Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Exp Bot ; 68(7): 1555-1567, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28369580

RESUMEN

FUSCA3 (FUS3) is a short-lived B3-domain transcription factor that regulates seed development and phase transitions in Arabidopsis thaliana. The mechanisms controlling FUS3 levels are currently poorly understood. Here we show that FUS3 interacts with the RING E3 ligase ABI3-INTERACTING PROTEIN2 (AIP2). AIP2-green fluorescent protein (GFP) is preferentially expressed in the protoderm during early embryogenesis, similarly to FUS3, suggesting that their interaction is biologically relevant. FUS3 degradation is delayed in the aip2-1 mutant and FUS3-GFP fluorescence is increased in aip2-1, but only during mid-embryogenesis, suggesting that FUS3 is negatively regulated by AIP2 at a specific time during embryogenesis. aip2-1 shows delayed flowering and therefore also functions post-embryonically to regulate developmental phase transitions. Plants overexpressing FUS3 post-embryonically in the L1 layer (ML1p:FUS3) show late flowering and other developmental phenotypes that can be rescued by ML1p:AIP2, further supporting a negative role for AIP2 in FUS3 accumulation. However, additional factors regulate FUS3 levels during embryogenesis, as ML1:AIP2 seeds do not resemble fus3-3. Lastly, targeted expression of a RING-inactive AIP2 variant to the protoderm/L1 layer causes FUS3 and ABI3 overexpression phenotypes and defects in cotyledon development. Taken together, these results indicate that AIP2 targets FUS3 for degradation and plays a role in cotyledon development and flowering time in Arabidopsis.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Proteínas Portadoras/genética , Cotiledón/crecimiento & desarrollo , Regulación de la Expresión Génica de las Plantas , Factores de Transcripción/genética , Arabidopsis/crecimiento & desarrollo , Proteínas de Arabidopsis/metabolismo , Proteínas Portadoras/metabolismo , Cotiledón/genética , Factores de Transcripción/metabolismo
2.
J Immunol ; 195(7): 3334-44, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26297762

RESUMEN

The secreted neurorepellent Slit2, acting through its transmembrane receptor, Roundabout (Robo)-1, inhibits chemotaxis of varied cell types, including leukocytes, endothelial cells, and vascular smooth muscle cells, toward diverse attractants. The role of Slit2 in regulating the steps involved in recruitment of monocytes in vascular inflammation is not well understood. In this study, we showed that Slit2 inhibited adhesion of monocytic cells to activated human endothelial cells, as well as to immobilized ICAM-1 and VCAM-1. Microfluidic live cell imaging showed that Slit2 inhibited the ability of monocytes tethered to endothelial cells to stabilize their actin-associated anchors and to resist detachment in response to increasing shear forces. Transfection of constitutively active plasmids revealed that Slit2 inhibited postadhesion stabilization of monocytes on endothelial cells by preventing activation of Rac1. We further found that Slit2 inhibited chemotaxis of monocytes toward CXCL12 and CCL2. To determine whether Slit2 and Robo-1 modulate pathologic monocyte recruitment associated with vascular inflammation and cardiovascular disease, we tested PBMC from patients with coronary artery disease. PBMC from these patients had reduced surface levels of Robo-1 compared with healthy age- and sex-matched subjects, and Slit2 failed to inhibit chemotaxis of PBMC of affected patients, but not healthy control subjects, toward CCL2. Furthermore, administration of Slit2 to atherosclerosis-prone LDL receptor-deficient mice inhibited monocyte recruitment to nascent atherosclerotic lesions. These results demonstrate that Slit2 inhibits chemotaxis of monocytes, as well as their ability to stabilize adhesions and resist detachment forces. Slit2 may represent a powerful new tool to inhibit pathologic monocyte recruitment in vascular inflammation and atherosclerosis.


Asunto(s)
Adhesión Celular/fisiología , Quimiotaxis de Leucocito/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Monocitos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Aterosclerosis/patología , Enfermedades Cardiovasculares/inmunología , Línea Celular , Quimiocina CCL2 , Quimiocina CXCL12 , Activación Enzimática , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos Mononucleares/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Monocitos/inmunología , Receptores de LDL/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas Roundabout
3.
J Am Soc Nephrol ; 27(9): 2609-15, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26869008

RESUMEN

Fibrosis and inflammation are closely intertwined injury pathways present in nearly all forms of CKD for which few safe and effective therapies exist. Slit glycoproteins signaling through Roundabout (Robo) receptors have been described to have anti-inflammatory effects through regulation of leukocyte cytoskeletal organization. Notably, cytoskeletal reorganization is also required for fibroblast responses to TGF-ß Here, we examined whether Slit2 also controls TGF-ß-induced renal fibrosis. In cultured renal fibroblasts, which we found to express Slit2 and Robo-1, the bioactive N-terminal fragment of Slit2 inhibited TGF-ß-induced collagen synthesis, actin cytoskeletal reorganization, and Smad2/3 transcriptional activity, but the inactive C-terminal fragment of Slit2 did not. In mouse models of postischemic renal fibrosis and obstructive uropathy, treatment with N-terminal Slit2 before or after injury inhibited the development of renal fibrosis and preserved renal function, whereas the C-terminal Slit2 had no effect. Our data suggest that administration of recombinant Slit2 may be a new treatment strategy to arrest chronic injury progression after ischemic and obstructive renal insults by not only attenuating inflammation but also, directly inhibiting renal fibrosis.


Asunto(s)
Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Enfermedades Renales/prevención & control , Riñón/patología , Proteínas del Tejido Nervioso/farmacología , Proteínas del Tejido Nervioso/uso terapéutico , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/fisiología , Animales , Fibrosis/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes
4.
Circulation ; 126(11): 1385-95, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22865890

RESUMEN

BACKGROUND: Vascular injury and atherothrombosis involve vessel infiltration by inflammatory leukocytes, migration of medial vascular smooth muscle cells to the intimal layer, and ultimately acute thrombosis. A strategy to simultaneously target these pathological processes has yet to be identified. The secreted protein, Slit2, and its transmembrane receptor, Robo-1, repel neuronal migration in the developing central nervous system. More recently, it has been appreciated that Slit2 impairs chemotaxis of leukocytes and vascular smooth muscle cells toward diverse inflammatory attractants. The effects of Slit2 on platelet function and thrombus formation have never been explored. METHODS AND RESULTS: We detected Robo-1 expression in human and murine platelets and megakaryocytes and confirmed its presence via immunofluorescence microscopy and flow cytometry. In both static and shear microfluidic assays, Slit2 impaired platelet adhesion and spreading on diverse extracellular matrix substrates by suppressing activation of Akt. Slit2 also prevented platelet activation on exposure to ADP. In in vivo studies, Slit2 prolonged bleeding times in murine tail bleeding assays. Using intravital microscopy, we found that after mesenteric arteriolar and carotid artery injury, Slit2 delayed vessel occlusion time and prevented the stable formation of occlusive arteriolar thrombi. CONCLUSIONS: These data demonstrate that Slit2 is a powerful negative regulator of platelet function and thrombus formation. The ability to simultaneously block multiple events in vascular injury may allow Slit2 to effectively prevent and treat thrombotic disorders such as myocardial infarction and stroke.


Asunto(s)
Plaquetas/fisiología , Movimiento Celular/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas del Tejido Nervioso/fisiología , Animales , Trombosis de las Arterias Carótidas/inducido químicamente , Trombosis de las Arterias Carótidas/fisiopatología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Cloruros/efectos adversos , Compuestos Férricos/efectos adversos , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Proteínas del Tejido Nervioso/farmacología , Adhesividad Plaquetaria/fisiología , Receptores Inmunológicos/fisiología , Factores de Riesgo , Proteínas Roundabout
5.
Life Sci Alliance ; 6(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37311584

RESUMEN

SLIT/ROBO signaling impacts many aspects of tissue development and homeostasis, in part, through the regulation of cell growth and proliferation. Recent studies have also linked SLIT/ROBO signaling to the regulation of diverse phagocyte functions. However, the mechanisms by which SLIT/ROBO signaling acts at the nexus of cellular growth control and innate immunity remain enigmatic. Here, we show that SLIT2-mediated activation of ROBO1 leads to inhibition of mTORC1 kinase activity in macrophages, leading to dephosphorylation of its downstream targets, including transcription factor EB and ULK1. Consequently, SLIT2 augments lysosome biogenesis, potently induces autophagy, and robustly promotes the killing of bacteria within phagosomes. Concordant with these results, we demonstrate decreased lysosomal content and accumulated peroxisomes in the spinal cords of embryos from Robo1 -/- , Robo2 -/- double knockout mice. We also show that impediment of auto/paracrine SLIT-ROBO signaling axis in cancer cells leads to hyperactivation of mTORC1 and inhibition of autophagy. Together, these findings elucidate a central role of chemorepellent SLIT2 in the regulation of mTORC1 activity with important implications for innate immunity and cancer cell survival.


Asunto(s)
Proteínas del Tejido Nervioso , Receptores Inmunológicos , Animales , Ratones , Proteínas del Tejido Nervioso/genética , Receptores Inmunológicos/genética , Lisosomas , Bacterias , Diana Mecanicista del Complejo 1 de la Rapamicina
6.
Elife ; 122023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37773612

RESUMEN

Neutrophils are essential for host defense against Staphylococcus aureus (S. aureus). The neuro-repellent, SLIT2, potently inhibits neutrophil chemotaxis, and might, therefore, be expected to impair antibacterial responses. We report here that, unexpectedly, neutrophils exposed to the N-terminal SLIT2 (N-SLIT2) fragment kill extracellular S. aureus more efficiently. N-SLIT2 amplifies reactive oxygen species production in response to the bacteria by activating p38 mitogen-activated protein kinase that in turn phosphorylates NCF1, an essential subunit of the NADPH oxidase complex. N-SLIT2 also enhances the exocytosis of neutrophil secondary granules. In a murine model of S. aureus skin and soft tissue infection (SSTI), local SLIT2 levels fall initially but increase subsequently, peaking at 3 days after infection. Of note, the neutralization of endogenous SLIT2 worsens SSTI. Temporal fluctuations in local SLIT2 levels may promote neutrophil recruitment and retention at the infection site and hasten bacterial clearance by augmenting neutrophil oxidative burst and degranulation. Collectively, these actions of SLIT2 coordinate innate immune responses to limit susceptibility to S. aureus.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Humanos , Ratones , Quimiotaxis de Leucocito , Inmunidad Innata , Neutrófilos , Infecciones Estafilocócicas/microbiología
7.
Sci Rep ; 11(1): 3614, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33574432

RESUMEN

Atherosclerosis is characterized by retention of modified lipoproteins, especially oxidized low density lipoprotein (oxLDL) within the sub-endothelial space of affected blood vessels. Recruited monocyte-derived and tissue-resident macrophages subsequently ingest oxLDL by binding and internalizing oxLDL via scavenger receptors, particularly CD36. The secreted neurorepellent, Slit2, acting through its transmembrane receptor, Roundabout-1 (Robo-1), was previously shown to inhibit recruitment of monocytes into nascent atherosclerotic lesions. The effects of Slit2 on oxLDL uptake by macrophages have not been explored. We report here that Slit2 inhibits uptake of oxLDL by human and murine macrophages, and the resulting formation of foam cells, in a Rac1-dependent and CD36-dependent manner. Exposure of macrophages to Slit2 prevented binding of oxLDL to the surface of cells. Using super-resolution microscopy, we observed that exposure of macrophages to Slit2 induced profound cytoskeletal remodeling with formation of a thick ring of cortical actin within which clusters of CD36 could not aggregate, thereby attenuating binding of oxLDL to the surface of cells. By inhibiting recruitment of monocytes into early atherosclerotic lesions, and the subsequent binding and internalization of oxLDL by macrophages, Slit2 could represent a potent new tool to combat individual steps that collectively result in progression of atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Lípidos/inmunología , Lipoproteínas LDL/genética , Proteínas del Tejido Nervioso/genética , Animales , Aterosclerosis/inmunología , Aterosclerosis/patología , Vasos Sanguíneos/inmunología , Antígenos CD36/genética , Antígenos CD36/inmunología , Modelos Animales de Enfermedad , Células Espumosas , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lípidos/genética , Lipoproteínas LDL/inmunología , Macrófagos/inmunología , Ratones , Monocitos/inmunología , Proteínas del Tejido Nervioso/metabolismo , Receptores Depuradores/genética , Receptores Depuradores/inmunología
8.
Nat Commun ; 11(1): 4112, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32807784

RESUMEN

Macropinocytosis is essential for myeloid cells to survey their environment and for growth of RAS-transformed cancer cells. Several growth factors and inflammatory stimuli are known to induce macropinocytosis, but its endogenous inhibitors have remained elusive. Stimulation of Roundabout receptors by Slit ligands inhibits directional migration of many cell types, including immune cells and cancer cells. We report that SLIT2 inhibits macropinocytosis in vitro and in vivo by inducing cytoskeletal changes in macrophages. In mice, SLIT2 attenuates the uptake of muramyl dipeptide, thereby preventing NOD2-dependent activation of NF-κB and consequent secretion of pro-inflammatory chemokine, CXCL1. Conversely, blocking the action of endogenous SLIT2 enhances CXCL1 secretion. SLIT2 also inhibits macropinocytosis in RAS-transformed cancer cells, thereby decreasing their survival in nutrient-deficient conditions which resemble tumor microenvironment. Our results identify SLIT2 as a physiological inhibitor of macropinocytosis and challenge the conventional notion that signals that enhance macropinocytosis negatively regulate cell migration, and vice versa.


Asunto(s)
Citoesqueleto/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Quimiocina CXCL1/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Péptidos y Proteínas de Señalización Intercelular/genética , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/genética , Fagocitos/metabolismo , Pinocitosis/genética , Pinocitosis/fisiología , Receptores Inmunológicos/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Proteínas Roundabout
9.
Mol Biol Cell ; 25(24): 3884-99, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25253723

RESUMEN

CX3CL1 is a unique chemokine that acts both as a transmembrane endothelial adhesion molecule and, upon proteolytic cleavage, a soluble chemoattractant for circulating leukocytes. The constitutive release of soluble CX3CL1 requires the interaction of its transmembrane species with the integral membrane metalloprotease ADAM10, yet the mechanisms governing this process remain elusive. Using single-particle tracking and subdiffraction imaging, we studied how ADAM10 interacts with CX3CL1. We observed that the majority of cell surface CX3CL1 diffused within restricted confinement regions structured by the cortical actin cytoskeleton. These confinement regions sequestered CX3CL1 from ADAM10, precluding their association. Disruption of the actin cytoskeleton reduced CX3CL1 confinement and increased CX3CL1-ADAM10 interactions, promoting the release of soluble chemokine. Our results demonstrate a novel role for the cytoskeleton in limiting membrane protein proteolysis, thereby regulating both cell surface levels and the release of soluble ligand.


Asunto(s)
Proteínas ADAM/metabolismo , Citoesqueleto de Actina/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Membrana Celular/metabolismo , Quimiocina CX3CL1/metabolismo , Proteínas de la Membrana/metabolismo , Proteína ADAM10 , Células Cultivadas , Quimiocina CX3CL1/genética , Endocitosis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Unión Proteica , Proteolisis , Factor de Necrosis Tumoral alfa/farmacología , Grabación de Cinta de Video
10.
J Clin Invest ; 122(2): 759-76, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22232208

RESUMEN

Hemolytic uremic syndrome (HUS) is a potentially life-threatening condition. It often occurs after gastrointestinal infection with E. coli O157:H7, which produces Shiga toxins (Stx) that cause hemolytic anemia, thrombocytopenia, and renal injury. Stx-mediated changes in endothelial phenotype have been linked to the pathogenesis of HUS. Here we report our studies investigating Stx-induced changes in gene expression and their contribution to the pathogenesis of HUS. Stx function by inactivating host ribosomes but can also alter gene expression at concentrations that minimally affect global protein synthesis. Gene expression profiling of human microvascular endothelium treated with Stx implicated a role for activation of CXCR4 and CXCR7 by their shared cognate chemokine ligand (stromal cell-derived factor-1 [SDF-1]) in Stx-mediated pathophysiology. The changes in gene expression required a catalytically active Stx A subunit and were mediated by enhanced transcription and mRNA stability. Stx also enhanced the association of CXCR4, CXCR7, and SDF1 mRNAs with ribosomes. In a mouse model of Stx-mediated pathology, we noted changes in plasma and tissue content of CXCR4, CXCR7, and SDF-1 after Stx exposure. Furthermore, inhibition of the CXCR4/SDF-1 interaction decreased endothelial activation and organ injury and improved animal survival. Finally, in children infected with E. coli O157:H7, plasma SDF-1 levels were elevated in individuals who progressed to HUS. Collectively, these data implicate the CXCR4/CXCR7/SDF-1 pathway in Stx-mediated pathogenesis and suggest novel therapeutic strategies for prevention and/or treatment of complications associated with E. coli O157:H7 infection.


Asunto(s)
Quimiocina CXCL12/metabolismo , Síndrome Hemolítico-Urémico/etiología , Síndrome Hemolítico-Urémico/fisiopatología , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Toxinas Shiga/toxicidad , Animales , Línea Celular , Quimiocina CXCL12/genética , Niño , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Infecciones por Escherichia coli/complicaciones , Escherichia coli O157/metabolismo , Escherichia coli O157/patogenicidad , Expresión Génica/efectos de los fármacos , Síndrome Hemolítico-Urémico/patología , Humanos , Riñón/patología , Riñón/fisiopatología , Ratones , Análisis por Micromatrices , Análisis de Secuencia por Matrices de Oligonucleótidos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores CXCR/genética , Receptores CXCR4/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
11.
Methods Mol Biol ; 748: 143-53, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21701972

RESUMEN

Fractalkine (CX(3)CL1) is a membrane-anchored chemokine whose N-terminus contains a unique CX(3)C motif that is cleaved and released. The membrane-bound form functions as an adhesion molecule and the secreted form as a chemotactic factor. Like other chemokines, CX(3)CL1 is regulated at the levels of transcription and translation. Recent evidence points to additional functional regulation by cellular trafficking owing to the unique transmembrane structure. CX(3)CL1 is the only chemokine known to undergo constitutive internalization. To understand mechanisms governing the regulation and processing of such membrane-bound proteins, it is vital to study their subcellular distribution and transport. The methods outlined in this chapter describe (1) transfection of mammalian cells with plasmids encoding the expression of green fluorescent protein-tagged CX(3)CL1; (2) immunofluorescence antibody labeling as well as fluorescence recovery after photobleaching to study internalization of CX(3)CL1 by endocytosis; and (3) acid-stripping assays to study the recycling of internalized CX(3)CL1 back to the plasma membrane. Together, these methods allow for the examination of subcellular distribution and traffic of recycling membrane proteins.


Asunto(s)
Quimiocina CX3CL1/metabolismo , Línea Celular , Quimiocina CX3CL1/genética , Endocitosis/genética , Endocitosis/fisiología , Exocitosis/genética , Exocitosis/fisiología , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA