Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Biochim Biophys Acta Mol Basis Dis ; 1867(11): 166212, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34311083

RESUMEN

Previous in vitro studies have shown that protein arginine N-methyltransferase 4 (PRMT4) is a co-activator for an array of cellular activities, including NF-κB-regulated pro-inflammatory responses. Here we investigated the effect of PRMT4 inhibitor TP-064 treatment on macrophage inflammation in vitro and in vivo. Exposure of RAW 264.7 monocyte/macrophages to TP-064 was associated with a significant decrease in the production of pro-inflammatory cytokines upon a lipopolysaccharide challenge. Similarly, thioglycollate-elicited peritoneal cells isolated from wildtype mice treated with TP-064 showed lowered mRNA expression levels and cytokine production of pro-inflammatory mediators interleukin (IL)-1ß, IL-6, IL-12p40, and tumor necrosis factor-α in response to lipopolysaccharide exposure. However, TP-064-treated mice exhibited an ongoing pro-inflammatory peritonitis after 5 days of thioglycollate exposure, as evident from a shift in the peritoneal macrophage polarization state from an anti-inflammatory LY6ClowCD206hi to a pro-inflammatory LY6ChiCD206low phenotype. In addition, TP-064-treated mice accumulated (activated) neutrophils within the peritoneum as well as in the blood (7-fold higher; P < 0.001) and major organs such as kidney and liver, without apparent tissue toxicity. TP-064 treatment downregulated hepatic mRNA expression levels of the PRMT4 target genes glucose-6-phosphatase catalytic subunit (-50%, P < 0.05) and the cyclin-dependent kinases 2 (-50%, P < 0.05) and 4 (-30%, P < 0.05), suggesting a direct transcriptional effect of PRMT4 also in hepatocytes. In conclusion, we have shown that the PRMT4 inhibitor TP-064 induces peritonitis-associated neutrophilia in vivo and inhibits the pro-inflammatory macrophage lipopolysaccharide response in vitro and ex vivo. Our findings suggest that TP-064 can possibly be applied as therapy in NF-κB-based inflammatory diseases.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Macrófagos Peritoneales/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Peritonitis/tratamiento farmacológico , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Humanos , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Neutrófilos/inmunología , Peritonitis/sangre , Peritonitis/inducido químicamente , Peritonitis/inmunología , Proteína-Arginina N-Metiltransferasas/metabolismo , Células RAW 264.7 , Tioglicolatos/administración & dosificación , Tioglicolatos/toxicidad
2.
Sci Rep ; 10(1): 21389, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33288796

RESUMEN

Menstrual blood-derived stromal cells (MenSCs) are emerging as a strong candidate for cell-based therapies due to their immunomodulatory properties. However, their direct impact on innate immune populations remains elusive. Since macrophages play a key role in the onset and development of inflammation, understanding MenSCs implication in the functional properties of these cells is required to refine their clinical effects during the treatment of inflammatory disorders. In this study, we assessed the effects that MenSCs had on the recruitment of macrophages and other innate immune cells in two mouse models of acute inflammation, a thioglycollate (TGC)-elicited peritonitis model and a monobacterial sepsis model. We found that, in the TGC model, MenSCs injection reduced the percentage of macrophages recruited to the peritoneum and promoted the generation of peritoneal immune cell aggregates. In the sepsis model, MenSCs exacerbated infection by diminishing the recruitment of macrophages and neutrophils to the site of infection and inducing defective bacterial clearance. Additional in vitro studies confirmed that co-culture with MenSCs impaired macrophage bactericidal properties, affecting bacterial killing and the production of reactive oxygen intermediates. Our findings suggest that MenSCs modulate the macrophage population and that this modulation must be taken into consideration when it comes to future clinical applications.


Asunto(s)
Macrófagos/citología , Menstruación/sangre , Células del Estroma/citología , Animales , Femenino , Humanos , Macrófagos/metabolismo , Ratones , Neutrófilos/metabolismo , Peritonitis/inducido químicamente , Peritonitis/metabolismo , Sepsis/inducido químicamente , Sepsis/metabolismo , Células del Estroma/metabolismo , Tioglicolatos/toxicidad
3.
Glycoconj J ; 37(3): 395-407, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32222873

RESUMEN

Through the catalysis of α2,6-linked sialylation, the enzyme ST6Gal1 is thought to play key roles in immune cell communication and homeostasis. Of particular importance, glycans with terminal α2,6-sialic acids are known to negatively regulate B cell receptor signaling and are associated with an immunosuppressive tumor microenvironment that promotes T cell anergy, suggesting that α2,6-sialic acids are a key immune inhibitory signal. Consistent with this model, mice harboring a hepatocyte-specific ablation of ST6Gal1 (H-cKO) develop a progressive and severe non-alcoholic fatty liver disease characterized by steatohepatitis. Using this H-cKO mouse, we have further discovered that loss of hepatocyte α2,6-sialylation not only increases the inflammatory state of the local tissue microenvironment, but also systemic T cell-dependent immune responses. H-cKO mice responded normally to innate and passively induced inflammation, but showed significantly increased morbidity in T cell-dependent house dust mite-antigen (HDM)-induced asthma and myelin oligodendrocyte glycoprotein (MOG) peptide-induced experimental autoimmune encephalomyelitis (EAE). We further discovered that H-cKO mice have a profound shift toward effector/memory T cells even among unchallenged mice, and that macrophages from both the liver and spleen expressed the inhibitory and α2,6-sialic acid-specific glycan binding molecule CD22. These findings align with previously reported pro-inflammatory changes in liver macrophages, and support a model in which the liver microenvironment sets a systemic immune tone that is regulated by tissue α2,6-sialylation and mediated by liver macrophages and systemic T cells.


Asunto(s)
Hepatocitos/metabolismo , Inmunidad Innata/fisiología , Sialiltransferasas/metabolismo , Linfocitos T/inmunología , Animales , Asma/etiología , Asma/inmunología , Colitis/inducido químicamente , Colitis/inmunología , Modelos Animales de Enfermedad , Hepatocitos/inmunología , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Hígado/inmunología , Pulmón/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Noqueados , Peritonitis/inducido químicamente , Peritonitis/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Sialiltransferasas/genética , Tioglicolatos/toxicidad
4.
Sci Rep ; 9(1): 1808, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755642

RESUMEN

PiT1/SLC20A1 is an inorganic phosphate transporter with additional functions including the regulation of TNFα-induced apoptosis, erythropoiesis, cell proliferation and insulin signaling. Recent data suggest a relationship between PiT1 and NF-κB-dependent inflammation: (i) Pit1 mRNA is up-regulated in the context of NF-κB pathway activation; (ii) NF-κB target gene transcription is decreased in PiT1-deficient conditions. This led us to investigate the role of PiT1 in lipopolysaccharide (LPS)-induced inflammation. MCP-1 and IL-6 concentrations were impaired in PiT1-deficient bone marrow derived macrophages (BMDMs) upon LPS stimulation. Lower MCP-1 and IL-6 serum levels were observed in Mx1-Cre; Pit1lox/lox mice dosed intraperitoneally with LPS. Lower PiT1 expression correlated with decreased in vitro wound healing and lower reactive oxygen species levels. Reduced IκB degradation and lower p65 nuclear translocation were observed in PiT1-deficient cells stimulated with LPS. Conversely, PiT1 expression was induced in vitro upon LPS stimulation. Addition of an NF-κB inhibitor abolished LPS-induced PiT1 expression. Furthermore, we showed that p65 expression activated Pit1 promoter activity. Finally, ChIP assays demonstrated that p65 directly binds to the mPit1 promoter in response to LPS. These data demonstrate a completely novel function of PiT1 in the response to LPS and provide mechanistic insights into the regulation of PiT1 expression by NF-κB.


Asunto(s)
Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Factor de Transcripción Pit-1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , NADPH Oxidasa 2/metabolismo , FN-kappa B/metabolismo , Peritonitis/inducido químicamente , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Tioglicolatos/toxicidad , Factor de Transcripción Pit-1/genética , Factor de Necrosis Tumoral alfa/metabolismo , Cicatrización de Heridas/efectos de los fármacos
5.
Nat Commun ; 9(1): 2416, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29925830

RESUMEN

Controlled macrophage differentiation and activation in the initiation and resolution of inflammation is crucial for averting progression to chronic inflammatory and autoimmune diseases. Here we show a negative feedback mechanism for proinflammatory IFN-γ activation of macrophages driven by macrophage-associated matrix metalloproteinase 12 (MMP12). Through C-terminal truncation of IFN-γ at 135Glu↓Leu136 the IFN-γ receptor-binding site was efficiently removed thereby reducing JAK-STAT1 signaling and IFN-γ activation of proinflammatory macrophages. In acute peritonitis this signature was absent in Mmp12 -/- mice and recapitulated in Mmp12 +/+ mice treated with a MMP12-specific inhibitor. Similarly, loss-of-MMP12 increases IFN-γ-dependent proinflammatory markers and iNOS+/MHC class II+ macrophage accumulation with worse lymphadenopathy, arthritic synovitis and lupus glomerulonephritis. In active human systemic lupus erythematosus, MMP12 levels were lower and IFN-γ higher compared to treated patients or healthy individuals. Hence, macrophage proteolytic truncation of IFN-γ attenuates classical activation of macrophages as a prelude for resolving inflammation.


Asunto(s)
Interferón gamma/metabolismo , Nefritis Lúpica/inmunología , Activación de Macrófagos/inmunología , Metaloproteinasa 12 de la Matriz/metabolismo , Animales , Artritis/inmunología , Artritis/patología , Biopsia , Línea Celular , Colágeno/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Riñón/patología , Nefritis Lúpica/patología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Metaloproteinasa 12 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peritonitis/inducido químicamente , Peritonitis/inmunología , Peritonitis/patología , Cultivo Primario de Células , Proteolisis , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células THP-1 , Tioglicolatos/toxicidad
6.
BMC Res Notes ; 10(1): 695, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29208049

RESUMEN

OBJECTIVE: In this study, we investigate the diversity and modulation of leukocyte populations represented in the gates defined by size and granularity at different time points of thioglycollate-induced peritonitis in mouse. RESULTS: The inflammatory cells were distributed into four regions (R1-R4) of a data plot graph defined by cell size and granularity. R1 and R2 contained agranular cells that were small in size and predominately included T (CD3+) lymphocytes along with B (B220+) lymphocytes. Macrophages (F4/80+) were the predominant cells found in the R3 region. However, these cells were present in all regions, albeit at a lower frequency in R1 and R2. Granulocytes (Gr1+) were mainly distributed in R3 and R4. The wide distribution of F4/80+ and Gr1+ cells may reflect the recruitment and activation state of the different macrophage and granulocyte populations. Based on these observations, size and granularity may contribute to an initial step in the analysis and sorting of thioglycollate-elicited peritoneal exudate cells. However, the developmental stage and cell activation state may interfere with cell segregation using size and granularity as parameters.


Asunto(s)
Exudados y Transudados , Peritonitis/patología , Tioglicolatos/toxicidad , Animales , Separación Celular , Granulocitos/patología , Macrófagos/patología , Ratones
7.
Mol Med Rep ; 13(4): 3335-41, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26936418

RESUMEN

Antimicrobial, antifungal and anti-inflammatory effects of essential oils extracted from Chamaecyparis obtusa (EOCO) have previously been reported. In the present study, the anti-inflammatory effects of EOCO were investigated in two murine models of inflammation: Carrageenan-induced paw edema and thioglycollate-induced peritonitis, and in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells. The expression levels of proinflammatory cytokines were analyzed by ELISA, the expression of nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) were determined by western blotting, and nitrite concentration was measured using Griess reagent. In mice with carrageenan-induced edema, paw thickness and the expression levels of interleukin (IL)­1ß and IL-6 in paw homogenates were significantly decreased in the EOCO (5 and 10 mg/kg) group, as compared with the control group. In mice with thioglycollate-induced peritonitis, treatment with EOCO (5 and 10 mg/kg) reduced the number of total cells and suppressed tumor necrosis factor­α (TNF­α), IL­1ß and IL­6 levels in peritoneal fluid. In addition, EOCO reduced nitric oxide, TNF­α and IL­6 production, and suppressed iNOS and COX­2 expression in LPS­stimulated RAW 264.7 cells. These results suggest that EOCO may exert anti­inflammatory effects in vivo and in vitro, and that these effects may be associated with the inhibition of inflammatory mediators. Therefore, EOCO may be considered an effective therapeutic agent for the treatment of inflammatory diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Chamaecyparis/química , Edema/tratamiento farmacológico , Aceites Volátiles/uso terapéutico , Peritonitis/tratamiento farmacológico , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Carragenina/toxicidad , Supervivencia Celular/efectos de los fármacos , Chamaecyparis/metabolismo , Ciclooxigenasa 2/metabolismo , Citocinas/análisis , Modelos Animales de Enfermedad , Edema/inducido químicamente , Edema/patología , Lipopolisacáridos/toxicidad , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/análisis , Óxido Nítrico Sintasa de Tipo II/metabolismo , Aceites Volátiles/química , Aceites Volátiles/farmacología , Peritonitis/inducido químicamente , Peritonitis/patología , Células RAW 264.7 , Tioglicolatos/toxicidad
8.
FASEB J ; 29(5): 1914-29, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25609430

RESUMEN

Models of microbe-elicited peritonitis have been invaluable to identify mechanisms underlying inflammation resolution, but whether resolution mechanisms differ from an inflammatory agent to another has not been determined. Thus, we analyzed the cellular and molecular components of the resolution phase of non-microbe-induced inflammation. In thioglycollate (TG)-induced peritonitis, resolution started at 12 h (Tmax) and displayed a 22 h resolution interval (Ri). During resolution, lipoxin A4, resolvin (Rv) D1 and RvD2, protectin D1 (PD1), and maresin 1 (MaR1) were transiently produced while RvD5 was continually generated. In addition, docosahexaenoic acid (DHA)-derived mediators were produced to a higher extent than in microbial peritonitis. We also investigated leukocyte infiltration and clearance in peritoneal tissues surrounding the inflammatory site. In the omentum, resolution parameters, neutrophil apoptosis, and efferocytosis were similar to those of the peritoneal cavity. However, we noticed long-term persistence of M2-polarized macrophages and B-lymphocytes in the omentum after TG administration, whereas zymosan injection caused M1/M2-macrophage and T-lymphocyte persistence regardless of the magnitude of the inflammatory response. Our study indicates that some aspects of resolution are shaped in a stimulus-specific manner, and it ultimately argues that the tissues surrounding the inflammatory site must also be considered to address the inflammatory response globally.


Asunto(s)
Linfocitos B/inmunología , Inflamación/inmunología , Leucocitos/inmunología , Macrófagos/inmunología , Peritonitis/inmunología , Peritonitis/metabolismo , Tioglicolatos/toxicidad , Animales , Apoptosis/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Western Blotting , Células Cultivadas , Ácidos Docosahexaenoicos/genética , Ácidos Docosahexaenoicos/metabolismo , Femenino , Citometría de Flujo , Técnicas para Inmunoenzimas , Inflamación/metabolismo , Inflamación/patología , Leucocitos/metabolismo , Leucocitos/patología , Lípidos/análisis , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Epiplón/inmunología , Epiplón/metabolismo , Epiplón/patología , Peritonitis/inducido químicamente , Fagocitosis/fisiología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Zimosan/toxicidad
9.
Mediators Inflamm ; 2014: 173403, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25294953

RESUMEN

INTRODUCTION: Macrophage reprogramming is vital for resolution of acute inflammation. Parenteral vitamin C (VitC) attenuates proinflammatory states in murine and human sepsis. However information about the mechanism by which VitC regulates resolution of inflammation is limited. METHODS: To examine whether physiological levels of VitC modulate resolution of inflammation, we used transgenic mice lacking L-gulono-γ-lactone oxidase. VitC sufficient/deficient mice were subjected to a thioglycollate-elicited peritonitis model of sterile inflammation. Some VitC deficient mice received daily parenteral VitC (200 mg/kg) for 3 or 5 days following thioglycollate infusion. Peritoneal macrophages harvested on day 3 or day 5 were examined for intracellular VitC levels, pro- and anti-inflammatory protein and lipid mediators, mitochondrial function, and response to lipopolysaccharide (LPS). The THP-1 cell line was used to determine the modulatory activities of VitC in activated human macrophages. RESULTS: VitC deficiency significantly delayed resolution of inflammation and generated an exaggerated proinflammatory response to in vitro LPS stimulation. VitC sufficiency and in vivo VitC supplementation restored macrophage phenotype and function in VitC deficient mice. VitC loading of THP-1 macrophages attenuated LPS-induced proinflammatory responses. CONCLUSION: VitC sufficiency favorably modulates macrophage function. In vivo or in vitro VitC supplementation restores macrophage phenotype and function leading to timely resolution of inflammation.


Asunto(s)
Ácido Ascórbico/metabolismo , Ácido Ascórbico/uso terapéutico , Inflamación/tratamiento farmacológico , Animales , Western Blotting , Línea Celular , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Peritonitis/inducido químicamente , Peritonitis/tratamiento farmacológico , Peritonitis/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Tioglicolatos/toxicidad
10.
Mediators Inflamm ; 2013: 931562, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23606798

RESUMEN

Understanding of spatiotemporal profiling of inflammatory mediators and their associations with MΦ accumulation is crucial to elucidate the complex immune properties. Here, we used murine thioglycollate elicited peritonitis to determine concentrations of 23 inflammatory mediators in peritoneal exudates and plasma before (day 0) and after (days 1 and 3) thioglycollate administration to peritoneal cavities; these mediators included TNF-α , FGF-9, IFN-γ , IP-10, RANTES, IL-1α , IL-6, IL-7, IL-10, IL-11, IL-12p70, IL-17A, lymphotactin, OSM, KC/GRO, SCF, MIP-1ß , MIP-2, TIMP-1, VEGF-A, MCP-1, MCP-3, and MCP-5. Our results showed that concentrations of most mediators in exudates and plasma reached peak levels on day 1 and were significantly reduced on day 3. Conversely, MΦ numbers started to increase on day 1 and reached peak levels on day 3. Moreover, LPS treatment in vitro significantly induced mediator productions in cell culture media and lysates from MΦ isolated on day 3. Our results also showed that on day 0, concentrations of many mediators in plasma were higher than those in exudates, whereas on day 1, the trend was reversed. Overall, the findings from thioglycollate elicited peritonitis reveal that reversible chemotactic gradients between peritoneal exudates and blood exist in basal and inflamed conditions and the inflammatory mediator production in vivo is disassociated with macrophage accumulation during inflammation resolution.


Asunto(s)
Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Peritonitis/inducido químicamente , Peritonitis/metabolismo , Tioglicolatos/toxicidad , Animales , Células Cultivadas , Citometría de Flujo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Peritonitis/inmunología
11.
Protein Cell ; 4(4): 259-65, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23549611

RESUMEN

Pannexin-1 (Panx1) forms nonselective large channel in cell plasma membrane and has been shown to be associated with NLRP3 inflammasome activation, ATP release and phagocytes recruitment. In the current study, by manipulation of Panx1 expression in human myeloid cells and application of Panx1 deficient mice, we failed to find a correlation between Panx1 and NLRP3 inflammasome activation, although an interaction between these two proteins was evident. However, in thioglycollate induced peritonitis, Panx1 deficient mice showed much more phagocytes infiltration. Further analyses showed that mice deficient for Panx1 exhibited enlarged F4/80(low)Gr1(-)Ly6C(-)cell population in the peritonea. Our study thus reveals an important role for Panx1 in regulation of peritoneal cell population and peritonitis development.


Asunto(s)
Proteínas Portadoras/metabolismo , Conexinas/metabolismo , Inflamasomas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Cavidad Peritoneal/citología , Animales , Línea Celular , Conexinas/antagonistas & inhibidores , Conexinas/deficiencia , Conexinas/genética , Células HEK293 , Humanos , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Peritonitis/inducido químicamente , Peritonitis/metabolismo , Peritonitis/patología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Tioglicolatos/toxicidad
12.
Circ Res ; 113(1): 52-61, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23584255

RESUMEN

RATIONALE: Apoptotic cell phagocytosis (efferocytosis) is mediated by specific receptors and is essential for resolution of inflammation. In chronic inflammation, apoptotic cell clearance is dysfunctional and soluble levels of several apoptotic cell receptors are elevated. Reports have identified proteolytic cleavage as a mechanism capable of releasing soluble apoptotic cell receptors, but the functional implications of their proteolysis are unclear. OBJECTIVE: To test the hypothesis that ADAM17-mediated cleavage of apoptotic cell receptors limits efferocytosis in vivo. METHODS AND RESULTS: In vivo comparison of macrophage efferocytosis in wild-type and Adam17-null hematopoietic chimeras demonstrates that ADAM17 deficiency leads to a 60% increase in efferocytosis and an enhanced anti-inflammatory phenotype in a model of peritonitis. In vitro uptake of phosphatidylserine liposomes identifies the dual-pass apoptotic cell receptor CD36 as a major contributor to enhanced efferocytosis, and CD36 surface levels are elevated on macrophages from Adam17-null mice. Further, temporal elevation of CD36 expression with inflammation may also contribute to its impact. Soluble CD36 from macrophage-conditioned media comprises 2 species based on Western blotting, and mass spectrometry identifies 3 N-terminal peptides that represent probable cleavage sites. Levels of soluble CD36 are decreased in Adam17-null conditioned media, providing evidence for involvement of ADAM17 in CD36 cleavage. Importantly, enhanced efferocytosis in vivo by macrophages lacking ADAM17 is CD36 dependent and accelerates macrophage clearance from the peritoneum, thus promoting resolution of inflammation and highlighting the impact of increased apoptotic cell uptake. CONCLUSIONS: Our studies demonstrate the importance of ADAM17-mediated proteolysis for in vivo efferocytosis regulation and suggest a possible mechanistic link between chronic inflammation and defective efferocytosis.


Asunto(s)
Proteínas ADAM/fisiología , Apoptosis/fisiología , Antígenos CD36/fisiología , Macrófagos Peritoneales/enzimología , Peritonitis/enzimología , Fagocitosis/fisiología , Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Proteína ADAM17 , Secuencia de Aminoácidos , Animales , Trasplante de Médula Ósea , Antígenos CD36/química , Quimera , Medios de Cultivo Condicionados/farmacología , Dexametasona/farmacología , Células Madre Embrionarias/trasplante , Liposomas , Macrófagos Peritoneales/fisiología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Peritonitis/inducido químicamente , Peritonitis/patología , Fenotipo , Tioglicolatos/toxicidad , Timocitos/patología , Timocitos/trasplante
13.
Int J Nanomedicine ; 7: 2631-40, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22679373

RESUMEN

Quantum dots (QDs) have many potential clinical and biological applications because of their advantages over traditional fluorescent dyes. However, the genotoxicity potential of QDs still remains unclear. In this paper, a plasmid-based system was designed to explore the genotoxic mechanism of QDs by detecting changes in DNA configuration and biological activities. The direct chemicobiological interactions between DNA and mercaptoacetic acid-coated CdSecore QDs (MAA-QDs) were investigated. After incubation with different concentrations of MAA-QDs (0.043, 0.13, 0.4, 1.2, and 3.6 µmol/L) in the dark, the DNA conversion of the covalently closed circular (CCC) DNA to the open circular (OC) DNA was significantly enhanced (from 13.9% ± 2.2% to 59.9% ± 12.8%) while the residual transformation activity of plasmid DNA was greatly decreased (from 80.7% ± 12.8% to 13.6% ± 0.8%), which indicated that the damages to the DNA structure and biological activities induced by MAA-QDs were concentration-dependent. The electrospray ionization mass spectrometry data suggested that the observed genotoxicity might be correlated with the cadmium-mercaptoacetic acid complex (Cd-MAA) that is formed in the solution of MAA-QDs. Circular dichroism spectroscopy and transformation assay results indicated that the Cd-MAA complex might interact with DNA through the groove-binding mode and prefer binding to DNA fragments with high adenine and thymine content. Furthermore, the plasmid transformation assay could be used as an effective method to evaluate the genotoxicities of nanoparticles.


Asunto(s)
Compuestos de Cadmio/toxicidad , Cadmio/toxicidad , ADN Circular/efectos de los fármacos , Mutágenos/toxicidad , Puntos Cuánticos , Compuestos de Selenio/toxicidad , Tioglicolatos/toxicidad , Animales , Composición de Base , Bovinos , Dicroismo Circular , ADN Circular/química , Escherichia coli/genética , Pruebas de Mutagenicidad , Conformación de Ácido Nucleico/efectos de los fármacos , Espectrometría de Fluorescencia , Espectrometría de Masa por Ionización de Electrospray , Transformación Bacteriana/efectos de los fármacos
14.
J Immunol ; 189(2): 980-7, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22706085

RESUMEN

Various heterotrimeric G(i) proteins are considered to be involved in cell migration and effector function of immune cells. The underlying mechanisms, how they control the activation of myeloid effector cells, are not well understood. To elucidate isoform-redundant and -specific roles for Gα(i) proteins in these processes, we analyzed mice genetically deficient in Gα(i2) or Gα(i3). First, we show an altered distribution of tissue macrophages and blood monocytes in the absence of Gα(i2) but not Gα(i3). Gα(i2)-deficient but not wild-type or Gα(i3)-deficient mice exhibited reduced recruitment of macrophages in experimental models of thioglycollate-induced peritonitis and LPS-triggered lung injury. In contrast, genetic ablation of Gα(i2) had no effect on Gα(i)-dependent peritoneal cytokine production in vitro and the phagocytosis-promoting function of the Gα(i)-coupled C5a anaphylatoxin receptor by liver macrophages in vivo. Interestingly, actin rearrangement and CCL2- and C5a anaphylatoxin receptor-induced chemotaxis but not macrophage CCR2 and C5a anaphylatoxin receptor expression were reduced in the specific absence of Gα(i2). Furthermore, knockdown of Gα(i2) caused decreased cell migration and motility of RAW 264.7 cells, which was rescued by transfection of Gα(i2) but not Gα(i3). These results indicate that Gα(i2), albeit redundant to Gα(i3) in some macrophage activation processes, clearly exhibits a Gα(i) isoform-specific role in the regulation of macrophage migration.


Asunto(s)
Inhibición de Migración Celular/inmunología , Subunidad alfa de la Proteína de Unión al GTP Gi2/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Macrófagos/inmunología , Macrófagos/patología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Animales , Inhibición de Migración Celular/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Peritonitis/inducido químicamente , Peritonitis/inmunología , Peritonitis/patología , Tioglicolatos/toxicidad
15.
Endocrinology ; 153(6): 2724-34, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22495675

RESUMEN

Prolactin (PRL) is mainly expressed in the pituitary in rodents, whereas in humans, expression is observed in many extrapituitary sites, including lymphocytes. Due to the lack of adequate experimental models, the function of locally produced PRL in the immune system is largely unknown. Using transgenic rats that express luciferase under the control of extensive human PRL regulatory regions, we characterized immune cell responses to thioglycollate (TG)-induced peritonitis. Resident populations of myeloid cells in the peritoneal cavity of untreated rats expressed barely detectable levels of luciferase. In contrast, during TG-induced peritonitis, cell-specific expression in both neutrophils and monocytes/macrophages in peritoneal exudates increased dramatically. Elevated luciferase expression was also detectable in peripheral blood and bone marrow CD11b(+) cells. Ex vivo stimulation of primary myeloid cells showed activation of the human extrapituitary promoter by TNF-α, lipopolysaccharide, or TG. These findings were confirmed in human peripheral blood monocytes, showing that the transgenic rat provided a faithful model for the human gene. Thus, the resolution of an inflammatory response is associated with dramatic activation of the PRL gene promoter in the myeloid lineage.


Asunto(s)
Células Mieloides/metabolismo , Peritonitis/genética , Prolactina/genética , Transcripción Genética , Animales , Células de la Médula Ósea/metabolismo , Antígeno CD11b/metabolismo , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Lipopolisacáridos/farmacología , Luciferasas/genética , Luciferasas/metabolismo , Macrófagos/metabolismo , Microscopía Fluorescente , Monocitos/metabolismo , Neutrófilos/metabolismo , Peritonitis/inducido químicamente , Peritonitis/metabolismo , Prolactina/metabolismo , Ratas , Ratas Transgénicas , Secuencias Reguladoras de Ácidos Nucleicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tioglicolatos/farmacología , Tioglicolatos/toxicidad , Factor de Necrosis Tumoral alfa/farmacología
16.
J Exp Med ; 209(2): 407-21, 2012 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-22291096

RESUMEN

Neutrophils trigger inflammation-induced acute kidney injury (AKI), a frequent and potentially lethal occurrence in humans. Molecular mechanisms underlying neutrophil recruitment to sites of inflammation have proved elusive. In this study, we demonstrate that SLP-76 (SH2 domain-containing leukocyte phosphoprotein of 76 kD) and ADAP (adhesion and degranulation promoting adaptor protein) are involved in E-selectin-mediated integrin activation and slow leukocyte rolling, which promotes ischemia-reperfusion-induced AKI in mice. By using genetically engineered mice and transduced Slp76(-/-) primary leukocytes, we demonstrate that ADAP as well as two N-terminal-located tyrosines and the SH2 domain of SLP-76 are required for downstream signaling and slow leukocyte rolling. The Tec family kinase Bruton tyrosine kinase is downstream of SLP-76 and, together with ADAP, regulates PI3Kγ (phosphoinositide 3-kinase-γ)- and PLCγ2 (phospholipase Cγ2)-dependent pathways. Blocking both pathways completely abolishes integrin affinity and avidity regulation. Thus, SLP-76 and ADAP are involved in E-selectin-mediated integrin activation and neutrophil recruitment to inflamed kidneys, which may underlie the development of life-threatening ischemia-reperfusion-induced AKI in humans.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Riñón/irrigación sanguínea , Riñón/inmunología , Rodamiento de Leucocito/inmunología , Neutrófilos/inmunología , Fosfoproteínas/metabolismo , Daño por Reperfusión/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Agammaglobulinemia Tirosina Quinasa , Animales , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Selectina E/metabolismo , Vectores Genéticos , Humanos , Integrinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Peritonitis/inducido químicamente , Peritonitis/inmunología , Fosfolipasa C gamma/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Proteínas Tirosina Quinasas/metabolismo , Retroviridae , Tioglicolatos/toxicidad , Transducción Genética
17.
Blood ; 118(20): 5622-30, 2011 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-21940822

RESUMEN

Localization of plasmin on macrophages and activation of pro-MMP-9 play key roles in macrophage recruitment in the inflammatory response. These functions are promoted by plasminogen receptors exposing C-terminal basic residues on the macrophage surface. Recently, we identified a novel transmembrane plasminogen receptor, Plg-R(KT), which exposes a C-terminal lysine on the cell surface. In the present study, we investigated the role of Plg-R(KT) in macrophage invasion, chemotactic migration, and recruitment. Plg-R(KT) was prominently expressed in membranes of human peripheral blood monocytes and monocytoid cells. Plasminogen activation by urokinase-type plasminogen activator (uPA) was markedly inhibited (by 39%) by treatment with anti-Plg-R(KT) mAb. Treatment of monocytes with anti-Plg-R(KT) mAb substantially inhibited invasion through the representative matrix, Matrigel, in response to MCP-1 (by 54% compared with isotype control). Furthermore, chemotactic migration was also inhibited by treatment with anti-Plg-R(KT) mAb (by 64%). In a mouse model of thioglycollate-induced peritonitis, anti-Plg-R(KT) mAb markedly inhibited macrophage recruitment (by 58%), concomitant with a reduction in pro-MMP-9 activation in the inflamed peritoneum. Treatment with anti-Plg-R(KT) mAb did not further reduce the low level of macrophage recruitment in plasminogen-null mice. We conclude that Plg-R(KT) plays a key role in the plasminogen-dependent regulation of macrophage invasion, chemotactic migration, and recruitment in the inflammatory response.


Asunto(s)
Movimiento Celular/inmunología , Macrófagos/citología , Peritonitis/inmunología , Plasminógeno/inmunología , Receptores de Superficie Celular/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Materiales Biocompatibles , Movimiento Celular/efectos de los fármacos , Colágeno , Modelos Animales de Enfermedad , Combinación de Medicamentos , Femenino , Fibrinolisina/metabolismo , Fibrinolisina/farmacología , Humanos , Laminina , Macrófagos/inmunología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Monocitos/citología , Monocitos/inmunología , Peritonitis/inducido químicamente , Peritonitis/metabolismo , Plasminógeno/genética , Plasminógeno/metabolismo , Proteoglicanos , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/inmunología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Tioglicolatos/toxicidad
18.
Pharmacology ; 88(1-2): 100-13, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21865767

RESUMEN

Bortezomib (Velcade®) is a proteasome inhibitor that has been approved for the treatment of multiple myeloma and mantle cell lymphoma. It has been shown to inhibit the expression of cell adhesion molecules, co-stimulatory molecules, and NFκB activation, to deplete alloreactive T lymphocytes, and to decrease Th1 cytokine production. The anti-inflammatory effects of bortezomib were further investigated in this current set of studies. Systemic treatment with bortezomib was efficacious in the thioglycolate-induced MCP-1 production model, and the dinitrofluorobenzene-induced delayed-type hypersensitivity model. Psoriasis is an autoimmune disease that affects about 2% of the world population. Many treatments have been reported with varying degrees of efficacy. A topical bortezomib formulation was developed to minimize systemic exposure. Its tolerability was investigated in a topical imiquimod (IMQ)-induced psoriasis model. Daily application of IMQ on mouse skin induced inflamed scaly skin lesions resembling plaque-type psoriasis. Fatality was observed in the 1-mg/ml dose group. At 0.1 and 0.01 mg/ml, bortezomib potentiated IMQ-induced erythema, scaling, skin thickening, and caused necrotic lesions. Lower doses had no effect on the clinical observations. Histologically, bortezomib dose-dependently increased parakeratosis, hyperkeratosis, acanthosis, and inflammatory cell infiltration. This study demonstrated that topical bortezomib is not suitable for the treatment of psoriasis.


Asunto(s)
Antiinflamatorios/farmacología , Ácidos Borónicos/farmacología , Factores Inmunológicos/farmacología , Pirazinas/farmacología , Adyuvantes Inmunológicos/toxicidad , Aminoquinolinas/toxicidad , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/uso terapéutico , Antiinflamatorios/toxicidad , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/uso terapéutico , Ácidos Borónicos/toxicidad , Bortezomib , Dinitrofluorobenceno/toxicidad , Modelos Animales de Enfermedad , Composición de Medicamentos , Evaluación Preclínica de Medicamentos , Estabilidad de Medicamentos , Femenino , Hipersensibilidad Tardía/inducido químicamente , Hipersensibilidad Tardía/tratamiento farmacológico , Imiquimod , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/uso terapéutico , Factores Inmunológicos/toxicidad , Irritantes/toxicidad , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Peritonitis/inducido químicamente , Peritonitis/tratamiento farmacológico , Psoriasis/inducido químicamente , Psoriasis/tratamiento farmacológico , Pirazinas/administración & dosificación , Pirazinas/uso terapéutico , Pirazinas/toxicidad , Distribución Aleatoria , Temperatura , Tioglicolatos/toxicidad
19.
Eur J Cancer ; 47(1): 151-61, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20850297

RESUMEN

Animal models provide an important tool for investigating the biology of cancer and for testing the efficacy of novel treatments. Here we describe several aspects of the transgenic MexTAg mouse that develops asbestos-induced mesothelioma. Targeted expression of the TAg transgene causes mesothelioma to develop more rapidly after asbestos exposure in wild-type mice with 100% incidence compared to 30% incidence in wild-type mice. MexTAg mice do not develop spontaneous mesothelioma and exhibit a very low incidence of other tumours. Here we show that TAg does not affect the aggressiveness or rate of progression of the mesotheliomas, suggesting that the oncogene alters only the rate at which disease is initiated. The instillation of an alternative inflammatory agent, thioglycollate, did not induce mesotheliomas, demonstrating acute inflammation is not sufficient for tumour development in MexTAg mice. We found that neither the age of a mouse at the time of exposure nor its gender were prognostic factors. MexTAg mice with mesotheliomas respond to treatment with a cytotoxic drug in a similar way to that of patients with mesothelioma, demonstrating the validity of the model. We also show that long-term treatment with a COX-2 inhibitor prior to the development of disease does not have a survival benefit, suggesting that this is not a useful cancer prevention therapy for asbestos-exposed individuals. The model is robust and suitable for testing a wide variety of protocols and a range of translational studies.


Asunto(s)
Amianto/toxicidad , Contactina 2/metabolismo , Modelos Animales de Enfermedad , Mesotelioma/etiología , Neoplasias Peritoneales/etiología , Animales , Antineoplásicos/uso terapéutico , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Femenino , Irritantes/toxicidad , Masculino , Mesotelioma/tratamiento farmacológico , Ratones , Ratones Transgénicos , Neoplasias Peritoneales/tratamiento farmacológico , Tioglicolatos/toxicidad
20.
Blood ; 116(7): 1136-46, 2010 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-20424186

RESUMEN

The plasminogen activation system plays an integral role in the migration of macrophages in response to an inflammatory stimulus, and the binding of plasminogen to its cell-surface receptor initiates this process. Although previous studies from our laboratory have shown the importance of the plasminogen receptor S100A10 in cancer cell plasmin production, the potential role of this protein in macrophage migration has not been investigated. Using thioglycollate to induce a peritoneal inflammatory response, we demonstrate, for the first time, that compared with wild-type (WT) mice, macrophage migration across the peritoneal membrane into the peritoneal cavity in S100A10-deficient (S100A10(-/-)) mice was decreased by up to 53% at 24, 48, and 72 hours. Furthermore, the number of S100A10-deficient macrophages that infiltrated Matrigel plugs was reduced by 8-fold compared with their WT counterpart in vivo. Compared with WT macrophages, macrophages from S100A10(-/-) mice demonstrated a 50% reduction in plasmin-dependent invasion across a Matrigel barrier and a 45% reduction in plasmin generation in vitro. This loss in plasmin-dependent invasion was in part the result of a decreased generation of plasmin and a decreased activation of pro-MMP-9 by S100A10-deficient macrophages. This study establishes a direct involvement of S100A10 in macrophage recruitment in response to inflammatory stimuli.


Asunto(s)
Anexina A2/fisiología , Inflamación/patología , Macrófagos Peritoneales/metabolismo , Plasminógeno/metabolismo , Proteínas S100/fisiología , Animales , Apoptosis , Western Blotting , Adhesión Celular , Movimiento Celular , Proliferación Celular , Colágeno/metabolismo , Combinación de Medicamentos , Femenino , Fibrinolisina/metabolismo , Citometría de Flujo , Técnicas para Inmunoenzimas , Inflamación/inducido químicamente , Inflamación/metabolismo , Laminina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteoglicanos/metabolismo , Tioglicolatos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA