Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Nutrients ; 14(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35057553

RESUMEN

As of late, evidence has been emerging that the Maillard reaction (MR, also referred to as glycation) affects the structure and function of food proteins. MR induces the conformational and chemical modification of food proteins, not only on the level of IgG/IgE recognition, but also by increasing the interaction and recognition of these modified proteins by antigen-presenting cells (APCs). This affects their biological properties, including digestibility, bioavailability, immunogenicity, and ultimately their allergenicity. APCs possess various receptors that recognize glycation structures, which include receptor for advanced glycation end products (RAGE), scavenger receptors (SRs), galectin-3 and CD36. Through these receptors, glycation structures may influence the recognition, uptake and antigen-processing of food allergens by dendritic cells (DCs) and monocytes. This may lead to enhanced cytokine production and maturation of DCs, and may also induce adaptive immune responses to the antigens/allergens as a result of antigen uptake, processing and presentation to T cells. Here, we aim to review the current literature on the immunogenicity of AGEs originating from food (exogenous or dietary AGEs) in relation to AGEs that are formed within the body (endogenous AGEs), their interactions with receptors present on immune cells, and their effects on the activation of the innate as well as the adaptive immune system. Finally, we review the clinical relevance of AGEs in food allergies.


Asunto(s)
Inmunidad Adaptativa , Hipersensibilidad a los Alimentos/inmunología , Productos Finales de Glicación Avanzada/inmunología , Inmunidad Innata , Alérgenos/inmunología , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/inmunología , Células Dendríticas/inmunología , Dieta/métodos , Alimentos , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Reacción de Maillard , Receptor para Productos Finales de Glicación Avanzada/inmunología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Linfocitos T/inmunología
2.
Biochem Biophys Res Commun ; 581: 38-45, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34653677

RESUMEN

A detrimental role of the receptor for the advanced glycation end product (RAGE) has been identified in the immune response, and various pathological conditions and its V and C1 domains in the extracellular region of RAGE are believed to be the main ligand-binding domains. Consequently, specific inhibitors targeting those domains could be of clinical value in fighting against the pathological condition associated with RAGE over-activation. Single-domain antibodies, also called nanobodies (Nbs), are antibody fragments engineered from the heavy-chain only antibodies found in camelids, which offer a range of advantages in therapy. In this study, we report the development and characterization of the V-C1 domain-specific Nbs. Three Nbs (3CNB, 4BNB, and 5ENB) targeting V-C1 domain of human RAGE were isolated from an immunized alpaca using a phage display. All of these Nbs revealed high thermostability. 3CNB, 4BNB, and 5ENB bind to V-C1 domain with a dissociation constant (KD) of 27.25, 39.37, and 47.85 nM, respectively, using Isothermal Titration Calorimetry (ITC). After homodimerization using human IgG1-Fc fusion, their binding affinity improved to 0.55, 0.62, and 0.41 nM, respectively, using Surface Plasmon Resonance (SPR). Flow cytometry showed all the Fc fusions Nbs can bind to human RAGE expressed on the cell surface. Competitive ELISA further confirmed their V-C1-hS100B blocking ability in solution, providing insights into the applicability of Nbs in treating RAGE-associated diseases.


Asunto(s)
Productos Finales de Glicación Avanzada/química , Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/química , Receptor para Productos Finales de Glicación Avanzada/química , Proteínas Recombinantes de Fusión/química , Anticuerpos de Dominio Único/biosíntesis , Secuencia de Aminoácidos , Animales , Sitios de Unión , Camélidos del Nuevo Mundo , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Productos Finales de Glicación Avanzada/genética , Productos Finales de Glicación Avanzada/inmunología , Células HEK293 , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Biblioteca de Péptidos , Unión Proteica , Dominios Proteicos , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/aislamiento & purificación
3.
Biomolecules ; 11(8)2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34439812

RESUMEN

Intra-amniotic infections (IAI) are one of the reasons for preterm birth. High mobility group box 1 (HMGB1) is a nuclear protein with various physiological functions, including tissue healing. Its excessive extracellular release potentiates inflammatory reaction and can revert its action from beneficial to detrimental. We infected the amniotic fluid of a pig on the 80th day of gestation with 1 × 104 colony forming units (CFUs) of E. coli O55 for 10 h, and evaluated the appearance of HMGB1, receptor for glycation endproducts (RAGE), and Toll-like receptor (TLR) 4 in the amniotic membrane and fluid. Sham-infected amniotic fluid served as a control. The expression and release of HMGB1 were evaluated by Real-Time PCR, immunofluorescence, immunohistochemistry, and ELISA. The infection downregulated HMGB1 mRNA expression in the amniotic membrane, changed the distribution of HMGB1 protein in the amniotic membrane, and increased its level in amniotic fluid. All RAGE mRNA, protein expression in the amniotic membrane, and soluble RAGE level in the amniotic fluid were downregulated. TLR4 mRNA and protein expression and soluble TLR4 were all upregulated. HMGB1 is a potential target for therapy to suppress the exaggerated inflammatory response. This controlled expression and release can, in some cases, prevent the preterm birth of vulnerable infants. Studies on suitable animal models can contribute to the development of appropriate therapy.


Asunto(s)
Infecciones por Escherichia coli/veterinaria , Escherichia coli/patogenicidad , Proteína HMGB1/genética , Complicaciones Infecciosas del Embarazo/veterinaria , ARN Mensajero/genética , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor Toll-Like 4/genética , Amnios/inmunología , Amnios/microbiología , Amnios/patología , Líquido Amniótico/inmunología , Líquido Amniótico/microbiología , Animales , Modelos Animales de Enfermedad , Escherichia coli/crecimiento & desarrollo , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Femenino , Regulación de la Expresión Génica , Proteína HMGB1/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Embarazo , Complicaciones Infecciosas del Embarazo/genética , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/microbiología , Nacimiento Prematuro/prevención & control , ARN Mensajero/inmunología , Receptor para Productos Finales de Glicación Avanzada/inmunología , Transducción de Señal , Porcinos , Receptor Toll-Like 4/inmunología
4.
Front Immunol ; 12: 697588, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34305934

RESUMEN

The Toll-interleukin-1 Receptor (TIR) domain-containing adaptor protein (TIRAP) represents a key intracellular signalling molecule regulating diverse immune responses. Its capacity to function as an adaptor molecule has been widely investigated in relation to Toll-like Receptor (TLR)-mediated innate immune signalling. Since the discovery of TIRAP in 2001, initial studies were mainly focused on its role as an adaptor protein that couples Myeloid differentiation factor 88 (MyD88) with TLRs, to activate MyD88-dependent TLRs signalling. Subsequent studies delineated TIRAP's role as a transducer of signalling events through its interaction with non-TLR signalling mediators. Indeed, the ability of TIRAP to interact with an array of intracellular signalling mediators suggests its central role in various immune responses. Therefore, continued studies that elucidate the molecular basis of various TIRAP-protein interactions and how they affect the signalling magnitude, should provide key information on the inflammatory disease mechanisms. This review summarizes the TIRAP recruitment to activated receptors and discusses the mechanism of interactions in relation to the signalling that precede acute and chronic inflammatory diseases. Furthermore, we highlighted the significance of TIRAP-TIR domain containing binding sites for several intracellular inflammatory signalling molecules. Collectively, we discuss the importance of the TIR domain in TIRAP as a key interface involved in protein interactions which could hence serve as a therapeutic target to dampen the extent of acute and chronic inflammatory conditions.


Asunto(s)
Inflamación/inmunología , Glicoproteínas de Membrana/inmunología , Receptores de Interleucina-1/inmunología , Agammaglobulinemia Tirosina Quinasa/inmunología , Agammaglobulinemia Tirosina Quinasa/metabolismo , Animales , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia/inmunología , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Humanos , Inmunidad Innata , Inflamación/metabolismo , Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , Mapas de Interacción de Proteínas , Proteína Quinasa C-delta/inmunología , Proteína Quinasa C-delta/metabolismo , Receptor para Productos Finales de Glicación Avanzada/inmunología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptores de Interleucina-1/metabolismo , Transducción de Señal/inmunología
5.
Diabetes ; 70(9): 2147-2158, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34155040

RESUMEN

Effective treatment of diabetic kidney disease (DKD) remains a large unmet medical need. Within the disease's complicated pathogenic mechanism, activation of the advanced glycation end products (AGEs)-receptor for AGE (RAGE) axis plays a pivotal role in the development and progression of DKD. To provide a new therapeutic strategy against DKD progression, we developed a vaccine against RAGE. Three rounds of immunization of mice with the RAGE vaccine successfully induced antigen-specific serum IgG antibody titers and elevated antibody titers were sustained for at least 38 weeks. In addition, RAGE vaccination significantly attenuated the increase in urinary albumin excretion in streptozotocin-induced diabetic mice (type 1 diabetes model) and leptin-receptor-deficient db/db mice (type 2 diabetes model). In microscopic analyses, RAGE vaccination suppressed glomerular hypertrophy and mesangial expansion in both diabetic models and significantly reduced glomerular basement membrane thickness in streptozotocin-induced diabetic mice. Results of an in vitro study indicated that the serum IgG antibody elicited by RAGE vaccination suppressed the expression of AGE-induced vascular cell adhesion molecule 1 and intracellular adhesion molecule 1 in endothelial cells. Thus, our newly developed RAGE vaccine attenuated the progression of DKD in mice and is a promising potential therapeutic strategy for patients with DKD.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/prevención & control , Receptor para Productos Finales de Glicación Avanzada/inmunología , Vacunación , Animales , Nefropatías Diabéticas/metabolismo , Progresión de la Enfermedad , Masculino , Ratones , Receptor para Productos Finales de Glicación Avanzada/metabolismo
6.
Biomolecules ; 11(4)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33915939

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) remains a very difficult cancer to treat. Recent in vitro and in vivo studies suggest that the activation of the receptor for advanced glycation end products (RAGE) by its ligands stimulates pancreatic cancer cell proliferation and tumor growth. Additional studies show that, in the RAGE ligand, the high mobility group box 1 (HMGB1) protein plays an important role in chemoresistance against the cytotoxic agent gemcitabine by promoting cell survival through increased autophagy. We hypothesized that blocking the RAGE/HMGB1 interaction would enhance the cytotoxic effect of gemcitabine by reducing cell survival and autophagy. Using a preclinical mouse model of PDAC and a monoclonal antibody (IgG 2A11) as a RAGE inhibitor, we demonstrate that RAGE inhibition concurrent with gemcitabine treatment enhanced the cytotoxic effect of gemcitabine. The combination of IgG 2A11 and gemcitabine resulted in decreased autophagy compared to treatment with gemcitabine combined with control antibodies. Notably, we also observed that RAGE inhibition protected against excessive weight loss during treatment with gemcitabine. Our data suggest that the combination of gemcitabine with a RAGE inhibitor could be a promising therapeutic approach for the treatment of pancreatic cancer and needs to be further investigated.


Asunto(s)
Autofagia/efectos de los fármacos , Desoxicitidina/análogos & derivados , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Proteína HMGB1/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/inmunología , Trasplante Homólogo , Gemcitabina
7.
Vet Immunol Immunopathol ; 234: 110216, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33636544

RESUMEN

The pathogenesis of chronic inflammatory enteropathies (CIE) in dogs involves dysregulated innate immune responses. The receptor for advanced glycation end products (RAGE), a pattern recognition receptor, plays a role in chronic inflammation. Abrogation of proinflammatory RAGE signaling by ligand binding (e.g., S100/calgranulins) to soluble RAGE (sRAGE) might also be a novel therapeutic avenue. Serum sRAGE levels are decreased in canine CIE, but gastrointestinal tissue RAGE expression has not been investigated in dogs. Thus, the study aimed to evaluate the gastrointestinal mucosal RAGE expression in dogs with CIE. Further, the potential binding of RAGE to canine S100/calgranulin ligands was investigated. Epithelial RAGE expression was quantified in gastrointestinal (gastric, duodenal, ileal, and colonic) biopsies from 12 dogs with CIE and 9 healthy control dogs using confocal laser scanning microscopy. RAGE expression was compared between both groups of dogs and was tested for an association with patient characteristics, clinical variables, histologic lesion severity, and biomarkers of extra-gastrointestinal disease, systemic or gastrointestinal inflammation, function, or protein loss. Statistical significance was set at p < 0.05. RAGE:S100/calgranulin binding was assessed by immunoassay and electrophoretic techniques. RAGE expression was detected in all 59 biopsies from diseased and healthy control dogs evaluated. Epithelial RAGE expression in the duodenum and colon was significantly higher in dogs with CIE than in healthy controls (p < 0.04). Compared to healthy controls, RAGE expression in dogs with CIE also tended to be higher in the ileum but lower in the stomach. A slight (statistically not significant) shift towards more basal intestinal epithelial RAGE expression was detected in CIE dogs. Serum sRAGE was proportional to epithelial RAGE expression in the duodenum (p < 0.04), and RAGE expression in the colon inversely correlated with biomarkers of protein loss in serum (both p < 0.04). Several histologic morphologic and inflammatory lesion criteria and markers of inflammation (serum C-reactive protein and fecal calprotectin concentration) were related to epithelial RAGE expression in the duodenum, ileum, and/or colon. in vitro canine RAGE:S100A12 binding appeared more pronounced than RAGE:S100A8/A9 binding. This study showed a dysregulation of epithelial RAGE expression along the gastrointestinal tract in dogs with CIE. Compensatory regulations in the sRAGE/RAGE axis are an alternative explanation for these findings. The results suggest that RAGE signaling plays a role in dogs with CIE, but higher anti-inflammatory decoy receptor sRAGE levels paralleled RAGE overexpression. Canine S100/calgranulins were demonstrated to be ligands for RAGE.


Asunto(s)
Biopsia/veterinaria , Enfermedades de los Perros/genética , Expresión Génica , Inflamación/veterinaria , Enfermedades Intestinales/genética , Enfermedades Intestinales/inmunología , Receptor para Productos Finales de Glicación Avanzada/genética , Animales , Enfermedades de los Perros/inmunología , Perros , Femenino , Tracto Gastrointestinal/patología , Humanos , Inflamación/genética , Masculino , Receptor para Productos Finales de Glicación Avanzada/inmunología
8.
Am J Physiol Endocrinol Metab ; 320(3): E539-E550, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33459180

RESUMEN

Association between receptor for advanced glycation end products (RAGE) and postmyocardial infarction (MI) ventricular arrhythmias (VAs) in diabetes was investigated. Correlation between premature ventricular contractions (PVCs) and serum advanced glycation end products (AGEs) content was analyzed in a cohort consisting of 101 patients with ST-segment elevated MI (STEMI). MI diabetic rats were treated with anti-receptor for AGE (RAGE) antibody. Electrocardiography was used to record VAs. Myocytes were isolated from adjacent area around infracted region. Immunofluorescent stains were used to evaluate the association between FKBP12.6 (FK506-bindingprotein 12.6) and ryanodine receptor 2 (RyR2). Calcium sparks were evaluated by confocal microscope. Protein expression and phosphorylation were assessed by Western blotting. Calcineurin (CaN) enzymatic activity and RyR2 channel activity were also determined. In the cohort study, significantly increased amount of PVC was found in STEMI patients with diabetes (P < 0.05). Serum AGE concentration was significantly positively correlated with PVC amount in patients with STEMI (r = 0.416, P < 0.001). Multivariate analysis showed that serum AGE concentration was independently and positively related to frequent PVCs (adjusted hazard ratio, 1.86; 95% CI, 1.09-3.18, P = 0.022). In the animal study, increased glucose-regulated protein 78 (GRP78) expression, protein kinase RNA-like ER kinase (PERK) phosphorylation, CaN enzymatic activity, FKBP12.6-RyR2 disassociation, RyR2 channel opening, and endoplasmic reticulum (ER) calcium releasing were found in diabetic MI animals, which were attenuated by anti-RAGE antibody treatment. This RAGE blocking also significantly lowered the VA amount in diabetic MI animals. Activation of RAGE-dependent ER stress-mediated PERK/CaN/RyR2 signaling participated in post-MI VAs in diabetes.NEW & NOTEWORTHY In this study, we proposed a possible mechanism interpreting the clinical scenario that after myocardial infarction (MI) patients were more vulnerable to ventricular arrhythmias (VAs) when complicated with diabetes. A cohort study revealed that advanced glycation end products (AGEs) accumulated in patients with diabetes and closely associated post-MI VAs. In vivo and in vitro studies indicated that receptor for AGEs (RAGE)-dependent endoplasmic reticulum (ER) stress protein kinase RNA-like ER kinase (PERK) pathway triggered VAs, via ER calcium releasing, through calcineurin/RyR2 mechanism.


Asunto(s)
Arritmias Cardíacas/patología , Diabetes Mellitus , Estrés del Retículo Endoplásmico/fisiología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Infarto del Miocardio con Elevación del ST , Animales , Anticuerpos/farmacología , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/terapia , Estudios de Casos y Controles , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Angiopatías Diabéticas/metabolismo , Angiopatías Diabéticas/patología , Angiopatías Diabéticas/terapia , Progresión de la Enfermedad , Chaperón BiP del Retículo Endoplásmico , Femenino , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada/agonistas , Receptor para Productos Finales de Glicación Avanzada/inmunología , Infarto del Miocardio con Elevación del ST/complicaciones , Infarto del Miocardio con Elevación del ST/metabolismo , Infarto del Miocardio con Elevación del ST/patología
9.
J Am Heart Assoc ; 10(1): e016696, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33327730

RESUMEN

Background Expression of receptor for advanced glycation end products (RAGE) plays an important role in diabetic peripheral artery disease. We proposed to show that treatment with an antibody blocking RAGE would improve hind limb perfusion and muscle viability in diabetic pig with femoral artery (FA) ligation. Methods and Results Purpose-bred diabetic Yucatan minipigs with average fasting blood sugar of 357 mg/dL on insulin to maintain a glucose range of 300 to 500 mg/dL were treated with either a humanized monoclonal anti-RAGE antibody (CR-3) or nonimmune IgG. All pigs underwent intravascular occlusion of the anterior FA. Animals underwent (201Tl) single-photon emission computed tomography/x-ray computed tomography imaging on days 1 and 28 after FA occlusion, angiogenesis imaging with [99mTc]dodecane tetra-acetic acid-polyethylene glycol-single chain vascular endothelial growth factor (scVEGF), muscle biopsies on day 7, and contrast angiogram day 28. Results showed greater increases in perfusion to the gastrocnemius from day 1 to day 28 in CR-3 compared with IgG treated pigs (P=0.0024), greater uptake of [99mTc]dodecane tetra-acetic acid-polyethylene glycol-scVEGF (scV/Tc) in the proximal gastrocnemius at day 7, confirmed by tissue staining for capillaries and vascular endothelial growth factor A, and less muscle loss and fibrosis at day 28. Contrast angiograms showed better reconstitution of the distal FA from collaterals in the CR-3 versus IgG treated diabetic pigs (P=0.01). The gastrocnemius on nonoccluded limb at necropsy had higher 201Tl uptake (percentage injected dose per gram) and reduced RAGE staining in arterioles in CR-3 treated compared with IgG treated animals (P=0.04). Conclusions A novel RAGE-blocking antibody improved hind limb perfusion and angiogenesis in diabetic pigs with FA occlusion. Contributing factors are increased collaterals and reduced vascular RAGE expression. CR-3 shows promise for clinical treatment in diabetic peripheral artery disease.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Angiopatías Diabéticas , Enfermedad Arterial Periférica , Receptor para Productos Finales de Glicación Avanzada , Angiografía/métodos , Animales , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/metabolismo , Angiopatías Diabéticas/tratamiento farmacológico , Angiopatías Diabéticas/metabolismo , Descubrimiento de Drogas/métodos , Productos Finales de Glicación Avanzada/metabolismo , Miembro Posterior/irrigación sanguínea , Músculo Esquelético/irrigación sanguínea , Enfermedad Arterial Periférica/tratamiento farmacológico , Enfermedad Arterial Periférica/etiología , Enfermedad Arterial Periférica/metabolismo , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/inmunología , Porcinos , Porcinos Enanos , Resultado del Tratamiento
10.
FASEB J ; 34(12): 15659-15674, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33131091

RESUMEN

Although the innate immune receptor protein, Receptor for Advanced Glycation End products (RAGE), has been extensively studied, there has been renewed interest in RAGE for its potential role in sepsis, along with a host of other inflammatory diseases of chronic, noninfectious origin. In contrast to other innate immune receptors, for example, Toll-like receptors (TLRs), that recognize ligands derived from pathogenic organisms that are collectively known as "pathogen-associated molecular patterns" (PAMPs) or host-derived "damage-associated molecular patterns" (DAMPs), RAGE has been shown to recognize a broad collection of DAMPs exclusively. Historically, these DAMPs have been shown to be pro-inflammatory in nature. Early studies indicated that the adaptor molecule, MyD88, might be important for this change. More recent studies have explored further the mechanisms underlying this inflammatory change. Overall, the newer results have shown that there is extensive crosstalk between RAGE and TLRs. The three canonical RAGE ligands, Advanced Glycation End products (AGEs), HMGB1, and S100 proteins, have all been shown to activate both TLRs and RAGE to varying degrees in order to induce inflammation in in vitro models. As with any field that delves deeply into innate signaling, obstacles of reagent purity may be a cause of some of the discrepancies in the literature, and we have found that commercial antibodies that have been widely used exhibit a high degree of nonspecificity. Nonetheless, the weight of published evidence has led us to speculate that RAGE may be physically interacting with TLRs on the cell surface to elicit inflammation via MyD88-dependent signaling.


Asunto(s)
Inmunidad Innata/inmunología , Receptor para Productos Finales de Glicación Avanzada/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Animales , Humanos , Inflamación/inmunología
11.
Front Immunol ; 11: 581300, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117389

RESUMEN

Damage-associated molecular patterns (DAMPs) are released from tubular and interstitial cells in the kidney after unilateral ureteral obstruction (UUO). DAMPs are recognized by pattern recognition receptors (PRRs), which mediate the initiation of an immune response and the release of inflammatory cytokines. The animal model of UUO is used for various purposes. UUO in adult mice serves as a model for accelerated renal fibrosis, which is a hallmark of progressive renal disease. UUO in adult mice enables to study cell death, inflammation, and extracellular matrix deposition in the kidney. Neonatal UUO is a model for congenital obstructive nephropathies. It studies inflammation, apoptosis, and interstitial fibrosis in the neonatal kidney, when nephrogenesis is still ongoing. Following UUO, several DAMPs as well as DAMP receptors are upregulated. In adult UUO, soluble uric acid is upregulated and activates the NOD-like receptor family, pyrin domain containing-3 (NLRP3) inflammasome, which promotes fibrosis, apoptosis, and reactive oxygen species (ROS) injury. Further DAMPs associated with UUO are uromodulin, members of the IL-1 family, and necrotic cell DNA, all of which promote sterile inflammation. In neonatal UUO, the receptor for advanced glycation endproducts (RAGE) is highly upregulated. RAGE is a ligand for several DAMPs, including high mobility group box 1 (HMGB1) and S100 proteins, which play an important role in renal fibrosis. Additionally, necroptosis is an important mechanism of cell death, besides apoptosis, in neonatal UUO. It is highly inflammatory due to release of cytokines and specific DAMPs. The release and recognition of DAMPs initiate sterile inflammation, which makes them good candidates to develop and improve diagnostic and therapeutic strategies in renal fibrosis and congenital obstructive nephropathies.


Asunto(s)
Alarminas/inmunología , Obstrucción Ureteral/inmunología , Animales , Animales Recién Nacidos , Muerte Celular , Citocinas/inmunología , Modelos Animales de Enfermedad , Fibrosis , Humanos , Inmunidad Innata , Inflamasomas/inmunología , Riñón/inmunología , Riñón/patología , Ratones , Receptor para Productos Finales de Glicación Avanzada/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Obstrucción Ureteral/patología
12.
Exp Parasitol ; 216: 107946, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32622941

RESUMEN

This study was aimed at investigating the involvement of Receptor for Advanced Glycation End Products (RAGE) during malaria infection and the effects of modulating RAGE on the inflammatory cytokines release and histopathological conditions of affected organs in malarial animal model. Plasmodium berghei (P. berghei) ANKA-infected ICR mice were treated with mRAGE/pAb and rmRAGE/Fc Chimera drugs from day 1 to day 4 post infection. Survival and parasitaemia levels were monitored daily. On day 5 post infection, mice were sacrificed, blood were drawn for cytokines analysis and major organs including kidney, spleen, liver, brain and lungs were extracted for histopathological analysis. RAGE levels were increased systemically during malaria infection. Positive correlation between RAGE plasma concentration and parasitaemia development was observed. Treatment with RAGE related drugs did not improve survival of malaria-infected mice. However, significant reduction on the parasitaemia levels were recorded. On the other hand, inhibition and neutralization of RAGE production during the infection significantly increased the plasma levels of interleukin (IL-4, IL-17A, IL-10 and IL-2) and reduced interferon (IFN)-γ secretion. Histopathological analysis revealed that all treated malarial mice showed a better outcome in histological assessment of affected organs (brain, liver, spleen, lungs and kidney). RAGE is involved in malaria pathogenesis and targeting RAGE could be beneficial in malaria infected host in which RAGE inhibition or neutralization increased the release of anti-inflammatory cytokines (IL-10 and IL-4) and reduce pro-inflammatory cytokine (IFNγ) which may help alleviate tissue injury and improve histopathological conditions of affected organs during the infection.


Asunto(s)
Citocinas/metabolismo , Malaria/inmunología , Malaria/patología , Plasmodium berghei/inmunología , Receptor para Productos Finales de Glicación Avanzada/fisiología , Animales , Encéfalo/parasitología , Encéfalo/patología , Citocinas/sangre , Riñón/parasitología , Riñón/patología , Modelos Lineales , Hígado/parasitología , Hígado/patología , Pulmón/parasitología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos ICR , Parasitemia/inmunología , Distribución Aleatoria , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/sangre , Receptor para Productos Finales de Glicación Avanzada/inmunología , Bazo/parasitología , Bazo/patología
13.
PLoS Pathog ; 16(7): e1008651, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32658914

RESUMEN

Type-2 immunity elicits tissue repair and homeostasis, however dysregulated type-2 responses cause aberrant tissue remodelling, as observed in asthma. Severe respiratory viral infections in infancy predispose to later asthma, however, the processes that mediate tissue damage-induced type-2 inflammation and the origins of airway remodelling remain ill-defined. Here, using a preclinical mouse model of viral bronchiolitis, we find that increased epithelial and mesenchymal high-mobility group box 1 (HMGB1) expression is associated with increased numbers of IL-13-producing type-2 innate lymphoid cell (ILC2s) and the expansion of the airway smooth muscle (ASM) layer. Anti-HMGB1 ablated lung ILC2 numbers and ASM growth in vivo, and inhibited ILC2-mediated ASM cell proliferation in a co-culture model. Furthermore, we identified that HMGB1/RAGE (receptor for advanced glycation endproducts) signalling mediates an ILC2-intrinsic IL-13 auto-amplification loop. In summary, therapeutic targeting of the HMGB1/RAGE signalling axis may act as a novel asthma preventative by dampening ILC2-mediated type-2 inflammation and associated ASM remodelling.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Proteína HMGB1/inmunología , Inflamación/inmunología , Linfocitos/inmunología , Músculo Liso/inmunología , Animales , Ratones , Músculo Liso/patología , Receptor para Productos Finales de Glicación Avanzada/inmunología
14.
J Immunol ; 205(3): 767-775, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32580932

RESUMEN

Receptor for advanced glycation end-products (RAGE) and TLR4 play an important role in the inflammatory response against High-mobility group box 1 protein (HMGB1), a late proinflammatory cytokine and a damage-associated molecular pattern. As cell surface receptors, both RAGE and TLR4 are constantly trafficking between the cytoplasm and plasma membrane. However, whether TLR4 is related to the intracellular transport of RAGE in HMGB1-induced inflammation remains unknown. In this study, we demonstrated that HMGB1 not only increased RAGE expression in both the cytoplasm and plasma membrane but also upregulated the expression of TLR4 in the plasma membrane. Knocking out of RAGE led to decreased MAPK activation, TLR4 cellular membrane expression, and corresponding inflammatory cytokine generation. Meanwhile, inhibiting MAPK activation also decreased TLR4 surface expression. These results indicated that HMGB1 may bind to cell surface RAGE receptors on the cell surface, leading to MAPK activation, thus promoting TLR4 translocation on the cell surface, but does not regulate its transcription and translation. In contrast, TLR4 can increase the transcription and translation of RAGE, which translocates to the cell surface and is able to bind to more HMGB1. The cell surface receptors TLR4 and RAGE bind to HMGB1, leading to the transcription and secretion of inflammatory cytokines. Finally, we also observed these results in the mice pseudofracture model, which is closely related to HMGB1-induced inflammatory response. All these results demonstrated that the interplay between RAGE and TLR4 are critical for HMGB1-induced inflammatory response.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Proteína HMGB1/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Receptor para Productos Finales de Glicación Avanzada/inmunología , Receptor Toll-Like 4/inmunología , Animales , Membrana Celular/genética , Membrana Celular/inmunología , Citoplasma/genética , Citoplasma/inmunología , Proteína HMGB1/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Noqueados , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor Toll-Like 4/genética , Transcripción Genética/inmunología
15.
Nat Rev Immunol ; 20(2): 95-112, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31558839

RESUMEN

The innate immune system has the capacity to detect 'non-self' molecules derived from pathogens, known as pathogen-associated molecular patterns, via pattern recognition receptors. In addition, an increasing number of endogenous host-derived molecules, termed damage-associated molecular patterns (DAMPs), have been found to be sensed by various innate immune receptors. The recognition of DAMPs, which are produced or released by damaged and dying cells, promotes sterile inflammation, which is important for tissue repair and regeneration, but can also lead to the development of numerous inflammatory diseases, such as metabolic disorders, neurodegenerative diseases, autoimmune diseases and cancer. Here we examine recent discoveries concerning the roles of DAMP-sensing receptors in sterile inflammation and in diseases resulting from dysregulated sterile inflammation, and then discuss insights into the cross-regulation of these receptors and their ligands.


Asunto(s)
Alarminas/inmunología , Inmunidad Innata/inmunología , Inflamación/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Humanos , Lectinas Tipo C/inmunología , Proteínas NLR/inmunología , Receptor para Productos Finales de Glicación Avanzada/inmunología , Receptores Acoplados a Proteínas G/inmunología , Autotolerancia/inmunología , Receptores Toll-Like/inmunología , Receptor Activador Expresado en Células Mieloides 1/inmunología
16.
J Neurochem ; 151(5): 595-607, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31520526

RESUMEN

High-mobility group box-1 (HMGB-1) acts as a pro-inflammatory cytokine contributing to the occurrence of many central inflammatory and infectious disorders. Brain mast cells (MCs) are the first responders to peripheral inflammatory stimulation because of their rapid response to external stimuli coupled with their release of preformed and newly synthesized reactive chemicals. Little is known about the involvement of brain MCs in the pro-inflammatory effects of HMGB-1 on the central nervous system (CNS). Thus, we investigated the activation process of MCs by HMGB-1 and explored whether this process is involved in the pro-inflammatory effects of HMGB-1 on the CNS. In this study, we used P815 cells to study the activating role of HMGB-1 on MCs and to explore its potential mechanism in vitro. In an in vivo study, adult male Sprague-Dawley rats received i.c.v. injection of sterile saline or cromoglycate (stabilizer of MCs) 30 min prior to i.p. injection of HMGB-1. Increased levels of tumor necrosis factor and IL-1ß were observed in the P815 cells, as well as in the rats' brains, after HMGB-1 treatment. Pretreatment with the receptor of advanced glycation endproducts (RAGE)-siRNA inhibited the HMGB-1-induced inflammatory process in the P815 cells. Activation of the RAGE/nuclear factor-κB (NF-κB) pathway was observed in both the P815 cells and rats' brains. In addition, HMGB-1 induced the accumulation of brain MCs in the hippocampal CA1 region, and the blood-brain barrier was disrupted. Pretreatment with cromoglycate, a stabilizer of MCs, mitigated these HMGB-1-induced pro-inflammatory processes in rats. These findings indicate that brain MCs are involved in the pro-inflammatory effect of HMGB-1 on the CNS, probably via activating the RAGE/NF-κB pathway.


Asunto(s)
Encéfalo/inmunología , Proteína HMGB1/inmunología , Mastocitos/inmunología , Transducción de Señal/inmunología , Animales , Encéfalo/metabolismo , Proteína HMGB1/metabolismo , Masculino , Mastocitos/metabolismo , Ratones , FN-kappa B/inmunología , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada/inmunología , Receptor para Productos Finales de Glicación Avanzada/metabolismo
17.
Front Immunol ; 10: 1904, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31440260

RESUMEN

Inflammasome activation can trigger an inflammatory and innate immune response through the release of cytokines and induction of pyroptosis. A dysfunctional inflammasome has been implicated in the development of human pathologies, including sepsis and septic shock. Here, we show that advanced glycosylation end-product specific receptor (AGER/RAGE) is required for caspase-11 inflammasome activation in macrophages. A nuclear damage-associated molecular pattern (nDAMP) complex, including high-mobility group box 1, histone, and DNA, can promote caspase-11-mediated gasdermin D cleavage, interleukin 1ß proteolytic maturation, and lactate dehydrogenase release. The inhibition of AGER-mediated lipid peroxidation via arachidonate 5-lipoxygenase (ALOX5) limits caspase-11 inflammasome activation and pyroptosis in macrophages in response to nDAMPs or cytosolic lipopolysaccharide. Importantly, the pharmacologic inhibition of the AGER-ALOX5 pathway or global depletion (Ager-/-) or conditional depletion of AGER in myeloid cells (AgerMye-/-) protects against lipopolysaccharide-induced septic death in poly(I:C)-primed mice. These data identify a molecular basis for caspase-11 inflammasome activation and provide a potential strategy to treat sepsis.


Asunto(s)
Caspasas Iniciadoras/inmunología , Muerte Celular/inmunología , Inflamasomas/inmunología , Peroxidación de Lípido/inmunología , Receptor para Productos Finales de Glicación Avanzada/inmunología , Sepsis/inmunología , Animales , Araquidonato 5-Lipooxigenasa/inmunología , Femenino , Inmunidad Innata/inmunología , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología
18.
Biochem Biophys Res Commun ; 517(2): 221-226, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31331643

RESUMEN

The brain is one of the earliest organs to be influenced during sepsis. Sepsis-associated encephalopathy (SAE) is frequent, but seldomly recognized and has no testified pharmacological therapy. In this study, we demonstrated that pentamidine, an antiprotozoal drug, is a good candidate since it blocks S100B/RAGE/NF-κB signaling pathway. Pentamidine ameliorated cecal ligation and puncture (CLP)-induced brain damage assessed by crystal violet staining and hematoxylin and eosin (H&E) staining. Moreover, pentamidine reduced neuroinflammation in mouse hippocampi. Immunofluorescence and Western blot analysis also showed that pentamidine inhibited CLP-induced gliosis and S100B/RAGE/NF-κB pathway activation. Interestingly, it could also attenuate oxidative stress indicated by decreased protein levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), and attenuation of malondialdehyde (MDA) accumulation and superoxide dismutase (SOD) consumption. Thus the S100B/RAGE/NF-κB pathway may be crucial in the pathogenesis of SAE and may be a promising pharmacological target to prevent SAE.


Asunto(s)
Antiprotozoarios/uso terapéutico , Lesiones Encefálicas/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Pentamidina/uso terapéutico , Subunidad beta de la Proteína de Unión al Calcio S100/antagonistas & inhibidores , Animales , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/patología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores , FN-kappa B/inmunología , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/inmunología , Subunidad beta de la Proteína de Unión al Calcio S100/inmunología , Transducción de Señal/efectos de los fármacos
19.
Int J Mol Sci ; 20(13)2019 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-31266174

RESUMEN

It is incompletely understood how self-antigens become targets of humoral immunity in antibody-mediated autoimmune diseases. In this context, alarmins are discussed as an important level of regulation. Alarmins are recognized by various receptors, such as receptor for advanced glycation end products (RAGE). As RAGE is upregulated under inflammatory conditions, strongly binds nucleic acids and mediates pro-inflammatory responses upon alarmin recognition, our aim was to examine its contribution to immune complex-mediated autoimmune diseases. This question was addressed employing RAGE-/- animals in murine models of pristane-induced lupus, collagen-induced, and serum-transfer arthritis. Autoantibodies were assessed by enzyme-linked immunosorbent assay, renal disease by quantification of proteinuria and histology, arthritis by scoring joint inflammation. The associated immune status was determined by flow cytometry. In both disease entities, we detected tendentiously decreased autoantibody levels in RAGE-/- mice, however no differences in clinical outcome. In accordance with autoantibody levels, a subgroup of the RAGE-/- animals showed a decrease in plasma cells, and germinal center B cells and an increase in follicular B cells. Based on our results, we suggest that RAGE deficiency alone does not significantly affect antibody-mediated autoimmunity. RAGE may rather exert its effects along with other receptors linking environmental factors to auto-reactive immune responses.


Asunto(s)
Artritis Experimental/inmunología , Autoanticuerpos/metabolismo , Nefritis Lúpica/inmunología , Receptor para Productos Finales de Glicación Avanzada/deficiencia , Animales , Artritis Experimental/genética , Autoanticuerpos/sangre , Linfocitos B/inmunología , Colágeno/efectos adversos , Modelos Animales de Enfermedad , Centro Germinal/inmunología , Nefritis Lúpica/genética , Ratones , Receptor para Productos Finales de Glicación Avanzada/inmunología , Terpenos/efectos adversos
20.
Nutrients ; 11(6)2019 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-31242665

RESUMEN

The effect of glycation and aggregation of thermally processed ß-lactoglobulin (BLG) on binding to sRAGE and specific immunoglobulin E (sIgE) from cow milk allergic (CMA) patients were investigated. BLG was heated under dry conditions (water activity < 0.7) and wet conditions (in phosphate buffer at pH 7.4) at low temperature (<73 °C) and high temperatures (>90 °C) in the presence or absence of the milk sugar lactose. Nε-(carboxymethyl)-l-lysine (CML) western blot and glycation staining were used to directly identify glycation structures on the protein fractions on SDS-PAGE. Western blot was used to specify sRAGE and sIgE binding fractions. sRAGE binding was highest under wet-heated BLG independent of the presence of the milk sugar lactose. Under wet heating, high-molecular-weight aggregates were most potent and did not require the presence of CML to generate sRAGE binding ligands. In the dry system, sRAGE binding was observed only in the presence of lactose. sIgE binding affinity showed large individual differences and revealed four binding profiles. Dependent on the individual, sIgE binding decreased or increased by wet heating independent of the presence of lactose. Dry heating required the presence of lactose to show increased binding to aggregates in most individuals. This study highlights an important role of heating condition-dependent protein aggregation and glycation in changing the immunogenicity and antigenicity of cow's milk BLG.


Asunto(s)
Epítopos , Productos Finales de Glicación Avanzada/metabolismo , Calor , Inmunoglobulina E/metabolismo , Lactoglobulinas/metabolismo , Lisina/análogos & derivados , Hipersensibilidad a la Leche/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Agua/química , Productos Finales de Glicación Avanzada/inmunología , Inmunoglobulina E/inmunología , Lactoglobulinas/inmunología , Lactosa/química , Ligandos , Lisina/inmunología , Lisina/metabolismo , Hipersensibilidad a la Leche/inmunología , Agregado de Proteínas , Unión Proteica , Conformación Proteica , Receptor para Productos Finales de Glicación Avanzada/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...