Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 370
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Immunol ; 195(1): 1-9, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30307604

RESUMEN

The objective of this study was to test the capacity of a newly developed fusion protein of interleukin 4 (IL-4) and IL-10 [IL4-10 fusion protein (FP)] to shift multiple pro-inflammatory pathways towards immune regulation, and to inhibit pro-inflammatory activity in arthritis models. The effects of IL4-10 FP in comparison with IL-4, IL-10 and IL-4 plus IL-10 on pro- and anti-inflammatory mediators, T cells and immunoglobulin (Ig) receptors in favour of immunoregulatory activity were studied. In addition, the capacity of IL4-10 FP to inhibit pro-inflammatory activity in ex-vivo and in-vivo arthritis models was investigated. IL4-10 FP robustly inhibited pro-inflammatory cytokine [IL-1ß, tumour necrosis factor (TNF)-α, IL-6 and IL-8] production in whole blood cultures, mediated by both the IL-10 and the IL-4 moiety. IL4-10 fusion protein induced IL-1 receptor antagonist (IL-1RA) production and preserved soluble TNF receptor (sTNFR) levels, strongly increasing IL-1RA/IL-1ß and sTNFR/TNF-α ratios. In addition, IL4-10 FP strongly inhibited T helper (Th) type 1 and 17 cytokine secretion, while maintaining FoxP3 expression and up-regulating Th2 activity. In addition, while largely leaving expression of activating Fc gamma receptor (FcγR)I, III and Fc epsilon receptor (FcεR) unaffected, it significantly shifted the FcγRIIa/FcγRIIb ratio in favour of the inhibitory FcγRIIb. Moreover, IL4-10 FP robustly inhibited secretion of pro-inflammatory cytokines by rheumatoid arthritis synovial tissue and suppressed experimental arthritis in mice, without inducing B cell hyperactivity. IL4-10 fusion protein is a novel drug, signalling cells to induce immunoregulatory activity that overcomes limitations of IL-4 and IL-10 stand-alone therapy, and therefore has therapeutic potential for inflammatory diseases such as rheumatoid arthritis.


Asunto(s)
Artritis Reumatoide/terapia , Inmunoterapia/métodos , Inflamación/terapia , Interleucina-10/inmunología , Interleucina-4/uso terapéutico , Leucocitos Mononucleares/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Artritis Reumatoide/inducido químicamente , Artritis Reumatoide/inmunología , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Humanos , Inmunomodulación , Inflamación/inmunología , Interleucina-4/genética , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos BALB C , Proteoglicanos , Proteínas Recombinantes de Fusión/genética , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
2.
Am J Transplant ; 18(9): 2274-2284, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29464832

RESUMEN

The presence of donor-specific anti-HLA antibodies (DSAs) is associated with increased risk of graft failure after kidney transplant. We hypothesized that DSAs against HLA class I, class II, or both classes indicate a different risk for graft loss between deceased and living donor transplant. In this study, we investigated the impact of pretransplant DSAs, by using single antigen bead assays, on long-term graft survival in 3237 deceased and 1487 living donor kidney transplants with a negative complement-dependent crossmatch. In living donor transplants, we found a limited effect on graft survival of DSAs against class I or II antigens after transplant. Class I and II DSAs combined resulted in decreased 10-year graft survival (84% to 75%). In contrast, after deceased donor transplant, patients with class I or class II DSAs had a 10-year graft survival of 59% and 60%, respectively, both significantly lower than the survival for patients without DSAs (76%). The combination of class I and II DSAs resulted in a 10-year survival of 54% in deceased donor transplants. In conclusion, class I and II DSAs are a clear risk factor for graft loss in deceased donor transplants, while in living donor transplants, class I and II DSAs seem to be associated with an increased risk for graft failure, but this could not be assessed due to their low prevalence.


Asunto(s)
Selección de Donante , Rechazo de Injerto/mortalidad , Antígenos HLA/inmunología , Isoanticuerpos/efectos adversos , Fallo Renal Crónico/cirugía , Trasplante de Riñón/mortalidad , Donadores Vivos , Adulto , Cadáver , Femenino , Estudios de Seguimiento , Rechazo de Injerto/etiología , Rechazo de Injerto/patología , Supervivencia de Injerto , Humanos , Trasplante de Riñón/efectos adversos , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Tasa de Supervivencia
3.
Osteoarthritis Cartilage ; 26(8): 1127-1135, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29775732

RESUMEN

OBJECTIVE: Effective disease-modifying drugs for osteoarthritis (DMOAD) should preferably have chondroprotective, anti-inflammatory, and analgesic activity combined in a single molecule. We developed a fusion protein of IL4 and IL10 (IL4-10 FP), in which the biological activity of both cytokines is preserved. The present study evaluates the chondroprotective, anti-inflammatory, and analgesic activity of IL4-10 FP in in vitro and in vivo models of osteoarthritis. METHODS: Human osteoarthritic cartilage tissue and synovial tissue were cultured with IL4-10 FP. Cartilage proteoglycan turnover and release of pro-inflammatory, catabolic, and pain mediators by cartilage and synovial tissue were measured. The analgesic effect of intra-articularly injected IL4-10 FP was evaluated in a canine model of osteoarthritis by force-plate analysis. RESULTS: IL4-10 FP increased synthesis (P = 0.018) and decreased release (P = 0.018) of proteoglycans by osteoarthritic cartilage. Release of pro-inflammatory IL6 and IL8 by cartilage and synovial tissue was reduced in the presence of IL4-10 FP (all P < 0.05). The release of MMP3 by osteoarthritic cartilage and synovial tissue was decreased (P = 0.018 and 0.028) whereas TIMP1 production was not significantly changed. Furthermore, IL4-10 FP protected cartilage against destructive properties of synovial tissue mediators shown by the increased cartilage proteoglycan synthesis (P = 0.0235) and reduced proteoglycan release (P = 0.0163). Finally, intra-articular injection of IL4-10 FP improved the deficient joint loading in dogs with experimentally induced osteoarthritis. CONCLUSION: The results of current preliminary study suggest that IL4-10 FP has DMOAD potentials since it shows chondroprotective and anti-inflammatory effects in vitro, as well as potentially analgesic effect in a canine in vivo model of osteoarthritis.


Asunto(s)
Analgésicos/uso terapéutico , Antiinflamatorios/uso terapéutico , Interleucina-10/uso terapéutico , Interleucina-4/uso terapéutico , Osteoartritis/tratamiento farmacológico , Animales , Cartílago Articular/citología , Cartílago Articular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Perros , Femenino , Humanos , Masculino , Proteoglicanos/metabolismo , Proteínas Recombinantes , Membrana Sinovial/citología , Membrana Sinovial/efectos de los fármacos
4.
J Neurosci ; 36(28): 7353-63, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27413147

RESUMEN

UNLABELLED: Chronic pain is a major clinical problem that is difficult to treat and requires novel therapies. Although most pain therapies primarily target neurons, neuroinflammatory processes characterized by spinal cord and dorsal root ganglion production of proinflammatory cytokines play an important role in persistent pain states and represent potential therapeutic targets. Anti-inflammatory cytokines are attractive candidates to regulate aberrant neuroinflammatory processes, but the therapeutic potential of these cytokines as stand-alone drugs is limited. Their optimal function requires concerted actions with other regulatory cytokines, and their relatively small size causes rapid clearance. To overcome these limitations, we developed a fusion protein of the anti-inflammatory cytokines interleukin 4 (IL4) and IL10. The IL4-10 fusion protein is a 70 kDa glycosylated dimeric protein that retains the functional activity of both cytokine moieties. Intrathecal administration of IL4-10 dose-dependently inhibited persistent inflammatory pain in mice: three IL4-10 injections induced full resolution of inflammatory pain in two different mouse models of persistent inflammatory pain. Both cytokine moieties were required for optimal effects. The IL4-10 fusion protein was more effective than the individual cytokines or IL4 plus IL10 combination therapy and also inhibited allodynia in a mouse model of neuropathic pain. Mechanistically, IL4-10 inhibited the activity of glial cells and reduced spinal cord and dorsal root ganglion cytokine levels without affecting paw inflammation. In conclusion, we developed a novel fusion protein with improved efficacy to treat pain, compared with wild-type anti-inflammatory cytokines. The IL4-10 fusion protein has potential as a treatment for persistent inflammatory pain. SIGNIFICANCE STATEMENT: The treatment of chronic pain is a major clinical and societal challenge. Current therapies to treat persistent pain states are limited and often cause major side effects. Therefore, novel analgesic treatments are urgently needed. In search of a novel drug to treat chronic pain, we developed a fusion protein consisting of two prototypic regulatory cytokines, interleukin 4 (IL4) and IL10. The work presented in this manuscript shows that this IL4-10 fusion protein overcomes some major therapeutic limitations of pain treatment with individual cytokines. The IL4-10 fusion protein induces full resolution of persistent inflammatory pain in two different mouse models. These novel findings are significant, as they highlight the IL4-10 fusion protein as a long-needed potential new drug to stop persistent pain states.


Asunto(s)
Analgésicos/uso terapéutico , Inflamación/complicaciones , Interleucina-10/uso terapéutico , Interleucina-4/uso terapéutico , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Animales , Carragenina/toxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Adyuvante de Freund/toxicidad , Humanos , Inflamación/inducido químicamente , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Manejo del Dolor , Umbral del Dolor/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Médula Espinal/citología , Resultado del Tratamiento
5.
Scand J Rheumatol ; 46(1): 11-16, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27250804

RESUMEN

OBJECTIVES: Thymus and activation-regulated chemokine (TARC) attracts cells that express the C-C chemokine receptor type 4 (CCR4), including CD4 T cells. As expression of CCR4 is increased on peripheral T cells and intra-articular interleukin (IL)-17-producing cells in patients with rheumatoid arthritis (RA), we investigated whether TARC plays a role in the attraction of T cells to the synovial compartment. In addition, we assessed the role of classical dendritic cells (cDCs) in the production of TARC in RA. METHOD: TARC was measured in synovial fluid (SF) samples from RA and osteoarthritis (OA) patients. Spontaneous and thymic stromal lymphopoietin (TSLP)-induced TARC production by mononuclear cells (MCs) and CD1c cDCs from peripheral blood (PB) and SF was assessed. The role of TARC in CD4 T-cell migration towards cDCs was assessed and the contribution of CD1c-expressing cells to TARC production was studied. RESULTS: TARC concentrations were higher in SF of RA patients compared to OA patients. MCs from SF produced TARC spontaneously and produced more TARC upon stimulation than paired PBMCs. Blocking TARC strongly inhibited CD4 T-cell chemotaxis by TSLP-stimulated cDCs, associated with decreased production of tumour necrosis factor (TNF)-α. Depletion of cDCs from SFMCs strongly reduced TARC production. CONCLUSIONS: TARC levels are increased in RA SF and our data indicate that this results from production by SFMCs and in particular CD1c cDCs. TARC attracts T cells and TARC secretion by MCs is crucially dependent on the presence of CD1c cDCs. Considering the potential of SF cDCs to activate T cells and induce pro-inflammatory cytokine secretion, targeting intra-articular cDCs constitutes a novel therapeutic approach in RA.


Asunto(s)
Artritis Reumatoide/metabolismo , Linfocitos T CD4-Positivos/fisiología , Quimiocina CCL17/metabolismo , Células Dendríticas/fisiología , Líquido Sinovial/metabolismo , Adulto , Anciano , Antígenos CD1/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Quimiotaxis , Femenino , Glicoproteínas/metabolismo , Humanos , Masculino , Persona de Mediana Edad
6.
Am J Transplant ; 16(3): 987-98, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26517734

RESUMEN

Complement activation leads primarily to membrane attack complex formation and subsequent target cell lysis. Protection against self-damage is regulated by complement regulatory proteins, including CD46, CD55, and CD59. Within their promoter regions, single-nucleotide polymorphisms (SNPs) are present that could influence transcription. We analyzed these SNPs and investigated their influence on protein expression levels. A single SNP configuration in the promoter region of CD59 was found correlating with lower CD59 expression on lung endothelial cells (p = 0.016) and monocytes (p = 0.013). Lung endothelial cells with this SNP configuration secreted more profibrotic cytokine IL-6 (p = 0.047) and fibroblast growth factor ß (p = 0.036) on exposure to sublytic complement activation than cells with the opposing configuration, whereas monocytes were more susceptible to antibody-mediated complement lysis (p < 0.0001). Analysis of 137 lung transplant donors indicated that this CD59 SNP configuration correlates with impaired long-term survival (p = 0.094) and a significantly higher incidence of bronchiolitis obliterans syndrome (p = 0.046) in the recipient. These findings support a role for complement in the pathogenesis of this posttransplant complication and are the first to show a deleterious association of a donor CD59 promoter polymorphism in lung transplantation.


Asunto(s)
Antígenos CD59/genética , Rechazo de Injerto/diagnóstico , Trasplante de Pulmón , Polimorfismo Genético/genética , Complicaciones Posoperatorias , Regiones Promotoras Genéticas/genética , Donantes de Tejidos , Adolescente , Adulto , Activación de Complemento , Femenino , Estudios de Seguimiento , Rechazo de Injerto/etiología , Supervivencia de Injerto , Humanos , Masculino , Persona de Mediana Edad , Monocitos/citología , Monocitos/metabolismo , Pronóstico , Tasa de Supervivencia , Adulto Joven
7.
Allergy ; 70(5): 506-13, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25640891

RESUMEN

BACKGROUND: Recommended management of attacks of hereditary angioedema (HAE) due to C1 esterase inhibitor (C1-INH) deficiency (C1-INH-HAE) includes therapy with exogenous C1INH. Thrombotic/thromboembolic events (TEE) have been reported with plasma-derived C1INH, but so far none with recombinant human C1INH (rhC1INH). This phase III, randomized, placebo (saline)-controlled study evaluated the safety of rhC1INH 50 IU/kg for the treatment of acute attacks in 74 patients with C1-INH-HAE. METHODS: Monitoring for TEE and assessment of risk of deep vein thrombosis (DVT) by the Wells prediction rule were performed, and levels of fibrin degradation products (plasma D-dimers) were assessed before study drug administration (baseline), 2 h, and 7 days posttreatment. RESULTS: Plasma D-dimer levels were elevated in 80% of the patients (median [25th-75th percentiles]: 2149 [480-5105] µg/l; normal ≤250 µg/l) and were higher in patients with submucosal (abdominal, oropharyngeal-laryngeal) attacks (3095 [890-10000] µg/l; n = 29) compared with subcutaneous (peripheral, facial) attacks (960 [450-4060] µg/l; n = 35). Median plasma D-dimer levels were comparable across treatment groups at baseline (1874 [475-4568] µg/l rhC1INH; 2259 [586-7533] µg/l saline) and 2 h postinfusion (2389 [760-4974] µg/l rhC1INH; 2550 [310-8410] µg/l saline); median plasma D-dimer levels were decreased by Day 7 in both groups (425 [232-3240] µg/l rhC1INH; 418 [246-2318] µg/l saline). No increased risk of DVT was identified, nor any TEE reported in rhC1INH treated or controls. CONCLUSION: Elevated plasma D-dimer levels were associated with acute C1-INH-HAE attacks, particularly with submucosal involvement. However, rhC1INH therapy was not associated with thrombotic events.


Asunto(s)
Productos de Degradación de Fibrina-Fibrinógeno/análisis , Angioedema Hereditario Tipos I y II/sangre , Angioedema Hereditario Tipos I y II/complicaciones , Tromboembolia/sangre , Trombosis de la Vena/sangre , Adolescente , Adulto , Anciano , Proteínas Inactivadoras del Complemento 1/deficiencia , Proteínas Inactivadoras del Complemento 1/uso terapéutico , Proteína Inhibidora del Complemento C1 , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/uso terapéutico , Tromboembolia/etiología , Trombosis de la Vena/etiología , Adulto Joven
8.
Clin Exp Immunol ; 177(1): 280-6, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24588117

RESUMEN

Hereditary angioedema (HAE) patients experience recurrent episodes of angioedema attacks that can be painful, disfiguring and even life-threatening. The disorder results from a mutation in the gene that controls the synthesis of C1-inhibitor (C1INH). C1INH is a major regulator of activation of the contact system. It is often assumed that attacks results from uncontrolled local activation of the contact system with subsequent formation of bradykinin. To evaluate the involvement of inflammatory reactions in HAE, we analysed C-reactive protein (CRP) levels. HAE patients included in a clinical database of recombinant human C1-inhibitor (rhC1INH) studies were evaluated. For the current study we analysed CRP levels when patients were asymptomatic, during a clinical attack and in a follow-up period, and correlated these with the clinical manifestations of the attack. Data from 68 HAE patients were analysed and included CRP levels on 273 occasions. While asymptomatic, 20% of the patients analysed had increased CRP. At the onset of the attack (P = 0·049) and during the next 24 h CRP rose significantly (P = 0·002) in patients with an abdominal location, and post-attack levels were significantly higher in these patients than in patients with attacks at other locations (P = 0·034). In conclusion, CRP levels are elevated in a substantial proportion of asymptomatic HAE patients. Levels of CRP increase significantly during an abdominal attack. These data suggest low-grade systemic inflammatory reactions in HAE patients as well as a triggering event for attacks that starts prior to symptom onset.


Asunto(s)
Angioedemas Hereditarios/sangre , Angioedemas Hereditarios/tratamiento farmacológico , Enfermedades Asintomáticas , Proteína C-Reactiva/metabolismo , Proteínas Inactivadoras del Complemento 1/uso terapéutico , Abdomen/fisiopatología , Enfermedad Aguda , Adolescente , Anciano , Bradiquinina/metabolismo , Proteína C-Reactiva/análisis , Proteína Inhibidora del Complemento C1 , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Proteínas Recombinantes/uso terapéutico , Adulto Joven
9.
Eur Surg Res ; 52(1-2): 50-62, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24642533

RESUMEN

BACKGROUND: Ischemia-reperfusion (I/R) models have shown that C-reactive protein (CRP) and immunoglobulin M (IgM) are involved in complement activation. Binding of CRP and IgM to damaged cell membranes initiates complement activation and aggravates I/R injury in various organs. However, the time course of CRP- and IgM-mediated complement activation and the relation to hepatocellular injury and inflammation in liver I/R are unknown. AIM: To evaluate the time course of IgM- and CRP-related complement activation and the relation to hepatocellular injury and inflammation in a hepatic I/R rat model. METHODS: Male Wistar rats were allocated to (1) five groups of animals exposed to 60 min of partial ischemia (70%) induced via clamping of the left segmental portal triad, followed by 0, 3, 6, 12 or 24 h of reperfusion (n = 6 in each group); (2) five groups of sham-operated animals with corresponding reperfusion times (n = 5), and (3) a control group sacrificed before ischemia (n = 5). Hepatocellular injury, inflammatory response, rat plasma CRP and IgM levels and immunohistochemical depositions of CRP, IgM and C3 were assessed for each group. RESULTS: Histopathological injury scores of hematoxylin and eosin sections of ischemic liver lobes demonstrated increasing values throughout the reperfusion time with a peak at 12 h. Plasma aminotransferases (alanine aminotransferase and aspartate aminotransferase) significantly increased after 3 h of reperfusion, peaking at 6 h (3,100 ± 800 U/l; p < 0.05). Hepatic neutrophil influx significantly increased from 3 to 6 h of reperfusion (p < 0.05) and demonstrated the highest value at 12 h (1.1 ± 0.2 U/mg of protein). Plasma IL-6 levels in the ischemia groups showed peak values after 6 h of reperfusion, decreasing significantly thereafter (p < 0.05). Plasma CRP values reached highest levels after 3 h of reperfusion (mean 91 ± 5% of control pool), decreasing significantly thereafter. Rat IgM concentrations in plasma did not significantly change throughout the reperfusion time. Immunohistochemical depositions of IgM, CRP and C3 in ischemic lobes demonstrated a similar pattern in time, reaching maximum values at 12 h of reperfusion. The percentages of depositions of CRP and IgM were significantly correlated [r(S) = 0.569; p < 0.001; Spearman test]. The time course of C3 and CRP depositions throughout reperfusion and C3 and IgM staining were significantly similar [r(S) = 0.797 and r(S) = 0.656, respectively; p < 0.0001; ANOVA]. CONCLUSIONS: CRP and IgM depositions demonstrate a parallel time course throughout the reperfusion to hepatocellular damage, inflammatory response and activated complement deposition in this rat hepatic I/R model. Furthermore, the time course of CRP and IgM depositions was significantly similar to that of activated complement depositions.


Asunto(s)
Proteína C-Reactiva/metabolismo , Activación de Complemento , Inmunoglobulina M/sangre , Hígado/inmunología , Hígado/lesiones , Daño por Reperfusión/sangre , Daño por Reperfusión/inmunología , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/patología , Interleucina-6/sangre , Hígado/patología , Masculino , Neutrófilos/inmunología , Neutrófilos/patología , Ratas , Ratas Wistar
10.
Dev Neurosci ; 35(2-3): 197-211, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23689428

RESUMEN

BACKGROUND: Neonatal encephalopathy induced by perinatal asphyxia is a serious condition associated with high mortality and morbidity. Inflammation after the insult is thought to contribute to brain injury. This inflammatory response to hypoxia-ischemia (HI) may not only occur in the brain but also in peripheral organs. The aim of the present study was to investigate the effect of neonatal HI on the inflammatory response in the liver in comparison to inflammation in the brain. METHODS: HI was induced in P7 Wistar rats by unilateral carotid artery occlusion and hypoxia. Cytokine and chemokine mRNA levels were determined in the brain and liver by quantitative PCR. Polarization of brain macrophages to the M1/M2-like phenotype and infiltration of neutrophils were characterized by immunohistochemistry. RESULTS: 3 h after HI, an upregulation of the proinflammatory cytokines TNF-α and IL-1ß and anti-inflammatory IL-10 was observed in the ipsilateral hemisphere of the brain compared to mRNA levels in sham-operated animals. Additionally, cerebral CINC-1 and MCP-1 mRNA expressions were increased. We also observed increased numbers of macrophages/microglia of the M1-like phenotype as well as a small increase in granulocyte influx in the ipsilateral hemisphere. Conversely, in the liver 3 h after HI, a downregulation of TNF-α, IL-1ß, and MCP-1 and a trend towards an upregulation of IL-10 were observed compared to mRNA levels of sham-operated animals. However, hepatic CINC-1 expression was increased compared to levels in sham-operated animals. Following systemic hypoxia only, no significant changes in the expression of TNF-α, CINC-1 or MCP-1 were observed in the liver compared to sham-operated littermates, except for an upregulation in hepatic IL-1ß expression 3 h after hypoxia. Twenty-four hours after insult, cerebral ipsilateral TNF-α, MCP-1 and CINC-1 mRNA expression was still increased, together with an increase in TGF-ß expression. Moreover, an increase in macrophages/microglia of the M1-like phenotype was observed together with the appearance of macrophages/microglia of the M2-like phenotype around the cerebral lesion as well as an increase in granulocyte influx in comparison to 3 h after HI. In the liver, 24 h after HI, cytokine and chemokine responses were similar to mRNA levels in sham-operated animals except for a decrease in IL-10 and MCP-1. CONCLUSION: We describe for the first time that brain damage following neonatal HI induces an early downregulation of the proinflammatory response in the liver. HI induces an early proinflammatory response in the brain with a concomitant increase in influx of neutrophils and polarization of macrophages/microglia to the M1-like phenotype starting at 3 h and increasing up to 24 h after HI. The inflammatory state of the brain changes after 24 h, with an increase in the anti-inflammatory cytokine TGF-ß together with the appearance of macrophages/microglia of the M2-like phenotype. The downregulation of proinflammatory cytokines in the liver is not due to systemic hypoxia only, but is induced by the cerebral damage.


Asunto(s)
Hepatitis/patología , Hipoxia-Isquemia Encefálica/patología , Hipoxia/complicaciones , Inflamación/patología , Animales , Animales Recién Nacidos , Citocinas/análisis , Citocinas/biosíntesis , Hepatitis/etiología , Hepatitis/metabolismo , Hipoxia/metabolismo , Hipoxia/patología , Hipoxia-Isquemia Encefálica/etiología , Hipoxia-Isquemia Encefálica/metabolismo , Inmunohistoquímica , Inflamación/etiología , Inflamación/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Nat Genet ; 1(5): 354-8, 1992 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1363816

RESUMEN

Heterozygosity for a mutant dysfunctional C1 inhibitor protein, a member of the serine proteinase inhibitor (serpin) superfamily, results in type II hereditary angioneurotic oedema. We identified a "hinge" region mutation in C1 inhibitor with a Val to Glu replacement at P14 Val-432. Recombinant C1 inhibitors P10 Ala-->Thr and P14Val-->Glu did not form stable complexes with fluid phase C1s or kallikrein. The P14 Val-->Glu mutant, however, was cleaved to a 96K form by C1s, while the P10 Ala-->Thr mutant was not. The recombinant P10 mutant also did not complex with C1s, kallikrein or beta-factor Xlla-Sepharose. The two mutations, therefore, result in dysfunction by different mechanisms: in one (P14 Val-->Glu), the inhibitor is converted to a substrate, while in the other (P10 Ala-->Thr), interaction with target protease is blocked.


Asunto(s)
Angioedema/genética , Proteínas Inactivadoras del Complemento 1/genética , Mutación Puntual , Alanina , Secuencia de Aminoácidos , Angioedema/sangre , Animales , Secuencia de Bases , Línea Celular , Células Cultivadas , Codón/genética , Proteínas Inactivadoras del Complemento 1/química , Proteínas Inactivadoras del Complemento 1/metabolismo , Complemento C1s/metabolismo , Fibroblastos/metabolismo , Glutamatos , Ácido Glutámico , Heterocigoto , Humanos , Calicreínas/metabolismo , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Treonina , Transfección , Valina
12.
Allergy ; 67(1): 123-30, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21923668

RESUMEN

BACKGROUND: Hereditary angioedema (HAE) is a heterozygous deficiency of first component of complement-inhibitor (C1INH). Insufficient C1INH activity leads to uncontrolled activation of plasma cascade systems, which results in acute angioedema attacks in patients with HAE. Plasma-derived or recombinant C1INH products are approved for the treatment of such angioedema attacks. The target level of C1INH activity needed to achieve optimal efficacy, however, remains unknown. We determined the plasma level of C1INH associated with optimal clinical efficacy in the treatment of angioedema attacks. METHODS: Efficacy and pharmacokinetic data were reviewed from recently published placebo-controlled randomized trials in the treatment of HAE with either plasma-derived or recombinant C1INH products, tested at various doses. RESULTS: A dose-dependent effect was observed on time to the beginning of relief of symptoms, on time to resolution of symptoms, and on the response rate within 4 h. Optimal efficacy of C1INH therapy is achieved at doses ≥50 U/kg. This dose increases plasma C1INH activity in almost all patients to values ≥0.7 U/ml (70% of normal), the lower limit of the normal range. The differences in half-lives of the various C1INH products do not have an obvious effect on clinical efficacy. CONCLUSION: A review of the efficacy and pharmacokinetic data from recently published controlled studies in the treatment of HAE attacks suggests that efficacy of C1INH therapy is optimal when C1INH activity levels are restored to the normal range.


Asunto(s)
Angioedemas Hereditarios/sangre , Angioedemas Hereditarios/tratamiento farmacológico , Proteínas Inactivadoras del Complemento 1/administración & dosificación , Proteínas Inactivadoras del Complemento 1/análisis , Proteínas Inactivadoras del Complemento 1/farmacocinética , Proteína Inhibidora del Complemento C1 , Relación Dosis-Respuesta a Droga , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto
13.
Perfusion ; 26(1): 21-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20965956

RESUMEN

OBJECTIVES: To investigate whether cell-derived microparticles play a role in complement activation in pericardial blood of patients undergoing cardiac surgery with cardiopulmonary bypass (CPB) and whether microparticles in pericardial blood contribute to systemic complement activation upon retransfusion. METHODS: Pericardial blood of 13 patients was retransfused in 9 and discarded in 4 cases. Microparticles were isolated from systemic blood collected before anesthesia (T1) and at the end of CPB (T2), and from pericardial blood. The microparticles were analyzed by flow cytometry for bound complement components C1q, C4 and C3, and bound complement activator molecules C-reactive protein (CRP), serum amyloid P-component (SAP), immunoglobulin (Ig)M and IgG. Fluid-phase complement activation products (C4b/c, C3b/c) and activator molecules were determined by ELISA. RESULTS: Compared with systemic T1 blood, pericardial blood contained increased C4b/c and C3b/c, and increased levels of microparticles with bound complement components. In systemic T1 samples, microparticle-bound CRP, whereas in pericardial blood, microparticle-bound SAP and IgM were associated with complement activation. At the end of CPB, increased C3b/c (but not C4b/c) was present in systemic T2 blood compared with T1, while concentrations of microparticles binding complement components and of those binding complement activator molecules were similar. Concentrations of fluid-phase complement activation products and microparticles were similar in patients whether or not retransfused with pericardial blood. CONCLUSIONS: In pericardial blood of patients undergoing cardiac surgery with CPB, microparticles contribute to activation of the complement system via bound SAP and IgM. Retransfusion of pericardial blood, however, does not contribute to systemic complement activation.


Asunto(s)
Transfusión de Sangre Autóloga , Procedimientos Quirúrgicos Cardíacos , Puente Cardiopulmonar , Micropartículas Derivadas de Células/fisiología , Activación de Complemento/fisiología , Pericardio/fisiopatología , Proteína C-Reactiva/metabolismo , Complemento C1q/metabolismo , Citometría de Flujo , Humanos , Inmunoglobulina M/metabolismo , Componente Amiloide P Sérico/metabolismo
14.
J Exp Med ; 174(3): 729-32, 1991 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-1714936

RESUMEN

Tumor necrosis factor (TNF) may be involved in the disturbance of the procoagulant-fibrinolytic balance in septicemia, leading to microvascular thrombosis. To assess the dynamics of the fibrinolytic response to TNF in humans, we performed a crossover saline-controlled study in six healthy men, investigating the effects of a bolus intravenous injection of recombinant human TNF (50 micrograms/m2) on the stimulation and inhibition of plasminogen activation as well as on plasmin activity and inhibition. TNF induced a brief fourfold increase in the overall plasma plasminogen activator (PA) activity peaking after 1 h (p less than 0.0001), which was associated with rises in the antigenic levels of urokinase-type plasminogen activator (p less than 0.0001) and tissue-type plasminogen activator (p less than 0.0001). Plasminogen activator inhibitor type I antigen remained unchanged in the first hour, but showed a rapid eightfold increase thereafter (p less than 0.0001), which coincided with the decrease in PA activity. Generation of plasmin activity in the first hour was signified by an 11-fold rise in D-dimer levels (p less than 0.0001); inhibition of plasmin was reflected by a 36-fold rise in plasmin-alpha 2 antiplasmin complexes (p less than 0.0001), as well as by a transient 16% decrease in alpha 2-antiplasmin activity (p less than 0.01). In conclusion, TNF induced an early activation of the fibrinolytic system becoming maximal in 1 h, with a rapid inhibition thereafter. Earlier observations in the same subjects showed sustained coagulation activation for 6-12 h. The observed disbalance between the procoagulant and fibrinolytic mechanisms after TNF injection confirms the in vivo relevance of the effects of TNF on vascular endothelium in vitro and may explain the tendency towards microvascular thrombosis in septicemia.


Asunto(s)
Fibrinólisis , Factor de Necrosis Tumoral alfa/farmacología , Adulto , Activación Enzimática , Fibrinolisina/metabolismo , Humanos , Masculino , Plasminógeno/metabolismo , Inactivadores Plasminogénicos/sangre , Proteínas Recombinantes , alfa-Macroglobulinas/metabolismo
15.
J Exp Med ; 179(4): 1253-9, 1994 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-8145042

RESUMEN

The role of interleukin 6 (IL-6) in the toxic sequelae of sepsis is controversial. To assess the part of IL-6 in inflammatory responses to endotoxin, we investigated eight chimpanzees after either a bolus intravenous injection of Escherichia coli endotoxin (n = 4; 4 ng/kg) or after the same dose of endotoxin with a simultaneous bolus intravenous injection of an anti-IL-6 mAb (30 mg; n = 4). Anti-IL-6 did not affect the induction of the cytokine network (tumor necrosis factor [TNF], soluble TNF receptors types I and II, and IL-8) by endotoxin, nor did it influence the occurrence of a neutrophilic leukocytosis and neutrophil degranulation, as monitored by the measurement of elastase-alpha 1-antitrypsin complexes. In contrast, anti-IL-6 markedly attenuated endotoxin-induced activation of coagulation, monitored with the plasma levels of the prothrombin fragment F1+2 and thrombin-antithrombin III complexes, whereas activation of fibrinolysis, determined with the plasma concentrations of plasmin-alpha 2-antiplasmin complexes, remained unaltered. We conclude that IL-6 does not have a feedback effect on the release of other cytokines after injection of endotoxin, and that it is not involved in endotoxin-induced neutrophilia or neutrophil degranulation. IL-6 is, however, an important intermediate factor in activation of coagulation in low grade endotoxemia in chimpanzees.


Asunto(s)
Coagulación Sanguínea , Interleucina-6/fisiología , Toxemia/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Recuento de Células , Endotoxinas , Fibrina/metabolismo , Humanos , Inyecciones Intravenosas , Interleucina-6/inmunología , Interleucina-8/metabolismo , Monocitos/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Pan troglodytes , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
16.
J Exp Med ; 185(6): 1143-8, 1997 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-9091588

RESUMEN

To determine the effect of a physiologically relevant elevation in the plasma concentrations of epinephrine on the activation of the hemostatic mechanism during endotoxemia, 17 healthy men were studied after intravenous injection of lipopolysaccharide (LPS, 2 ng/kg), while receiving a continuous infusion of epinephrine (30 ng/kg/min) started either 3 h (n = 5) or 24 h (n = 6) before LPS injection, or an infusion of normal saline (n = 6). Activation of the coagulation system (plasma concentrations of thrombin-antithrombin III complexes and prothrombin fragment F1+2) was significantly attenuated in the groups treated with epinephrine when compared with subjects injected with LPS only (P <0.05). Epinephrine enhanced LPS-induced activation of fibrinolysis (plasma levels of tissue-type plasminogen activator and plasmin-alpha2-antiplasmin complexes; P <0.05), but did not influence inhibition of fibrinolysis (plasminogen activator inhibitor type I). In subjects infused with epinephrine, the ratio of maximal activation of coagulation and maximal activation of fibrinolysis was reduced by >50%. Hence, epinephrine exerts antithrombotic effects during endotoxemia by concurrent inhibition of coagulation, and stimulation of fibrinolysis. Epinephrine, whether endogenously produced or administered as a component of treatment, may limit the development of disseminated intravascular coagulation during systemic infection.


Asunto(s)
Anticoagulantes , Coagulación Sanguínea/efectos de los fármacos , Endotoxemia/sangre , Epinefrina/farmacología , Fibrinólisis/efectos de los fármacos , Lipopolisacáridos/toxicidad , Adulto , Epinefrina/administración & dosificación , Epinefrina/sangre , Escherichia coli , Fibrinolisina/metabolismo , Hemostasis , Humanos , Infusiones Intravenosas , Masculino , Inhibidor 1 de Activador Plasminogénico/sangre , Activador de Tejido Plasminógeno/sangre , alfa 2-Antiplasmina/metabolismo
17.
Clin Immunol ; 135(3): 490-5, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20138585

RESUMEN

BACKGROUND: C-reactive protein (CRP) is elevated in patients with acute myocardial infarction (AMI). When CRP binds to membrane phospholipids or Fc receptors, it activates the complement system. Recent studies show that CRP can be exposed on cell-derived microparticles (MP) and is associated complement activation. OBJECTIVES: We studied complement activation on circulating MP in AMI patients and healthy controls. METHODS: MP were isolated from plasma of AMI patients (n=21) and sex- and age-matched healthy individuals (n=10), and analyzed by flow cytometry for bound complement components (C1q, C4, C3) and complement inhibitor and activator molecules (C4bp, CRP, serum amyloid P component, immunoglobulins IgM and IgG). Concurrently, the levels of fluid phase complement activation products and inhibitor and activator molecules were determined. RESULTS: Fluid phase CRP, MP with bound CRP (CRP + MP), and C3 activation products were elevated in AMI patients compared to controls (P=0.032, P=0.031 and P=0.023, respectively), and fluid phase CRP correlated with CRP+ MP (r=0.84, P<0.001). Although CRP+ MP were elevated, they were not associated with C1q+ MP (r=0.32, P=0.174). In contrast, IgG+ MP were associated with C1q+ MP (r=0.73, P<0.001), C4+ MP and C3+ MP (r=0.78 and r=0.87, respectively; both P<0.001), and C4bp (r=0.63, P=0.004). In healthy individuals, CRP+ MP were strongly associated with C1q+ MP (r=0.82, P=0.007), which in turn were associated with C4+ MP and C3+ MP (r=0.68, P=0.032 and r=0.68, P=0.031, respectively). CONCLUSIONS: Despite CRP-associated complement activation on the surface of MP in healthy individuals and a strong correlation between MP-bound CRP and fluid phase CRP in AMI patients, the MP-associated complement activation is IgG- but not CRP-dependent in AMI patients.


Asunto(s)
Proteína C-Reactiva/metabolismo , Micropartículas Derivadas de Células/metabolismo , Infarto del Miocardio/metabolismo , Separación Celular , Activación de Complemento , Femenino , Citometría de Flujo , Humanos , Inmunoglobulina G/metabolismo , Masculino , Persona de Mediana Edad
18.
PLoS One ; 14(7): e0219587, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31295306

RESUMEN

OBJECTIVE: An ideal disease modifying osteoarthritis drug (DMOAD) has chondroprotective, anti-inflammatory, and analgesic effects. This study describes the production and characterization of a canine IL4-10 fusion protein (IL4-10 FP) and evaluates its in vivo DMOAD activity in a canine model of osteoarthritis (OA). DESIGN: The canine Groove model was used as an in vivo model of degenerative knee OA. Six weeks after OA induction dogs were intra-articularly injected weekly, for ten weeks, with either IL4-10 FP or phosphate buffered saline (PBS). In addition to the use of human IL4-10 FP, canine IL4-10 FP was developed and characterized in vitro, and tested in vivo. Force plate analysis (FPA) was performed to analyze joint loading as a proxy measure for pain. After ten weeks dogs were euthanized and cartilage and synovial tissue samples were analyzed by histochemistry (OARSI scores) and biochemistry (cartilage proteoglycan turnover). RESULTS: Repetitive intra-articular injections with human IL4-10 FP led to antibody formation, that blocked its functional activity. Therefore, a canine IL4-10 FP was developed, which completely inhibited LPS-induced TNFα production by canine blood cells, and increased proteoglycan synthesis of canine cartilage in vitro (p = 0.043). In vivo, canine IL4-10 FP restored the, by OA impaired, joint loading (p = 0.002) and increased cartilage proteoglycan content (p = 0.029). CONCLUSIONS: This first study on the potential DMOAD activity upon prolonged repeated treatment with IL4-10 FP demonstrates that a species-specific variant has anti-inflammatory and chondroprotective effects in vitro and chondroprotective and analgesic effects in vivo. These data warrant further research on the DMOAD potential of the IL4-10 FP.


Asunto(s)
Enfermedades de los Perros/genética , Interleucina-10/genética , Interleucina-4/genética , Osteoartritis de la Rodilla/genética , Dolor/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Cartílago Articular/metabolismo , Cartílago Articular/fisiopatología , Modelos Animales de Enfermedad , Enfermedades de los Perros/tratamiento farmacológico , Enfermedades de los Perros/fisiopatología , Perros , Humanos , Inyecciones Intraarticulares , Articulación de la Rodilla/efectos de los fármacos , Articulación de la Rodilla/patología , Osteoartritis de la Rodilla/tratamiento farmacológico , Osteoartritis de la Rodilla/patología , Dolor/genética , Proteoglicanos , Proteínas Recombinantes de Fusión/genética , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
19.
Cell Death Differ ; 14(8): 1486-96, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17479112

RESUMEN

Apoptosis is a highly controlled process, whose triggering is associated with the activation of caspases. Apoptosis can be induced via a subgroup of the tumor necrosis factor (TNF) receptor superfamily, which recruit and activate pro-caspase-8 and -10. Regulation of apoptosis is achieved by several inhibitors, including c-FLICE-inhibitory protein, which prevents apoptosis by inhibiting the pro-apoptotic activation of upstream caspases. Here we show that the human intracellular serine protease inhibitor (serpin), protease inhibitor 9 (PI9), inhibits TNF-, TNF-related apoptosis-inducing ligand- and Fas ligand-mediated apoptosis in certain TNF-sensitive cell lines. The reactive center P1 residue of PI9 was required for this inhibition since PI9 harboring a Glu --> Ala mutation in its reactive center failed to impair death receptor-induced cell death. This suggests a classical serpin-protease interaction. Indeed, PI9 inhibited apoptotic death by directly interacting with the intermediate active forms of caspase-8 and -10. This indicates that PI9 can regulate pro-apoptotic apical caspases.


Asunto(s)
Apoptosis/fisiología , Receptores de Muerte Celular/fisiología , Inhibidores de Serina Proteinasa/genética , Inhibidores de Serina Proteinasa/fisiología , Serpinas/genética , Serpinas/fisiología , Animales , Caspasa 10/metabolismo , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Línea Celular Tumoral , Proteína Ligando Fas/fisiología , Humanos , Ligandos , Ratones , Modelos Biológicos , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Transducción Genética , Factor de Necrosis Tumoral alfa/fisiología
20.
Eur J Clin Invest ; 38(4): 205-10, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18339001

RESUMEN

BACKGROUND: Secretory type-II phospholipase A(2) (sPLA(2)-II) is a cardiovascular risk marker since higher levels of this acute phase protein imply an increased risk for coronary artery disease. Moreover, it is hypothesized that local activity of sPLA(2)-II in the atherosclerotic plaque facilitates an inflammatory response to induce plaque instability or rupture. We have studied the presence of sPLA(2)-II in culprit lesions in the coronary arteries of patients with acute myocardial infarction (AMI) or angina pectoris. MATERIALS AND METHODS: We performed a histological examination of culprit lesions in 41 patients with stable (SAP) or unstable angina pectoris (UAP), or AMI using directed coronary atherectomy (DCA). Frozen slides were analysed immuno-histochemically for the presence of sPLA(2)-II, macrophages and smooth muscle cells. Immunopositive areas were calculated as a percentage of the total tissue area using image analysis software. RESULTS: Intracellular sPLA(2)-II was found in atherosclerotic lesions in the macrophages of the intima as well as in vascular smooth muscle cells. Next to this, extracellular sPLA(2)-II depositions were also found. These depositions were significantly more extensive in patients with AMI, i.e. 26%(median)[6%(25th(percentile))-44%(75th(percentile))] of the intima area, than in patients with SAP 0%(median) (0%(25th)-10%(75th); P = 0.013) or UAP 0%(median) (0%(25th)-0%(75th); P = 0.04). CONCLUSIONS: Extracellular sPLA(2)-II is more abundantly present in atherosclerotic culprit lesions that have led to myocardial infarction. This suggests a role for extracellular sPLA(2)-II in the development of complications of atherosclerotic lesions in coronary arteries.


Asunto(s)
Angina de Pecho/diagnóstico , Enfermedad de la Arteria Coronaria/diagnóstico , Infarto del Miocardio/diagnóstico , Fosfolipasas A2/análisis , Actinas/metabolismo , Adulto , Anciano , Biomarcadores/análisis , Femenino , Humanos , Inmunohistoquímica , Macrófagos/patología , Masculino , Persona de Mediana Edad , Miocitos del Músculo Liso/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA