Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Spectrochim Acta A Mol Biomol Spectrosc ; 237: 118387, 2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32416513

RESUMEN

In this study, the effect of a glycoprotein obtained from Fupenzi (FPZ) (Rubus chingii Hu.) on the fibrillation of bovine serum album (BSA) was investigated by multi-spectroscopic methods and transmission electron microscopy. Moreover, the cytotoxicity of the glycoprotein and the effect of it on H2O2-induced cell viability were investigated by cell counting kit and ß-galactosidase kit, respectively. The experimental results indicated that the glycoprotein showed very low toxicity to NRK-52E cells and could obviously delay cell senescence and improve cell viability. Moreover, the glycoprotein could effectively inhibit the formation of BSA fibrils and destroy the stability of preformed BSA fibrils in a concentration-dependent manner. Generally, antioxidant capacities are thought to be related to the anti-amyloidogenic activity of inhibitors; therefore, to reveal the inhibitory mechanism, the anti-oxidative property of the glycoprotein was examined by DPPH and ABTS assays. The results demonstrated that FPZ glycoprotein had a remarkable antioxidant activity and the IC50 values of DPPH and ABTS were 0.249 mg mL-1 and 0.092 mg mL-1, respectively. This work suggested that the FPZ glycoprotein had the potential to be designed a new therapeutic agent for attenuating aging and preventing the age-related diseases.


Asunto(s)
Glicoproteínas/química , Rubus/química , Albúmina Sérica Bovina/química , Amiloide/química , Amiloide/metabolismo , Animales , Antioxidantes/química , Antioxidantes/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Dicroismo Circular , Glicoproteínas/toxicidad , Peróxido de Hidrógeno/farmacología , Interacciones Hidrofóbicas e Hidrofílicas , Microscopía Electrónica de Transmisión , Estructura Secundaria de Proteína , Ratas
2.
Food Chem Toxicol ; 133: 110762, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31421212

RESUMEN

Miraculin is a glycoprotein with the ability to make sour substances taste sweet. The safety of miraculin has been evaluated using an approach proposed by the Food and Agriculture Organization of the United Nations and the World Health Organization for assessing the safety of novel proteins. Miraculin was shown to be fully and rapidly digested by pepsin in an in vitro digestibility assay. The proteomic analysis of miraculin's pepsin digests further corroborated that it is highly unlikely that any of the protein will remain intact within the gastrointestinal tract for potential absorption. The potential allergenicity and toxigenicity of miraculin, investigated using in silico bioinformatic analyses, demonstrated that miraculin does not represent a risk of allergy or toxicity to humans with low potential for cross-reactivity with other allergens. The results of a sensory study, characterizing the taste receptor activity of miraculin, showed that the taste-modifying effect of miraculin at the concentration intended for product development has a rapid onset and disappearance with no desensitizing impact on the receptor. Overall, the results of this study demonstrate that the use of miraculin to impact the sensory qualities of orally administered products with a bitter/sour taste profile is not associated with any safety concerns.


Asunto(s)
Glicoproteínas/toxicidad , Edulcorantes/toxicidad , Alérgenos/química , Alérgenos/aislamiento & purificación , Alérgenos/toxicidad , Simulación por Computador , Frutas/química , Glicoproteínas/química , Glicoproteínas/aislamiento & purificación , Humanos , Pepsina A/química , Proteolisis , Edulcorantes/química , Edulcorantes/aislamiento & purificación , Synsepalum/química , Gusto/efectos de los fármacos
4.
J Am Heart Assoc ; 6(8)2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28751543

RESUMEN

BACKGROUND: GPVI (Glycoprotein VI) is the essential platelet collagen receptor in atherothrombosis. Dimeric GPVI-Fc (Revacept) binds to GPVI binding sites on plaque collagen. As expected, it did not increase bleeding in clinical studies. GPVI-Fc is a potent inhibitor of atherosclerotic plaque-induced platelet aggregation at high shear flow, but its inhibition at low shear flow is limited. We sought to increase the platelet inhibitory potential by fusing GPVI-Fc to the ectonucleotidase CD39 (fusion protein GPVI-CD39), which inhibits local ADP accumulation at vascular plaques, and thus to create a lesion-directed dual antiplatelet therapy that is expected to lack systemic bleeding risks. METHODS AND RESULTS: GPVI-CD39 effectively stimulated local ADP degradation and, compared with GPVI-Fc alone, led to significantly increased inhibition of ADP-, collagen-, and human plaque-induced platelet aggregation in Multiplate aggregometry and plaque-induced platelet thrombus formation under arterial flow conditions. GPVI-CD39 did not increase bleeding time in an in vitro assay simulating primary hemostasis. In a mouse model of ferric chloride-induced arterial thrombosis, GPVI-CD39 effectively delayed vascular thrombosis but did not increase tail bleeding time in vivo. CONCLUSIONS: GPVI-CD39 is a novel approach to increase local antithrombotic activity at sites of atherosclerotic plaque rupture or injury. It enhances GPVI-Fc-mediated platelet inhibition and presents a potentially effective and safe molecule for the treatment of acute atherothrombotic events, with a favorable risk-benefit ratio.


Asunto(s)
Antígenos CD/farmacología , Apirasa/farmacología , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Fibrinolíticos/farmacología , Glicoproteínas/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/farmacología , Trombosis/prevención & control , Animales , Antígenos CD/toxicidad , Apirasa/farmacocinética , Apirasa/toxicidad , Enfermedades de las Arterias Carótidas/sangre , Enfermedades de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/inducido químicamente , Traumatismos de las Arterias Carótidas/patología , Cloruros , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Compuestos Férricos , Fibrinolíticos/farmacocinética , Fibrinolíticos/toxicidad , Glicoproteínas/farmacocinética , Glicoproteínas/toxicidad , Hemorragia/inducido químicamente , Humanos , Fragmentos Fc de Inmunoglobulinas/toxicidad , Masculino , Ratones Endogámicos C57BL , Placa Aterosclerótica , Inhibidores de Agregación Plaquetaria/farmacocinética , Inhibidores de Agregación Plaquetaria/toxicidad , Glicoproteínas de Membrana Plaquetaria/farmacocinética , Glicoproteínas de Membrana Plaquetaria/toxicidad , Proteínas Recombinantes de Fusión/farmacología , Trombosis/sangre , Trombosis/inducido químicamente , Trombosis/patología
5.
Thromb Haemost ; 117(8): 1651-1659, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28569920

RESUMEN

The efficiency of current dual antiplatelet therapy might be further improved by its combination with a glycoprotein (GP) VI-targeting strategy without increasing bleeding. GPVI-Fc, a recombinant dimeric fusion protein binding to plaque collagen and concealing binding sites for platelet GPVI, acts as a lesion-focused antiplatelet drug, and does not increase bleeding in vivo. We investigated, whether GPVI-Fc added in vitro on top of acetylsalicylic acid (ASA), the P2Y12 antagonist ticagrelor, and the fibrinogen receptor antagonist abciximab alone or in combination would increase inhibition of platelet activation by atherosclerotic plaque. Under static conditions, GPVI-Fc inhibited plaque-induced platelet aggregation by 53 %, and increased platelet inhibition by ASA (51 %) and ticagrelor (64 %) to 66 % and 80 %, respectively. Under arterial flow, GPVI-Fc inhibited plaque-induced platelet aggregation by 57 %, and significantly increased platelet inhibition by ASA (28 %) and ticagrelor (47 %) to about 81 % each. The triple combination of GPVI-Fc, ASA and ticagrelor achieved almost complete inhibition of plaque-induced platelet aggregation (93 %). GPVI-Fc alone or in combination with ASA or ticagrelor did not increase closure time measured by the platelet function analyzer (PFA)-200. GPVI-Fc added on top of abciximab, a clinically used anti-fibrinogen receptor antibody which blocks platelet aggregation, strongly inhibited total (81 %) and stable (89 %) platelet adhesion. We conclude that GPVI-Fc added on top of single or dual antiplatelet therapy with ASA and/or a P2Y12 antagonist is likely to improve anti-atherothrombotic protection without increasing bleeding risk. In contrast, the strong inhibition of platelet adhesion by GPVI-Fc in combination with GPIIb/IIIa inhibitors could be harmful.


Asunto(s)
Adenosina/análogos & derivados , Anticuerpos Monoclonales/farmacología , Aspirina/farmacología , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Glicoproteínas/farmacología , Fragmentos Fab de Inmunoglobulinas/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Placa Aterosclerótica , Inhibidores de Agregación Plaquetaria/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Trombosis/prevención & control , Abciximab , Adenosina/farmacología , Adenosina/toxicidad , Anticuerpos Monoclonales/toxicidad , Aspirina/toxicidad , Plaquetas/metabolismo , Quimioterapia Combinada , Glicoproteínas/toxicidad , Hemorragia/inducido químicamente , Humanos , Fragmentos Fab de Inmunoglobulinas/toxicidad , Fragmentos Fc de Inmunoglobulinas/toxicidad , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/toxicidad , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Trombosis/sangre , Trombosis/patología , Ticagrelor , Factores de Tiempo
6.
Toxicon ; 118: 141-8, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27155562

RESUMEN

Fish venom cytolysins are multifunctional proteins that in addition to their cytolytic/hemolytic effects display neurotoxic, cardiotoxic and inflammatory activities, being described as "protein lethal factors". A pore-forming cytolysin called Sp-CTx (Scorpaena plumieriCytolytic Toxin) has been recently purified from the venom of the scorpionfish Scorpaena plumieri. It is a glycoprotein with dimeric constitution, comprising subunits of approximately 65 kDa. Previous studies have revealed that this toxin has a vasorelaxant activity that appears to involve the L-arginine-nitric oxide synthase pathway; however its cardiovascular effects have not been fully comprehended. The present study examined the cardiovascular effects of Sp-CTx in vivo and in vitro. In anesthetized rats Sp-CTx (70 µg/kg i.v) produced a biphasic response which consisted of an initial systolic and diastolic pressure increase followed by a sustained decrease of these parameters and the heart rate. In isolated rats hearts Sp-CTx (10(-9) to 5 × 10(-6) M) produced concentration-dependent and transient ventricular positive inotropic effect and vasoconstriction response on coronary bed. In papillary muscle, Sp-CTx (10(-7) M) also produced an increase in contractile isometric force, which was attenuated by the catecholamine releasing agent tyramine (100 µM) and the ß-adrenergic antagonist propranolol (10 µM). On isolated ventricular cardiomyocytes Sp-CTx (1 nM) increased the L-type Ca(2+) current density. The results show that Sp-CTx induces disorders in the cardiovascular system through increase of sarcolemmal calcium influx, which in turn is partially caused by the release of endogenous noradrenaline.


Asunto(s)
Cardiotoxinas/toxicidad , Circulación Coronaria/efectos de los fármacos , Venenos de los Peces/química , Corazón/efectos de los fármacos , Músculos Papilares/efectos de los fármacos , Perciformes , Perforina/toxicidad , Animales , Presión Sanguínea/efectos de los fármacos , Brasil , Cardiotoxinas/administración & dosificación , Cardiotoxinas/aislamiento & purificación , Células Cultivadas , Proteínas de Peces/administración & dosificación , Proteínas de Peces/aislamiento & purificación , Proteínas de Peces/toxicidad , Glicoproteínas/administración & dosificación , Glicoproteínas/aislamiento & purificación , Glicoproteínas/toxicidad , Corazón/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Técnicas In Vitro , Inyecciones Intravenosas , Masculino , Contracción Muscular/efectos de los fármacos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Músculos Papilares/fisiología , Técnicas de Placa-Clamp , Perforina/administración & dosificación , Perforina/aislamiento & purificación , Ratas Wistar , Vasoconstrictores/administración & dosificación , Vasoconstrictores/aislamiento & purificación , Vasoconstrictores/toxicidad
7.
Glycobiology ; 25(11): 1142-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26224786

RESUMEN

Chagas' disease is a potentially life-threatening disease caused by the protozoan parasite Trypanosoma cruzi. Since the description of Chagas'disease in 1909 extensive research has identified important events in the disease in order to understand the biochemical mechanism that modulates T. cruzi-host cell interactions and the ability of the parasite to ensure its survival in the infected host. Exactly 30 years ago, we presented evidence for the first time of a trans-sialidase activity in T. cruzi (T. cruzi-TS). This enzyme transfers sialic acid from the host glycoconjugates to the terminal ß-galactopyranosyl residues of mucin-like molecules on the parasite's cell surface. Thenceforth, many articles have provided convincing data showing that T. cruzi-TS is able to govern relevant mechanisms involved in the parasite's survival in the mammalian host, such as invasion, escape from the phagolysosomal vacuole, differentiation, down-modulation of host immune responses, among others. The aim of this review is to cover the history of the discovery of T. cruzi-TS, as well as some well-documented biological effects encompassed by this parasite's virulence factor, an enzyme with potential attributes to become a drug target against Chagas disease.


Asunto(s)
Enfermedad de Chagas/parasitología , Glicoproteínas/toxicidad , Neuraminidasa/toxicidad , Proteínas Protozoarias/toxicidad , Trypanosoma cruzi/patogenicidad , Factores de Virulencia/toxicidad , Animales , Enfermedad de Chagas/inmunología , Glicoproteínas/inmunología , Humanos , Neuraminidasa/inmunología , Proteínas Protozoarias/inmunología , Trypanosoma cruzi/enzimología , Trypanosoma cruzi/inmunología , Factores de Virulencia/inmunología
8.
J Neuroimmunol ; 243(1-2): 43-51, 2012 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-22264993

RESUMEN

Current treatments used in Multiple Sclerosis (MS) are partly effective in the early stages of the disease but display very limited benefits in patients affected by progressive MS. One possible explanation is that these therapies are unable to target the inflammatory component most active during the progressive phase of the disease, and compartmentalized behind the blood-brain barrier. Our findings show that Rapamycin ameliorates clinical and histological signs of chronic EAE when administered during ongoing disease. Moreover, Rapamycin significantly reduced the hyperalgesia observed before clinical development of EAE which, in turn, is completely abolished by the administration of the drug.


Asunto(s)
Encefalomielitis Autoinmune Experimental/complicaciones , Inmunosupresores/uso terapéutico , Neuralgia/tratamiento farmacológico , Sirolimus/uso terapéutico , Análisis de Varianza , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Glicoproteínas/toxicidad , Hiperalgesia/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito , Neuralgia/patología , Umbral del Dolor/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Toxina del Pertussis/toxicidad , ARN Mensajero/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Factores de Tiempo
9.
Neurochem Int ; 60(3): 215-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22226840

RESUMEN

The role of glia maturation factor (GMF) in myelin oligodendrocyte glycoprotein (MOG) 35-55 peptide-induced experimental autoimmune encephalomyelitis (EAE) was investigated using GMF-deficient (GMF-KO) mice. We demonstrate that GMF-KO mice were resistant to the MOG 35-55 peptide-induced EAE as compared to wild type (Wt) mice (two in eight versus 10 in 10). Next, we examined the effect of administration of recombinant human GMF (rGMF) on MOG 35-55 peptide-induced EAE in mice. Daily administration of rGMF, staring days 1-14, resulted in significant exacerbation of clinical symptoms. Following rGMF injections, both GMF-KO (six in eight) and Wt mice (eight in eight) developed severe EAE (maximal clinical score of 3.5-4.0) with high frequency. The histological examination revealed severe infiltration of inflammatory cells in the spinal cord of MOG-immunized Wt mice while the resistance to EAE in GMF-KO mice was characterized by the absence of inflammatory cells. Administration of rGMF in Wt mice and GMF-KO mice resulted in a significant increase in infiltrating cells in the spinal cord following MOG-immunizations. We also evaluated cytokines and chemokines production as parameters of severity of inflammation in the spinal cord of Wt versus GMF-KO mice with and without GMF-reconstitution following MOG-immunizations. Cytokines (TNF-α, IFN-γ, IL-1ß, IL-6) and chemokines (CCL2, CCL3, CXCL10, GM-CSF) production were significantly greater in Wt mice than in GMF-KO mice following MOG-immunization. Furthermore, the reconstitution experiment with rGMF showed that the administration of rGMF in both, Wt mice and GMF-KO mice produced significant increase in the GMF-mediated cytokine/chemokine production.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Factor de Maduración de la Glia/fisiología , Glicoproteínas/toxicidad , Fragmentos de Péptidos/toxicidad , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Factor de Maduración de la Glia/genética , Humanos , Inmunización , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito , Proteínas Recombinantes/farmacología , Médula Espinal/patología
10.
J Immunol ; 188(5): 2084-92, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22279107

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) is a widely used model of multiple sclerosis. In NOD mice, EAE develops as a relapsing-remitting disease that transitions to a chronic progressive disease, making the NOD model the only mouse model that recapitulates the full clinical disease course observed in most multiple sclerosis patients. We have generated a TCR transgenic mouse that expresses the α- and ß-chains of a myelin oligodendrocyte glycoprotein (MOG) 35-55-reactive TCR (1C6) on the NOD background. 1C6 TCR transgenic mice spontaneously generate both CD4(+) and CD8(+) T cells that recognize MOG and produce proinflammatory cytokines, allowing for the first time to our knowledge the simultaneous examination of myelin-reactive CD4(+) and CD8(+) T cells in the same host. 1C6 CD8(+) T cells alone can induce optic neuritis and mild EAE with delayed onset; however, 1C6 CD4(+) T cells alone induce severe EAE and predominate in driving disease when both cell types are present. When 1C6 mice are crossed with mice bearing an IgH specific for MOG, the mice develop spontaneous EAE with high incidence, but surprisingly the disease pattern does not resemble the neuromyelitis optica-like disease observed in mice bearing CD4(+) T cells and B cells reactive to MOG on the C57BL/6 background. Collectively, our data show that although myelin-reactive CD8(+) T cells contribute to disease, disease is primarily driven by myelin-reactive CD4(+) T cells and that the coexistence of myelin-reactive T and B cells does not necessarily result in a distinct pathological phenotype.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Secuencia de Aminoácidos , Animales , Subgrupos de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/patología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Femenino , Técnicas de Sustitución del Gen , Glicoproteínas/administración & dosificación , Glicoproteínas/toxicidad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Datos de Secuencia Molecular , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/toxicidad
11.
J Neurosci Res ; 90(1): 84-95, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21922514

RESUMEN

During immune-mediated demyelinating lesions, the endocannabinoid system is involved in the pathogenesis of both neuroinflammation and neurodegeneration through different mechanisms. Here we explored the cellular distribution of the CB1 receptor (CB1R) in the central nervous system (CNS) and detected the level of CB1R expression during experimental autoimmune encephalomyelitis (EAE) by RT-qPCR, Western blotting, and immunostaining. Expression of CB1R was observed in neurons and microglia/macrophages but was barely detected in astrocytes. During EAE, the expression of CB1R in spinal cords was reduced at days 9, 17, and 28 postimmunization (p.i.), but the level of CB1R expression in spleens did not show a significant difference compared with complete Freund's adjuvant (CFA)-immunized mice. A selective CB1R antagonist (SR141716A) increased EAE clinical score, accompanied by weight loss. Unexpectedly, SR141716A inhibited the expression of CB1R but increased the expression of CB2R in brains, spinal cords, and spleens simultaneously. The administration of SR141716A increased interferon-γ, interleukin-17 (IL-17), and inflammatory cytokines such as IL-1ß, IL-6, and tumor necrosis factor-α in brains and/or spinal cords. A similar increase was observed in spontaneous and specific antigen-stimulated splenic mononuclear cells compared with vehicle controls. Interestingly, the expression of CX3CL1 was increased in brains and spinal cords but declined in spleens of EAE mice treated with SR141716A. These results indicate that manipulation of the CB1R may have therapeutic value in MS, but its complexity remains to be carefully considered and studied in further clinical application.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Piperidinas/farmacología , Pirazoles/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Análisis de Varianza , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Glicoproteínas/toxicidad , Hipocampo/citología , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito , Neuroglía/efectos de los fármacos , Neuroglía/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fragmentos de Péptidos/toxicidad , ARN Mensajero/metabolismo , Receptor Cannabinoide CB2/metabolismo , Rimonabant , Médula Espinal/metabolismo , Bazo/metabolismo
12.
Invest Ophthalmol Vis Sci ; 53(1): 406-12, 2012 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-22167100

RESUMEN

PURPOSE: Optic neuritis (ON) is a condition involving primary inflammation, demyelination, and axonal injury in the optic nerve and leads to apoptotic retinal ganglion cell (RGC) death, which contributes to the persistence of visual loss. Currently, ON has no effective treatment. The goal was to determine the effectiveness of immunotherapy with recombinant T-cell receptor ligand (RTL) in preventing ON in humanized HLA-DR2 transgenic mice. METHODS: Experimental autoimmune encephalomyelitis (EAE) was induced with myelin oligodendrocyte glycoprotein in humanized HLA-DR2 (DRß1*1501) transgenic mice. Five consecutive doses of RTL342M were administrated at the onset of ON. The development of autoimmune ON was assessed by histopathology at different time points. The levels of myelin loss, axonal loss, and RGC damage were examined by immunofluorescence. RESULTS: HLA-DR2 mice developed chronic ON 2 days before EAE characterized by progressive neurodegeneration in both organs. RTL342M significantly suppressed inflammation in the optic nerve and spinal cord and provided protection for at least 30 days. Examination of myelin loss showed a marked suppression of demyelination and an increase in myelin recovery in the optic nerve. Moreover, RTL342M treatment revealed a neuroprotective effect on optic nerve axons and RGCs in retinas at postimmunization (PI) day 62. CONCLUSIONS: RTL342M suppressed clinical and histologic signs of EAE/ON by preventing the recruitment of inflammatory cells into the optic nerve and showed neuroprotective effects against ON. However, to achieve full therapeutic benefit, more doses may be needed. These findings suggest a possible clinical application of this novel class of T-cell-tolerizing drugs for patients with optic neuritis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/prevención & control , Inmunoterapia , Ligandos , Neuritis Óptica/prevención & control , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Axones/patología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Glicoproteínas/toxicidad , Cadenas HLA-DRB1/genética , Masculino , Ratones , Ratones Transgénicos , Vaina de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito , Neuritis Óptica/inducido químicamente , Neuritis Óptica/patología , Fragmentos de Péptidos/toxicidad , Proteínas Recombinantes de Fusión/uso terapéutico , Células Ganglionares de la Retina/patología , Transgenes
13.
J Immunol ; 187(2): 791-804, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21685327

RESUMEN

The potential roles of TLRs in the cause and pathogenesis of autoimmune CNS inflammation remain contentious. In this study, we examined the effects of targeted deletions of TLR1, TLR2, TLR4, TLR6, TLR9, and MyD88 on the induction of myelin oligodendrocyte glycoprotein 35-55 (MOG(35-55)) peptide/CFA/pertussis toxin-induced autoimmune encephalomyelitis. Although C57BL/6.Tlr1(-/-), C57BL/6.Tlr4(-/-) and C57BL/6.Tlr6(-/-) mice showed normal susceptibility to disease, signs were alleviated in female C57BL/6.Tlr2(-/-) and C57BL/6.Tlr9(-/-) mice and C57BL/6.Tlr2/9(-/-) mice of both sexes. C57BL/6.Myd88(-/-) mice were completely protected. Lower clinical scores were associated with reduced leukocyte infiltrates. These results were confirmed by passive adoptive transfer of disease into female C57BL/6.Tlr2(-/-) and C57BL/6.Tlr9(-/-) mice, where protection in the absence of TLR2 was associated with fewer infiltrating CD4(+) cells in the CNS, reduced prevalence of detectable circulating IL-6, and increased proportions of central (CD62L(+)) CD4(+)CD25(+)Foxp3(+) regulatory T cells. These results provide a potential molecular mechanism for the observed effects of TLR signaling on the severity of autoimmune CNS inflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Factor 88 de Diferenciación Mieloide/fisiología , Receptor Toll-Like 1/deficiencia , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 6/deficiencia , Receptor Toll-Like 9/fisiología , Animales , Movimiento Celular/genética , Movimiento Celular/inmunología , Células Cultivadas , Encefalomielitis Autoinmune Experimental/genética , Femenino , Silenciador del Gen , Predisposición Genética a la Enfermedad , Glicoproteínas/administración & dosificación , Glicoproteínas/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito , Factor 88 de Diferenciación Mieloide/deficiencia , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/toxicidad , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/toxicidad , Índice de Severidad de la Enfermedad , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 9/deficiencia
14.
Restor Neurol Neurosci ; 29(3): 177-85, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21586824

RESUMEN

PURPOSE: Hematopoietic stem cell transplantation (HSCT) has been proposed as a novel therapy for multiple sclerosis (MS). CD4 + CD25 + regulatory T cells (Tregs) expressing Foxp3 play an important role in the maintenance of immune tolerance to self. Our study was conducted to confirm the efficiency of nonmyeloablative conditioning and syngeneic bone marrow transplantation (BMT) on experimental autoimmune encephalomyelitis (EAE) mice and to determine whether Tregs plays a role in the underlying mechanism. METHODS: EAE were induced in C57BL/6 mice and were randomly divided into 4 groups: the Conditioning group received the conditioning regimen, the Normal-EAE BMT group received conditioning and bone marrow (BM) grafts from normal mice, the EAE-EAE BMT group received conditioning and BM grafts from EAE mice and the EAE control group received no further therapy. The cumulative clinical score was used to assess the efficacy of the different treatments, and the proportion of Tregs in the spleen was measured by flow cytometry on day 40, 80 and 120 after BMT. Foxp3 mRNA expression was assessed by real-time PCR, and the expression of Foxp3 protein was tested by western blot on day 120 after BMT. RESULTS: Conditioning and conditioning with BMT led to a significant clinical improvement on day 80 after BMT compared with EAE without further treatment. On day 120 after BMT, the clinical score of the Conditioning group showed no significant difference from that of the EAE control group, whereas BMT led to a further amelioration of the disease score. On day 80 and day 120 after BMT, the proportions of Tregs of the two BMT groups were significantly higher than that in EAE control group, whereas no statistically significant difference was found between the Conditioning group and the EAE control group. On day 120 after BMT, the Foxp3 mRNA level and Foxp3 protein expression was higher in the two BMT groups than in EAE control group or Conditioning group. CONCLUSIONS: Nonmyeloablative conditioning could temporarily reverse already established EAE, but it was not sufficient for the induction of long-term EAE remission. Transplantation by BM cells from healthy or diseased donors was necessary and responsible for complete and long-time remission of EAE, and these beneficial effects may be the result of the induction of Tregs and the Treg-related factor Foxp3.


Asunto(s)
Trasplante de Médula Ósea/métodos , Ciclofosfamida/uso terapéutico , Encefalomielitis Autoinmune Experimental/terapia , Inmunosupresores/uso terapéutico , Análisis de Varianza , Animales , Antígenos CD4/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Citometría de Flujo/métodos , Regulación de la Expresión Génica/inmunología , Glicoproteínas/toxicidad , Factor Nuclear 3-gamma del Hepatocito/genética , Factor Nuclear 3-gamma del Hepatocito/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/toxicidad , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores , Factores de Tiempo
15.
J Agric Food Chem ; 58(19): 10356-63, 2010 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-20831249

RESUMEN

Soybean toxin (SBTX) is a 44 kDa glycoprotein that is lethal to mice (LD(50) = 5.6 mg/kg). This study reports the toxicity of SBTX on pathogenic fungi and yeasts and the mechanism of its action. SBTX inhibited spore germination of Aspergillus niger and Penicillium herguei and was toxic to Candida albicans, Candida parapsilosis, Kluyveromyces marxiannus , Pichia membranifaciens, and Saccharomyces cerevisiae. In addition, SBTX hampered the growth of C. albicans and K. marxiannus and inhibited the glucose-stimulated acidification of the incubation medium by S. cerevisiae, suggesting that SBTX interferes with intracellular proton transport to the external medium. Moreover, SBTX caused cell-wall disruption, condensation/shrinkage of cytosol, pseudohyphae formation, and P. membranifaciens and C. parapsilosis cell death. SBTX is toxic to fungi at concentrations far below the dose lethal to mice and has potential in the design of new antifungal drugs or in the development of transgenic crops resistant to pathogens.


Asunto(s)
Antifúngicos/farmacología , Fungicidas Industriales/farmacología , Glicoproteínas/farmacología , Proteínas de Soja/farmacología , Animales , Antifúngicos/toxicidad , Aspergillus niger/efectos de los fármacos , Aspergillus niger/crecimiento & desarrollo , Candida/efectos de los fármacos , Candida/crecimiento & desarrollo , Candida albicans/efectos de los fármacos , Candida albicans/crecimiento & desarrollo , Fungicidas Industriales/toxicidad , Glicoproteínas/toxicidad , Humanos , Kluyveromyces/efectos de los fármacos , Kluyveromyces/crecimiento & desarrollo , Dosificación Letal Mediana , Ratones , Penicillium/efectos de los fármacos , Pichia/efectos de los fármacos , Pichia/crecimiento & desarrollo , Enfermedades de las Plantas/microbiología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Proteínas de Soja/toxicidad , Esporas Fúngicas/efectos de los fármacos
16.
J Neurochem ; 115(4): 930-40, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20807308

RESUMEN

Photoreceptor degeneration is the hallmark of a group of inherited blinding diseases collectively termed retinitis pigmentosa (RP); a major cause of blindness in humans. RP is at present untreatable and the underlying neurodegenerative mechanisms are largely unknown, even though the genetic causes are often established. The activation of calpain-type proteases may play an important role in cell death in various neuronal tissues, including the retina. We therefore tested the efficacy of two different calpain inhibitors in preventing cell death in the retinal degeneration (rd1) human homologous mouse model for RP. Pharmacological inhibition of calpain activity in rd1 organotypic retinal explants had ambiguous effects on photoreceptor viability. Calpain inhibitor XI had protective effects when applied for short periods of time (16 h) but demonstrated substantial levels of toxicity in both wild-type and rd1 retina when used over several days. In contrast, the highly specific calpain inhibitor calpastatin peptide reduced photoreceptor cell death in vitro after both short and prolonged exposure, an effect that was also evident after in vivo application via intravitreal injection. These findings highlight the importance of calpain activation for photoreceptor cell death but also for photoreceptor survival and propose the use of highly specific calpain inhibitors to prevent or delay RP.


Asunto(s)
Calpaína/antagonistas & inhibidores , Glicoproteínas/toxicidad , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/enzimología , Retinitis Pigmentosa/inducido químicamente , Retinitis Pigmentosa/prevención & control , Animales , Proteínas de Unión al Calcio/uso terapéutico , Calpaína/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Glicoproteínas/uso terapéutico , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/inducido químicamente , Degeneración Retiniana/enzimología , Degeneración Retiniana/patología , Degeneración Retiniana/prevención & control , Retinitis Pigmentosa/enzimología , Retinitis Pigmentosa/patología
17.
J Neuroimmunol ; 223(1-2): 55-64, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20434781

RESUMEN

Clinical symptoms in MOG-induced EAE mice significantly exacerbated following chondroitin sulfate A (CS-A) injection, whereas administration of a degraded product, CSPG-DS, caused dramatic inhibition of EAE development. Also, administration of CSPG-DS but not CS-A, after the onset of clinical symptoms of EAE, was able to suppress the disease. Further studies demonstrated that CS-A up-regulated STAT4 expression and thus, induced IFN-gamma production and Th1 CD4 T cell differentiation. CS-A also up-regulated STAT3 and IL-23 expression and thus increased IL-17 producing T cells. CSPG-DS treatment both in vivo and in vitro decreased TNFalpha production from splenocytes. In vitro and in vivo studies indicated that CSPG-DS treatment in EAE mice significantly blocked migration of lymphocytes, whereas CS-A treatment increased lymphocyte infiltration in the brain.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antiinflamatorios no Esteroideos/administración & dosificación , Proteoglicanos Tipo Condroitín Sulfato/administración & dosificación , Disacáridos/administración & dosificación , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Adyuvantes Inmunológicos/metabolismo , Adyuvantes Inmunológicos/fisiología , Secuencia de Aminoácidos , Animales , Antiinflamatorios no Esteroideos/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/fisiología , Disacáridos/metabolismo , Disacáridos/fisiología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Femenino , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/toxicidad , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/toxicidad , Toxoides/antagonistas & inhibidores , Toxoides/toxicidad
18.
Nat Prod Res ; 24(7): 599-609, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20401791

RESUMEN

Consumption of fruits and vegetables has been associated with a low incidence of cardiovascular and other chronic diseases. The present study was aimed at evaluating the protective effects of fresh apple extract (AE) on human umbilical vein endothelial cells (HUVEC) exposed to cytotoxic glycated protein (GFBS)/iron (FeCl(3)) chelate. The experimental design comprised 10 groups with 5 flasks in each group. Group I was treated with 15% foetal bovine serum (FBS). Groups II, III and IV were treated with GFBS (70 microM), FBS + FeCl(3) (20 microM), and GFBS + FeCl(3), respectively. The other six groups were as follows: Group V, GFBS + AE (100 microg); Group VI, FBS + FeCl(3) + AE (100 microg); Group VII, GFBS + FeCl(3) + AE (100 microg); Group VIII, GFBS + AE (250 microg); Group IX, FBS + FeCl(3) + AE (250 microg); and Group X, GFBS + FeCl(3) + AE (250 microg). After 24 h incubation, cells were collected from all the experimental groups and assessed for lipid peroxidation (LPO) and activities of the antioxidant enzymes cytochrome c reductase and glutathione S-transferase (GST). HUVEC incubated with glycated protein (GFBS) either alone or combined with iron chelate showed a significant (p < 0.001) elevation of LPO accompanied by depletion of superoxide dismutase, catalase, glutathione peroxidase (GPx) and glutathione reductase (GR), in addition to increased microsomal cytochrome c reductase and decreased GST activities. Treatment of GFBS- or GFBS + FeCl(3)-exposed HUVEC with AE at 100 or 250 microg significantly decreased the level of LPO and returned the levels of antioxidants cytochrome c reductase and GST to near normal in a dose-dependent manner. The extracts recovered viability of HUVEC damaged by GFBS-iron treatment in a concentration-dependent manner. These findings suggest a protective effect of AE on HUVEC against glycated protein/iron chelate-induced toxicity, which suggests that AE could exert a beneficial effect by preventing diabetic angiopathies.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Glicoproteínas/toxicidad , Malus/química , Extractos Vegetales/farmacología , Venas Umbilicales/citología , Línea Celular , Productos Finales de Glicación Avanzada/toxicidad , Humanos , Peróxidos Lipídicos/metabolismo , Extractos Vegetales/química
19.
Eur J Immunol ; 40(5): 1486-95, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20186879

RESUMEN

DC are professional APC that instruct T cells during the inflammatory course of EAE. We have previously shown that MAPK3 (Erk1) is important for the induction of T-cell anergy. Our goal was to determine the influence of MAPK3 on the capacity of DC to arm T-cell responses in autoimmunity. We report that DC from Mapk3(-/-) mice have a significantly higher membrane expression of CD86 and MHC-II and--when loaded with the myelin oligodendrocyte glycoprotein--show a superior capacity to prime naïve T cells towards an inflammatory phenotype than Mapk3(+/+) DC. Nonetheless and as previously described, Mapk3(-/-) mice were only slightly but not significantly more susceptible to myelin oligodendrocyte glycoprotein-induced EAE than WT littermate mice. However, Mapk3(+/+) mice engrafted with Mapk3(-/-) BM (KO-->WT) developed a severe form of EAE, in direct contrast to WT-->KO mice, which were even less sick than control WT-->WT mice. An infiltration of DC and accumulation of Th17 cells was also observed in the CNS of KO-->WT mice. Therefore, triggering of MAPK3 in the periphery might be a therapeutic option for the treatment of neuroinflammation since absence of this kinase in the immune system leads to severe EAE.


Asunto(s)
Autoinmunidad/fisiología , Células Dendríticas/enzimología , Encefalomielitis Autoinmune Experimental/enzimología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Subgrupos de Linfocitos T/inmunología , Animales , Antígeno B7-2/metabolismo , Citocinas/biosíntesis , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Glicoproteínas/inmunología , Glicoproteínas/toxicidad , Antígenos de Histocompatibilidad Clase II/inmunología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Quinasa 3 Activada por Mitógenos/deficiencia , Proteína Quinasa 3 Activada por Mitógenos/genética , Glicoproteína Mielina-Oligodendrócito , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Quimera por Radiación , Organismos Libres de Patógenos Específicos , Especificidad del Receptor de Antígeno de Linfocitos T
20.
J Immunol ; 184(3): 1536-42, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20028655

RESUMEN

Calcium (Ca(2+)) signaling in T lymphocytes is essential for a variety of functions, including the regulation of differentiation, gene transcription, and effector functions. A major Ca(2+) entry pathway in nonexcitable cells, including T cells, is store-operated Ca(2+) entry (SOCE), wherein depletion of intracellular Ca(2+) stores upon receptor stimulation causes subsequent influx of extracellular Ca(2+) across the plasma membrane. Stromal interaction molecule (STIM) 1 is the Ca(2+) sensor in the endoplasmic reticulum, which controls this process, whereas the other STIM isoform, STIM2, coregulates SOCE. Although the contribution of STIM molecules and SOCE to T lymphocyte function is well studied in vitro, their significance for immune processes in vivo has remained largely elusive. In this study, we studied T cell function in mice lacking STIM1 or STIM2 in a model of myelin-oligodendrocyte glycoprotein (MOG(35-55))-induced experimental autoimmune encephalomyelitis (EAE). We found that STIM1 deficiency significantly impaired the generation of neuroantigen-specific T cell responses in vivo with reduced Th1/Th17 responses, resulting in complete protection from EAE. Mice lacking STIM2 developed EAE, but the disease course was ameliorated. This was associated with a reduced clinical peak of disease. Deficiency of STIM2 was associated with an overall reduced proliferative capacity of lymphocytes and a reduction of IFN-gamma/IL-17 production by neuroantigen-specific T cells. Neither STIM1 nor STIM2 deficiency altered the phenotype or function of APCs. These findings reveal a crucial role of STIM-dependent pathways for T cell function and activation under autoimmune inflammatory conditions, establishing them as attractive new molecular therapeutic targets for the treatment of inflammatory and autoimmune disorders.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Mediadores de Inflamación/fisiología , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/fisiología , Subgrupos de Linfocitos T/inmunología , Animales , Canales de Calcio , Señalización del Calcio/genética , Señalización del Calcio/inmunología , Sistemas de Liberación de Medicamentos , Encefalomielitis Autoinmune Experimental/mortalidad , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Glicoproteínas/administración & dosificación , Glicoproteínas/toxicidad , Inmunidad Innata/genética , Activación de Linfocitos/genética , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Noqueados , Ratones Mutantes , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/toxicidad , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Molécula de Interacción Estromal 1 , Molécula de Interacción Estromal 2 , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA