Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neuromolecular Med ; 20(1): 63-72, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29299869

RESUMEN

Acute ischemic stroke causes a high rate of deaths and permanent neurological deficits in survivors. Current interventional treatment, in the form of enzymatic thrombolysis, benefits only a small percentage of patients. Brain ischemia triggers mobilization of innate immunity, specifically the complement system and Toll-like receptors (TLRs), ultimately leading to an exaggerated inflammatory response. Here we demonstrate that intravenous immunoglobulin (IVIG), a scavenger of potentially harmful complement fragments, and C1-esterase inhibitor (C1-INH), an inhibitor of complement activation, exert a beneficial effect on the outcome of experimental brain ischemia (I) and reperfusion (R) injury induced by transient occlusion of middle cerebral artery in mice. Both IVIG and C1-INH significantly and in a dose-responsive manner reduced brain infarction size, neurological deficit and mortality when administered to male mice 30 min before ischemia or up to 6 h after the onset of reperfusion. When combined, suboptimal doses of IVIG and C1-INH potentiated each other's neuroprotective therapeutic effects. Complement C3 and TLR2 signals were colocalized and significantly greater in brain cells adjacent to infracted brain lesions when compared to the corresponding regions of the contralateral hemisphere and to control (sham) mice. Treatment with IVIG and C1-INH effectively reduced deposition of C3b and downregulated excessive TLR2 and p-JNK1 expression at the site of I/R injury. Taken together, these results provide a rationale for potential use of IVIG and C1-INH, alone or in combination with ischemic stroke and other neurological conditions that involve inappropriately activated components of the innate immune system.


Asunto(s)
Proteína Inhibidora del Complemento C1/uso terapéutico , Inactivadores del Complemento/uso terapéutico , Trastornos Neurológicos de la Marcha/prevención & control , Inmunoglobulinas Intravenosas/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Activación de Complemento/efectos de los fármacos , Proteína Inhibidora del Complemento C1/administración & dosificación , Complemento C3b/análisis , Inactivadores del Complemento/administración & dosificación , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Femenino , Trastornos Neurológicos de la Marcha/etiología , Inmunoglobulinas Intravenosas/administración & dosificación , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Método Simple Ciego , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/genética , Regulación hacia Arriba
2.
Ann Clin Transl Neurol ; 3(7): 495-511, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27386499

RESUMEN

OBJECTIVE: Traumatic spinal cord injury (SCI) elicits immediate neural cell death, axonal damage, and disruption of the blood-spinal cord barrier, allowing circulating immune cells and blood proteins into the spinal parenchyma. The inflammatory response to SCI involves robust complement system activation, which contributes to secondary injury and impairs neurological recovery. This study aimed to determine whether intravenous immunoglobulin (IVIg), an FDA-approved treatment for inflammatory conditions, can scavenge complement activation products and improve recovery from contusive SCI. METHODS: We used functional testing, noninvasive imaging, and detailed postmortem analysis to assess whether IVIg therapy is effective in a mouse model of severe contusive SCI. RESULTS: IVIg therapy at doses of 0.5-2 g/kg improved the functional and histopathological outcomes from SCI, conferring protection against lesion enlargement, demyelination, central canal dilation, and axonal degeneration. The benefits of IVIg were detectable through noninvasive diffusion tensor imaging (DTI), with IVIg treatment counteracting the progressive SCI-induced increase in radial diffusivity (RD) in white matter. Diffusion indices significantly correlated with the functional performance of individual mice and accurately predicted the degree of myelin preservation. Further experiments revealed that IVIg therapy reduced the presence of complement activation products and phagocytically active macrophages at the lesion site, providing insight as to its mechanisms of action. INTERPRETATION: Our findings highlight the potential of using IVIg as an immunomodulatory treatment for SCI, and the value of DTI to assess tissue damage and screen for the efficacy of candidate intervention strategies in preclinical models of SCI, both quantitatively and noninvasively.

3.
J Neuroinflammation ; 12: 73, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25886362

RESUMEN

BACKGROUND: Ischemic stroke causes a high rate of deaths and permanent neurological damage in survivors. Ischemic stroke triggers the release of damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1), which activate toll-like receptors (TLRs) and receptor for advanced glycation endproducts (RAGE) in the affected area, leading to an exaggerated inflammatory response and cell death. Both TLRs and RAGE are transmembrane pattern recognition receptors (PRRs) that have been shown to contribute to ischemic stroke-induced brain injury. Intravenous immunoglobulin (IVIg) preparations obtained by fractionating human blood plasma are increasingly being used as an effective therapeutic agent in the treatment of several inflammatory diseases. Its use as a potential therapeutic agent for treatment of stroke has been proposed, but little is known about the direct neuroprotective mechanisms of IVIg. We therefore investigate whether IVIg exerts its beneficial effects on the outcome of neuronal injury by modulating HMGB1-induced TLR and RAGE expressions and activations. METHODS: Primary cortical neurons were subjected to glucose deprivation or oxygen and glucose deprivation conditions and treated with IVIg and recombinant HMGB1. C57/BL6J mice were subjected to middle cerebral artery occlusion, followed by reperfusion, and IVIg was administered intravenously 3 h after the start of reperfusion. Expression of TLRs, RAGE and downstream signalling proteins in neurons and brain tissues were evaluated by immunoblot. RESULTS: Treatment of cultured neurons with IVIg reduced simulated ischemia-induced TLR2, TLR4, TLR8 and RAGE expressions, pro-apoptotic caspase-3 cleavage and phosphorylation of the cell death-associated kinases such as c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (MAPK) as well as the p65 subunit of nuclear factor kappa B (NF-κB). These results were recapitulated in an in vivo model of stroke. IVIg treatment also upregulated the anti-apoptotic protein B-cell lymphoma 2 (Bcl-2) in cortical neurons under ischemic conditions. Finally, IVIg protected neurons against HMGB1-induced neuronal cell death by modulating TLR and RAGE expressions and signalling pathways. CONCLUSIONS: Taken together, these results provide a rationale for the potential use of IVIg to target inappropriately activated components of the innate immune system following ischemic stroke.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Inmunoglobulinas Intravenosas/farmacología , Factores Inmunológicos/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Neuronas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Glucosa/deficiencia , Hipoxia , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-24991401

RESUMEN

BACKGROUND: The brain endothelium is a key component of the blood brain barrier which is compromised following ischemia, allowing infiltration of damaging immune cells and other inflammatory molecules into the brain. Intravenous immunoglobulin (IVIg) is known to reduce infarct size in a mouse model of experimental stroke. FINDINGS: Flow cytometry analysis showed that the protective effect of IVIg in ischemia and reperfusion injury in vivo is associated with reduced leukocyte infiltration, suggesting an involvement of the endothelium. In an in vitro model of ischemia, permeability analysis of the mouse brain endothelial cell line bEnd.3 revealed that IVIg prevented the loss of permeability caused by oxygen and glucose deprivation (OGD). In addition, western blot analysis of these brain endothelial cells showed that IVIg prevented the down-regulation of tight junction proteins claudin 5 and occludin and the decline in anti-apoptotic proteins Bcl-2 and Bcl-XL caused by OGD. CONCLUSION: IVIg protects endothelial cells from ischemic insult. These studies support the use of IVIg as a pharmacological intervention for stroke therapy.

5.
Pediatr Res ; 75(5): 612-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24488088

RESUMEN

BACKGROUND: Neonatal asphyxia is one of the leading causes of death in newborn and permanent neurological disabilities in surviving children. The underlying hypoxic-ischemic (HI) injury triggers an inflammatory response leading to neuronal damage. Here, we tested the hypothesis that high-dose intravenous immunoglobulin (IVIG) could exert immunomodulatory effect in rat pups subjected to HI injury. METHODS: HI injury was induced in 7-d-old pups by ligating the common carotid artery followed by exposure to 8% oxygen for 2 h. Brain infarction was evaluated by imaging stained coronal brain sections. Neurological deficits were assessed in weeks 1 through 4 after HI. Western blotting and immunohistochemistry were used to assess complement fragment deposition in the brain tissue. RESULTS: Treatment with IVIG at 2 g/kg significantly and in a dose-responsive manner reduced brain infarction size as well as mortality and neurological deficits caused by HI. Anatomical and functional improvements in IVIG-treated pups correlated with decreased deposition of C3b complement fragments in the injured brain hemisphere. CONCLUSION: IVIG significantly improved the outcome of HI injury in rat pups and could potentially be used for the treatment of human neonatal asphyxia to target proinflammatory complement fragments.


Asunto(s)
Asfixia Neonatal/terapia , Infarto Encefálico/tratamiento farmacológico , Inmunoglobulinas Intravenosas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Animales Recién Nacidos , Asfixia Neonatal/fisiopatología , Peso Corporal , Encéfalo/fisiopatología , Infarto Encefálico/patología , Activación de Complemento , Complemento C3b/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Ratas , Ratas Wistar , Resultado del Tratamiento
6.
J Neurochem ; 122(2): 321-32, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22494053

RESUMEN

Intravenous immunoglobulin (IVIg) preparations obtained by fractionating blood plasma, are increasingly being used increasingly as an effective therapeutic agent in treatment of several inflammatory diseases. Its use as a potential therapeutic agent for treatment of stroke and Alzheimer's disease has been proposed, but little is known about the neuroprotective mechanisms of IVIg. In this study, we investigated the effect of IVIg on downstream signaling pathways that are involved in neuronal cell death in experimental models of stroke and Alzheimer's disease. Treatment of cultured neurons with IVIg reduced simulated ischemia- and amyloid ßpeptide (Aß)-induced caspase 3 cleavage, and phosphorylation of the cell death-associated kinases p38MAPK, c-Jun NH2 -terminal kinase and p65, in vitro. Additionally, Aß-induced accumulation of the lipid peroxidation product 4-hydroxynonenal was attenuated in neurons treated with IVIg. IVIg treatment also up-regulated the anti-apoptotic protein, Bcl2 in cortical neurons under ischemia-like conditions and exposure to Aß. Treatment of mice with IVIg reduced neuronal cell loss, apoptosis and infarct size, and improved functional outcome in a model of focal ischemic stroke. Together, these results indicate that IVIg acts directly on neurons to protect them against ischemic stroke and Aß-induced neuronal apoptosis by inhibiting cell death pathways and by elevating levels of the anti-apoptotic protein Bcl2.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/toxicidad , Isquemia Encefálica/prevención & control , Muerte Celular/efectos de los fármacos , Inmunoglobulinas Intravenosas/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/prevención & control , Péptidos beta-Amiloides/farmacología , Animales , Western Blotting , Isquemia Encefálica/patología , Mapeo Encefálico , Hipoxia de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glucosa/deficiencia , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Infarto de la Arteria Cerebral Media/patología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Accidente Cerebrovascular/patología , Resultado del Tratamiento , Regulación hacia Arriba
7.
Mol Immunol ; 45(16): 4073-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18706699

RESUMEN

Pathogen and self-specific antibodies are known for their ability to trigger generation of active complement fragments that then serve as effectors of cell damage. The remainder of the immunoglobulin pool of the host has the capacity to quench harmful effects of activated complement cascade by preventing active complement fragments from binding to their specific receptors. This scavenging function is mediated by different acceptor sites within the immunoglobulin molecule. Large fragments, such as C3b and C4b are preferentially bound to the Fc region, while biologically potent C3a and C5a anaphylatoxins get neutralized by low-affinity interaction with the constant domain of the F(ab)(2.) The ambivalent effect of immunoglobulins on the complement system implies their role in homeostasis as well as expansion of use of high-dose intravenous immunoglobulins in diseases and states mediated by inappropriate complement activation.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Inmunoglobulinas Intravenosas/farmacología , Animales , Relación Dosis-Respuesta a Droga , Humanos , Modelos Inmunológicos
8.
Proc Natl Acad Sci U S A ; 104(35): 14104-9, 2007 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-17715065

RESUMEN

Stroke is among the three leading causes of death worldwide and the most frequent cause of permanent disability. Brain ischemia induces an inflammatory response involving activated complement fragments. Here we show that i.v. Ig (IVIG) treatment, which scavenges complement fragments, protects brain cells against the deleterious effects of experimental ischemia and reperfusion (I/R) and prevents I/R-induced mortality in mice. Animals administered IVIG either 30 min before ischemia or after 3 h of reperfusion exhibited a 50-60% reduction of brain infarct size and a 2- to 3-fold improvement of the functional outcome. Even a single low dose of IVIG given after stroke was effective. IVIG was protective in the nonreperfusion model of murine stroke as well and did not exert any peripheral effects. Human IgG as well as intrinsic murine C3 levels were significantly higher in the infarcted brain region compared with the noninjured side, and their physical association was demonstrated by immuno-coprecipitation. C5-deficient mice were significantly protected from I/R injury compared with their wild-type littermates. Exposure of cultured neurons to oxygen/glucose deprivation resulted in increased levels of C3 associated with activation of caspase 3, a marker of apoptosis; both signals were attenuated with IVIG treatment. Our data suggest a major role for complement-mediated cell death in ischemic brain injury and the prospect of using IVIG in relatively low doses as an interventional therapy for stroke.


Asunto(s)
Encéfalo/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Infarto Cerebral/prevención & control , Inmunoglobulinas Intravenosas/uso terapéutico , Neuronas/patología , Animales , Encéfalo/inmunología , Encéfalo/patología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/inmunología , Corteza Cerebral/patología , Infarto Cerebral/inmunología , Complemento C5/deficiencia , Modelos Animales de Enfermedad , Esquema de Medicación , Factores Inmunológicos/uso terapéutico , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos
9.
Clin Immunol ; 114(2): 137-46, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15639647

RESUMEN

Intravenous immunoglobulin (IVIG) has been found useful in the treatment of various clinical entities and its effect has been associated with inhibition of complement-mediated tissue damage. The aim of this study was to determine the ability of IVIG to protect against mesenteric ischemia-reperfusion (IR)-induced local and remote injury. Rats received vehicle or IVIG (150-600 mg/kg) 5 min prior to sham operation or 30 min of superior mesenteric artery occlusion, followed by 5, 120, or 240 min of reperfusion. IVIG reduced IR-induced mucosal injury without altering IR-induced increases in PMN infiltration or LTB(4) generation. At 5 min post IR, the deposition of IgG and C3 in the lamina propria and surface epithelial cells was attenuated by IVIG. The increased capillary leak, evident at 240 min, was inhibited by IVIG and coincided with a reduction in C3 deposition in lung tissue. The beneficial effects of IVIG may be related to the ability to scavenge deleterious products.


Asunto(s)
Inmunoglobulinas Intravenosas/farmacología , Intestino Delgado/irrigación sanguínea , Daño por Reperfusión/tratamiento farmacológico , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Complemento C3/inmunología , Complemento C5/inmunología , Inmunohistoquímica , Mucosa Intestinal/irrigación sanguínea , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Intestino Delgado/inmunología , Intestino Delgado/patología , Leucotrieno B4/análisis , Leucotrieno B4/inmunología , Pulmón/inmunología , Masculino , Infiltración Neutrófila/inmunología , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Estadísticas no Paramétricas
10.
Nat Med ; 9(4): 431-8, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12612546

RESUMEN

High-dose intravenous immunoglobulin (IVIG) prevents immune damage by scavenging complement fragments C3b and C4b. We tested the hypothesis that exogenous immunoglobulin molecules also bind anaphylatoxins C3a and C5a, thereby neutralizing their pro-inflammatory effects. Single-cell calcium measurements in HMC-1 human mast cells showed that a rise in intracellular calcium caused by C3a and C5a was inhibited in a concentration-dependent manner by IVIG, F(ab)2-IVIG and irrelevant human monoclonal antibody. C3a- and C5a-induced thromboxane (TXB2) generation and histamine release from HMC-1 cells and whole-blood basophils were also suppressed by exogenous immunoglobulins. In a mouse model of asthma, immunoglobulin treatment reduced cellular migration to the lung. Lethal C5a-mediated circulatory collapse in pigs was prevented by pretreatment with F(ab)2-IVIG. Molecular modeling, surface plasmon resonance (SPR) and western blot analyses suggested a physical association between anaphylatoxins and the constant region of F(ab)2. This binding could interfere with the role of C3a and C5a in inflammation.


Asunto(s)
Complemento C3a/antagonistas & inhibidores , Complemento C5a/antagonistas & inhibidores , Inmunoglobulinas Intravenosas/farmacología , gammaglobulinas/farmacología , Animales , Asma/metabolismo , Presión Sanguínea/efectos de los fármacos , Calcio , Línea Celular , Inhibición de Migración Celular , Complemento C3a/metabolismo , Complemento C5a/metabolismo , Relación Dosis-Respuesta a Droga , Liberación de Histamina/efectos de los fármacos , Humanos , Mastocitos/metabolismo , Ratones , Síndrome de Dificultad Respiratoria/prevención & control , Porcinos , Tromboxano B2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA