Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Autoimmun ; 62: 11-21, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26094774

RESUMEN

Autoantibodies are a hallmark of autoimmune diseases, such as rheumatoid arthritis, autoimmune hepatitis, and systemic lupus erythematosus (SLE). High titers of anti-nuclear antibodies are used as surrogate marker for SLE, however their contribution to pathogenesis remains unclear. Using murine model of SLE and human samples, we studied the effect of immune stimulation on relapsing of SLE. Although autoantibodies bound to target cells in vivo, only additional activation of CD8(+) T cells converted this silent autoimmunity into overt disease. In mice as well as in humans CD8(+) T cells derived IFN-γ enhanced expression of Fc-receptors on CD11b(+) cells. High expression of Fc-receptors allowed CD11b(+) cells to bind to antibody covered target cells and to destroy them in vivo. We found that autoantibodies induce clinically relevant disease when adaptive immunity, specific for disease non-related antigen, is activated.


Asunto(s)
Antígeno CD11b/metabolismo , Interferón gamma/metabolismo , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Animales , Autoanticuerpos/inmunología , Autoinmunidad/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Ratones , Ratones Noqueados , Receptores de IgG/genética , Recurrencia
2.
J Allergy Clin Immunol ; 131(3): 815-24, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23141740

RESUMEN

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is a rare X-linked primary immunodeficiency caused by absence of Wiskott-Aldrich syndrome protein (WASP) expression, resulting in defective function of many immune cell lineages and susceptibility to severe bacterial, viral, and fungal infections. Despite a significant proportion of patients with WAS having recurrent viral infections, surprisingly little is known about the effects of WASP deficiency on antiviral immunity. OBJECTIVE: We sought to evaluate the antiviral immune response in patients with WASP deficiency in vivo. METHODS: Viral clearance and associated immunopathology were measured after infection of WASP-deficient (WAS KO) mice with lymphocytic choriomeningitis virus (LCMV). Induction of antiviral CD8(+) T-cell immunity and cytotoxicity was documented in WAS KO mice by means of temporal enumeration of total and antigen-specific T-cell numbers. Type I interferon (IFN-I) production was measured in serum in response to LCMV challenge and characterized in vivo by using IFN-I reporter mice crossed with WAS KO mice. RESULTS: WAS KO mice showed reduced viral clearance and enhanced immunopathology during LCMV infection. This was attributed to both an intrinsic CD8(+) T-cell defect and defective priming of CD8(+) T cells by dendritic cells (DCs). IFN-I production by WAS KO DCs was reduced both in vivo and in vitro. CONCLUSIONS: These studies use a well-characterized model of persistence-prone viral infection to reveal a critical deficiency of CD8(+) T-cell responses in murine WASP deficiency, in which abrogated production of IFN-I by DCs might play an important contributory role. These findings might help us to understand the immunodeficiency of WAS.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Infecciones por Rhabdoviridae/inmunología , Proteína del Síndrome de Wiskott-Aldrich/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Humanos , Virus de la Coriomeningitis Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de la Estomatitis Vesicular Indiana , Síndrome de Wiskott-Aldrich/inmunología , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/genética
3.
Int J Lab Hematol ; 46(3): 510-514, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38284270

RESUMEN

INTRODUCTION: A previously published web-based App using Gradient-boosted models (GBMs) of eight laboratory parameters was established by Oster et al. to facilitate diagnosis or exclusion of myelodysplastic syndromes (MDS) in patients. METHODS: To validate their algorithm, we compared 175 anemic patients with MDS diagnosis from our German MDS Registry with 1378 non-MDS anemic patients who consulted various specialties in the Düsseldorf university hospital. RESULTS: Based on hemoglobin level, leukocyte and platelet count, mean corpuscular volume, absolute neutrophil count, absolute monocyte count, glucose and creatinine, plus the patients' gender and age, we could not reproduce a high negative predictive value (NPV), but confirmed a useful specificity of 90.9% and a positive predictive value (PPV) of 77.1%. 1192 of 1378 controls were correctly categorized as "probably not MDS (pnMDS)" patients. A total of 65 patients were wrongly classified as "probable MDS (pMDS)," of whom 48 had alternative explanations for their altered laboratory results. In a second analysis, we included 29 patients with chronic myelomonocytic leukemia (CMML) resulting in only one label as possible MDS, suggesting that highly proliferative bone marrow disorders are correctly excluded. CONCLUSION: The possibility of reliably excluding MDS from differential diagnosis based on peripheral blood lab work appears to be attractive for patients and physicians alike while the confirmation of MDS diagnosis still requires a bone marrow biopsy.


Asunto(s)
Algoritmos , Síndromes Mielodisplásicos , Humanos , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/sangre , Femenino , Masculino , Anciano , Persona de Mediana Edad , Diagnóstico Diferencial , Anciano de 80 o más Años , Sensibilidad y Especificidad
4.
ESC Heart Fail ; 10(2): 1258-1269, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36717981

RESUMEN

AIMS: A causal link between non-ischaemic heart failure (HF) and humoral autoimmunity against G-protein-coupled receptors (GPCR) remains unclear except for Chagas' cardiomyopathy. Uncertainty arises from ambiguous reports on incidences of GPCR autoantibodies, spurious correlations of autoantibody levels with disease activity, and lack of standardization and validation of measuring procedures for putatively cardio-pathogenic GPCR autoantibodies. Here, we use validated and certified immune assays presenting native receptors as binding targets. We compared candidate GPCR autoantibody species between HF patients and healthy controls and tested associations of serum autoantibody levels with serological, haemodynamic, metabolic, and functional parameters in HF. METHODS: Ninety-five non-ischaemic HF patients undergoing transcatheter endomyocardial biopsy and 60 healthy controls were included. GPCR autoantibodies were determined in serum by IgG binding to native receptors or a cyclic peptide (for ß1AR autoantibodies). In patients, cardiac function, volumes, and myocardial structural properties were assessed by cardiac magnetic resonance imaging; right heart catheterization served for determination of cardiac haemodynamics; endomyocardial biopsies were used for histological assessment of cardiomyopathy and determination of cardiac mitochondrial oxidative function by high-resolution respirometry. RESULTS: Autoantibodies against ß1 adrenergic (ß1 AR) , M5-muscarinic (M5AR), and angiotensin II type 2 receptors (AT2R) were increased in HF (all P < 0.001). Autoantibodies against α1 -adrenergic (α1 AR) and angiotensin II type 1 receptors (AT1R) were decreased in HF (all P < 0.001). Correlation of alterations of GPCR autoantibodies with markers of cardiac or systemic inflammation or cardiac damage, haemodynamics, myocardial histology, or left ventricular inflammation (judged by T2 mapping) were weak, even when corrected for total IgG. ß1 AR autoantibodies were related inversely to markers of left ventricular fibrosis indicated by T1 mapping (r = -0.362, P < 0.05) and global longitudinal strain (r = -0.323, P < 0.05). AT2R autoantibodies were associated with improved myocardial mitochondrial coupling as measured by high-resolution respirometry in myocardial biopsies (r = -0.352, P < 0.05). In insulin-resistant HF patients, AT2R autoantibodies were decreased (r = -.240, P < 0.05), and AT1R autoantibodies were increased (r = 0.212, P < 0.05). CONCLUSIONS: GPCR autoantibodies are markedly altered in HF. However, they are correlated poorly or even inversely to haemodynamic, metabolic, and functional markers of disease severity, myocardial histology, and myocardial mitochondrial efficiency. These observations do not hint towards a specific cardio-pathogenic role of GPCR autoantibodies and suggest that further investigations are required before specific therapies directed at GPCR autoantibodies can be clinically tested in non-ischaemic HF.


Asunto(s)
Autoanticuerpos , Insuficiencia Cardíaca , Humanos , Angiotensina II , Insuficiencia Cardíaca/patología , Receptores Acoplados a Proteínas G/metabolismo , Inflamación , Inmunoglobulina G
5.
J Surg Res ; 178(2): 833-41, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22622111

RESUMEN

BACKGROUND: The perioperative morbidity and mortality of abdominal aortic aneurysm repair is linked to systemic inflammation. Important triggers of the latter are Toll-like receptors (TLRs), which play a central role in innate immunity. Ischemia/reperfusion (I/R) injury can be influenced by either TLR stimulation before I/R (preconditioning) or TLR dysfunction (deficiency or polymorphism). The influence of TLR2 stimulation or deficiency on systemic cytokine release and organ damage after aortic cross clamping has not been evaluated yet. METHODS: Wild type (WT) and TLR2-deficient mice were subjected to 1 h ischemia and 2 or 4 h reperfusion of the infrarenal aorta. One group of WT mice was preconditioned with the synthetic TLR2 agonist Pam(3)Cys-Ser-Lys(4) (Pam(3)CSK(4)). Sham-operated animals without I/R served as controls. Plasma levels of interleukin (IL)-1ß, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, granulocyte macrophage-colony stimulating factor, tumor necrosis factor-α, alanine transaminase (ALT), aspartate transaminase (AST), lactate dehydrogenase (LDH), and creatinine were measured. RESULTS: I/R injury caused by transient clamping of the infrarenal aorta led to a time-dependent increase of all measured cytokines except IL-4, IL-5, and granulocyte macrophage-colony stimulating factor. This was accompanied by elevated markers of organ damage (ALT, AST, and LDH) except creatinine. Preconditioning with Pam(3)CSK(4) led to lower plasma concentrations of all cytokines, with the exception of anti-inflammatory IL-10 which was significantly upregulated. Furthermore, ALT, AST, and LDH plasma concentrations were lower in the preconditioning group. TLR2-deficient mice similarly showed reduced signs of systemic inflammation and organ damage, although less distinctive. IL-2, IL-6, IL-12, ALT, and LDH were time dependently lower compared with WT mice. IL-10 concentration was higher in TLR2-deficient mice compared with WT. CONCLUSIONS: Both, preconditioning via TLR2 and TLR2 deficiency, ameliorate systemic inflammation and organ damage during I/R injury caused by clamping of the infrarenal aorta. Compared with untreated WT animals, Pam(3)CSK(4) preconditioned and TLR2-deficient mice showed lower concentrations of pro-inflammatory cytokines, whereas anti-inflammatory IL-10 was elevated in both groups. This was accompanied by reduced organ dysfunction parameters.


Asunto(s)
Aorta Abdominal/cirugía , Inflamación/etiología , Precondicionamiento Isquémico , Receptor Toll-Like 2/fisiología , Animales , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Lipopéptidos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor Toll-Like 2/deficiencia
6.
Eur J Immunol ; 40(9): 2401-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20683833

RESUMEN

T cells directed against self antigens play an important role in several autoimmune diseases. The available immunosuppressive compounds used to treat autoimmune diseases are limited, and often they have side effects that limit their application. T cells express ATP receptors, which could be new target molecules to treat autoimmune disease. Here we analyzed the effect of oxidized ATP (oxATP), an inhibitor of the ATP receptor P2rx7, in different murine models of T-cell-mediated autoimmune diseases. Treatment with oxATP inhibited proliferation and effector function of T cells. In the systems we used, oxATP did not obviously interfere with the innate immune response, but strongly reduced antigen-specific T-cell responses. This treatment ameliorated T-cell-mediated autoimmune type I diabetes and autoimmune encephalitis in mice. In conclusion, oxATP was found to strongly inhibit activated T cells and could thus be used to target T-cell-mediated autoimmune disease.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Diabetes Mellitus Tipo 1/metabolismo , Médula Espinal/efectos de los fármacos , Adenosina Trifosfato/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Autoinmunidad/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/fisiopatología , Femenino , Interferón gamma/biosíntesis , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X7 , Médula Espinal/patología
7.
J Clin Virol ; 74: 32-6, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26655265

RESUMEN

BACKGROUND: Previous studies have indicated that the absence of serum antibodies to influenza A H1N1 virus on day 4 after onset of symptoms predicted a fatal outcome in patients critically ill with influenza. The underlying mechanism was suggested to be the trapping of anti-influenza antibodies in pulmonary immune complexes. OBJECTIVES: To study serum and mucosal antibodies as prognostic markers in patients with severe influenza A H1N1 infection. STUDY DESIGN: Blood and respiratory samples (n=324) from 12 patients with severe influenza were analysed for anti-H1N1 antibodies with and without immune complex dissociation from symptom onset until convalescence or death (follow up 14-169 days). Eleven healthy subjects were analysed for comparison. RESULTS: One of the 12 patients died from influenza pneumonia and had no detectable anti-H1N1 serum antibodies. However, also 2 of the 11 surviving patients remained negative for anti-H1N1 serum antibodies during follow-up (20 and 41 days, respectively). In six of the 11 survivors serum antibodies on day 4 were negative, but turned positive between day 7 and 23. In the remaining 3 patients antibodies were detected in the first 4 days of illness. Mucosal IgG or IgA was detected in all of the patients regardless of their clinical outcome and in 4 of 11 healthy subjects. No mucosal immune complexes were found in the patient who died but were detected in 3 of the 11 survivors. CONCLUSIONS: This study suggests that no prognostic conclusions can be drawn from anti-H1N1 serum and mucosal antibodies in patients with severe influenza.


Asunto(s)
Anticuerpos Antivirales/análisis , Biomarcadores/análisis , Inmunidad Mucosa , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Suero/inmunología , Adulto , Femenino , Humanos , Lactante , Gripe Humana/virología , Masculino , Persona de Mediana Edad , Pronóstico , Adulto Joven
9.
Shock ; 41(5): 413-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24430541

RESUMEN

Preconditioning with noble gases serves as an effective strategy to diminish tissue injury in different organs. The aim of this study was to investigate the influence of pretreatment with the nonanesthetic noble gas helium on hepatic injury after warm ischemia and reperfusion (IR) in comparison to ischemic preconditioning (IPC). Anesthetized and ventilated rats were randomized into six groups (n = 8/group): sham: after laparotomy, the portal triad was exposed without clamping; IPC was performed with 10 min of partial liver ischemia and 10 min of reperfusion; HePC: three cycles of 5 min with inhalation of helium 70 vol% and intermittent washout; IR: 45 min of ischemia followed by 240 min of reperfusion; IPC-IR: IPC followed by hepatic IR; HePC-IR: pretreatment with helium 70 vol% followed by hepatic IR. Hepatic injury was evaluated by measurement of serum enzymes aspartate aminotransferase and alanine aminotransferase. Hepatic mRNA expression and serum levels of tumor necrosis factor α (TNF-α) and interleukin 10 (IL-10) were measured with real-time quantitative polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. Myeloperoxidase in liver tissue was assessed spectrophotometrically as a marker of neutrophil accumulation. mRNA levels of heme oxygenase 1 in liver tissue were assessed to investigate a protein of the most abundant protective system in the liver. Aspartate aminotransferase and alanine aminotransferase serum activities increased after hepatic IR (sham vs. IR; P < 0.05). The serum levels of liver enzymes after IR were significantly diminished with IPC (P < 0.05), whereas helium pretreatment had no effect. mRNA expression of TNF-α increased in all groups except IPC-IR compared with sham, whereas mRNA expression of IL-10 increased only after helium pretreatment. Serum levels of IL-10 were not affected by any intervention, whereas serum levels of TNF-α and liver myeloperoxidase were increased after IR, but not after HePC-IR. In conclusion, pretreatment with inhaled helium does not attenuate hepatic injury after warm IR of the liver, although there is evidence for a modulation of the inflammatory response.


Asunto(s)
Helio/uso terapéutico , Precondicionamiento Isquémico , Hígado/lesiones , Daño por Reperfusión/prevención & control , Animales , Ensayo de Inmunoadsorción Enzimática , Interleucina-10/sangre , Masculino , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/sangre , Isquemia Tibia
10.
Shock ; 36(1): 45-53, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21478813

RESUMEN

Remote ischemic preconditioning (RIPC) and local ischemic preconditioning (IPC) protect the myocardium from subsequent ischemia/reperfusion (I/R) injury. In this study, the protective effects of early RIPC, IPC, and the combination of both (RIPC-IPC) were characterized. Furthermore, the hypothesis was tested that protein kinase C (PKC) and mitogen-activated protein kinases (MAPKs), important mediators of IPC, are activated in RIPC. Infarct size, serum troponin T, and creatine kinase levels were assessed after 4 × 5-min noninvasive RIPC, local IPC, or a combination of both and 35 min of regional ischemia and 120 min of reperfusion. Protein kinase C ε and the MAPKs extracellular signal-regulated MAPK (ERK), c-jun N-terminal kinase (JNK), and p38 MAPK were analyzed by Western blot analysis and activity assays in the myocardium and skeletal muscle immediately after the preconditioning protocol. Remote ischemic preconditioning, IPC, and RIPC-IPC significantly reduced myocardial infarct size (RIPC-I/R: 54% ± 15%; IPC-I/R: 33% ± 15%; RIPC-IPC-I/R: 33% ± 15%; P < 0.05 vs. I/R [76% ± 14%]) and troponin T release (RIPC-I/R: 15.4 ± 6.4 ng/mL; IPC-I/R: 10.9 ± 7.0 ng/mL; RIPC-IPC-I/R: 9.8 ± 5.6 ng/mL; P < 0.05 vs. I/R [27.1 ± 12.0 ng/mL]) after myocardial I/R. Ischemic preconditioning led to an activation of PKCε and ERK 1/2, whereas RIPC did not lead to a translocation of PKCε to the mitochondria or phosphorylation of the MAPKs ERK 1/2, JNK 1/2, and p38 MAPK. Remote ischemic preconditioning did not induce translocation of PKCε to the mitochondria or phosphorylation of MAPKs in the preconditioned muscle tissue. Remote ischemic preconditioning, IPC, and RIPC-IPC exert early protection against myocardial I/R injury. Remote ischemic preconditioning and local IPC exhibit different activation dynamics of signal transducers in the myocardium. The studied PKC-MAPK pathway is likely not involved in the protective effects of RIPC.


Asunto(s)
Precondicionamiento Isquémico , Infarto del Miocardio/metabolismo , Animales , Western Blotting , Creatina Quinasa/sangre , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hemodinámica/efectos de los fármacos , Precondicionamiento Isquémico Miocárdico , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Infarto del Miocardio/sangre , Fosforilación , Proteína Quinasa C/metabolismo , Ratas , Ratas Wistar , Troponina T/sangre , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Shock ; 33(5): 507-12, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20395771

RESUMEN

Pharmacological protection from myocardial reperfusion injury, despite plenty of approaches, has still not been realized in humans. We studied the putative infarct size (IS)-sparing capacity of poly(ADP-ribose)polymerase inhibitor, INO-1001, and focused on cardiac functional recovery during reperfusion. Male farm-bred Landrace pigs were subjected to 1-h left anterior descending coronary artery occlusion followed by 3 h of reperfusion (control). Infarct size was determined by triphenyltetrazolium chloride/Evans blue staining. Plasma markers of myocardial injury (troponin T, creatine kinase, lactate dehydrogenase) were determined upon protocol completion. Cardiac function was continuously assessed via pulmonary and femoral artery catheters. INO-1001 (1 mg/kg) was administered upon reperfusion in the treatment group. As a positive control, untreated pigs were subjected to ischemic preconditioning (10-min left anterior descending coronary artery occlusion followed by 15-min reperfusion before the intervention). Ischemic preconditioning reduced myocardial damage reflected by a smaller IS and lower plasma markers of myocardial injury. INO-1001 did not reduce IS but significantly improved functional recovery (increased stroke volume, cardiac index, and mixed venous oxygen saturation) during reperfusion compared with vehicle-treated control and ischemic preconditioning. Although we could not confirm the IS-sparing capacities of poly(ADP-ribose)polymerase inhibitor, INO-1001, the drug holds the potential of hemodynamic improvement during reperfusion.


Asunto(s)
Cardiotónicos/uso terapéutico , Indoles/uso terapéutico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , Hemodinámica/efectos de los fármacos , Precondicionamiento Isquémico Miocárdico , Masculino , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Porcinos
12.
Crit Care Med ; 35(7): 1730-5, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17522584

RESUMEN

OBJECTIVE: The fibrin-derived peptide Bbeta15-42 has been shown to reduce infarct size in rodent models of ischemia-reperfusion injury. To increase its potential for translation into the clinic, we studied the effects of Bbeta15-42 in pigs, whose coronary anatomy is similar to that of humans. In addition, we evaluated the pharmacokinetics and safety of Bbeta15-42 in several species, including humans. DESIGN: Animal study and phase I trial. SETTING: University hospital and contract research laboratories. SUBJECTS: Pigs/healthy volunteers. INTERVENTIONS: Male farm-bred Landrace pigs were subjected to 1 hr of left anterior descending coronary artery occlusion followed by 3 hrs of reperfusion. At the time of reperfusion, Bbeta15-42 (2.4 mg/kg, n = 6) or random peptide (control; 2.4 mg/kg, n = 6) was administered as an intravenous bolus. As a positive control, pigs were subjected to ischemic preconditioning (n = 6). Cardiac damage and hemodynamics were recorded. Biodistribution and pharmacokinetics of Bbeta15-42 were determined in rats and dogs. In a phase I trial involving 30 male healthy volunteers, pharmacokinetics and safety were tested in a randomized, double-blinded, placebo-controlled, parallel-group, single ascending dose study. MEASUREMENTS AND MAIN RESULTS: Bbeta15-42 and ischemic preconditioning significantly reduced myocardial infarct size and troponin I levels. Bbeta15-42 also reduces interleukin-6 levels, underlining its anti-inflammatory properties. Furthermore, in humans, the pharmacokinetics of the peptide Bbeta15-42 were comparable to those of animals, and no serious adverse effects were observed. CONCLUSIONS: Bbeta15-42 elicits cardioprotection in pigs and is clinically safe in phase I testing of humans. This study confirms the new concept of a pathogenic role of fibrin derivatives in myocardial reperfusion injury, which can be inhibited by peptide Bbeta15-42.


Asunto(s)
Cardiotónicos/uso terapéutico , Productos de Degradación de Fibrina-Fibrinógeno/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Daño por Reperfusión/prevención & control , Adolescente , Adulto , Análisis de Varianza , Animales , Cardiotónicos/efectos adversos , Cardiotónicos/farmacocinética , Perros , Método Doble Ciego , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Productos de Degradación de Fibrina-Fibrinógeno/efectos adversos , Productos de Degradación de Fibrina-Fibrinógeno/farmacocinética , Semivida , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/patología , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/farmacocinética , Ratas , Porcinos , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA