Platelets also respond to the IL-1ß they make, which is exclusively associated with shed microparticles. Specific blockade of IL-1R1 with IL-1R antagonist suppressed platelet stimulation by IL-1, so IL-1ß stimulates its own synthesis in an autocrine signaling loop. Strikingly, IL-1R antagonist inhibition, pharmacologic or genetic suppression of pro-IL-1ß processing to active cytokine by caspase-1, or blockade of de novo protein synthesis also blocked LPS-induced IL-1ß mRNA production. Robust stimulation of platelets by LPS therefore also required IL-1ß amplification. Activated platelets made IL-1ß in vivo as IL-1ß rapidly accumulated in occluded murine carotid arteries by posttranscriptional RNA splicing unique to platelets. We conclude that IL-1ß is a platelet agonist, that IL-1ß acts through an autocrine stimulatory loop, that an IL-1ß autocrine loop is required to amplify platelet activation by LPS, and that platelets immobilized in occlusive thrombi are activated over time to produce IL-1ß. IL-1 is a new platelet agonist that promotes its own synthesis, connecting thrombosis with immunity.


Asunto(s)
Plaquetas/inmunología , Interleucina-1beta/metabolismo , Lipopolisacáridos/metabolismo , Activación Plaquetaria/inmunología , Animales , Plaquetas/metabolismo , Caspasa 1 , Células Cultivadas , Humanos , Inflamación/inmunología , Interleucina-1alfa/metabolismo , Ratones , Empalme del ARN , ARN Mensajero , Receptores Tipo I de Interleucina-1/antagonistas & inhibidores , Receptores Tipo I de Interleucina-1/biosíntesis , Transducción de Señal , Trombosis/inmunología , Receptor Toll-Like 4/metabolismo
20.
Blood ; 119(25): 6136-44, 2012 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-22431576

RESUMEN

Diabetes mellitus has been associated with platelet hyperreactivity, which plays a central role in the hyperglycemia-related prothrombotic phenotype. The mechanisms responsible for this phenomenon are not established. In the present study, we investigated the role of CD36, a class-B scavenger receptor, in this process. Using both in vitro and in vivo mouse models, we demonstrated direct and specific interactions of platelet CD36 with advanced glycation end products (AGEs) generated under hyperglycemic conditions. AGEs bound to platelet CD36 in a specific and dose-dependent manner, and binding was inhibited by the high-affinity CD36 ligand NO(2)LDL. Cd36-null platelets did not bind AGE. Using diet- and drug-induced mouse models of diabetes, we have shown that cd36-null mice had a delayed time to the formation of occlusive thrombi compared with wild-type (WT) in a FeCl(3)-induced carotid artery injury model. Cd36-null mice had a similar level of hyperglycemia and a similar level of plasma AGEs compared with WT mice under this condition, but WT mice had more AGEs incorporated into thrombi. Mechanistic studies revealed that CD36-dependent JNK2 activation is involved in this prothrombotic pathway. Therefore, the results of the present study couple vascular complications in diabetes mellitus with AGE-CD36-mediated platelet signaling and hyperreactivity.


Asunto(s)
Enfermedades Asintomáticas , Plaquetas/metabolismo , Antígenos CD36/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Productos Finales de Glicación Avanzada/fisiología , Trombosis/etiología , Animales , Plaquetas/efectos de los fármacos , Antígenos CD36/genética , Antígenos CD36/fisiología , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Angiopatías Diabéticas/sangre , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/metabolismo , Angiopatías Diabéticas/patología , Dieta Aterogénica , Productos Finales de Glicación Avanzada/farmacología , Hiperglucemia/sangre , Hiperglucemia/etiología , Hiperglucemia/genética , Hiperglucemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Agregación Plaquetaria/genética , Agregación Plaquetaria/fisiología , Unión Proteica , Estreptozocina , Trombosis/metabolismo , Trombosis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA