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










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(15)2023 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-37569901

RESUMEN

The major proteins involved in Alzheimer's disease (AD) are amyloid precursor protein (APP) and Tau. We demonstrate that APP1 (390-412) and Tau1 (19-34), linked together with either a flexible or a rigid peptide bridge, are able to inhibit, in vitro, the interaction between APP and Tau proteins. Furthermore, nasal administration of biotin-labelled Flex peptide for two weeks indicated the localization of the peptide around and close to plaques in the hippocampus area. In vivo studies in 5xFAD transgenic (Tg) mice, which exhibit plaque load and mild cognitive decline at four months of age, show that nasal administration of the flexible linked peptide reduced amyloid plaque burden. Additionally, nasal treatment with either flexible or rigid linked peptides prevented cognitive function deterioration. A significant treatment effect was achieved when either treatment was initiated at the age of three months, before severe cognitive deficiency is evident, or at five months, when such deficiency is already observed. The nasally treated mice demonstrated a cognitive ability not significantly different from the non-Tg littermate controls. Testing the effect of the flexible peptide by gavage feeding on the cognitive function of 5xFAD Tg mice demonstrated that feeding as well as nasal treatment significantly improves the cognitive ability of Tg mice compared to control PBS-treated mice.

2.
Int J Mol Sci ; 21(9)2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32380752

RESUMEN

The two major proteins involved in Alzheimer's disease (AD) are the amyloid precursor protein (APP) and Tau. Here, we demonstrate that these two proteins can bind to each other. Four possible peptides APP1 (390-412), APP2 (713-730), Tau1 (19-34) and Tau2 (331-348), were predicted to be involved in this interaction, with actual binding confirmed for APP1 and Tau1. In vivo studies were performed in an Alzheimer Disease animal model-APP double transgenic (Tg) 5xFAD-as well as in 5xFAD crossed with Tau transgenic 5xFADXTau (FT), which exhibit declined cognitive reduction at four months of age. Nasal administration of APP1 and Tau1 mixture, three times a week for four or five months, reduced amyloid plaque burden as well as the level of soluble Aß 1-42 in the brain. The treatment prevented the deterioration of cognitive functions when initiated at the age of three months, before cognitive deficiency was evident, and also at the age of six months, when such deficiencies are already observed, leading to a full regain of cognitive function.


Asunto(s)
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Biomarcadores , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/etiología , Placa Amiloide/patología , Unión Proteica
3.
Biochem Biophys Res Commun ; 513(1): 219-225, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30952434

RESUMEN

By year 2025 pancreatic ductal adenocarcinoma (PDAC) is expected to become the second leading cause of cancer related death. However, other than improved chemotherapy and a small molecule inhibitor of the epidermal growth factor receptor (EGFR), no targeted drugs are currently available. Repurposing of approved drugs might offer a rapid solution. We employed an animal PDAC model, expressing a mutant and a wild type form of p53 and KRAS, respectively. Cetuximab, a clinically approved anti-EGFR monoclonal antibody (mAb) weakly inhibited PDAC xenografts, similar to trastuzumab, a mAb against HER2, a co-receptor of EGFR. Because the combination of cetuximab and trastuzumab only moderately enhanced the anti-tumor effects, we combined each with a home-made mAb to the same receptor and identified two cooperative pairs. The pair of trastuzumab and a murine anti-HER2 mAb better than the anti-EGFR pair inhibited PDAC xenografts, although HER2's abundance in our model is 15-fold lower than the level of EGFR. In vitro studies attribute cooperation to forced receptor endocytosis/degradation and inhibition of both DNA synthesis and cell migration. Taken together, our results identify cooperative pairs of anti-PDAC antibodies and highlight potential mechanisms of anti-tumor effects.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Cetuximab/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Trastuzumab/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Ratones Desnudos , Neoplasias Pancreáticas/patología
4.
Biochem Biophys Res Commun ; 465(2): 218-24, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26248137

RESUMEN

Aptamers represent a promising new treatment modality for cancer. Specificity and high affinity are two parameters that characterize aptamers. In this work, we elucidated physicochemical parameters of an ERBB2/HER2 specific aptamer and determined an optimal multimerization state, leading to higher binding and improved avidity. We applied biochemical, immunochemical and biophysical methodologies to characterize binding behaviors of multimerized versions of an ERBB2/HER2 specific aptamer and demonstrate structural integrity. Finally, we show that the trimeric ERBB2/HER2 specific aptamer instigates no immunogenic response in vivo. In summary, the set of methodologies we employed establishes a way to enhance activity of a model HER2-aptamer.


Asunto(s)
Antineoplásicos/administración & dosificación , Aptámeros de Nucleótidos/administración & dosificación , Tolerancia Inmunológica , Polimerizacion , Receptor ErbB-2/antagonistas & inhibidores , Animales , Antineoplásicos/síntesis química , Antineoplásicos/inmunología , Aptámeros de Nucleótidos/síntesis química , Aptámeros de Nucleótidos/inmunología , Secuencia de Bases , Línea Celular Tumoral , Expresión Génica , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Unión Proteica , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(38): 15389-94, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-24003140

RESUMEN

Due to intrinsic aggressiveness and lack of effective therapies, prognosis of pancreatic cancer remains dismal. Because the only molecular targeted drug approved for pancreatic ductal adenocarcinoma is a kinase inhibitor specific to the epidermal growth factor receptor (EGFR), and this receptor collaborates with another kinase, called HER2 (human EGF-receptor 2), we assumed that agents targeting EGFR and/or HER2 would effectively retard pancreatic ductal adenocarcinoma. Accordingly, two immunological strategies were tested in animal models: (i) two antibodies able to engage distinct epitopes of either EGFR or HER2 were separately combined, and (ii) pairs of one antibody to EGFR and another to HER2. Unlike the respective single monoclonal antibodies, which induced weak effects, both types of antibody combinations synergized in animals in terms of tumor inhibition. Immunological cooperation may not depend on receptor density, antigenic sites, or the presence of a mutant RAS protein. Nevertheless, both types of antibody combinations enhanced receptor degradation. Future efforts will examine the feasibility of each strategy and the potential of combining them to achieve sustained tumor inhibition.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Receptores ErbB/inmunología , Receptor ErbB-2/inmunología , Transducción de Señal/inmunología , Análisis de Varianza , Animales , Anticuerpos Monoclonales/inmunología , Carcinoma Ductal Pancreático/inmunología , Sinergismo Farmacológico , Femenino , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Desnudos
6.
Proc Natl Acad Sci U S A ; 110(20): 8170-5, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23630281

RESUMEN

Aptamers, oligonucleotides able to avidly bind cellular targets, are emerging as promising therapeutic agents, analogous to monoclonal antibodies. We selected from a DNA library an aptamer specifically recognizing human epidermal growth factor receptor 2 (ErbB-2/HER2), a receptor tyrosine kinase, which is overexpressed in a variety of human cancers, including breast and gastric tumors. Treatment of human gastric cancer cells with a trimeric version (42 nucleotides) of the selected aptamer (14 nucleotides) resulted in reduced cell growth in vitro, but a monomeric version was ineffective. Likewise, when treated with the trimeric aptamer, animals bearing tumor xenografts of human gastric origin reflected reduced rates of tumor growth. The antitumor effect of the aptamer was nearly twofold stronger than that of a monoclonal anti-ErbB-2/HER2 antibody. Consistent with aptamer-induced intracellular degradation of ErbB-2/HER2, incubation of gastric cancer cells with the trimeric aptamer promoted translocation of ErbB-2/HER2 from the cell surface to cytoplasmic puncta. This translocation was associated with a lysosomal hydrolase-dependent clearance of the ErbB-2/HER2 protein from cell extracts. We conclude that targeting ErbB-2/HER2 with DNA aptamers might retard the tumorigenic growth of gastric cancer by means of accelerating lysosomal degradation of the oncoprotein. This work exemplifies the potential pharmacological utility of aptamers directed at cell surface proteins, and it highlights an endocytosis-mediated mechanism of tumor inhibition.


Asunto(s)
Antineoplásicos/farmacología , Aptámeros de Nucleótidos/química , Receptor ErbB-2/química , Neoplasias Gástricas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Proliferación Celular , Reactivos de Enlaces Cruzados/química , Citoplasma/metabolismo , ADN/química , Femenino , Humanos , Lisosomas/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Nucleótidos/química , Unión Proteica
7.
Proc Natl Acad Sci U S A ; 110(5): 1815-20, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23319610

RESUMEN

Breast tumors lacking expression of human epidermal growth factor receptor 2 (HER2) and the estrogen and the progesterone receptors (triple negative; TNBC) are more aggressive than other disease subtypes, and no molecular targeted agents are currently available for their treatment. Because TNBC commonly displays EGF receptor (EGFR) expression, and combinations of monoclonal antibodies to EGFR effectively inhibit other tumor models, we addressed the relevance of this strategy to treatment of TNBC. Unlike a combination of the clinically approved monoclonal antibodies, cetuximab and panitumumab, which displaced each other and displayed no cooperative effects, several other combinations resulted in enhanced inhibition of TNBC's cell growth both in vitro and in animals. The ability of certain antibody mixtures to remove EGFR from the cell surface and to promote its intracellular degradation correlated with the inhibitory potential. However, unlike EGF-induced sorting of EGFR to lysosomal degradation, the antibody-induced pathway displayed independence from the intrinsic kinase activity and dimer formation ability of EGFR, and it largely avoided the recycling route. In conclusion, although TNBC clinical trials testing EGFR inhibitors reported lack of benefit, our results offer an alternative strategy that combines noncompetitive antibodies to achieve robust degradation of EGFR and tumor inhibition.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cetuximab , Receptores ErbB/metabolismo , Femenino , Células HeLa , Humanos , Immunoblotting , Ratones , Ratones Desnudos , Panitumumab , Proteolisis/efectos de los fármacos , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Carga Tumoral/efectos de los fármacos
8.
J Immunol ; 185(6): 3401-7, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20720210

RESUMEN

Mucosal (nasal or oral) administration of anti-CD3 mAb is effective in ameliorating animal models of autoimmunity (experimental autoimmune encephalomyelitis, diabetes, and lupus) by inducing LAP(+) regulatory T cells. We tested this approach in an arthritis model using type II collagen. We found that nasal anti-CD3 was more effective than oral anti-CD3 in attenuating the development of arthritis. Nasal anti-CD3 induced a LAP(+) regulatory T cell that secreted high levels of IL-10 and suppressed collagen-specific T cell proliferation and anti-collagen Ab production. However, neither nasal nor oral anti-CD3 attenuated disease when given to animals with ongoing arthritis, and this was associated with a lack of induction of LAP(+) regulatory T cells. We found, however, that coadministration of a novel emulsome adjuvant, which enhances Th2 responses, resulted in the induction of LAP(+) regulatory T cells and suppression of ongoing arthritis by both nasal and oral anti-CD3. Suppression of arthritis by mucosal anti-CD3 was associated with less joint damage, a decrease of TNF-alpha and IFN-gamma mRNA expression in joints, and a reduction in anti-collagen Abs. These results demonstrate that mucosal anti-CD3 therapy may serve as a therapeutic approach in arthritis and that the biologic effect is enhanced by an emulsome-based adjuvant.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Artritis Experimental/prevención & control , Complejo CD3/inmunología , Colágeno Tipo II/toxicidad , Precursores de Proteínas/biosíntesis , Linfocitos T Reguladores/inmunología , Células Th2/patología , Factor de Crecimiento Transformador beta/biosíntesis , Regulación hacia Arriba/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Monoclonales/fisiología , Artritis Experimental/inmunología , Artritis Experimental/patología , Diferenciación Celular/inmunología , Células Cultivadas , Emulsiones , Masculino , Ratones , Ratones Endogámicos DBA , Mucosa Bucal/inmunología , Mucosa Bucal/metabolismo , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Péptidos/fisiología , Precursores de Proteínas/fisiología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Células Th2/inmunología , Factor de Crecimiento Transformador beta/fisiología
9.
Proc Natl Acad Sci U S A ; 107(21): 9765-70, 2010 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-20445103

RESUMEN

Leptin-deficient ob/ob mice are overweight, develop insulin resistance, and serve as a model for type 2 diabetes (T2D). Studies suggest that inflammatory pathways are linked to the development of insulin resistance and T2D both in animals and humans. We asked whether the induction of regulatory T cells (Tregs) could alleviate the pathological and metabolic abnormalities in ob/ob mice. We induced TGF-beta-dependent CD4(+) latency-associated peptide (LAP)-positive Tregs by oral administration of anti-CD3 antibody plus beta-glucosylceramide. We found a decrease in pancreatic islet cell hyperplasia, fat accumulation in the liver, and inflammation in adipose tissue, accompanied by lower blood glucose and liver enzymes. In addition, treated animals had decreased CD11b(+)F4/80(+) macrophages and TNF-alpha in adipose tissue. Adoptive transfer of orally induced CD4(+)LAP(+) Tregs ameliorated metabolic and cytokine abnormalities. Our results demonstrate the importance of inflammation in T2D and identify a unique immunological approach for treatment of T2D by the induction of Tregs.


Asunto(s)
Tejido Adiposo/inmunología , Resistencia a la Insulina , Linfocitos T Reguladores/inmunología , Animales , Glucemia/metabolismo , Proliferación Celular , Colesterol/sangre , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/inmunología , Hiperplasia/inmunología , Inflamación/inmunología , Hígado/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Células T Asesinas Naturales/inmunología , Páncreas/inmunología , Linfocitos T Reguladores/citología
10.
J Neurol Sci ; 274(1-2): 9-12, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18804221

RESUMEN

One of the major goals for the immunotherapy of autoimmune diseases is the induction of regulatory T cells that mediate immunologic tolerance. Parenteral administration of anti-CD3 monoclonal antibody is an approved therapy for transplantation in humans and is effective in autoimmune diabetes. We have found that oral administration of anti-CD3 monoclonal antibody is biologically active in the gut and suppresses experimental autoimmune encephalomyelitis both prior to disease induction and at the height of disease. Oral anti-CD3 antibody acts by inducing a unique type of regulatory T cell characterized by latency-associated peptide (LAP) on its cell surface that functions in vivo and in vitro via TGF-beta dependent mechanism. Orally delivered antibody would not have side effects including cytokine release syndromes, thus oral anti-CD3 antibody is clinically applicable for chronic therapy. These findings identify a novel and powerful immunologic approach that is widely applicable for the treatment of human autoimmune conditions.


Asunto(s)
Anticuerpos/administración & dosificación , Autoinmunidad/efectos de los fármacos , Complejo CD3/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Administración Oral , Animales , Autoinmunidad/inmunología , Diabetes Mellitus Tipo 1/terapia , Humanos , Ratones
11.
Diabetes ; 56(8): 2103-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17456848

RESUMEN

Anti-CD3 monoclonal antibody (mAb) has been shown to induce tolerance and to be an effective treatment for diabetes both in animal models and in human trials. We have shown that anti-CD3 mAb given orally is biologically active in the gut and suppresses experimental autoimmune encephalitis by the induction of a regulatory T-cell that expresses latency-associated peptide (LAP) on its surface. In the present study, we investigated the effect of oral anti-CD3 mAb on the prevention of autoimmune diabetes in AKR mice in which the low-dose streptozocin (STZ) model induces autoimmunity to the beta-cells of the islets. We found that oral anti-CD3 mAb given at doses of 50 and 250 microg/feeding suppressed the incidence of diabetes in this model with the best effects seen at the 50 microg/dose. Associated with suppression, we observed decreased cell proliferation in the spleen and conversion of T-helper (Th)1 responses into Th2/Th3 responses in the periphery, including the pancreatic lymph nodes. Oral anti-CD3 mAb increased the expression of LAP on CD4(+) T-cells, and these cells could adoptively transfer protection. Protection by oral anti-CD3 was transforming growth factor-beta dependent. Our results demonstrate that oral anti-CD3 is effective in the model of STZ-induced diabetes and may be a useful form of therapy for type 1 diabetes in humans.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Complejo CD3/inmunología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Inmunoterapia , Administración Oral , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/biosíntesis , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/prevención & control , Insulina/metabolismo , Absorción Intestinal , Intestinos/efectos de los fármacos , Intestinos/inmunología , Masculino , Ratones , Ratones Endogámicos AKR , Páncreas/metabolismo , Bazo/efectos de los fármacos , Bazo/metabolismo , Estreptozocina/farmacología , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
12.
Nat Med ; 12(6): 627-35, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16715091

RESUMEN

A major goal of immunotherapy for autoimmune diseases and transplantation is induction of regulatory T cells that mediate immunologic tolerance. The mucosal immune system is unique, as tolerance is preferentially induced after exposure to antigen, and induction of regulatory T cells is a primary mechanism of oral tolerance. Parenteral administration of CD3-specific monoclonal antibody is an approved therapy for transplantation in humans and is effective in autoimmune diabetes. We found that orally administered CD3-specific antibody is biologically active in the gut and suppresses autoimmune encephalomyelitis both before induction of disease and at the height of disease. Orally administered CD3-specific antibody induces CD4+ CD25- LAP+ regulatory T cells that contain latency-associated peptide (LAP) on their surface and that function in vitro and in vivo through a TGF-beta-dependent mechanism. These findings identify a new immunologic approach that is widely applicable for the treatment of human autoimmune conditions.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Péptidos/inmunología , Receptores de Interleucina-2/inmunología , Subgrupos de Linfocitos T/inmunología , Administración Oral , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Linfocitos T CD4-Positivos/citología , Cricetinae , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Péptidos/genética , Receptores de Interleucina-2/genética , Subgrupos de Linfocitos T/citología , Factor de Crecimiento Transformador beta/inmunología
13.
J Clin Invest ; 115(9): 2423-33, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16100572

RESUMEN

Amyloid beta-peptide (Abeta) appears to play a key pathogenic role in Alzheimer disease (AD). Immune therapy in mouse models of AD via Abeta immunization or passive administration of Abeta antibodies markedly reduces Abeta levels and reverses behavioral impairment. However, a human trial of Abeta immunization led to meningoencephalitis in some patients and was discontinued. Here we show that nasal vaccination with a proteosome-based adjuvant that is well tolerated in humans plus glatiramer acetate, an FDA-approved synthetic copolymer used to treat multiple sclerosis, potently decreases Abeta plaques in an AD mouse model. This effect did not require the presence of antibody, as it was observed in B cell-deficient (Ig mu-null) mice. Vaccinated animals developed activated microglia that colocalized with Abeta fibrils, and the extent of microglial activation correlated strongly with the decrease in Abeta fibrils. Activation of microglia and clearing of Abeta occurred with the adjuvant alone, although to a lesser degree. Our results identify a novel approach to immune therapy for AD that involves clearing of Abeta through the utilization of compounds that have been safely tested on or are currently in use in humans.


Asunto(s)
Adyuvantes Inmunológicos , Administración Intranasal , Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Cisteína Endopeptidasas , Lipopolisacáridos , Péptidos/uso terapéutico , Vacunación , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Antígenos CD/metabolismo , Barrera Hematoencefálica/citología , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/patología , Cisteína Endopeptidasas/administración & dosificación , Cisteína Endopeptidasas/uso terapéutico , Modelos Animales de Enfermedad , Combinación de Medicamentos , Femenino , Acetato de Glatiramer , Humanos , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/uso terapéutico , Masculino , Ratones , Ratones Transgénicos , Complejo de la Endopetidasa Proteasomal
14.
J Neurol Sci ; 233(1-2): 125-32, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15894335

RESUMEN

Mucosal tolerance has been used successfully to treat animal models of autoimmune diseases and is being tested in human diseases. In this work we demonstrate the reduction of infarct size following mucosal tolerance by myelin oligodendrocyte glycoprotein (MOG) (35-55) peptide in mouse stroke model. Nasal MOG was most efficacious and reduced ischemic infarct size by 70% at 24 h as well as improving behavior score. Using immunohistological methods and IL-10 -/- mice, we demonstrate the importance of IL-10-producing CD4+ T cells in the reduction of the ischemic infarct volume following middle cerebral artery occlusion (MCAO). Furthermore, adoptive transfer of CD4+ T cells from nasally tolerized mice to untreated mice prior to MCAO surgery significantly decreased stroke size (p<0.001 vs. control), whereas CD4+ T cells from nasally tolerized IL-10-deficient mice had no significant effect. Based on these results, modulation of cerebral inflammation by mucosal tolerance to myelin antigens may have applicability both as prophylactic therapy and treatment following ischemia attacks.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Glicoproteínas/administración & dosificación , Interleucina-10/metabolismo , Interleucina-10/uso terapéutico , Fragmentos de Péptidos/administración & dosificación , Accidente Cerebrovascular/terapia , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Conducta Animal , Infarto Encefálico/etiología , Infarto Encefálico/prevención & control , Linfocitos T CD4-Positivos/fisiología , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Esquema de Medicación , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/fisiología , Inmunohistoquímica/métodos , Infarto de la Arteria Cerebral Media/complicaciones , Interleucina-10/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Inmunológicos , Glicoproteína Mielina-Oligodendrócito , Índice de Severidad de la Enfermedad , Accidente Cerebrovascular/etiología , Factores de Tiempo
15.
J Immunol ; 171(12): 6549-55, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14662856

RESUMEN

Inflammation plays an important role in ischemic stroke and in humans IL-10 may have a beneficial effect in stroke. We mucosally administered myelin oligodendrocyte glycoprotein (MOG) 35-55 peptide to C57BL/6 mice before middle cerebral artery occlusion (MCAO) to induce an anti-inflammatory T cell response directed at CNS myelin. Nasal and oral administration of MOG(35-55) peptide decreased ischemic infarct size at 24 and 72 h after MCAO surgery. Nasal MOG(35-55) peptide was most efficacious and reduced infarct size by 70% at 24 h and by 50% at 72 h (p

Asunto(s)
Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Glicoproteínas/administración & dosificación , Interleucina-10/biosíntesis , Fragmentos de Péptidos/administración & dosificación , Vacunas/administración & dosificación , Administración Intranasal , Administración Oral , Traslado Adoptivo , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/inmunología , Linfocitos T CD4-Positivos/trasplante , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Glicoproteínas/inmunología , Tolerancia Inmunológica/genética , Inmunidad Mucosa/genética , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/prevención & control , Interleucina-10/deficiencia , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/inmunología , Fragmentos de Péptidos/inmunología , Vacunas/inmunología
16.
FASEB J ; 17(8): 860-8, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12724345

RESUMEN

Anti-glomerular basement membrane (GBM) disease is a rapidly progressive glomerulonephritis (GN) resulting from autoimmunity against the Goodpasture antigen alpha3(IV)NC1. In addition to the well-characterized antibody contribution, a T helper 1 (Th1) response has been suspected as the culprit for glomerular injury. We induced anti-GBM disease in DBA/1, C57BL/6, AKR, and NOD mice with recombinant human alpha3(IV)NC1 to investigate the involvement of humoral and cellular autoimmunity. DBA/1 mice had crescentic GN 11 wk postimmunization with alpha3(IV)NC1. C57BL/6 and AKR mice developed a chronic disease course resulting in comparable kidney injury to DBA/1 mice within 6 months. NOD revealed only minor glomerular changes. The rapid course and the severity of the disease in DBA/1 mice can be explained by our immunological findings in their sera and splenocytes: 1) high antibody titers specific for the putative clinically relevant epitope of alpha3(IV)NC1 with Th1-type isotypes, and 2) a strong proliferative response and high amounts of the inflammatory cytokine IFN-gamma, secreted by splenocytes stimulated in vitro with alpha3(IV)NC1, with only low amounts of the anti-inflammatory cytokine IL-10. Our in vivo and in vitro results provide direct evidence that the balance between Th1 and Th2 responses associates with the outcome of anti-GBM disease in mice.


Asunto(s)
Enfermedad por Anticuerpos Antimembrana Basal Glomerular/inmunología , Inmunidad Celular/inmunología , Animales , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/patología , Western Blotting , Línea Celular , Colágeno Tipo IV/genética , Colágeno Tipo IV/inmunología , Humanos , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Glomérulos Renales/ultraestructura , Masculino , Ratones , Ratones Endogámicos AKR , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos NOD , Microscopía Electrónica , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Esclerosis , Índice de Severidad de la Enfermedad , Especificidad de la Especie , Factores de Tiempo
17.
J Autoimmun ; 20(2): 135-45, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12657527

RESUMEN

Oral administration of antigen leads to specific immune hyporesponsiveness termed as oral tolerance. Different doses and feeding regimens have been demonstrated to induce different types of tolerance and degrees of immune suppression. Herein, we compare distinct different regimens of feeding using equivalent final doses of antigen in order to investigate the role of frequency of antigen uptake in the induction of oral tolerance. We demonstrate that continuous feeding of antigen in the drinking water, as compared to a single feeding or feeding once per day over several days enhances suppression to both Th1 and Th2 type responses in B6D2F1 and BALB/c mice. Continuous feeding suppresses antibody responses in aged B6D2F1 mice, which are otherwise refractory to oral tolerance induction. Continuous feeding of ovalbumin (OVA) in high or low doses, as compared to control or single daily feeding over several days, up-regulates interleukin-10 (IL-10) and transforming growth factor-beta (TGF-beta) production in both OVA TCR transgenic and BALB/c mice. In all regimens tested in wild type mice, low doses were more efficacious than high doses in inducing IL-10 and TGF-beta. Serial feeding (multiple low dose daily gavages) using OVA or myelin basic protein (MBP), also led to up-regulation of TGF-beta and IL-10 production in OVA TCR and MBP TCR transgenic mice, as well as enhanced inhibition of MBP-induced experimental autoimmune encephalomyelitis (EAE) in (PLxSJL) F1 mice. We did not find differences in the cytokine profile between serial (multiple low dose daily gavages) and continuous feeding regimens, suggesting that repetitive discrete delivery of oral antigen provides a sustained signal for the induction of oral tolerance. Thus, using different regimens of feeding that resemble natural feeding with equivalent final doses of antigen, we found enhancement of oral tolerance utilizing regimens that resemble natural feeding. Such feeding regimens may be advantageous in the application of oral tolerance for clinical purposes in the treatment of autoimmune and other inflammatory conditions.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Tolerancia Inmunológica/inmunología , Interleucina-10/farmacología , Factor de Crecimiento Transformador beta/farmacología , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ovalbúmina/farmacología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Regulación hacia Arriba
18.
Circulation ; 106(13): 1708-15, 2002 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-12270867

RESUMEN

BACKGROUND: Increasing evidence supports the involvement of inflammation and immunity in atherogenesis as well as the role of autoimmunity to heat shock proteins (HSPs) in the progression of atherosclerosis. Mucosal administration of autoantigens decreases organ-specific inflammation and disease in several models of autoimmunity (diabetes, arthritis, and encephalomyelitis) and is also being tested in human clinical trials. METHODS AND RESULTS: We examined the effect of nasal or oral administration of mycobacterial HSP-65 on atherosclerotic lesion formation in mice lacking the receptor for LDL that were maintained on a high-cholesterol diet. Animals were nasally or orally treated for 1 week with HSP-65, and a high-cholesterol diet was started after the last treatment. The mice were mucosally treated once a week for 8 or 12 weeks, at which time pathological analysis was performed. We found a significant decrease in the size of atherosclerotic plaques, a reduction in macrophage-positive area in the aortic arch, increased interleukin-10 expression, and a reduced number of T cells in nasally treated animals compared with control animals. A similar trend was observed in orally treated mice, but it was not significant. CONCLUSIONS: Our results demonstrate that nasal vaccination with HSP reduces the inflammatory process associated with atherosclerosis and provides a new immunologic approach for the treatment of atherosclerosis.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Arteriosclerosis/prevención & control , Proteínas Bacterianas , Chaperoninas/administración & dosificación , Inflamación/prevención & control , Receptores de LDL/deficiencia , Administración Intranasal , Administración Oral , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/efectos adversos , Aorta Torácica/metabolismo , Aorta Torácica/patología , Arteriosclerosis/inmunología , Arteriosclerosis/patología , Recuento de Células , División Celular/efectos de los fármacos , División Celular/inmunología , Chaperonina 60 , Chaperoninas/efectos adversos , Chaperoninas/inmunología , Colesterol en la Dieta , Citocinas/metabolismo , Femenino , Adyuvante de Freund/administración & dosificación , Inflamación/patología , Ganglios Linfáticos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium/inmunología , Mucosa Nasal/citología , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Receptores de LDL/genética , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/patología
19.
J Clin Invest ; 109(7): 951-60, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11927622

RESUMEN

IL-15, a T cell growth factor, has been linked to exacerbating autoimmune diseases and allograft rejection. To test the hypothesis that IL-15-deficient (IL-15-/-) mice would be protected from T cell-dependent nephritis, we induced nephrotoxic serum nephritis (NSN) in IL-15-/- and wild-type (IL-15+/+) C57BL/6 mice. Contrary to our expectations, IL-15 protects the kidney during this T cell-dependent immunologic insult. Tubular, interstitial, and glomerular pathology and renal function are worse in IL-15-/- mice during NSN. We detected a substantial increase in tubular apoptosis in IL-15-/- kidneys. Moreover, macrophages and CD4 T cells are more abundant in the interstitia and glomeruli in IL-15-/- mice. This led us to identify several mechanisms responsible for heightened renal injury in the absence of IL-15. We now report that IL-15 and the IL-15 receptor (alpha, beta, gamma chains) are constitutively expressed in normal tubular epithelial cells (TECs). IL-15 is an autocrine survival factor for TECs. TEC apoptosis induced with anti-Fas or actinomycin D is substantially greater in IL-15-/- than in wild-type TECs. Moreover, IL-15 decreases the induction of a nephritogenic chemokine, MCP-1, that attracts leukocytes into the kidney during NSN. Taken together, we suggest that IL-15 is a therapeutic for tubulointerstitial and glomerular kidney diseases.


Asunto(s)
Apoptosis/inmunología , Interleucina-15/inmunología , Riñón/lesiones , Nefritis Intersticial/inmunología , Animales , Supervivencia Celular , Quimiocina CCL2/genética , Dactinomicina/farmacología , Regulación hacia Abajo , Células Epiteliales/citología , Células Epiteliales/inmunología , Expresión Génica , Isotipos de Inmunoglobulinas , Interleucina-15/genética , Riñón/citología , Riñón/inmunología , Túbulos Renales/inmunología , Túbulos Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-15 , Receptores de Interleucina-2/genética , Ovinos , Receptor fas/inmunología
20.
Int Immunol ; 14(2): 131-8, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11809732

RESUMEN

Oral tolerance to myelin basic protein (MBP) is an effective antigen-specific method to suppress experimental allergic encephalomyelitis (EAE). Glatiramer acetate [copolymer 1 (Cop1)] is a synthetic copolymer designed to mimic MBP which suppresses EAE, is used parenterally to treat multiple sclerosis (MS) and is being tested orally for efficacy in MS. We investigated the immunologic properties of Cop1 to determine the degree to which its effects were antigen specific using MBP TCR transgenic mice. Immunization of MBP TCR transgenic mice fed Cop1, MBP or MBP Ac1-11 resulted in decreased proliferation, and IL-2, IL-6 and IFN-gamma production, and increased secretion of IL-10 and transforming growth factor (TGF)-beta in Cop1-fed animals. IFN-gamma was decreased, and IL-10 and TGF-beta were increased in non-immunized mice fed Cop1 and stimulated in vitro with MBP. No such effects were observed in ovalbumin TCR transgenic mice. To determine if the effects of Cop1 were specific to MBP TCR-bearing cells, MBP TCR transgenic Rag2(-/-) mice were immunized and re-stimulated in vitro with Cop1. We found a marked increase in IL-4 and similar increases in IL-4 after feeding Cop1. In disease models, feeding Cop1 suppressed EAE in MBP TCR transgenic mice, (PL/J x SJL)F(1) mice, and in myelin oligodendrocyte glycoprotein-induced EAE in NOD mice. Oral Cop1 had no effect on collagen-induced arthritis. These results demonstrate that Cop1 is active orally in an antigen-specific fashion, and may function as an altered peptide ligand for MBP-specific TCR-bearing cells by decreasing pro-inflammatory cytokines (IFN-gamma) and increasing anti-inflammatory cytokines (IL-4, IL-10 and TGF-beta).


Asunto(s)
Citocinas/biosíntesis , Tolerancia Inmunológica , Proteína Básica de Mielina/inmunología , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/fisiología , Animales , Células Cultivadas , Reacciones Cruzadas , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Acetato de Glatiramer , Inmunización , Interleucina-10/biosíntesis , Interleucina-4/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Factor de Crecimiento Transformador beta/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...