Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Aging Brain ; 3: 100066, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36911262

RESUMEN

Background: To investigate how changes in expression of glial genes relate to a progression of Alzheimer's disease (AD) pathology, and how anti-Aß immunotherapy impact these changes, we conducted a transcriptomic analysis for brains from cohorts of 2-, 10-, and 20 month old 3xTg-AD mice, and a cross-sectional study in groups of 20 month-old mice treated with active DNA Aß42 immunization, passive immunotherapy, untreated, and wild-type (wt) controls. Methods: Twenty-four Formalin-Fixed Paraffin-Embedded (FFPE) mouse brain sections were used for the gene expression analyses (nanostring). Adjacent sections from these and additional mouse brains were stained for microglia using antibodies detecting IbaI and Gal3. For a semi-quantitative analysis of increased tau and amyloid pathology with aging and disease progression, a comparison of ELISA results from brains of 12 and 20 months old 3xTg-AD mice were shown. Results: Based on the different comparisons of transcript numbers found the 3xTg-AD age groups with the senescent 20 months old wt control mouse brains, and the 20 months old 3xTg-AD mouse brains with the 20 months old wt control mouse brains, genes were assigned as upregulated due to aging, or due to disease progression, or due to both. The immunohistochemistry of microglia markers revealed that Gal3 might be an important marker for phagocytosing microglia around amyloid plaques. The comparison of the two anti-Aß immunotherapy approaches showed a differential downregulation of inflammatory glial genes. Conclusion: These results are relevant for future clinical trials using active anti-amyloid immunotherapy.

2.
J Neurol Sci ; 446: 120564, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36731358

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia found in the elderly and disease progression is associated with accumulation of Amyloid beta 1-42 (Aß42) in brain. An immune-mediated approach as a preventive intervention to reduce amyloid plaques without causing brain inflammation is highly desirable for future clinical use. Genetic immunization, in which the immunizing agent is DNA encoding Aß42, has great potential because the immune response to DNA delivered into the skin is generally non-inflammatory, and thus differs quantitatively and qualitatively from immune responses elicited by peptides, which are inflammatory with production of IFNγ and IL-17 cytokines by activated T cells. DNA immunization has historically been proven difficult to apply to larger mammals. A potential barrier to use DNA immunization in large mammals is the method for delivery of the DNA antigen. We tested jet injection in mice and rabbits and found good antibody production and safe immune responses (no inflammatory cytokines). We found significant reduction of amyloid plaques and Aß peptides in brains of the DNA Aß42 immunized 3xTg-AD mouse model. This study was designed to optimize DNA delivery for possible testing of the DNA Aß42 vaccine for AD prevention in a clinical trial.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Ratones , Conejos , Animales , Péptidos beta-Amiloides/metabolismo , Placa Amiloide , Fragmentos de Péptidos , Inmunoterapia , Encéfalo/metabolismo , Citocinas , Inmunización/métodos , Inyecciones a Chorro , ADN , Ratones Transgénicos , Modelos Animales de Enfermedad , Mamíferos/genética , Mamíferos/metabolismo
3.
Artículo en Inglés | MEDLINE | ID: mdl-34848502

RESUMEN

BACKGROUND AND OBJECTIVES: Patients with Alzheimer dementia display evidence of amyloid-related neurodegeneration. Our focus was to determine whether such patients also display evidence of a disease-targeting adaptive immune response mediated by CD4+ T cells. To test this hypothesis, we evaluated the CSF immune profiles of patients with Alzheimer clinical syndrome (ACS), who display clinically defined dementia. METHODS: Innate and adaptive immune profiles of patients with ACS were measured using multicolor flow cytometry. CSF-derived CD4+ and CD8+ T-cell receptor repertoire genetics were measured using next-generation sequencing. Brain-specific autoantibody signatures of CSF-derived antibody pools were measured using array technology or ELISA. CSF from similar-age healthy controls (HCs) was used as a comparator cohort. RESULTS: Innate cells were expanded in the CSF of patients with ACS in comparison to HCs, and innate cell expansion increased with age in the patients with ACS, but not HCs. Despite innate cell expansion in the CSF, the frequency of total CD4+ T cells reduced with age in the patients with ACS. T-cell receptor repertoire genetics indicated that T-cell clonal expansion is enhanced, and diversity is reduced in the patients with ACS compared with similar-age HCs. DISCUSSION: Examination of CSF indicates that CD4+ T cell-mediated adaptive immune responses are altered in patients with ACS. Understanding the underlying mechanisms affecting adaptive immunity will help move us toward the goal of slowing cognitive decline.


Asunto(s)
Inmunidad Adaptativa/inmunología , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/inmunología , Autoanticuerpos/líquido cefalorraquídeo , Linfocitos T CD4-Positivos/metabolismo , Inmunidad Innata/inmunología , Anciano , Linfocitos T CD8-positivos/metabolismo , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Síndrome
4.
Neurobiol Dis ; 148: 105221, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33316368

RESUMEN

Alzheimer's disease (AD) has been associated with accumulation of amyloid beta (Aß) peptides in brain, and immunotherapy targeting Aß provides potential for AD prevention. We have used a DNA Aß42 trimer construct for immunization of 3xTg-AD mice and found previously significant reduction of amyloid and tau pathology due to the immunotherapy. We show here that DNA Aß42 immunized 3xTg-AD mice showed better performance in nest building activities and had a higher 24 months survival rate compared to the non-treated AD controls. The analysis of differently expressed genes in brains from 24 months old mice showed significant increases transcript levels between non-immunized AD mice and wild-type controls for genes involved in microglia and astrocyte function, cytokine and inflammatory signaling, apoptosis, the innate and adaptive immune response and are consistent with an inflammatory phenotype in AD. Most of these upregulated genes were downregulated in the DNA Aß42 immunized 3xTg-AD mice due to the vaccine. Transcript numbers for the immediate early genes, Arc, Bdnf, Homer1, Egr1 and cfos, involved in neuronal and neurotransmission pathways which were much lower in the non-immunized 3xTg-AD mice, were restored to wild-type mouse brain levels in DNA Aß42 immunized 3xTg-AD mice indicating positive effects of DNA Aß42 immunotherapy on synapse stability and plasticity. The immune response after immunization is complex, but the multitude of changes after DNA Aß42 immunization shows that this response moves beyond the amyloid hypothesis and into direction of disease prevention.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/inmunología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Fragmentos de Péptidos/inmunología , Transcriptoma/efectos de los fármacos , Vacunas de ADN/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Inmunización , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Estimación de Kaplan-Meier , Longevidad/efectos de los fármacos , Ratones , Ratones Transgénicos , Comportamiento de Nidificación/efectos de los fármacos , Polímeros , Presenilina-1/genética , Bazo/citología , Bazo/metabolismo , Tasa de Supervivencia , Transcriptoma/genética , Proteínas tau/genética
6.
Alzheimers Res Ther ; 10(1): 115, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30454039

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most well-known and most common type of age-related dementia. Amyloid deposition and hyperphosphorylation of tau protein are both pathological hallmarks of AD. Using a triple-transgenic mouse model (3xTg-AD) that develops plaques and tangles in the brain similar to human AD, we provide evidence that active full-length DNA amyloid-ß peptide 1-42 (Aß42) trimer immunization leads to reduction of both amyloid and tau aggregation and accumulation. METHODS: Immune responses were monitored by enzyme-linked immunosorbent assay (ELISA) (antibody production) and enzyme-linked immunospot (cellular activation, cytokine production). Brains from 20-month-old 3x Tg-AD mice that had received DNA Aß42 immunotherapy were compared with brains from age- and gender-matched transgenic Aß42 peptide-immunized and control mice by histology, Western blot analysis, and ELISA. Protein kinase activation and kinase levels were studied in Western blots from mouse hemibrain lysates. RESULTS: Quantitative ELISA showed a 40% reduction of Aß42 peptide and a 25-50% reduction of total tau and different phosphorylated tau molecules in the DNA Aß42 trimer-immunized 3xTg-AD mice compared with nonimmunized 3xTg-AD control animals. Plaque and Aß peptide reductions in the brain were due to the anti-Aß antibodies generated following the immunizations. Reductions of tau were likely due to indirect actions such as less Aß in the brain resulting in less tau kinase activation. CONCLUSIONS: The significance of these findings is that DNA Aß42 trimer immunotherapy targets two major pathologies in AD-amyloid plaques and neurofibrillary tangles-in one vaccine without inducing inflammatory T-cell responses, which carry the danger of autoimmune inflammation, as found in a clinical trial using active Aß42 peptide immunization in patients with AD (AN1792).


Asunto(s)
Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Vacunas contra el Alzheimer/administración & dosificación , Péptidos beta-Amiloides/inmunología , Fragmentos de Péptidos/inmunología , Vacunación/métodos , Proteínas tau/inmunología , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Transgénicos , Fragmentos de Péptidos/metabolismo , Proteínas tau/metabolismo
7.
J Neuroimmunol ; 322: 15-25, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29958693

RESUMEN

Aß immunotherapies with anti-Aß antibody responses have high potential as possible prevention treatment for Alzheimer's disease. We have previously shown that active DNA Aß1-42 immunization via gene gun delivery led to a non-inflammatory immune response resulting in decreased Aß levels in brains of an immunized AD mouse model. To make DNA vaccination more applicable for clinical use, we used here intradermal electroporation. With fine tuning of the electropulse parameters, high antibody levels and low levels of inflammatory cytokines in the cellular immunoassays were observed. Full-length DNA Aß1-42 immunization delivered via electroporation has potential to be used in the clinical setting.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Electroporación/métodos , Inmunización/métodos , Fragmentos de Péptidos/inmunología , Vacunas de ADN/inmunología , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/genética , Animales , Formación de Anticuerpos , Biolística , Relación Dosis-Respuesta Inmunológica , Fenómenos Electromagnéticos , Factores de Transcripción Forkhead/genética , Genes Reporteros , Humanos , Inmunogenicidad Vacunal , Inyecciones Intradérmicas , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Fragmentos de Péptidos/genética , Placa Amiloide/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Vacunas de ADN/administración & dosificación
8.
J Neuroimmunol ; 309: 100-110, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28601278

RESUMEN

BACKGROUND: Laquinimod is an anti-inflammatory agent with good central nervous system (CNS) bioavailability, and neuroprotective and myelorestorative properties. A clinical trial in patients with multiple sclerosis demonstrated that laquinimod significantly reduced loss of brain volume. The cellular substrate or molecular events underlying that treatment effect are unknown. In this study, we aimed to explore laquinimod's potential effects on brain volume, animal behavior, cellular numbers and composition of CNS-intrinsic cells and mononuclear cells within the CNS, amyloid beta (Aß) accumulation and tau phosphorylation in the F1 3xTg-AD/C3H mouse model of Alzheimer's disease. METHODS: Utilizing a dose response study design, four months old F1 3xTg-AD/C3H mice were treated for 10months between ages 4 and 14months with laquinimod (5, 10, or 25mg/kg), or PBS administered by oral gavage. Brain volumes were measured in a 7 Tesla magnetic resonance imager (MRI) at ages 4 and 14months. Behavioral testing included locomotor and rearing activity and the Morris water maze task. Cell numbers and immunophenotypes were assessed by multiparameter flow cytometry. Aß deposition and tau phosphorylation were determined by immunohistochemistry. RESULTS: In the F1 3xTg-AD/C3H animal model of AD, there was no detectable reduction of brain volume over a period of 10months of treatment, as there was not brain atrophy in any of the placebo or treatment groups. Laquinimod had no detectable effects on most neurobehavioral outcomes. The number or composition of CNS intrinsic cells and mononuclear subsets isolated from the CNS were not altered by laquinimod. CONCLUSION: This is the first demonstration that there are no age-associated brain volume changes in the F1 3xTg-AD/C3H mouse model of Alzheimer's disease. Consequently, laquinimod had no effect on that outcome of this study. Most secondary outcomes on the effects of laquinimod on behavior and the number and composition of CNS-intrinsic cells and mononuclear cells within the CNS were also negative.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Modelos Animales de Enfermedad , Quinolonas/uso terapéutico , Animales , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Tamaño de los Órganos/efectos de los fármacos , Quinolonas/farmacología , Resultado del Tratamiento
9.
Alzheimers Res Ther ; 9(1): 30, 2017 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-28441965

RESUMEN

BACKGROUND: Aggregated amyloid-ß peptide 1-42 (Aß42), derived from the cellular amyloid precursor protein, is one of the pathological hallmarks of Alzheimer's disease (AD). Although active immunization against Aß42 peptide was successful in AD mouse models and led to removal of plaques and improved memory, a similar clinical trial in humans (Aß42 peptide immunization with QS-21 adjuvant) was stopped in phase II, when 6% of the treated patients developed encephalitis. Currently ongoing passive immunizations with the injection of preformed monoclonal antibodies against different epitopes within the Aß1-42 peptide, which do not lead to activation of the immune system, have shown some effects in slowing AD pathology. Active DNA Aß42 immunizations administered with the gene gun into the skin are noninflammatory because they activate a different T-cell population (Th2) with different cytokine responses eliciting a different humoral immune response. We present our findings in rhesus macaques that underwent the DNA Aß42 immunization via gene gun delivery into the skin. METHODS: Six rhesus monkeys received two different doses of a DNA Aß42 trimer vaccine. The humoral immune response was analyzed from blood throughout the study, and cellular immune responses were determined in peripheral blood mononuclear cells (PBMCs) after three and six immunizations. RESULTS: DNA Aß42 trimer immunization led to high titer antibody responses in the nonhuman primate (NHP) model. Antibodies generated in the rhesus monkeys following DNA Aß42 immunization detected amyloid plaques consisting of human Aß42 peptide in the brain of the triple-transgenic AD mouse model. T-cell responses showed no interferon (IFN)-γ- and interleukin (IL)-17-producing cells from PBMCs in Enzyme-Linked ImmunoSpot assays after three immunization time points. At six immunization time points, IFN-γ- and IL-17-producing cells were found in immunized animals as well as in control animals and were thus considered nonspecific and not due to the immunization regimen. IFN-γ and IL-17 secretion in response to Aß42 peptide restimulation became undetectable after a 3-month rest period. CONCLUSIONS: Intradermal DNA Aß42 immunization delivered with the gene gun produces a high antibody response in NHPs and is highly likely to be effective and safe in a clinical AD prevention trial in patients.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Antígenos/inmunología , Autoanticuerpos/administración & dosificación , Autoanticuerpos/inmunología , Inmunidad Innata/inmunología , Inmunización/métodos , Fragmentos de Péptidos/inmunología , Vacunas de ADN/administración & dosificación , Animales , Inmunidad Innata/efectos de los fármacos , Inyecciones Intradérmicas , Cinética , Macaca mulatta , Ratones , Resultado del Tratamiento
10.
J Alzheimers Dis ; 57(1): 97-112, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28222511

RESUMEN

A pathological hallmark of Alzheimer's disease (AD) are amyloid plaques in the brain consisting of aggregated amyloid-ß 42 peptide (Aß42) derived from cellular amyloid-ß protein precursor (AßPP). Based on successful experiments in mouse AD models, active immunization with Aß42 peptide and passive immunizations with anti-Aß42 antibodies were started in clinical trials. Active Aß42 peptide immunization in humans had led to an inflammatory autoimmune response, and the trial was stopped. Passive immunizations had shown some effects in slowing AD pathology. Active DNA Aß42 immunizations administered with the gene gun into the skin elicits a different immune response and is non-inflammatory. While in rodents, good responses had been found for this type of immunization, positive results in larger mammals are missing. We present here results from sixteen New Zealand White Rabbits, which underwent intradermal DNA Aß42 immunization via gene gun. The humoral immune response was analyzed from blood throughout the study, and cellular immune responses were determined from spleens at the end of the study. A good anti-Aß antibody response was found in the rabbit model. The T cell response after re-stimulation in cell culture showed no IFNγ producing cells when ELISPOT assays were analyzed from PBMC, but low numbers of IFNγ and IL-17 producing cells were found in ELISPOTS from spleens (both 5 immunizations). Brains from immunized rabbits showed no signs of encephalitis. Based on these results, DNA Aß42 immunization is highly likely to be safe and effective to test in a possible clinical AD prevention trial in patients.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Fragmentos de Péptidos/inmunología , Vacunas de ADN/inmunología , Envejecimiento/inmunología , Envejecimiento/patología , Enfermedad de Alzheimer/inmunología , Animales , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Biolística , Encéfalo/inmunología , Encéfalo/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Epítopos de Linfocito B/inmunología , Femenino , Humanos , Inyecciones Intradérmicas , Masculino , Ratones Transgénicos , Placa Amiloide/metabolismo , Placa Amiloide/patología , Placa Amiloide/prevención & control , Conejos , Linfocitos T/inmunología , Vacunación , Vacunas de ADN/administración & dosificación , alfa-Sinucleína/metabolismo
11.
JAMA Neurol ; 73(7): 867-74, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27135718

RESUMEN

IMPORTANCE: To provide a comprehensive review of knowledge of the genomics of Alzheimer disease (AD) and DNA amyloid ß 42 (Aß42) vaccination as a potential therapy. OBSERVATIONS: Genotype-phenotype correlations of AD are presented to provide a comprehensive appreciation of the spectrum of disease causation. Alzheimer disease is caused in part by the overproduction and lack of clearance of Aß protein. Oligomer Aß, the most toxic species of Aß, causes direct injury to neurons, accompanied by enhanced neuroinflammation, astrocytosis and gliosis, and eventually neuronal loss. The strongest genetic evidence supporting this hypothesis derives from mutations in the amyloid precursor protein (APP) gene. A detrimental APP mutation at the ß-secretase cleavage site linked to early-onset AD found in a Swedish pedigree enhances Aß production, in contrast to a beneficial mutation 2 residues away in APP that reduces Aß production and protects against the onset of sporadic AD. A number of common variants associated with late-onset AD have been identified including apolipoprotein E, BIN1, ABC7, PICALM, MS4A4E/MS4A6A, CD2Ap, CD33, EPHA1, CLU, CR1, and SORL1. One or 2 copies of the apolipoprotein E ε4 allele are a major risk factor for late-onset AD. With DNA Aß42 vaccination, a Th2-type noninflammatory immune response was achieved with a downregulation of Aß42-specific effector (Th1, Th17, and Th2) cell responses at later immunization times. DNA Aß42 vaccination upregulated T regulator cells (CD4+, CD25+, and FoxP3+) and its cytokine interleukin 10, resulting in downregulation of T effectors. CONCLUSIONS AND RELEVANCE: Mutations in APP and PS-1 and PS-2 genes that are associated with early-onset, autosomal, dominantly inherited AD, in addition to the at-risk gene polymorphisms responsible for late-onset AD, all indicate a direct and early role of Aß in the pathogenesis of AD. A translational result of genomic research has been Aß-reducing therapies including DNA Aß42 vaccination as a promising approach to delay or prevent this disease.


Asunto(s)
Enfermedad de Alzheimer/genética , Predisposición Genética a la Enfermedad/genética , Genómica , Mutación/genética , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Estudios de Asociación Genética , Humanos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Presenilina-1/genética , Presenilina-2/genética
12.
PLoS One ; 11(4): e0152720, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27054574

RESUMEN

BACKGROUND: Aquaporin 4 (AQP4) is considered a putative autoantigen in patients with Neuromyelitis optica (NMO), an autoinflammatory disorder of the central nervous system (CNS). HLA haplotype analyses of patients with NMO suggest a positive association with HLA-DRB1* 03:01. We previously showed that the human (h) AQP4 peptide 281-300 is the dominant immunogenic determinant of hAQP4 in the context of HLA-DRB1*03:01. This immunogenic peptide stimulates a strong Th1 and Th17 immune response. AQP4281-300-specific encephalitogenic CD4+ T cells should initiate CNS inflammation that results in a clinical phenotype in HLA-DRB1*03:01 transgenic mice. METHODS: Controlled study with humanized experimental animals. HLA-DRB1*03:01 transgenic mice were immunized with hAQP4281-300, or whole-length hAQP4 protein emulsified in complete Freund's adjuvant. Humoral immune responses to both antigens were assessed longitudinally. In vivo T cell frequencies were assessed by tetramer staining. Mice were followed clinically, and the anterior visual pathway was tested by pupillometry. CNS tissue was examined histologically post-mortem. Flow cytometry was utilized for MHC binding assays and to immunophenotype T cells, and T cell frequencies were determined by ELISpot assay. RESULTS: Immunization with hAQP4281-300 resulted in an in vivo expansion of antigen-specific CD4+ T cells, and an immunoglobulin isotype switch. HLA-DRB1*03:01 TG mice actively immunized with hAQP4281-300, or with whole-length hAQP4 protein were resistant to developing a neurological disease that resembles NMO. Experimental mice show no histological evidence of CNS inflammation, nor change in pupillary responses. Subsequent analysis reveals that a single amino acid substitution from aspartic acid in hAQP4 to glutamic acid in murine (m)AQP4 at position 290 prevents the recognition of hAQP4281-300 by the murine T cell receptor (TCR). CONCLUSION: Induction of a CNS inflammatory autoimmune disorder by active immunization of HLA-DRB1*03:01 TG mice with human hAQP4281-300 will be complex due to a single amino acid substitution. The pathogenic role of T cells in this disorder remains critical despite these observations.


Asunto(s)
Acuaporina 4/genética , Acuaporina 4/inmunología , Cadenas HLA-DRB1/genética , Neuromielitis Óptica/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Acuaporina 4/metabolismo , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Cadenas HLA-DRB1/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Neuromielitis Óptica/genética , Vacunación
13.
Neurobiol Aging ; 36(3): 1274-81, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25725942

RESUMEN

Aging in the immune system results in tendency to proinflammatory responses. Intradermal DNA immunization showed Th2 polarized noninflammatory immune responses. We tested here 18-month-old mice which were immunized with Aß42 peptide, DNA Aß42 trimer, or 2 different prime boost protocols identical to previous experiments. High Aß42 antibody levels were found in aged mice which had received peptide immunizations (900 µg/mL plasma), and in mice which had received peptide prime and DNA boost immunizations (500 µg/mL), compared with antibodies in DNA Aß42 immunized mice with 50 µg/mL. Although we found T-cell proliferation and inflammatory cytokines in mice which had received peptide or prime boost immunization, these were not found in DNA-immunized mice. The results are concordant with proinflammatory responses because of immunosenescence and contraindicate the use of Aß42 peptide immunizations or prime boost immunization protocols for the use in elderly Alzheimer's disease patients. DNA Aß42 immunization only on the other hand does lead to effective levels of antibodies without inflammatory cytokine or T-cell responses in the aged animal model tested.


Asunto(s)
Envejecimiento/genética , Envejecimiento/inmunología , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/inmunología , ADN/inmunología , Inmunoterapia/métodos , Fragmentos de Péptidos/inmunología , Animales , Anticuerpos/sangre , Modelos Animales de Enfermedad , Mediadores de Inflamación/sangre , Ratones Endogámicos , Linfocitos T/inmunología
14.
J Neuroimmunol ; 278: 90-9, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25595257

RESUMEN

The study was designed to test DNA Aß42 immunization in mice as alternative approach for possible active immunotherapy in Alzheimer patients. As results, we found polarized Th2 immune responses, efficient Aß42 antibody levels, and disappearance of antigen specific T cells. In-vivo TNFRSF4/25 antibody co-stimulation enhanced Aß42 specific T cell responses with initial Th2 expansion and subsequent development of Aß42 specific CD4+CD25+Foxp3+ cells. It showed that Th2 biased responses due to gene gun immunizations propagate the development of regulatory T cells. In conclusion, full-length DNA Aß42 immunization into skin results in a regulatory response with minimal risk of inflammation and autoimmunity.


Asunto(s)
Péptidos beta-Amiloides/genética , Anticuerpos/farmacología , Citocinas/metabolismo , ADN/inmunología , Fragmentos de Péptidos/genética , Receptores OX40/inmunología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/inmunología , Animales , Anticuerpos/sangre , Células Cultivadas , ADN/química , Proteínas de Unión al ADN/metabolismo , Femenino , Factores de Transcripción Forkhead/genética , Factor de Transcripción GATA3/metabolismo , Humanos , Inmunoterapia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Fragmentos de Péptidos/inmunología , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo , Linfocitos T Reguladores/citología , Factores de Transcripción/metabolismo
15.
Discov Med ; 15(84): 319-26, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23725605

RESUMEN

One of the challenges of our society is to find a treatment or cure for Alzheimer's disease (AD). AD is the leading form of age-related dementia and with the increase of life expectancy worldwide, the social and economic burden from this disease will increase dramatically. It is a progressive and, in regard to clinical scores, a highly variable disease. AD pathogenesis has been associated with the accumulation, aggregation, and deposition of amyloid beta (Abeta) peptides in the brain. Hallmarks of AD are the amyloid plaques consisting of fibrillar Abeta and neurofibrillary tangles which are intracellular fibrils of hyperphosphorylated tau protein that develop later in this disease. The amyloid cascade hypothesis postulates that Abeta deposition is an initial event in the multifactorial pathogenesis and Abeta deposition may precede AD symptoms in some patients by at least 20 years. Amyloid beta therapy with active and passive immunizations against Abeta has a high possibility to be effective in removing Abeta from brain and might thus prevent the downstream pathology. Since 2000 a number of clinical trials for AD immunotherapy have started, have failed, and are continuing to be pursued. This article will review these clinical trials and ongoing research in this regard.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/inmunología , Vacunas/inmunología , Humanos
17.
Immunotargets Ther ; 2013(2): 105-114, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24926455

RESUMEN

Immunotherapy might provide an effective treatment for Alzheimer disease (AD). A unique feature of AD immunotherapies is that an immune response against a self antigen needs to be elicited without causing adverse autoimmune reactions. Current research is focussed on two possible targets in this regard: One is the inhibition of accumulation and deposition of Amyloid beta 1-42 (Aß42), which is one of the major peptides found in senile plaques and the second target is hyperphosphorylated tau, which forms neurofibrillary tangles inside the nerve cell and shows association with the progression of dementia. Mouse models have shown that immunotherapy targeting Aß42 as well as tau with the respective anti-Aß or anti-tau antibodies can provide significant improvements in these mice. While anti-Aß immunotherapy (active and passive immunizations) is already in several stages of clinical trials, tau based immunizations have been analyzed only in mouse models. Recently, as a significant correlation of progression of dementia and levels of phoshorylated tau was found, high interest has again focussed on further development of tau based therapies. While Aß immunotherapy might delay the onset of AD, immunotherapy targeting tau might provide benefits in later stages of this disease. And last but not least, targeting Aß and tau simultaneously with immunotherapy might provide additional therapeutic effects as these two pathologies are likely synergistic; an approach which has not been tested yet. In this review, we will summarize animal models used to test possible therapies for AD, some of the facts about Aß42 and tau biology, present on overview on halted, ongoing and upcoming clinical trials together with ongoing preclinical studies targeting tau or Aß42.

18.
J Neuroimmunol ; 254(1-2): 63-8, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23036592

RESUMEN

Immunotherapy has the potential to provide a possible treatment therapy to prevent or delay Alzheimer disease. In a clinical trial (AN1792) in which patients received this immunotherapy and received active Aß1-42 peptide immunizations, treatment was stopped when 6% of patients showed signs of meningoencephalitis. Follow up on these patients led to the conclusion that the antibody response was beneficial in removing Aß1-42 from brain but an accompanying inflammatory Th1 T cell response was harmful. As a safe alternative treatment targeting the same self protein, Aß1-42, in brain, we and others are working on a DNA Aß1-42 immunization protocol as the immune response to DNA immunizations differs in many aspects from immunizations with peptide antigens. Because the immune response to DNA vaccination has different kinetics and has a significantly lower antibody production, we evaluated two different prime boost regimens, Aß1-42 DNA prime/Aß1-42 peptide boost and Aß1-42 peptide prime/Aß1-42 DNA boost for their effectiveness in antibody production and possible side effects due to inflammatory T cell responses. While both boost regimes significantly enhanced the specific antibody production with comparable antibody concentrations, the absence of the Aß1-42 T cell response (no proliferation and no cytokine production) is consistent with our previous findings using this DNA Aß1-42 trimer immunization and greatly enhances the safety aspect for possible clinical use.


Asunto(s)
Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/inmunología , Formación de Anticuerpos/inmunología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Vacunación/métodos , Vacunas de ADN/inmunología , Animales , Anticuerpos/sangre , Anticuerpos/metabolismo , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Humanos , Ratones , Ratones Endogámicos , Fragmentos de Péptidos/administración & dosificación , Bazo/citología , Bazo/inmunología , Linfocitos T/clasificación , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Células Th2/metabolismo
19.
Arch Neurol ; 69(9): 1125-31, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22751865

RESUMEN

OBJECTIVE To identify linear determinants of human aquaporin 4 (hAQP4) in the context of HLA-DRB1*03:01. DESIGN In this controlled study with humanized experimental animals, HLA-DRB1*03:01 transgenic mice were immunized with whole-protein hAQP4 emulsified in complete Freund adjuvant. To test T-cell responses, lymph node cells and splenocytes were cultured in vitro with synthetic peptides 20 amino acids long that overlap by 10 amino acids across the entirety of hAQP4. The frequency of interferon γ, interleukin (IL) 17, granulocyte-macrophage colony-stimulating factor, and IL-5-secreting CD4+ T cells was determined by the enzyme-linked immunosorbent sport assay. Quantitative immunofluorescence microscopy was performed to determine whether hAQP4281-300 inhibits the binding of anti-hAQP4 recombinant antibody to surface full-length hAQP4. SETTING Academic neuroimmunology laboratories. SUBJECTS Humanized HLA-DRB1*03:01+/+ H-2b-/- transgenic mice on a B10 background. RESULTS Peptide hAQP4281-300 generated a significantly (P <.01) greater TH1 and TH17 immune response than any of the other linear peptides screened. This 20mer peptide contains 2 dominant immunogenic 15mer peptides. hAQP4284-298 induced predominantly an IL-17 and granulocyte-macrophage colony-stimulating factor TH cell phenotype, whereas hAQP4285-299 resulted in a higher frequency of TH1 cells. hAQP4281-300 did not interfere with recombinant AQP4 autoantibody binding. CONCLUSIONS hAQP4281-330 is the dominant linear immunogenic determinant of hAQP4 in the context of HLA-DRB1*03:01. Within hAQP4281-330 are 2 dominant immunogenic determinants that induce differential TH phenotypes. hAQP4 determinants identified in this study can serve as diagnostic biomarkers in patients with neuromyelitis optica and may facilitate the monitoring of treatment responses to pharmacotherapies.


Asunto(s)
Acuaporina 4/genética , Epítopos de Linfocito T/genética , Cadenas HLA-DRB1/genética , Epítopos Inmunodominantes/genética , Neuromielitis Óptica/inmunología , Animales , Acuaporina 4/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Cadenas HLA-DRB1/metabolismo , Epítopos Inmunodominantes/inmunología , Epítopos Inmunodominantes/metabolismo , Ratones , Ratones Transgénicos , Neuromielitis Óptica/diagnóstico , Neuromielitis Óptica/genética , Neuromielitis Óptica/metabolismo
20.
Transl Neurosci ; 3(4): 307-313, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23741624

RESUMEN

As a neurodegenerative disorder, Alzheimer disease (AD) is the most common form of dementia found in the aging population. Immunotherapy with passive or active immunizations targeting amyloid beta (Aß) build-up in the brain may provide a possible treatment option and may help prevent AD from progressing. A number of passive immunizations with anti-Aß42 antibodies are in different phases of clinical trials. One active immunization approach, AN-1792, was stopped after the development of autoimmune encephalitis in 6% of patients and a second one, CAD106, in which a small Aß epitope is used, is currently in safety and tolerability studies. Besides active immunizations with proteins or peptides, active immunizations using DNA which codes for the protein against which the immune response will be directed, so called genetic immunizations, provide additional safety as the immune response in DNA immunizations differs quantitatively and qualitatively from the response elicited by peptide immunizations. In this review, we summarize our data using DNA Aß42 immunizations in mouse models and discuss the results together with the results presented by other groups working on a DNA vaccine as treatment option for AD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...