Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 128(4): 1397-1412, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29369823

RESUMEN

Lupus nephritis (LN) often results in progressive renal dysfunction. The inactive rhomboid 2 (iRhom2) is a newly identified key regulator of A disintegrin and metalloprotease 17 (ADAM17), whose substrates, such as TNF-α and heparin-binding EGF (HB-EGF), have been implicated in the pathogenesis of chronic kidney diseases. Here, we demonstrate that deficiency of iRhom2 protects the lupus-prone Fcgr2b-/- mice from developing severe kidney damage without altering anti-double-stranded DNA (anti-dsDNA) Ab production by simultaneously blocking HB-EGF/EGFR and TNF-α signaling in the kidney tissues. Unbiased transcriptome profiling of kidneys and kidney macrophages revealed that TNF-α and HB-EGF/EGFR signaling pathways are highly upregulated in Fcgr2b-/- mice, alterations that were diminished in the absence of iRhom2. Pharmacological blockade of either TNF-α or EGFR signaling protected Fcgr2b-/- mice from severe renal damage. Finally, kidneys from LN patients showed increased iRhom2 and HB-EGF expression, with interstitial HB-EGF expression significantly associated with chronicity indices. Our data suggest that activation of iRhom2/ADAM17-dependent TNF-α and EGFR signaling plays a crucial role in mediating irreversible kidney damage in LN, thereby uncovering a target for selective and simultaneous dual inhibition of 2 major pathological pathways in the effector arm of the disease.


Asunto(s)
Proteínas Portadoras/biosíntesis , Receptores ErbB/metabolismo , Riñón/metabolismo , Nefritis Lúpica/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Receptores ErbB/genética , Regulación de la Expresión Génica , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Riñón/patología , Nefritis Lúpica/genética , Nefritis Lúpica/patología , Ratones , Ratones Noqueados , Receptores de IgG/genética , Receptores de IgG/metabolismo , Factor de Necrosis Tumoral alfa/genética
2.
Eur J Immunol ; 46(12): 2737-2748, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27601030

RESUMEN

CSF1R (colony stimulating factor 1 receptor) is the main receptor for CSF1 and has crucial roles in regulating myelopoeisis. CSF1R can be proteolytically released from the cell surface by ADAM17 (A disintegrin and metalloprotease 17). Here, we identified CSF1R as a major substrate of ADAM17 in an unbiased degradomics screen. We explored the impact of CSF1R shedding by ADAM17 and its upstream regulator, inactive rhomboid protein 2 (iRhom2, gene name Rhbdf2), on homeostatic development of mouse myeloid cells. In iRhom2-/- mice, we found constitutive accumulation of membrane-bound CSF1R on myeloid cells at steady state, although cell numbers of these populations were not altered. However, in the context of mixed bone marrow (BM) chimera, under competitive pressure, iRhom2-/- BM progenitor-derived monocytes, tissue macrophages and lung DCs showed a repopulation advantage over those derived from wild-type (WT) BM progenitors, suggesting enhanced CSF1R signaling in the absence of iRhom2. In vitro experiments indicate that iRhom2-/- Lin- SCA-1+ c-Kit+ (LSKs) cells, but not granulocyte-macrophage progenitors (GMPs), had faster growth rates than WT cells in response to CSF1. Our results shed light on an important role of iRhom2/ADAM17 pathway in regulation of CSF1R shedding and repopulation of monocytes, macrophages and DCs.


Asunto(s)
Proteína ADAM17/metabolismo , Células de la Médula Ósea/fisiología , Proteínas Portadoras/metabolismo , Células Progenitoras Mieloides/fisiología , Mielopoyesis , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteína ADAM17/genética , Animales , Proteínas Portadoras/genética , Células Cultivadas , Células Dendríticas/fisiología , Femenino , Regulación de la Expresión Génica , Pulmón/patología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Transducción de Señal , Quimera por Trasplante
3.
Arthritis Rheumatol ; 68(3): 730-739, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26474194

RESUMEN

OBJECTIVE: Pregnancies in women with the antiphospholipid syndrome (APS) are frequently complicated by fetal loss and intrauterine growth restriction (IUGR). How circulating antiphospholipid antibodies (aPL) cause pregnancy complications in APS is poorly understood. We sought to determine whether the low-density lipoprotein receptor family member apolipoprotein E receptor 2 (ApoER2) mediates trophoblast dysfunction and pregnancy complications induced by aPL. METHODS: Placental and trophoblast ApoER2 expression was evaluated by immunohistochemistry and immunoblotting. Normal human IgG and aPL were purified from healthy individuals and APS patients, respectively. The role of ApoER2 in aPL-induced changes in trophoblast proliferation and migration and in kinase activation was assessed using RNA interference in HTR-8/SVneo cells. The participation of ApoER2 in aPL-induced pregnancy loss and IUGR was evaluated in pregnant ApoER2(+/+) and ApoER2(-/-) mice injected with aPL or normal human IgG. RESULTS: We found that ApoER2 is abundant in human and mouse placental trophoblasts and in multiple trophoblast-derived cell lines, including HTR-8/SVneo cells. ApoER2 and its interaction with the cell surface protein ß2 -glycoprotein I were required for aPL-induced inhibition of cultured trophoblast proliferation and migration. In parallel, aPL antagonism of Akt kinase activation by epidermal growth factor in trophoblasts was mediated by ApoER2. Furthermore, in a murine passive-transfer model of pregnancy complications of APS, ApoER2(-/-) mice were protected from both aPL-induced fetal loss and aPL-induced IUGR. CONCLUSION: ApoER2 plays a major role in the attenuation of trophoblast function by aPL, and the receptor mediates aPL-induced pregnancy complications in vivo in mice. ApoER2-directed interventions can now potentially be developed to combat the pregnancy complications associated with APS.


Asunto(s)
Síndrome Antifosfolípido/inmunología , Proteínas Relacionadas con Receptor de LDL/inmunología , Complicaciones del Embarazo/inmunología , Trofoblastos/inmunología , Aborto Espontáneo/inmunología , Animales , Línea Celular , Femenino , Humanos , Immunoblotting , Inmunohistoquímica , Queratina-7/inmunología , Ratones , Placenta/inmunología , Embarazo , Interferencia de ARN , Transfección , Trofoblastos/citología , beta 2 Glicoproteína I/inmunología
4.
J Immunol ; 195(3): 1129-38, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26071558

RESUMEN

Defective placentation and subsequent placental insufficiency lead to maternal and fetal adverse pregnancy outcome, but their pathologic mechanisms are unclear, and treatment remains elusive. The mildly hypertensive BPH/5 mouse recapitulates many features of human adverse pregnancy outcome, with pregnancies characterized by fetal loss, growth restriction, abnormal placental development, and defects in maternal decidual arteries. Using this model, we show that recruitment of neutrophils triggered by complement activation at the maternal/fetal interface leads to elevation in local TNF-α levels, reduction of the essential angiogenic factor vascular endothelial growth factor, and, ultimately, abnormal placentation and fetal death. Blockade of complement with inhibitors specifically targeted to sites of complement activation, depletion of neutrophils, or blockade of TNF-α improves spiral artery remodeling and rescues pregnancies. These data underscore the importance of innate immune system activation in the pathogenesis of placental insufficiency and identify novel methods for treatment of pregnancy loss mediated by abnormal placentation.


Asunto(s)
Aborto Espontáneo/prevención & control , Activación de Complemento/inmunología , Inmunidad Innata , Neutrófilos/inmunología , Insuficiencia Placentaria/prevención & control , Placentación/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Aborto Espontáneo/inmunología , Animales , Línea Celular , Proteínas Inactivadoras de Complemento/farmacología , Proteínas del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Femenino , Muerte Fetal , Retardo del Crecimiento Fetal/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/inmunología , Placenta/citología , Insuficiencia Placentaria/inmunología , Embarazo , Trofoblastos/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Arthritis Rheumatol ; 67(4): 977-87, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25603823

RESUMEN

OBJECTIVE: To investigate whether an elevated interferon-α (IFNα) level early in pregnancy is associated with poor pregnancy outcomes and to examine the relationship of an elevated IFNα level to angiogenic imbalance. METHODS: Women were enrolled in a longitudinal case-control study of pregnant patients with lupus. Serum samples obtained monthly throughout pregnancy were assayed for IFNα and for the antiangiogenic factor soluble Flt-1 and the proangiogenic factor placenta growth factor (PlGF). Each of 28 patients with systemic lupus erythematosus (SLE) with a poor pregnancy outcome was matched to an SLE patient with an uncomplicated pregnancy and to a pregnant healthy control. The effects of IFNα and/or soluble Flt-1 on human endothelial cells and endothelial cell-trophoblast interactions were assessed. RESULTS: Compared to SLE patients with uncomplicated pregnancies, patients with preeclampsia had increased IFNα levels before clinical symptoms. Patients without autoimmune disease who developed preeclampsia did not have increased IFNα levels. In SLE patients with low IFNα levels, marked angiogenic imbalance (higher soluble Flt-1, lower PlGF, and higher soluble Flt-1:PlGF ratios) preceded maternal manifestations of preeclampsia, whereas in SLE patients with high IFNα levels, preeclampsia occurred without evidence of systemic angiogenic imbalance. Treatment of human endothelial cells with soluble Flt-1 induced expression of sFLT1 messenger RNA, and IFNα dramatically amplified responses to soluble Flt-1. In a model of spiral artery transformation, only the combination of IFNα and soluble Flt-1 disrupted the ability of trophoblast cells to remodel endothelial tube structures. CONCLUSION: Our findings identify a new mechanism by which IFNα induces an antiangiogenic milieu and increases the sensitivity of endothelial cells to soluble Flt-1, and suggest that elevated IFNα levels may contribute to the pathogenesis of preeclampsia in some pregnant patients with SLE.


Asunto(s)
Interferón-alfa/sangre , Lupus Eritematoso Sistémico/sangre , Preeclampsia/sangre , Proteínas Gestacionales/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Lupus Eritematoso Sistémico/complicaciones , Factor de Crecimiento Placentario , Embarazo , Resultado del Embarazo
6.
J Immunol ; 191(4): 1800-7, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23851693

RESUMEN

Cholinergic neural output has been shown to modulate innate immune responses to infection, injury and ischemia through stimulation of α7 nicotinic acetylcholine receptors (α7nAChR) on mononuclear phagocytes. We tested the hypothesis that cholinergic neurotransmitters, similar to those released through activation of a neural reflex, regulate responses to products of the adaptive immune system, specifically immune complex (IC)-mediated activation of effector cells. In this study, we show that stimulation of α7nAChR on human polymorphonuclear neutrophils (PMNs) and blood mononuclear phagocytes in vitro attenuates C5aR- and FcγR-triggered generation of reactive oxygen species, expression of leukocyte markers involved in cell recruitment and adhesion, and release of TNF-α and other proinflammatory cytokines. We show that this pathway is operative in vivo. Ligation of cholinergic receptors blunts IC-triggered responses in the reverse peritoneal Arthus reaction in mice. The selective 7nAChR agonist GTS21 decreased PMN accumulation and release of cytokines and chemokines at sites of IC deposition. In addition, mice lacking α7nAChR had exaggerated responses to reverse peritoneal Arthus reaction characterized by increased infiltration of PMNs and elevated of levels of TNF-α and CXCL1 in peritoneal fluid compared with wild-type mice. Taken together, these findings suggest that cholinergic output has the potential to exert tonic inhibitory activity that dampens responses to ICs and C5a and thus may be a target to minimize tissue damage in autoimmune diseases.


Asunto(s)
Enfermedades del Complejo Inmune/inmunología , Neutrófilos/inmunología , Fagocitos/inmunología , Receptor Nicotínico de Acetilcolina alfa 7/inmunología , Acetilcolina/farmacología , Animales , Líquido Ascítico/química , Líquido Ascítico/inmunología , Compuestos de Bencilideno/farmacología , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL1/biosíntesis , Quimiocina CXCL1/genética , Quimiotaxis de Leucocito/efectos de los fármacos , Complemento C5a/genética , Complemento C5a/farmacología , Citocinas/metabolismo , Femenino , Humanos , Quinasa I-kappa B/metabolismo , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Fagocitos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Piridinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptores de IgG/antagonistas & inhibidores , Proteínas Recombinantes/farmacología , Estallido Respiratorio/efectos de los fármacos , Estallido Respiratorio/fisiología , Organismos Libres de Patógenos Específicos , Receptor Nicotínico de Acetilcolina alfa 7/agonistas
7.
Arthritis Rheum ; 63(9): 2764-73, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21560114

RESUMEN

OBJECTIVE: To construct a humanized mouse model of systemic lupus erythematosus (SLE) that resembles the human disease in order to define the pathophysiology and targets for treatments. METHODS: We infused peripheral blood mononuclear cells (PBMCs) from SLE patients into BALB- RAG-2-/- IL-2Rγ-/- double-knockout (DKO) mice, which lack T cells, B cells, and natural killer cells. PBMCs from 5 SLE patients and 4 normal donors were infused intravenously/intraperitoneally at a density of 3-5×10(6) cells per animal into nonirradiated 4-5-week-old mice. We evaluated the engraftment of human CD45+ cells and monitored the plasma levels of human IgG, anti-double-stranded DNA (anti-dsDNA) antibody, and anticardiolipin antibody (aCL), as well as proteinuria and kidney histology. RESULTS: There was 100% successful engraftment in 40 DKO mice infused with human PBMCs. In the PBMC fraction from SLE PBMC-infused DKO (SLE-DKO) mice and normal donor PBMC-infused DKO (ND-DKO) mice, an average of 41% and 53% human CD45+ cells, respectively, were observed at 4 weeks postengraftment, with 70-90% CD3+ cells. There were fewer CD3+CD4+ cells (mean±SEM 5.5±2.1%) and more CD3+CD8+ cells (79.4±3.6%) in the SLE-DKO mice as in the SLE patients from which the PBMCs were derived. CD19+ B cells and CD11c+ monocytic cells were found in the spleen, lung, liver, and bone marrow. There was no significant difference in plasma levels of human IgG and anti-dsDNA antibodies between SLE-DKO and ND-DKO mice. Levels of aCL were significantly higher in all SLE-DKO mice infused with PBMCs from an SLE patient who had high titers of aCL. SLE-DKO mice had proteinuria, human IgG deposits in the kidneys, and a shorter life span. In SLE-DKO mice engrafted with PBMCs from the aCL-positive patient, we found microthrombi and infiltration of CD3+, CD8+, and CD19+ cells in the glomeruli, recapitulating the human antiphospholipid syndrome in these mice. CONCLUSION: We established a novel humanized SLE-DKO mouse exhibiting many of the immunologic and clinical features of human SLE.


Asunto(s)
Síndrome Antifosfolípido/inmunología , Leucocitos Mononucleares/trasplante , Lupus Eritematoso Sistémico/inmunología , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Leucocitos Mononucleares/inmunología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad
8.
Arthritis Rheum ; 63(1): 212-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20954190

RESUMEN

OBJECTIVE: Systemic lupus erythematosus (SLE) is a systemic inflammatory disease characterized by autoantibody production and immune complex deposition. The level of interleukin-10 (IL-10), predominantly an antiinflammatory cytokine, is paradoxically elevated in patients with SLE. The aim of this study was to examine the hypothesis that the antiinflammatory function of IL-10 is impaired in monocytes from patients with SLE with long-term exposure to immune complexes. METHODS: CD14+ monocytes were isolated from healthy donors and patients with SLE. Cultured CD14+ cells were treated with heat-aggregated human IgG (325 µg/ml) in the presence or absence of IL-10 (20 ng/ml). To study gene expression, RNA was extracted 3 hours after treatment. To study cytokine production, supernatants were harvested after 8 hours. To study IL-10 signaling, cell lysates were obtained from CD14+ cells treated with human IgG (325 µg/ml) for 1 hour followed by IL-10 (20 ng/ml) treatment for 10 minutes. Western blot analysis was used to assess STAT-3 phosphorylation. All experiments were performed in pairs. RESULTS: When stimulated with human IgG, SLE monocytes produced more tumor necrosis factor α (TNFα) and IL-6 than did control cells. The suppressive effect of IL-10 on human IgG-induced TNFα and IL-6 production was lower in SLE monocytes compared with control monocytes, although IL-10 receptor expression was similar in SLE and control monocytes. Human IgG suppressed IL-10 receptor expression and altered IL-10 signaling in control monocytes. Like SLE monocytes, interferon-α (IFNα)-primed control monocytes stimulated with human IgG were also less responsive to IL-10. CONCLUSION: Human IgG and IFNα modulate IL-10 function. In SLE monocytes, which are considered to be IFNα primed and persistently exposed to immune complexes, responses to IL-10 are abnormal, limiting the antiinflammatory effect of this cytokine.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Interleucina-10/farmacología , Lupus Eritematoso Sistémico/metabolismo , Monocitos/metabolismo , Complejo Antígeno-Anticuerpo/inmunología , Western Blotting , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Expresión Génica , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Inmunoglobulina G/farmacología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Lupus Eritematoso Sistémico/inmunología , Masculino , Monocitos/efectos de los fármacos , Monocitos/inmunología , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Kidney Int ; 79(3): 331-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20944547

RESUMEN

Preeclampsia is a major cause of maternal and neonatal morbidity and mortality. In mouse models, complement activation in the placenta is associated with abnormal placental development and miscarriage, and inhibiting complement prevents fetal injury. We mated two mouse strains, DBA/2 and CBA/J, expecting that the pregnancies might show features of preeclampsia and of immunologically mediated pregnancy loss. Along with placental dysfunction, these matings resulted in proteinuria, elevated BUN, fibrin deposition, and glomerular endotheliosis. We blocked placental complement activation throughout pregnancy by administering a single dose of the C3 inhibitor CR2-Crry given on day 5 of the pregnancy. This procedure specifically targets the sites of complement activation without inducing any systemic effects. Placental complement inhibition prevented oxidative stress and placental dysfunction, as well as proteinuria and renal pathologic features of preeclampsia. Thus, local blockade of complement activation at the maternal-fetal interface rescues preeclampsia in mice, and identifies new treatments. Hence, complement triggers a feed-forward cycle of placental damage, antiangiogenic factor production, and maternal vascular damage in patients.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Riñón/efectos de los fármacos , Placenta/efectos de los fármacos , Preeclampsia/tratamiento farmacológico , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Nitrógeno de la Urea Sanguínea , Modelos Animales de Enfermedad , Femenino , Fibrina/metabolismo , Inyecciones Intravenosas , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Neovascularización Fisiológica/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Placenta/inmunología , Placenta/metabolismo , Placenta/fisiopatología , Preeclampsia/inmunología , Preeclampsia/metabolismo , Preeclampsia/fisiopatología , Embarazo , Proteinuria/inmunología , Proteinuria/prevención & control , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
10.
Blood ; 114(8): 1675-83, 2009 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-19535796

RESUMEN

Using different mouse monoclonal and human antiphospholipid (aPL) antibodies, we developed a new animal model of renal injury that shares many features with thrombotic microangiopathy (TMA). We found that more than 1 mechanism/signaling pathway is involved in glomerular injury induced by aPL antibodies in this model. Both complement-dependent and complement-independent pathways were identified that lead to glomerular endothelial cell damage and renal function impairment. We also found that C5a-C5aR interaction is a crucial step for the activation of the coagulation cascade and glomerular injury induced by complement-activating antibodies. In addition, our studies demonstrated complement-independent mechanisms in which reactivity with beta(2) glycoprotein I (beta2GPI) plays an important role in aPL-induced glomerular damage and renal failure. Independently of the mechanism responsible for aPL-induced TMA, mice that express low levels of tissue factor (TF) were protected from glomerular injury. That genetic reduction of TF prevents renal injury induced by different aPL antibodies indicates that TF is a common mediator of glomerular damage and a possible target for selective pharmacologic intervention. Treatment with pravastatin, which down-regulates glomerular TF synthesis, prevents aPL-induced TMA in this mouse model, thus emphasizing that targeting TF might be a good therapeutic intervention in patients with TMA.


Asunto(s)
Anticuerpos Antifosfolípidos/efectos adversos , Tromboplastina/fisiología , Trombosis/etiología , Adulto , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Reacciones Antígeno-Anticuerpo/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Glomérulos Renales/inmunología , Glomérulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/patología , Persona de Mediana Edad , Insuficiencia Renal/etiología , Insuficiencia Renal/genética , Insuficiencia Renal/inmunología , Insuficiencia Renal/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo , Trombosis/genética , Trombosis/metabolismo , Trombosis/patología
11.
Blood ; 113(17): 4101-9, 2009 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-19234141

RESUMEN

Pregnancy loss and intrauterine growth restriction (IUGR) are serious pregnancy complications, and the triggers and mediators of placental and fetal damage are not completely understood. Using a mouse model of recurrent spontaneous miscarriages (DBA/2-mated CBA/J mice) that shares features with human recurrent miscarriage and fetal growth restriction, we identified tissue factor (TF) as an essential participating factor in placental and fetal injury. We have previously shown that C5a releases antiangiogenic molecule sFlt-1 in monocytes that causes defective placental development and fetal death in DBA/2-mated CBA/J mice. In this study, we found that TF not only activates the coagulation pathway, but it also mediates sFlt-1 release in monocytes causing defective placental development and fetal death. Blockade of TF with a monoclonal antibody inhibited sFlt-1 release, prevented the pathological activation of the coagulation pathway, restored placental blood flow, prevented placental oxidative stress, and rescued pregnancies. We also demonstrated that pravastatin, by down-regulating TF expression on monocytes and trophoblasts, prevented placental damage and protected pregnancies in DBA/2-mated CBA/J mice. These studies indicate that TF is an important mediator in fetal death and growth restriction and that statins may be a good treatment for women with recurrent miscarriages and IUGR.


Asunto(s)
Aborto Espontáneo/metabolismo , Aborto Espontáneo/prevención & control , Placenta/metabolismo , Pravastatina/farmacología , Tromboplastina/metabolismo , Animales , Animales Recién Nacidos/lesiones , Animales Recién Nacidos/metabolismo , Anticoagulantes/farmacología , Antitrombina III/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Masculino , Ratones , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Óxidos de Nitrógeno/metabolismo , Estrés Oxidativo , Placenta/irrigación sanguínea , Embarazo , Unión Proteica , Trombina/metabolismo , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
J Clin Invest ; 118(10): 3453-61, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18802482

RESUMEN

Women with antiphospholipid syndrome (APS), a condition characterized by the presence of antiphospholipid antibodies (aPL), often suffer pregnancy-related complications, including miscarriage. We have previously shown that C5a induction of tissue factor (TF) expression in neutrophils contributes to respiratory burst, trophoblast injury, and pregnancy loss in mice treated with aPL. Here we analyzed how TF contributes to neutrophil activation and trophoblast injury in this model. Neutrophils from aPL-treated mice expressed protease-activated receptor 2 (PAR2), and stimulation of this receptor led to neutrophil activation, trophoblast injury, and fetal death. An antibody specific for human TF that has little impact on coagulation, but potently inhibits TF/Factor VIIa (FVIIa) signaling through PAR2, inhibited aPL-induced neutrophil activation in mice that expressed human TF. Genetic deletion of the TF cytoplasmic domain, which allows interaction between TF and PAR2, reduced aPL-induced neutrophil activation in aPL-treated mice. Par2-/- mice treated with aPL exhibited reduced neutrophil activation and normal pregnancies, which indicates that PAR2 plays an important role in the pathogenesis of aPL-induced fetal injury. We also demonstrated that simvastatin and pravastatin decreased TF and PAR2 expression on neutrophils and prevented pregnancy loss. Our results suggest that TF/FVIIa/PAR2 signaling mediates neutrophil activation and fetal death in APS and that statins may be a good treatment for women with aPL-induced pregnancy complications.


Asunto(s)
Síndrome Antifosfolípido/complicaciones , Síndrome Antifosfolípido/fisiopatología , Modelos Animales de Enfermedad , Factor VIIa/metabolismo , Muerte Fetal/etiología , Activación Neutrófila , Receptor PAR-2/metabolismo , Tromboplastina/metabolismo , Animales , Anticuerpos Antifosfolípidos/inmunología , Anticuerpos Antifosfolípidos/farmacología , Síndrome Antifosfolípido/inmunología , Femenino , Muerte Fetal/inmunología , Muerte Fetal/prevención & control , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoglobulina G/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fagocitosis/efectos de los fármacos , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Receptor PAR-1/deficiencia , Receptor PAR-2/genética , Estallido Respiratorio/efectos de los fármacos , Transducción de Señal , Simvastatina/farmacología , Tromboplastina/genética , Tromboplastina/inmunología
13.
Arthritis Rheum ; 58(2): 384-8, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18240208

RESUMEN

OBJECTIVE: Human neutrophils express both activating and inhibitory Fcgamma receptors (FcgammaR), and their relative expression determines the inflammatory response to immune complexes. Tumor necrosis factor alpha (TNFalpha) up-regulates the expression of stimulatory FcgammaRIIa on neutrophils in vitro, and amplifies immune complex-induced activation of neutrophils in vivo. This study was undertaken to determine whether TNFalpha blockade in patients with rheumatoid arthritis (RA) alters the balance of activating FcgammaR and inhibitory FcgammaR and thereby decreases inflammation. METHODS: We used fluorescence-activated cell sorting and Western blotting to examine FcgammaR expression on neutrophils in 24 patients with RA, preceding their first infusion of infliximab and immediately prior to >or=3 subsequent infusions. RESULTS: In 13 of 24 patients (54.2%), there was a decrease in the expression of the predominant activating FcgammaR, FcgammaRIIa, after treatment with infliximab, an effect that persisted over >or=3 months of treatment. Although prior to initiation of infliximab therapy the inhibitory FcgammaR, FcgammaRIIb, was undetectable in neutrophils from 23 of 24 patients with RA, FcgammaRIIb protein was detected by Western blotting in 9 patients (37.5%) at the time of the third infliximab infusion. The induction of inhibitory FcgammaRIIb was always associated with decreased levels of FcgammaRIIa, and improvement following infliximab therapy, measured using the Health Assessment Questionnaire, was significantly associated with down-regulation of FcgammaRIIa. CONCLUSION: Our findings indicate that TNFalpha inhibition may reduce inflammation in patients with RA by restoring the balance of activating and inhibitory FcgammaR and thereby raising the threshold for immune complex-mediated activation of neutrophils.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antirreumáticos/administración & dosificación , Artritis Reumatoide/tratamiento farmacológico , Neutrófilos/efectos de los fármacos , Receptores de IgG/metabolismo , Adulto , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Antígenos CD/inmunología , Antígenos CD/metabolismo , Artritis Reumatoide/inmunología , Western Blotting , Femenino , Citometría de Flujo , Humanos , Infliximab , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de IgG/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
14.
Clin Immunol ; 127(1): 56-65, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18201931

RESUMEN

PURPOSE: In systemic lupus erythematosus (SLE), a disease of immune complex (IC) deposition, interleukin-10 (IL-10) is thought to promote B-lymphocyte hyperactivity and autoantibody production. Both ICs and Toll-like receptor (TLR) ligands have been shown to stimulate the production of IL-10 by human monocytes. Using an in vitro model, we studied how IC solubility, complement activation products, and TLR ligands could affect IL-10 production by human monocytes stimulated with ICs. METHODS: Human monocytes were stimulated with soluble or insoluble heat-aggregated human IgG with or without TLR ligands or C5a. Cytokine levels in cell culture supernatants were measured by ELISA. To study cytokine signaling, cell lysates were analyzed by Western blot for total or tyrosine-phosphorylated STAT3. RESULTS: Insoluble ICs were most effective at stimulating production of IL-10, and costimulation LPS enhanced synthesis of IL-10. In addition, stimulation with insoluble ICs together with C5a enhanced the production of IL-10 by 2-4 fold in either the presence or absence of TLR ligands. Increased STAT3 phosphorylation correlated temporally with enhanced IL-10 production and was reduced by an IL-10 receptor blocking antibody, suggesting that IL-10 was responsible for observed STAT3 phosphorylation. CONCLUSIONS: Because the immune deposits of SLE are, by definition, insoluble; and because IL-10 is thought to be important for B-cell hyperactivity and autoantibody production, these observations provide a critical link, bridging current views of B-cell hyperactivity with the early concept that SLE may arise from defective clearance of immune complexes.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Complemento C5a/metabolismo , Interleucina-10/biosíntesis , Monocitos/metabolismo , Receptores Toll-Like/metabolismo , Complejo Antígeno-Anticuerpo/inmunología , Western Blotting , Complemento C5a/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Ligandos , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Solubilidad , Receptores Toll-Like/inmunología
15.
Blood ; 110(7): 2423-31, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17536017

RESUMEN

Fetal loss in patients with antiphospholipid (aPL) antibodies has been ascribed to thrombosis of placental vessels. However, we have shown that inflammation, specifically activation of complement with generation of the anaphylotoxin C5a, is an essential trigger of fetal injury. In this study, we analyzed the role of the procoagulant molecule tissue factor (TF) in a mouse model of aPL antibody-induced pregnancy loss. We found that either blockade of TF with a monoclonal antibody in wild-type mice or a genetic reduction of TF prevented aPL antibody-induced inflammation and pregnancy loss. In response to aPL antibody-generated C5a, neutrophils express TF potentiating inflammation in the deciduas and leading to miscarriages. Importantly, we showed that TF in myeloid cells but not fetal-derived cells (trophoblasts) was associated with fetal injury, suggesting that the site for pathologic TF expression is neutrophils. We found that TF expression in neutrophils contributes to respiratory burst and subsequent trophoblast injury and pregnancy loss induced by aPL antibodies. The identification of TF as an important mediator of C5a-induced oxidative burst in neutrophils in aPL-induced fetal injury provides a new target for therapy to prevent pregnancy loss in the antiphospholipid syndrome.


Asunto(s)
Anticuerpos Antifosfolípidos/inmunología , Complemento C5a/metabolismo , Activación Neutrófila/inmunología , Lesiones Prenatales/inmunología , Lesiones Prenatales/metabolismo , Tromboplastina/metabolismo , Animales , Pérdida del Embrión/inmunología , Pérdida del Embrión/metabolismo , Pérdida del Embrión/patología , Femenino , Humanos , Ratones , Neutrófilos/inmunología , Neutrófilos/metabolismo , Estrés Oxidativo , Embarazo , Lesiones Prenatales/inducido químicamente , Lesiones Prenatales/patología , Receptor de Anafilatoxina C5a/metabolismo , Transducción de Señal , Tromboplastina/deficiencia , Tromboplastina/genética , Tromboplastina/inmunología
16.
J Immunol ; 177(12): 8440-7, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17142741

RESUMEN

Receptors for IgG (FcgammaR) expressed in dendritic cells (DCs) influence the initiation of Ab-mediated immunity. Dynamic variations in FcgammaR expression allow DCs to adjust their capacity to capture Ab-opsonized Ag. The current paradigm predicts a progressive decline in FcgammaR-mediated phagocytic function upon DC maturation. Surprisingly, we find that expression of the phagocytic receptor FcgammaRIIa is preserved in immature and mature DCs at comparable levels with macrophages. Moreover, phagocytosis of antigenic peptides directed to FcgammaRIIa on DCs leads to dramatic increases in Ag cross-presentation and T cell activation. In immature DCs, high expression of inhibitory FcgammaRIIb correlates with decreased uptake and cross-presentation of Ab-Ag complexes. In contrast, engagement of FcgammaRIIb is not associated with changes in cross-presentation in mature DCs. We provide evidence that FcgammaRIIb expression is patently reduced in mature DCs, an effect that is modulated by treatment with cytokines. The regulated expression of activating and inhibitory FcgammaRs in DCs emerges as a critical checkpoint in the process of Ag uptake and cross-presentation.


Asunto(s)
Presentación de Antígeno/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Reacciones Cruzadas/inmunología , Células Dendríticas/inmunología , Receptores de IgG/fisiología , Antígenos CD/genética , Antígenos CD/fisiología , Regulación de la Expresión Génica/inmunología , Humanos , Activación de Linfocitos/inmunología , Fagocitosis/inmunología , Receptores de IgG/genética , Linfocitos T/inmunología
17.
Clin Immunol ; 117(1): 78-86, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16084773

RESUMEN

Activation of neutrophils by the interaction of immune complexes with Fc gamma receptors (FcgammaR) is amplified in tumor necrosis factor-alpha (TNFalpha)-primed cells, whereas interleukin-10 (IL-10) has been reported to suppress cytokine-mediated neutrophil activation. We examined whether the expression and function of FcgammaR in human neutrophils is modulated by TNFalpha and IL-10 in vitro, and whether FcgammaRIIa expression is altered following treatment with the TNFalpha inhibitor infliximab in rheumatoid arthritis (RA) patients in vivo. TNFalpha treatment induced upregulation of expression and function of the major activating Fc receptor, FcgammaRIIa, in neutrophils from healthy donors. Unexpectedly, treatment with IL-10 led to gain of FcgammaRIIa function in TNFalpha-primed neutrophils. In neutrophils from RA patients initiating infliximab therapy and followed longitudinally through consecutive treatments, FcgammaRIIa protein decreased during the course of TNFalpha blockade, indicating that FcgammaRIIa is a target of TNFalpha modulation in human neutrophils in vivo.


Asunto(s)
Antígenos CD/efectos de los fármacos , Antígenos CD/inmunología , Artritis Reumatoide/inmunología , Neutrófilos/inmunología , Receptores de IgG/efectos de los fármacos , Receptores de IgG/inmunología , Factor de Necrosis Tumoral alfa/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Western Blotting , Citometría de Flujo , Humanos , Inmunoprecipitación , Infliximab , Interleucina-10/inmunología , Interleucina-10/farmacología , Neutrófilos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/inmunología
18.
Hum Genet ; 117(2-3): 220-7, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15895258

RESUMEN

The role for inhibitory Fc gamma receptors class IIb (FcgammaRIIb) in the onset, progression and severity of several animal models of autoimmune diseases is well established. By contrast, the pathogenic potential of FcgammaRIIb in human autoimmune diseases remains largely unknown. Here we report the identification of a polymorphism in the human FCGR2B promoter (dbSNP no. rs3219018) that is associated in homozygosity with systemic lupus erythematosus (SLE) phenotype in European-Americans (OR=11.1, P=0.003). Experimental evidence correlates the polymorphism (a G-C substitution at position -343 relative to the start of transcription) with altered FcgammaRIIb expression and function. The G-C substitution correlated with decreased transcription of the FCGR2B promoter, and resulted in decreased binding of the AP1 transcription complex to the mutant promoter sequence. The surface expression of FcgammaRIIb receptors was significantly reduced in activated B cells from (-343 C/C) SLE patients. These findings suggest that genetic defects may lead to deregulated expression of the FCGR2B gene in -343 C/C homozygous subjects, and may play a role in the pathogenesis of human SLE.


Asunto(s)
Lupus Eritematoso Sistémico/genética , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Receptores de IgG/genética , Transcripción Genética/genética , Linfocitos B/metabolismo , Regulación hacia Abajo/genética , Ligamiento Genético/genética , Homocigoto , Humanos , Lupus Eritematoso Sistémico/metabolismo , Activación de Linfocitos/genética , Receptores de IgG/biosíntesis , Factor de Transcripción AP-1/metabolismo
19.
Nat Med ; 10(11): 1222-6, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15489858

RESUMEN

The antiphospholipid syndrome (APS) is defined by thrombosis and recurrent pregnancy loss in the presence of antiphospholipid (aPL) antibodies and is generally treated with anticoagulation therapy. Because complement activation is essential and causative in aPL antibody-induced fetal injury, we hypothesized that heparin protects pregnant APS patients from complications through inhibition of complement. Treatment with heparin (unfractionated or low molecular weight) prevented complement activation in vivo and in vitro and protected mice from pregnancy complications induced by aPL antibodies. Neither fondaparinux nor hirudin, other anticoagulants, inhibited the generation of complement split products or prevented pregnancy loss, demonstrating that anticoagulation therapy is insufficient protection against APS-associated miscarriage. Our data indicate that heparins prevent obstetrical complications in women with APS because they block activation of complement induced by aPL antibodies targeted to decidual tissues, rather than by their anticoagulant effects.


Asunto(s)
Anticuerpos Antifosfolípidos/inmunología , Síndrome Antifosfolípido/inmunología , Activación de Complemento/efectos de los fármacos , Muerte Fetal/inmunología , Heparina/farmacología , Animales , Síndrome Antifosfolípido/tratamiento farmacológico , Western Blotting , Cromatografía de Afinidad , Activación de Complemento/inmunología , Electroforesis en Gel de Poliacrilamida , Femenino , Heparina/uso terapéutico , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Embarazo
20.
J Clin Invest ; 112(11): 1644-54, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14660741

RESUMEN

Antiphospholipid syndrome (APS) is defined by recurrent pregnancy loss and thrombosis in the presence of antiphospholipid (aPL) Ab's. Currently, therapy for pregnant women with APS is focused on preventing thrombosis, but anticoagulation is only partially successful in averting miscarriage. We hypothesized that complement activation is a central mechanism of pregnancy loss in APS and tested this in a model in which pregnant mice receive human IgG containing aPL Ab's. Here we identify complement component C5 (and particularly its cleavage product C5a) and neutrophils as key mediators of fetal injury, and we show that Ab's or peptides that block C5a-C5a receptor interactions prevent pregnancy complications. The fact that F(ab)'2 fragments of aPL Ab's do not mediate fetal injury and that C4-deficient mice are protected from fetal injury suggests that activation of the complement cascade is initiated via the classical pathway. Studies in factor B-deficient mice, however, indicate that alternative pathway activation is required and amplifies complement activation. In contrast, activating Fc gamma Rs do not play an important role in mediating aPL Ab-induced fetal injury. Our findings identify the key innate immune effectors engaged by pathogenic autoantibodies that mediate poor pregnancy outcomes in APS and provide novel and important targets for prevention of pregnancy loss in APS.


Asunto(s)
Aborto Espontáneo/etiología , Síndrome Antifosfolípido/complicaciones , Neutrófilos/fisiología , Complicaciones del Embarazo/inmunología , Receptor de Anafilatoxina C5a/fisiología , Animales , Síndrome Antifosfolípido/inmunología , Complemento C4/fisiología , Complemento C5/fisiología , Vía Alternativa del Complemento , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...