Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
J Immunol ; 206(12): 2989-2999, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34088769

RESUMEN

The respiratory epithelium forms the first line of defense against inhaled pathogens and acts as an important source of innate cytokine responses to environmental insults. One critical mediator of these responses is the IL-1 family cytokine IL-33, which is rapidly secreted upon acute epithelial injury as an alarmin and induces type 2 immune responses. Our recent work highlighted the importance of the NADPH oxidase dual oxidase 1 (DUOX1) in acute airway epithelial IL-33 secretion by various airborne allergens associated with H2O2 production and reduction-oxidation-dependent activation of Src kinases and epidermal growth factor receptor (EGFR) signaling. In this study, we show that IL-33 secretion in response to acute airway challenge with house dust mite (HDM) allergen critically depends on the activation of Src by a DUOX1-dependent oxidative mechanism. Intriguingly, HDM-induced epithelial IL-33 secretion was dramatically attenuated by small interfering RNA- or Ab-based approaches to block IL-33 signaling through its receptor IL1RL1 (ST2), indicating that HDM-induced IL-33 secretion includes a positive feed-forward mechanism involving ST2-dependent IL-33 signaling. Moreover, activation of type 2 cytokine responses by direct airway IL-33 administration was associated with ST2-dependent activation of DUOX1-mediated H2O2 production and reduction-oxidation-based activation of Src and EGFR and was attenuated in Duox1 -/- and Src +/- mice, indicating that IL-33-induced epithelial signaling and subsequent airway responses involve DUOX1/Src-dependent pathways. Collectively, our findings suggest an intricate relationship between DUOX1, Src, and IL-33 signaling in the activation of innate type 2 immune responses to allergens, involving DUOX1-dependent epithelial Src/EGFR activation in initial IL-33 secretion and in subsequent IL-33 signaling through ST2 activation.


Asunto(s)
Alérgenos/inmunología , Oxidasas Duales/inmunología , Interleucina-33/inmunología , Mucosa Respiratoria/inmunología , Familia-src Quinasas/inmunología , Enfermedad Aguda , Animales , Células Cultivadas , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucosa Respiratoria/patología , Transducción de Señal/inmunología , Familia-src Quinasas/deficiencia
2.
J Biol Chem ; 295(39): 13474-13487, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32690605

RESUMEN

Yes-associated protein (YAP) signaling has emerged as a crucial pathway in several normal and pathological processes. Although the main upstream effectors that regulate its activity have been extensively studied, the role of the endosomal system has been far less characterized. Here, we identified the late endosomal/lysosomal adaptor MAPK and mTOR activator (LAMTOR) complex as an important regulator of YAP signaling in a preosteoblast cell line. We found that p18/LAMTOR1-mediated peripheral positioning of late endosomes allows delivery of SRC proto-oncogene, nonreceptor tyrosine kinase (SRC) to the plasma membrane and promotes activation of an SRC-dependent signaling cascade that controls YAP nuclear shuttling. Moreover, ß1 integrin engagement and mechano-sensitive cues, such as external stiffness and related cell contractility, controlled LAMTOR targeting to the cell periphery and thereby late endosome recycling and had a major impact on YAP signaling. Our findings identify the late endosome recycling pathway as a key mechanism that controls YAP activity and explains YAP mechano-sensitivity.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Endosomas/metabolismo , Integrina beta1/metabolismo , Factores de Transcripción/metabolismo , Familia-src Quinasas/metabolismo , Animales , Proteínas de Ciclo Celular/deficiencia , Línea Celular , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Proto-Oncogenes Mas , Transducción de Señal , Factores de Transcripción/deficiencia , Familia-src Quinasas/deficiencia
3.
Circ Res ; 126(10): e61-e79, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32151196

RESUMEN

RATIONALE: Monocytes are key effectors of the mononuclear phagocyte system, playing critical roles in regulating tissue homeostasis and coordinating inflammatory reactions, including those involved in chronic inflammatory diseases such as atherosclerosis. Monocytes have traditionally been divided into 2 major subsets termed conventional monocytes and patrolling monocytes (pMo) but recent systems immunology approaches have identified marked heterogeneity within these cells, and much of what regulates monocyte population homeostasis remains unknown. We and others have previously identified LYN tyrosine kinase as a key negative regulator of myeloid cell biology; however, LYN's role in regulating specific monocyte subset homeostasis has not been investigated. OBJECTIVE: We sought to comprehensively profile monocytes to elucidate the underlying heterogeneity within monocytes and dissect how Lyn deficiency affects monocyte subset composition, signaling, and gene expression. We further tested the biological significance of these findings in a model of atherosclerosis. METHODS AND RESULTS: Mass cytometric analysis of monocyte subsets and signaling pathway activation patterns in conventional monocytes and pMos revealed distinct baseline signaling profiles and far greater heterogeneity than previously described. Lyn deficiency led to a selective expansion of pMos and alterations in specific signaling pathways within these cells, revealing a critical role for LYN in pMo physiology. LYN's role in regulating pMos was cell-intrinsic and correlated with an increased circulating half-life of Lyn-deficient pMos. Furthermore, single-cell RNA sequencing revealed marked perturbations in the gene expression profiles of Lyn-/- monocytes with upregulation of genes involved in pMo development, survival, and function. Lyn deficiency also led to a significant increase in aorta-associated pMos and protected Ldlr-/- mice from high-fat diet-induced atherosclerosis. CONCLUSIONS: Together our data identify LYN as a key regulator of pMo development and a potential therapeutic target in inflammatory diseases regulated by pMos.


Asunto(s)
Aterosclerosis/genética , Citometría de Flujo , Heterogeneidad Genética , Monocitos/enzimología , RNA-Seq , Transducción de Señal/genética , Análisis de la Célula Individual , Transcriptoma , Familia-src Quinasas/genética , Animales , Aterosclerosis/enzimología , Aterosclerosis/inmunología , Aterosclerosis/patología , Supervivencia Celular , Células Cultivadas , Senescencia Celular , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Fenotipo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Familia-src Quinasas/deficiencia
4.
Sci Signal ; 12(602)2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31594855

RESUMEN

CARD9 is an immune adaptor protein in myeloid cells that is involved in C-type lectin signaling and antifungal immunity. CARD9 is implicated in autoimmune and inflammatory-related diseases, such as rheumatoid arthritis, IgA nephropathy, ankylosing spondylitis, and inflammatory bowel disease (IBD). Given that Lyn-deficient (Lyn-/-) mice are susceptible to both autoimmunity and IBD, we investigated the immunological role of CARD9 in the development of these diseases using the Lyn-/- mouse model. We found that genetic deletion of CARD9 was sufficient to reduce the development of both spontaneous autoimmune disease as well as DSS- or IL-10 deficiency-associated colitis in Lyn-/- mice. Mechanistically, CARD9 was a vital component of the Lyn-mediated regulation of Toll-like receptor (TLR2 and TLR4) signaling in dendritic cells, but not in macrophages. In the absence of Lyn, signaling through a CD11b-Syk-PKCδ-CARD9 pathway was amplified, leading to increased TLR-induced production of inflammatory cytokines. Dendritic cell-specific deletion of CARD9 reversed the development of autoimmune and experimental colitis observed in dendritic cell-specific, Lyn-deficient mice. These findings suggest that targeting CARD9 may suppress the development of colitis and autoimmunity by reducing dendritic cell-driven inflammation.


Asunto(s)
Autoinmunidad , Proteínas Adaptadoras de Señalización CARD/deficiencia , Células Dendríticas/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Familia-src Quinasas/deficiencia , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Modelos Animales de Enfermedad , Enfermedades Inflamatorias del Intestino/genética , Interleucina-10/deficiencia , Interleucina-10/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo , Interferencia de ARN , Transducción de Señal , Quinasa Syk/genética , Quinasa Syk/metabolismo , Receptores Toll-Like/metabolismo , Familia-src Quinasas/genética
5.
Artículo en Inglés | MEDLINE | ID: mdl-30826459

RESUMEN

Much evidence suggests that hypofunction of the N-methyl-d-aspartate glutamate receptor (NMDAR) may contribute broadly towards a subset of molecular, cognitive and behavioral abnormalities common among psychiatric and developmental diseases. However, little is known about the specific molecular changes that lead to NMDAR dysfunction. As such, personalized approaches to remediating NMDAR dysfunction based on a specific etiology remains a challenge. Sarcoma tyrosine kinase (Src) serves as a hub for multiple signaling mechanisms affecting GluN2 phosphorylation and can be disrupted by convergent alterations of various signaling pathways. We recently showed reduced Src signaling in post mortem tissue from schizophrenia patients, despite increased MK-801 binding and NMDA receptor complex expression in the postsynaptic density (PSD). These data suggest that Src dysregulation may be an important underlying mechanism responsible for reduced glutamate signaling. Despite this evidence for a central role of Src in NMDAR signaling, little is known about how reductions in Src activity might regulate phenotypic changes in cognition and behavior. As such, the current study sought to characterize behavioral and electrophysiological phenotypes in mice heterozygous for the Src Acl gene (Src+/- mice). Src+/- mice demonstrated decreased sociability and working memory relative to Src+/+ (WT) mice while no significant differences were seen on locomotive activity and anxiety-related behavior. In relation to WT mice, Src+/- mice showed decreased mid-latency P20 auditory event related potential (aERP) amplitudes, decreased mismatch negativity (MMN) and decreased evoked gamma power, which was only present in males. These data indicate that Src+/- mice are a promising new model to help understand the pathophysiology of these electrophysiological, behavioral and cognitive changes. As such, we propose that Src+/- mice can be used in the future to evaluate potential therapeutic approaches by targeting increased Src activity as a common final pathway for multiple etiologies of SCZ and other diseases characterized by reduced glutamate function.


Asunto(s)
Memoria a Corto Plazo , Conducta Social , Familia-src Quinasas/deficiencia , Animales , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Electroencefalografía , Electrofisiología , Potenciales Evocados/fisiología , Femenino , Masculino , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esquizofrenia/enzimología , Esquizofrenia/fisiopatología , Familia-src Quinasas/fisiología
6.
J Immunol ; 201(5): 1353-1358, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30021765

RESUMEN

Genetic deletion of the Src family tyrosine kinase Lyn in mice recapitulates human systemic lupus erythematosus, characterized by hyperactive BCR signaling, splenomegaly, autoantibody generation, and glomerulonephritis. However, the molecular regulators of autoimmunity in Lyn-deficient mice and in human lupus remain poorly characterized. In this study, we report that conditional deletion of the membrane-cytoskeleton linker protein ezrin in B cells of Lyn-deficient mice (double knockout [DKO] mice) ameliorates B cell activation and lupus pathogenesis. B cells from DKO mice respond poorly to BCR stimulation, with severe downregulation of major signaling pathways. DKO mice exhibit reduced splenomegaly as well as significantly lower levels of autoantibodies against a variety of autoantigens, including dsDNA, histone, and chromatin. Leukocyte infiltration and deposition of IgG and complement component C3 in the kidney glomeruli of DKO mice are markedly reduced. Our data demonstrate that ezrin is a novel molecular regulator of B cell-associated lupus pathology.


Asunto(s)
Linfocitos B , Proteínas del Citoesqueleto/deficiencia , Lupus Eritematoso Sistémico , Activación de Linfocitos/genética , Transducción de Señal , Familia-src Quinasas/deficiencia , Animales , Autoantígenos/genética , Autoantígenos/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Proteínas del Citoesqueleto/inmunología , Regulación hacia Abajo/inmunología , Eliminación de Gen , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Familia-src Quinasas/inmunología
7.
J Cell Mol Med ; 22(9): 4317-4327, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29974666

RESUMEN

The Src family kinases (SFK) are a group of signalling molecules with important regulatory functions in inflammation and haemostasis. Leucocytes and platelets express multiple isoforms of the SFKs. Previous studies used broad-spectrum pharmacological inhibitors, or murine models deficient in multiple SFK isoforms, to demonstrate the functional consequences of deficiencies in SFK signalling. Here, we hypothesized that individual SFK operate in a non-redundant fashion in the thrombo-inflammatory recruitment of monocyte during atherosclerosis. Using in vitro adhesion assays and single SFK knockout mice crossed with the ApoE-/- model of atherosclerosis, we find that SFK signalling regulates platelet-dependent recruitment of monocytes. However, loss of a single SFK, Fgr or Lyn, reduced platelet-mediated monocyte recruitment in vitro. This translated into a significant reduction in the burden of atherosclerotic disease in Fgr-/- /ApoE-/- or Lyn-/- /ApoE-/- animals. SFK signalling is not redundant in thrombo-inflammatory vascular disease and individual SFK may represent targets for therapeutic intervention.


Asunto(s)
Apolipoproteínas E/genética , Enfermedad de la Arteria Coronaria/genética , Monocitos/metabolismo , Proteínas Proto-Oncogénicas/genética , Familia-src Quinasas/genética , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/deficiencia , Adhesión Celular , Enfermedad de la Arteria Coronaria/etiología , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Dieta Alta en Grasa/efectos adversos , Femenino , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/patología , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/deficiencia , Transducción de Señal , Familia-src Quinasas/deficiencia
8.
Med Sci Monit ; 24: 1282-1294, 2018 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-29500337

RESUMEN

BACKGROUND Src and Fn14 are implicated in the aggressiveness of non-small cell lung cancer (NSCLC) cells, yet the molecular mechanism is not fully understood. MATERIAL AND METHODS The proliferation, migration, and invasion of HCC827 cells with Src knockdown were examined in vitro. The expression of Fn14 and the activation of NF-κB signaling pathway in Src-silenced HCC827 cells were detected by western blot. The role of Fn14 in Src-regulated cell migration/invasion and activation of NF-κB signaling was investigated by overexpressing Fn14 in Src knockdown NSCLC cells. Furthermore, the pro-metastatic role of Src was validated in a NSCLC metastasis mouse model. RESULTS Knockdown of Src inhibited the proliferation, migration, and invasion of HCC827 cells, which was associated with reduced levels of Fn14, p-IκBα, p-IKKß, and nuclear NF-κB p65. Overexpression of Fn14 restored the potential of migration and invasion as well as the activation of NF-κB signaling in Src-silenced NSCLC cells. In addition, silencing of Src suppressed lung metastasis of HCC827 cells in mice, and inhibited the expression of Fn14 and nuclear translocation of NF-κB p65 in vivo. CONCLUSIONS The data demonstrated that the Src/Fn14/NF-κB axis plays a critical role in NSCLC metastasis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , FN-kappa B/metabolismo , Receptor de TWEAK/metabolismo , Familia-src Quinasas/metabolismo , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Proteínas I-kappa B/metabolismo , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Receptor de TWEAK/biosíntesis , Receptor de TWEAK/genética , Factor de Transcripción ReIA/metabolismo , Familia-src Quinasas/deficiencia , Familia-src Quinasas/genética
9.
J Bone Miner Metab ; 36(3): 264-273, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-28589412

RESUMEN

Src knockout (KO) and RANKL KO mice both exhibit near complete osteopetrosis in terms of 3D-bone volume (BV) fraction by micro-CT, whereas the serum CTX concentration of Src KO is apparently normal and that of RANKL KO is 30% of wild-type (WT) despite the fact that they lack osteoclasts. By histomorphometry we found that, whereas eroded surface (ES) and osteoid surface (OS) are zero values in RANKL KO, they are indistinguishable from WT in Src KO; because of marked increase in bone surface (BS), ES/BS and OS/BS of Src KO are 30-40% of WT. While RANKL KO lack both osteoclasts and osteoblasts, Src KO reveal increased numbers of osteoclasts and indistinguishable numbers of osteoblasts compared with WT; again, on the basis of BS, N.Oc/BS is comparable to WT and N.Ob/BS is markedly decreased in Src KO. The apparently increased number of total osteoclasts may be due to increased expression of RANKL found in Src KO bone in vivo. Src has a gene dosage-dependent effect on osteoclast function in vitro, with Src-/- osteoclasts completely lacking bone-resorbing function as determined by CTX release on dentin. Thus, Src KO osteoclasts retain some bone-resorbing function in vivo. The number of osteocytes is proportionally increased in RANKL KO, while Src KO mice have relative osteocyte deficiency, raising the possibility that RANKL and Src has an unrecognized role in osteocyte survival.


Asunto(s)
Colágeno Tipo I/sangre , Péptidos/sangre , Ligando RANK/deficiencia , Familia-src Quinasas/deficiencia , Animales , Biomarcadores/metabolismo , Resorción Ósea/sangre , Resorción Ósea/genética , Resorción Ósea/metabolismo , Regulación de la Expresión Génica , Imagenología Tridimensional , Ratones , Ratones Noqueados , Osteoblastos/metabolismo , Osteocalcina/sangre , Osteoclastos/metabolismo , Osteocitos/metabolismo , Fosfatasa Ácida Tartratorresistente/sangre , Tibia/diagnóstico por imagen , Tibia/patología , Microtomografía por Rayos X
10.
Bone ; 103: 216-223, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28716550

RESUMEN

Osteopetrotic bone shows dissociation between bone mineral density (BMD) and bone strength. In this study, volumetric BMD; preferential orientation of the extracellular matrix (ECM), which is composed of collagen fibers and apatite crystals as bone material quality; and mechanical properties of the src-/- osteopetrotic and normal mouse femoral cortical bone were analyzed and compared with each other at a bone tissue level. The degree of preferential orientation of ECM along the femoral long axis was significantly decreased in the src-/- mice femur, suggesting deteriorated bone quality. Young's modulus, as a tissue-level mechanical property analyzed by nano-indentation technique along the long bone direction, also was decreased in the src-/- mice cortical femur, in spite of the similar volumetric cortical BMD. To the best of our knowledge, this is the first report to demonstrate the synchronous deterioration of Young's modulus and anisotropic ECM organization in the src-/- osteopetrotic mouse bone. These results indicate that the deterioration of the preferential ECM orientation is one major cause of the impaired mechanical property in the src-/- mouse bone.


Asunto(s)
Matriz Extracelular/patología , Fémur/fisiopatología , Osteopetrosis/fisiopatología , Animales , Módulo de Elasticidad , Ratones , Ratones Noqueados , Estrés Mecánico , Familia-src Quinasas/deficiencia
11.
J Cell Mol Med ; 21(11): 2937-2949, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28598023

RESUMEN

Mechanical ventilation (MV) used in patients with acute respiratory distress syndrome (ARDS) can increase lung inflammation and pulmonary fibrogenesis. Src is crucial in mediating the transforming growth factor (TGF)-ß1-induced epithelial-mesenchymal transition (EMT) during the fibroproliferative phase of ARDS. Nintedanib, a multitargeted tyrosine kinase inhibitor that directly blocks Src, has been approved for the treatment of idiopathic pulmonary fibrosis. The mechanisms regulating interactions among MV, EMT and Src remain unclear. In this study, we suggested hypothesized that nintedanib can suppress MV-augmented bleomycin-induced EMT and pulmonary fibrosis by inhibiting the Src pathway. Five days after administrating bleomycin to mimic acute lung injury (ALI), C57BL/6 mice, either wild-type or Src-deficient were exposed to low tidal volume (VT ) (6 ml/kg) or high VT (30 ml/kg) MV with room air for 5 hrs. Oral nintedanib was administered once daily in doses of 30, 60 and 100 mg/kg for 5 days before MV. Non-ventilated mice were used as control groups. Following bleomycin exposure in wild-type mice, high VT MV induced substantial increases in microvascular permeability, TGF-ß1, malondialdehyde, Masson's trichrome staining, collagen 1a1 gene expression, EMT (identified by colocalization of increased staining of α-smooth muscle actin and decreased staining of E-cadherin) and alveolar epithelial apoptosis (P < 0.05). Oral nintedanib, which simulated genetic downregulation of Src signalling using Src-deficient mice, dampened the MV-augmented profibrotic mediators, EMT profile, epithelial apoptotic cell death and pathologic fibrotic scores (P < 0.05). Our data indicate that nintedanib reduces high VT MV-augmented EMT and pulmonary fibrosis after bleomycin-induced ALI, partly by inhibiting the Src pathway.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Indoles/farmacología , Fibrosis Pulmonar/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Familia-src Quinasas/antagonistas & inhibidores , Actinas/genética , Actinas/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Administración Oral , Animales , Bleomicina/toxicidad , Cadherinas/genética , Cadherinas/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Modelos Animales de Enfermedad , Esquema de Medicación , Transición Epitelial-Mesenquimal/genética , Regulación de la Expresión Génica , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Respiración Artificial/efectos adversos , Transducción de Señal , Volumen de Ventilación Pulmonar , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/etiología , Lesión Pulmonar Inducida por Ventilación Mecánica/genética , Lesión Pulmonar Inducida por Ventilación Mecánica/metabolismo , Familia-src Quinasas/deficiencia , Familia-src Quinasas/genética
12.
Biomed Res ; 38(2): 123-134, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28442663

RESUMEN

Since osteoblastic activities are believed to be coupled with osteoclasts, we have attempted to histologically verify which of the distinct cellular circumstances, the presence of osteoclasts themselves or bone resorption by osteoclasts, is essential for coupled osteoblastic activity, by examining c-fos-/- or c-src-/- mice. Osteopetrotic c-fos deficient (c-fos-/-) mice have no osteoclasts, while c-src deficient (c-src-/-) mice, another osteopetrotic model, develop dysfunctional osteoclasts due to a lack of ruffled borders. c-fos-/- mice possessed no tartrate-resistant acid phosphatase (TRAPase)-reactive osteoclasts, and showed very weak tissue nonspecific alkaline phosphatase (TNALPase)-reactive mature osteoblasts. In contrast, c-src-/- mice had many TNALPase-positive osteoblasts and TRAPase-reactive osteoclasts. Interestingly, the parallel layers of TRAPase-reactive/osteopontin-positive cement lines were observed in the superficial region of c-src-/- bone matrix. This indicates the possibility that in c-src-/- mice, osteoblasts were activated to deposit new bone matrices on the surfaces that osteoclasts previously passed along, even without bone resorption. Transmission electron microscopy demonstrated cell-to-cell contacts between mature osteoblasts and neighboring ruffled border-less osteoclasts, and osteoid including many mineralized nodules in c-src-/- mice. Thus, it seems likely that osteoblastic activities would be maintained in the presence of osteoclasts, even if they are dysfunctional.


Asunto(s)
Osteoblastos/fisiología , Osteoclastos/metabolismo , Familia-src Quinasas/genética , Animales , Biomarcadores , Resorción Ósea/genética , Resorción Ósea/metabolismo , Proteína Tirosina Quinasa CSK , Calcificación Fisiológica , Comunicación Celular , Microambiente Celular , Inmunohistoquímica , Ratones , Ratones Noqueados , Osteoblastos/ultraestructura , Osteoclastos/ultraestructura , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Familia-src Quinasas/deficiencia
13.
Int J Biochem Cell Biol ; 75: 123-30, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27102411

RESUMEN

Endothelin-1 (ET-1) plays an important role in the pathophysiology of diabetes-associated cardiovascular disorders. The molecular mechanisms leading to ET-1 upregulation in diabetes are not entirely defined. c-Src tyrosine kinase regulates important pathophysiological aspects of vascular response to insults. In this study, we aimed to elucidate whether high glucose-activated c-Src signaling plays a role in the regulation of ET-1 expression. Human endothelial cells EAhy926 (ECs) were exposed to normal or high levels of glucose for 24h. Male C57BL/6J mice were rendered diabetic with streptozotocin and then treated with a specific c-Src inhibitor (Src I1) or c-Src siRNA. Real-time PCR, Western blot, and ELISA, were used to investigate ET-1 regulation. The c-Src activity and expression were selectively downregulated by pharmacological inhibition and siRNA-mediated gene silencing, respectively. High glucose dose-dependently up-regulated c-Src phosphorylation and ET-1 gene and protein expression levels in human ECs. Chemical inhibition or silencing of c-Src significantly decreased the high-glucose augmented ET-1 expression in cultured ECs. In vivo studies showed significant elevations in the aortic ET-1 mRNA expression and plasma ET-1 concentration in diabetic mice compared to non-diabetic animals. Treatment with Src I1, as well as in vivo silencing of c-Src, significantly reduced the upregulated ET-1 expression in diabetic mice. These data provide new insights into the regulation of ET-1 expression in endothelial cells in diabetes. Pharmacological targeting of c-Src activity and/or expression may represent a potential therapeutic strategy to reduce ET-1 level and to counteract diabetes-induced deleterious vascular effects.


Asunto(s)
Endotelina-1/genética , Endotelina-1/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Familia-src Quinasas/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Línea Celular , Diabetes Mellitus Experimental/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Silenciador del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/deficiencia , Familia-src Quinasas/genética
14.
Immunity ; 43(4): 764-75, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26453377

RESUMEN

Sheep red blood cells (SRBCs) have long been used as a model antigen for eliciting systemic immune responses, yet the basis for their adjuvant activity has been unknown. Here, we show that SRBCs failed to engage the inhibitory mouse SIRPα receptor on splenic CD4(+) dendritic cells (DCs), and this failure led to DC activation. Removal of the SIRPα ligand, CD47, from self-RBCs was sufficient to convert them into an adjuvant for adaptive immune responses. DC capture of Cd47(-/-) RBCs and DC activation occurred within minutes in a Src-family-kinase- and CD18-integrin-dependent manner. These findings provide an explanation for the adjuvant mechanism of SRBCs and reveal that splenic DCs survey blood cells for missing self-CD47, a process that might contribute to detecting and mounting immune responses against pathogen-infected RBCs.


Asunto(s)
Inmunidad Adaptativa , Antígeno CD47/sangre , Células Dendríticas/inmunología , Eritrocitos/inmunología , Receptores Inmunológicos/inmunología , Autotolerancia/inmunología , Bazo/inmunología , Adyuvantes Inmunológicos , Animales , Secuencia de Bases , Antígenos CD18/fisiología , Antígenos CD4/análisis , Antígeno CD47/inmunología , Movimiento Celular , Células Dendríticas/metabolismo , Eritrocitos/química , Integrinas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Quimera por Radiación , Receptores Inmunológicos/antagonistas & inhibidores , Ovinos , Organismos Libres de Patógenos Específicos , Subgrupos de Linfocitos T/inmunología , Familia-src Quinasas/deficiencia , Familia-src Quinasas/fisiología
15.
Am J Physiol Lung Cell Mol Physiol ; 309(11): L1376-81, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26453518

RESUMEN

Overwhelming acute inflammation often leads to tissue damage during endotoxemia. In the present study, we investigated the role of Lyn, a member of the Src family tyrosine kinases, in modulating inflammatory responses in a murine model of endotoxemia. We examined lung inflammatory signaling in Lyn knockout (Lyn(-/-)) mice and wild-type littermates (Lyn(+/+)) during endotoxemia. Our data indicate that Lyn deletion aggravates endotoxin-induced pulmonary inflammation and proinflammatory signaling. We found increased activation of proinflammatory transcription factor NF-κB in the lung tissues of Lyn(-/-) mice after endotoxin challenge. Furthermore, during endotoxemia, the lung tissues of Lyn(-/-) mice showed increased inflammasome activation indicated by augmented caspase-1 and IL-1ß cleavage and activation. The aggravated lung inflammatory signaling in Lyn(-/-) mice was associated with increased production of proinflammatory mediators and elevated matrix metallopeptidase 9 and reduced VE-cadherin levels. Our results suggest that Lyn kinase modulates inhibitory signaling to suppress endotoxin-induced lung inflammation.


Asunto(s)
Eliminación de Gen , Neumonía/enzimología , Neumonía/patología , Familia-src Quinasas/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Mediadores de Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos , Pulmón/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Noqueados , FN-kappa B/metabolismo , Familia-src Quinasas/deficiencia
16.
Respir Res ; 16: 112, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-26377087

RESUMEN

BACKGROUND: Mechanical ventilation and concomitant administration of hyperoxia in patients with acute respiratory distress syndrome can damage the alveolar epithelial and capillary endothelial barrier by producing inflammatory cytokines and reactive oxygen species. The Src tyrosine kinase and Smad3 are crucial inflammatory regulators used for ventilator-induced lung injury (VILI). The mechanisms regulating interactions between high-tidal-volume mechanical ventilation, hyperoxia, and acute lung injury (ALI) are unclear. We hypothesized that high-tidal-volume mechanical stretches and hyperoxia augment lung inflammation through upregulation of the Src and Smad3 pathways. METHODS: Wild-type or Src-deficient C57BL/6 mice, aged between 6 and 8 weeks, were exposed to high-tidal-volume (30 mL/kg) ventilation with room air or hyperoxia for 1-4 h after 2-mg/kg Smad3 inhibitor (SIS3) administration. Nonventilated mice were used as control subjects. RESULTS: We observed that the addition of hyperoxia to high-tidal-volume mechanical ventilation further induced microvascular permeability, neutrophil infiltration, macrophage inflammatory protein-2 and matrix metalloproteinase-9 (MMP-9) production, malondialdehyde, nicotinamide adenine dinucleotide phosphate oxidase activity, MMP-9 mRNA expression, hypoxemia, and Src and Smad3 activation (P < 0.05). Hyperoxia-induced augmentation of VILI was attenuated in Src-deficient mice and mice with pharmacological inhibition of Smad3 activity by SIS3 (P < 0.05). Mechanical ventilation of Src-deficient mice with hyperoxia further reduced the activation of Smad3. CONCLUSIONS: Our data suggest that hyperoxia-increased high-tidal-volume ventilation-induced ALI partially depends on the Src and Smad3 pathways.


Asunto(s)
Hiperoxia/complicaciones , Pulmón/enzimología , Neutrófilos/enzimología , Estrés Oxidativo , Neumonía/etiología , Respiración Artificial/efectos adversos , Transducción de Señal , Proteína smad3/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/etiología , Familia-src Quinasas/metabolismo , Animales , Permeabilidad Capilar , Quimiocina CXCL2/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Isoquinolinas/farmacología , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/metabolismo , Infiltración Neutrófila , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Estrés Oxidativo/efectos de los fármacos , Fenotipo , Neumonía/enzimología , Neumonía/genética , Neumonía/inmunología , Neumonía/patología , Neumonía/prevención & control , Piridinas/farmacología , Pirroles/farmacología , Transducción de Señal/efectos de los fármacos , Proteína smad3/antagonistas & inhibidores , Volumen de Ventilación Pulmonar , Lesión Pulmonar Inducida por Ventilación Mecánica/enzimología , Lesión Pulmonar Inducida por Ventilación Mecánica/genética , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Familia-src Quinasas/deficiencia , Familia-src Quinasas/genética
17.
Circulation ; 132(6): 490-501, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26068045

RESUMEN

BACKGROUND: Leukocyte migration is critical for the infiltration of monocytes and accumulation of monocyte-derived macrophages in inflammation. Considering that Hck and Fgr are instrumental in this process, their impact on atherosclerosis and on lesion inflammation and stability was evaluated. METHODS AND RESULTS: Hematopoietic Hck/Fgr-deficient, LDLr(-/-) chimeras, obtained by bone marrow transplantation, had smaller but, paradoxically, less stable lesions with reduced macrophage content, overt cap thinning, and necrotic core expansion as the most prominent features. Despite a Ly6C(high)-skewed proinflammatory monocyte phenotype, Hck/Fgr deficiency led to disrupted adhesion of myeloid cells to and transmigration across endothelial monolayers in vitro and atherosclerotic plaques in vivo, as assessed by intravital microscopy, flow cytometry, and histological examination of atherosclerotic arteries. Moreover, Hck/Fgr-deficient macrophages showed blunted podosome formation and mesenchymal migration capacity. In consequence, transmigrated double-knockout macrophages were seen to accumulate in the fibrous cap, potentially promoting its focal erosion, as observed for double-knockout chimeras. CONCLUSIONS: The hematopoietic deficiency of Hck and Fgr led to attenuated atherosclerotic plaque formation by abrogating endothelial adhesion and transmigration; paradoxically, it also promoted plaque instability by causing monocyte subset imbalance and subendothelial accumulation, raising a note of caution regarding src kinase-targeted intervention in plaque inflammation.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Macrófagos Peritoneales/patología , Monocitos/patología , Placa Aterosclerótica/patología , Proteínas Proto-Oncogénicas c-hck/deficiencia , Proteínas Proto-Oncogénicas/deficiencia , Familia-src Quinasas/deficiencia , Animales , Apoptosis , Adhesión Celular , Extensiones de la Superficie Celular/ultraestructura , Células Cultivadas , Células Endoteliales , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Rodamiento de Leucocito , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Placa Aterosclerótica/enzimología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-hck/genética , Proteínas Proto-Oncogénicas c-hck/fisiología , Quimera por Radiación , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores de LDL/fisiología , Migración Transendotelial y Transepitelial , Familia-src Quinasas/genética , Familia-src Quinasas/fisiología
19.
PLoS One ; 9(10): e109953, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25310015

RESUMEN

BACKGROUND: High tidal volume (VT) mechanical ventilation (MV) can induce the recruitment of neutrophils, release of inflammatory cytokines and free radicals, and disruption of alveolar epithelial and endothelial barriers. It is proposed to be the triggering factor that initiates ventilator-induced lung injury (VILI) and concomitant hyperoxia further aggravates the progression of VILI. The Src protein tyrosine kinase (PTK) family is one of the most critical families to intracellular signal transduction related to acute inflammatory responses. The anti-inflammatory abilities of induced pluripotent stem cells (iPSCs) have been shown to improve acute lung injuries (ALIs); however, the mechanisms regulating the interactions between MV, hyperoxia, and iPSCs have not been fully elucidated. In this study, we hypothesize that Src PTK plays a critical role in the regulation of oxidants and inflammation-induced VILI during hyperoxia. iPSC therapy can ameliorate acute hyperoxic VILI by suppressing the Src pathway. METHODS: Male C57BL/6 mice, either wild-type or Src-deficient, aged between 2 and 3 months were exposed to high VT (30 mL/kg) ventilation with or without hyperoxia for 1 to 4 h after the administration of Oct4/Sox2/Parp1 iPSCs at a dose of 5×10(7) cells/kg of mouse. Nonventilated mice were used for the control groups. RESULTS: High VT ventilation during hyperoxia further aggravated VILI, as demonstrated by the increases in microvascular permeability, neutrophil infiltration, macrophage inflammatory protein-2 (MIP-2) and plasminogen activator inhibitor-1 (PAI-1) production, Src activation, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, and malaldehyde (MDA) level. Administering iPSCs attenuated ALI induced by MV during hyperoxia, which benefited from the suppression of Src activation, oxidative stress, acute inflammation, and apoptosis, as indicated by the Src-deficient mice. CONCLUSION: The data suggest that iPSC-based therapy is capable of partially suppressing acute inflammatory and oxidant responses that occur during hyperoxia-augmented VILI through the inhibition of Src-dependent signaling pathway.


Asunto(s)
Hiperoxia/complicaciones , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Trasplante de Células Madre , Lesión Pulmonar Inducida por Ventilación Mecánica/etiología , Lesión Pulmonar Inducida por Ventilación Mecánica/terapia , Familia-src Quinasas/metabolismo , Animales , Apoptosis , Reprogramación Celular , Quimiocina CXCL2/metabolismo , Activación Enzimática , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibroblastos/metabolismo , Hiperoxia/fisiopatología , Inflamación/patología , Pulmón/patología , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Estrés Oxidativo , Fosforilación , Inhibidor 1 de Activador Plasminogénico/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Respiración Artificial/efectos adversos , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Volumen de Ventilación Pulmonar , Lesión Pulmonar Inducida por Ventilación Mecánica/fisiopatología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/deficiencia
20.
J Immunol ; 193(10): 5249-63, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25339668

RESUMEN

The Lyn tyrosine kinase governs the development and function of various immune cells, and its dysregulation has been linked to malignancy and autoimmunity. Using models of chemically induced colitis and enteric infection, we show that Lyn plays a critical role in regulating the intestinal microbiota and inflammatory responses as well as protection from enteric pathogens. Lyn(-/-) mice were highly susceptible to dextran sulfate sodium (DSS) colitis, characterized by significant wasting, rectal bleeding, colonic pathology, and enhanced barrier permeability. Increased DSS susceptibility in Lyn(-/-) mice required the presence of T but not B cells and correlated with dysbiosis and increased IFN-γ(+) and/or IL-17(+) colonic T cells. This dysbiosis was characterized by an expansion of segmented filamentous bacteria, associated with altered intestinal production of IL-22 and IgA, and was transmissible to wild-type mice, resulting in increased susceptibility to DSS. Lyn deficiency also resulted in an inability to control infection by the enteric pathogens Salmonella enterica serovar Typhimurium and Citrobacter rodentium. Lyn(-/-) mice exhibited profound cecal inflammation, bacterial dissemination, and morbidity following S. Typhimurium challenge and greater colonic inflammation throughout the course of C. rodentium infection. These results identify Lyn as a key regulator of the mucosal immune system, governing pathophysiology in multiple models of intestinal disease.


Asunto(s)
Colitis/inmunología , Disbiosis/inmunología , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Salmonella/inmunología , Familia-src Quinasas/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/microbiología , Citrobacter rodentium/inmunología , Citrobacter rodentium/patogenicidad , Colitis/inducido químicamente , Colitis/microbiología , Colitis/patología , Sulfato de Dextran , Susceptibilidad a Enfermedades , Disbiosis/genética , Disbiosis/microbiología , Disbiosis/patología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Femenino , Expresión Génica , Inmunoglobulina A/genética , Inmunoglobulina A/metabolismo , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucinas/genética , Interleucinas/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Ratones , Ratones Noqueados , Microbiota/inmunología , Infecciones por Salmonella/genética , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/patología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Índice de Severidad de la Enfermedad , Linfocitos T/inmunología , Linfocitos T/microbiología , Familia-src Quinasas/deficiencia , Familia-src Quinasas/genética , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...