Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
J Biol Chem ; 296: 100334, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33508322

RESUMO

Systemic light chain (AL) amyloidosis is a fatal protein misfolding disease in which excessive secretion, misfolding, and subsequent aggregation of free antibody light chains eventually lead to deposition of amyloid plaques in various organs. Patient-specific mutations in the antibody VL domain are closely linked to the disease, but the molecular mechanisms by which certain mutations induce misfolding and amyloid aggregation of antibody domains are still poorly understood. Here, we compare a patient VL domain with its nonamyloidogenic germline counterpart and show that, out of the five mutations present, two of them strongly destabilize the protein and induce amyloid fibril formation. Surprisingly, the decisive, disease-causing mutations are located in the highly variable complementarity determining regions (CDRs) but exhibit a strong impact on the dynamics of conserved core regions of the patient VL domain. This effect seems to be based on a deviation from the canonical CDR structures of CDR2 and CDR3 induced by the substitutions. The amyloid-driving mutations are not necessarily involved in propagating fibril formation by providing specific side chain interactions within the fibril structure. Rather, they destabilize the VL domain in a specific way, increasing the dynamics of framework regions, which can then change their conformation to form the fibril core. These findings reveal unexpected influences of CDR-framework interactions on antibody architecture, stability, and amyloid propensity.


Assuntos
Amiloide/ultraestrutura , Regiões Determinantes de Complementaridade/genética , Amiloidose de Cadeia Leve de Imunoglobulina/genética , Placa Amiloide/genética , Sequência de Aminoácidos/genética , Amiloide/genética , Amiloide/imunologia , Proteínas Amiloidogênicas/genética , Proteínas Amiloidogênicas/imunologia , Proteínas Amiloidogênicas/ultraestrutura , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/ultraestrutura , Humanos , Amiloidose de Cadeia Leve de Imunoglobulina/imunologia , Amiloidose de Cadeia Leve de Imunoglobulina/metabolismo , Mutação/genética , Placa Amiloide/imunologia , Placa Amiloide/patologia , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/imunologia , Agregação Patológica de Proteínas/patologia , Conformação Proteica , Dobramento de Proteína
2.
J Neuropathol Exp Neurol ; 79(11): 1147-1162, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-33011810

RESUMO

ß-Amyloid (Aß) plaques can trigger chronic inflammation in the cellular environment that recruits infiltrating macrophages during the course of Alzheimer disease (AD). Activated macrophages release pro-inflammatory cytokines that increase neurotoxicity associated with AD. A major impediment to investigating neuroinflammation involving macrophage activity is the inability to discriminate resident microglial macrophages (mMϕ) from hematogenous macrophages (hMϕ), as they are morphologically and phenotypically similar when activated. To distinguish between mMϕ and hMϕ and to determine their respective roles in chronic inflammation associated with the progression of amyloidosis, we used lys-EGFP-ki transgenic mice that express enhanced green fluorescent protein in hMϕ, but not in mMϕ. These mice were crossed with 5XFAD mice. The offspring demonstrated robust AD pathology and enabled visual discrimination of mMϕ from hMϕ. Mutant mice demonstrated robust increases in Aß1-42, area of Aß plaques, gliosis and deficits in spatial learning by age 5 months. The time-course of Aß accumulation, paralleled by the accumulation of hMϕ around Aß plaques, was more robust in female compared with male mice and preceded behavioral changes. Thus, the accumulation of infiltrating hMϕ around Aß plaques was age- and sex-dependent and preceded cognitive impairment.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Macrófagos/patologia , Placa Amiloide/patologia , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/imunologia , Modelos Animais de Doenças , Feminino , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Transgênicos , Placa Amiloide/imunologia
3.
Front Immunol ; 11: 1029, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32582162

RESUMO

Genetic, clinical, biochemical and histochemical data indicate a crucial involvement of inflammation in Alzheimer's disease (AD), but harnessing the immune system to cure or prevent AD has so far proven difficult. Clarifying the cellular heterogeneity and signaling pathways associated with the presence of the AD hallmarks beta-amyloid and tau in the brain, would help to identify potential targets for therapy. While much attention has been so far devoted to microglia and their homeostatic phagocytic activity, additional cell types and immune functions might be affected in AD. Beyond microglia localized in the brain parenchyma, additional antigen-presenting cell (APC) types might be affected by beta-amyloid toxicity. Here, we investigated potential immunomodulatory properties of oligomeric species of beta-amyloid-peptide (Aß) on microglia and putative APCs. We performed a comprehensive characterization of time- and pathology-dependent APC and T-cell alterations in a model of AD-like brain beta-amyloidosis, the APP-PS1-dE9 mouse model. We show that the deposition of first beta-amyloid plaques is accompanied by a significant reduction in MHC class II surface levels on brain APCs. Furthermore, taking advantage of customized in vitro systems and RNAseq, we demonstrate that a preparation containing various forms of oligomeric Aß1-42 inhibits antigen presentation by altering the transcription of key immune mediators in dendritic cells. These results suggest that, beyond their neurotoxic effects, certain oligomeric Aß forms can act as immunomodulatory agents on cerebral APCs and interfere with brain antigen presentation. Impaired brain immune surveillance might be one of the factors that facilitate Aß and tau spreading in AD.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/imunologia , Apresentação de Antígeno , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Amiloidose/genética , Amiloidose/imunologia , Amiloidose/metabolismo , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/patologia , Encéfalo/imunologia , Encéfalo/patologia , Modelos Animais de Doenças , Expressão Gênica , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunomodulação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fragmentos de Peptídeos/genética , Placa Amiloide/imunologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia
4.
Proc Natl Acad Sci U S A ; 117(12): 6844-6854, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32144141

RESUMO

Chronic inflammation during Alzheimer's disease (AD) is most often attributed to sustained microglial activation in response to amyloid-ß (Aß) plaque deposits and cell death. However, cytokine release and microgliosis are consistently observed in AD transgenic animal models devoid of such pathologies, bringing into question the underlying processes that may be at play during the earliest AD-related immune response. We propose that this plaque-independent inflammatory reaction originates from neurons burdened with increasing levels of soluble and oligomeric Aß, which are known to be the most toxic amyloid species within the brain. Laser microdissected neurons extracted from preplaque amyloid precursor protein (APP) transgenic rats were found to produce a variety of potent immune factors, both at the transcript and protein levels. Neuron-derived cytokines correlated with the extent of microglial activation and mobilization, even in the absence of extracellular plaques and cell death. Importantly, we identified an inflammatory profile unique to Aß-burdened neurons, since neighboring glial cells did not express similar molecules. Moreover, we demonstrate within disease-vulnerable regions of the human brain that a neuron-specific inflammatory response may precede insoluble Aß plaque and tau tangle formation. Thus, we reveal the Aß-burdened neuron as a primary proinflammatory agent, implicating the intraneuronal accumulation of Aß as a significant immunological component in the AD pathogenesis.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/patologia , Inflamação/patologia , Neurônios/imunologia , Placa Amiloide/patologia , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Amiloidose , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Neurônios/metabolismo , Neurônios/patologia , Placa Amiloide/imunologia , Placa Amiloide/metabolismo , Ratos , Ratos Transgênicos
5.
Biomacromolecules ; 20(11): 4208-4217, 2019 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-31600059

RESUMO

Alzheimer's disease (AD) is a primary neurological disease with no effective cure. A hallmark of AD is the presence of intracellular tangles and extracellular plaques derived from the aberrant aggregation of tau- and beta-amyloid (Aß). Aß presents in the brain as well as in cerebrospinal fluid and the circulation, and Aß toxicity has been attributed to amyloidosis and inflammation, among other causes. In this study, the effects of the plasma protein corona have been investigated with regard to the blood cell association and cytokine secretion of oligomeric (Aßo) and fibrillar Aß1-42(Aßf), two major forms of the peptide aggregates. Aßo displayed little change in membrane association in whole blood or washed blood (i.e., cells in the absence of plasma proteins) at 37 °C, while Aßf showed a clear preference for binding with all cell types sans plasma proteins. Immune cells exposed to Aßo, but not to Aßf, resulted in significant expression of cytokines IL-6 and TNF measured in real-time by a localized surface plasmon resonance sensor. These observations indicate greater immune cell association and cytokine stimulation of Aßo than Aßf and shed new light on the contrasting toxicities of Aßo and Aßf resulting from their differential capacities in acquiring a plasma protein corona. These results further implicate a close connection between Aß amyloidosis and immunopathology in AD.


Assuntos
Doença de Alzheimer/imunologia , Amiloide/imunologia , Fragmentos de Peptídeos/química , Placa Amiloide/imunologia , Coroa de Proteína/química , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Amiloide/química , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/imunologia , Encéfalo/imunologia , Encéfalo/patologia , Citocinas/biossíntese , Citocinas/química , Humanos , Microglia/imunologia , Neurônios/imunologia , Neurônios/patologia , Fragmentos de Peptídeos/imunologia , Placa Amiloide/tratamento farmacológico , Placa Amiloide/patologia , Coroa de Proteína/imunologia , Transporte Proteico/imunologia
7.
Sci Rep ; 8(1): 9813, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29955078

RESUMO

Impairment of neuronal proteostasis is a hallmark of Alzheimer's and other neurodegenerative diseases. However, the underlying molecular mechanisms leading to pathogenic protein aggregation, and the role of secretory chaperone proteins in this process, are poorly understood. We have previously shown that the neural-and endocrine-specific secretory chaperone 7B2 potently blocks in vitro fibrillation of Aß42. To determine whether 7B2 can function as a chaperone in vivo, we measured plaque formation and performed behavioral assays in 7B2-deficient mice in an hAPPswe/PS1dE9 Alzheimer's model mouse background. Surprisingly, immunocytochemical analysis of cortical levels of thioflavin S- and Aß-reactive plaques showed that APP mice with a partial or complete lack of 7B2 expression exhibited a significantly lower number and burden of thioflavin S-reactive, as well as Aß-immunoreactive, plaques. However, 7B2 knockout did not affect total brain levels of either soluble or insoluble Aß. While hAPP model mice performed poorly in the Morris water maze, their brain 7B2 levels did not impact performance. Since 7B2 loss reduced amyloid plaque burden, we conclude that brain 7B2 can impact Aß disposition in a manner that facilitates plaque formation. These results are reminiscent of prior findings in hAPP model mice lacking the ubiquitous secretory chaperone clusterin.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Proteína Secretora Neuroendócrina 7B2/deficiência , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Animais , Benzotiazóis/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Clusterina/metabolismo , Modelos Animais de Doenças , Feminino , Genótipo , Heterozigoto , Humanos , Memória , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Secretora Neuroendócrina 7B2/genética , Proteína Secretora Neuroendócrina 7B2/metabolismo , Placa Amiloide/imunologia , Placa Amiloide/fisiopatologia , Solubilidade , Transgenes
8.
Molecules ; 23(5)2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29751505

RESUMO

Passive immunotherapy has emerged as a very promising approach for the treatment of Alzheimer's disease and other neurodegenerative disorders, which are characterized by the misfolding and deposition of amyloid peptides. On the basis of the amyloid hypothesis, the majority of antibodies in clinical development are directed against amyloid ß (Aß), the primary amyloid component in extracellular plaques. This review focuses on the current status of Aß antibodies in clinical development, including their characteristics and challenges that came up in clinical trials with these new biological entities (NBEs). Emphasis is placed on the current view of common side effects observed with passive immunotherapy, so-called amyloid-related imaging abnormalities (ARIAs), and potential ways to overcome this issue. Among these new ideas, a special focus is placed on molecules that are directed against post-translationally modified variants of the Aß peptide, an emerging approach for development of new antibody molecules.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/antagonistas & inibidores , Anticorpos Monoclonais/uso terapêutico , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Ensaios Clínicos como Assunto , Citotoxicidade Imunológica , Diagnóstico por Imagem , Modelos Animais de Doenças , Descoberta de Drogas , Humanos , Imunoterapia , Placa Amiloide/tratamento farmacológico , Placa Amiloide/imunologia , Placa Amiloide/metabolismo , Processamento de Proteína Pós-Traducional
9.
Neurodegener Dis ; 18(1): 49-56, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29402847

RESUMO

BACKGROUND: Strong genetic and epidemiological evidence points to a crucial role of the immune system in the development of Alzheimer disease (AD). CD3+ T lymphocytes have been described in brains of postmortem AD patients and in transgenic models of AD-like cerebral amyloidosis and tau pathology. However, the occurrence of T cells in AD brains is still controversial; furthermore, the relationship between T cells and hallmarks of AD pathology (amyloid plaques and neurofibrillary tangles) remains to be established. OBJECTIVES: We have studied the occurrence of T cells in postmortem hippocampi and mid frontal gyrus (MFG) samples of AD patients (Braak stage V-VI) and nondemented control subjects and correlated it with amyloid and tau pathology burden. METHODS: Confocal microscopy and bright-field immunohistochemistry were used to identify brain-associated T cells. Extravascular CD3+ T cells were quantified and compared to nondemented controls. In addition, numbers of extravascular CD3+ T cells were correlated with amyloid (6E10 staining) and tau pathology (AT8 staining) in the same sections. RESULTS: Several CD3+, extravascular T cells were observed in the brains of AD patients, mostly of the CD8+ subtype. AD hippocampi harbored significantly increased numbers of extravascular CD3+ T cells compared to nondemented controls. CD3+ T cells significantly correlated with tau pathology but not with amyloid plaques in AD samples. CONCLUSIONS: Our data support the notion of T-cell occurrence in AD brains and suggest that, in advanced stages of AD, T-cell extravasation is driven by tau-related neurodegenerative changes rather than by cerebral amyloidosis. T cells could be crucial for driving the amyloid-independent phase of the AD pathology.


Assuntos
Doença de Alzheimer/imunologia , Encéfalo/imunologia , Complexo CD3/imunologia , Placa Amiloide/imunologia , Linfócitos T/imunologia , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Complexo CD3/metabolismo , Feminino , Humanos , Masculino , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Linfócitos T/metabolismo
10.
Sci Transl Med ; 10(422)2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29298867

RESUMO

Systemic amyloidosis is a fatal disorder caused by pathological extracellular deposits of amyloid fibrils that are always coated with the normal plasma protein, serum amyloid P component (SAP). The small-molecule drug, miridesap, [(R)-1-[6-[(R)-2-carboxy-pyrrolidin-1-yl]-6-oxo-hexanoyl]pyrrolidine-2-carboxylic acid (CPHPC)] depletes circulating SAP but leaves some SAP in amyloid deposits. This residual SAP is a specific target for dezamizumab, a fully humanized monoclonal IgG1 anti-SAP antibody that triggers immunotherapeutic clearance of amyloid. We report the safety, pharmacokinetics, and dose-response effects of up to three cycles of miridesap followed by dezamizumab in 23 adult subjects with systemic amyloidosis (ClinicalTrials.gov identifier: NCT01777243). Amyloid load was measured scintigraphically by amyloid-specific radioligand binding of 123I-labeled SAP or of 99mTc-3,3-diphosphono-1,2-propanodicarboxylic acid. Organ extracellular volume was measured by equilibrium magnetic resonance imaging and liver stiffness by transient elastography. The treatment was well tolerated with the main adverse event being self-limiting early onset rashes after higher antibody doses related to whole body amyloid load. Progressive dose-related clearance of hepatic amyloid was associated with improved liver function tests. 123I-SAP scintigraphy confirmed amyloid removal from the spleen and kidneys. No adverse cardiac events attributable to the intervention occurred in the six subjects with cardiac amyloidosis. Amyloid load reduction by miridesap treatment followed by dezamizumab has the potential to improve management and outcome in systemic amyloidosis.


Assuntos
Amiloidose/tratamento farmacológico , Anticorpos/uso terapêutico , Componente Amiloide P Sérico/imunologia , Amiloidose/imunologia , Anticorpos/imunologia , Humanos , Imageamento por Ressonância Magnética , Placa Amiloide/tratamento farmacológico , Placa Amiloide/imunologia , Cintilografia
11.
ASN Neuro ; 9(4): 1759091417724915, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28817954

RESUMO

Genetic and pathologic data suggest that amyloid beta (Aß), produced by processing of the amyloid precursor protein, is a major initiator of Alzheimer's disease (AD). To gain new insights into Aß modulation, we sought to harness the power of the coevolution between the neurotropic parasite Toxoplasma gondii and the mammalian brain. Two prior studies attributed Toxoplasma-associated protection against Aß to increases in anti-inflammatory cytokines (TGF-ß and IL-10) and infiltrating phagocytic monocytes. These studies only used one Toxoplasma strain making it difficult to determine if the noted changes were associated with Aß protection or simply infection. To address this limitation, we infected a third human amyloid precursor protein AD mouse model (J20) with each of the genetically distinct, canonical strains of Toxoplasma (Type I, Type II, or Type III). We then evaluated the central nervous system (CNS) for Aß deposition, immune cell responses, global cytokine environment, and parasite burden. We found that only Type II infection was protective against Aß deposition despite both Type II and Type III strains establishing a chronic CNS infection and inflammatory response. Compared with uninfected and Type I-infected mice, both Type II- and Type III-infected mice showed increased numbers of CNS T cells and microglia and elevated pro-inflammatory cytokines, but neither group showed a >2-fold elevation of TGF-ß or IL-10. These data suggest that we can now use our identification of protective (Type II) and nonprotective (Type III) Toxoplasma strains to determine what parasite and host factors are linked to decreased Aß burden rather than simply with infection.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/metabolismo , Placa Amiloide/imunologia , Toxoplasmose Animal/imunologia , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Hipocampo/imunologia , Hipocampo/patologia , Humanos , Macrófagos/imunologia , Macrófagos/patologia , Camundongos Transgênicos , Microglia/imunologia , Microglia/patologia , Neuroproteção/fisiologia , Placa Amiloide/patologia , Especificidade da Espécie , Linfócitos T/imunologia , Linfócitos T/patologia , Toxoplasma , Toxoplasmose Animal/patologia
12.
Neurobiol Aging ; 55: 115-122, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28434692

RESUMO

Alzheimer's disease (AD) is strongly associated with microglia-induced neuroinflammation. Particularly, Aß plaque-associated microglia take on an "activated" morphology. However, the function and phenotype of these Aß plaque-associated microglia are not well understood. We show hyperreactivity of Aß plaque-associated microglia upon systemic inflammation in transgenic AD mouse models (i.e., 5XFAD and APP23). Gene expression profiling of Aß plaque-associated microglia (major histocompatibility complex II+ microglia) isolated from 5XFAD mice revealed a proinflammatory phenotype. The upregulated genes involved in the biological processes (gene ontology terms) included: "immune response to external stimulus" such as Axl, Cd63, Egr2, and Lgals3, "cell motility", such as Ccl3, Ccl4, Cxcr4, and Sdc3, "cell differentiation", and "system development", such as St14, Trpm1, and Spp1. In human AD tissue with similar Braak stages, expression of phagocytic markers and AD-associated genes, including HLA-DRA, APOE, AXL, TREM2, and TYROBP, was higher in laser-captured early-onset AD (EOAD) plaques than in late-onset AD plaques. Interestingly, the nonplaque parenchyma of both EOAD and late-onset AD brains, the expression of above-mentioned markers were similarly low. Here, we provide evidence that Aß plaque-associated microglia are hyperreactive in their immune response and phagocytosis in the transgenic AD mice as well as in EOAD brain tissue. We suggest that Aß plaque-associated microglia are the primary source of neuroinflammation related to AD pathology.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Microglia/imunologia , Placa Amiloide/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Animais , Apolipoproteínas E , Encéfalo/imunologia , Diferenciação Celular/genética , Movimento Celular/genética , Movimento Celular/imunologia , Modelos Animais de Doenças , Feminino , Expressão Gênica , Humanos , Inflamação/genética , Inflamação/imunologia , Masculino , Glicoproteínas de Membrana , Camundongos Transgênicos , Pessoa de Meia-Idade , Fagocitose/genética , Fagocitose/imunologia , Proteínas Proto-Oncogênicas , Receptores Proteína Tirosina Quinases , Receptores Imunológicos , Receptor Tirosina Quinase Axl
13.
J Alzheimers Dis ; 55(1): 101-113, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27662314

RESUMO

BACKGROUND: The immune system is increasingly mentioned as a potential target for Alzheimer's disease (AD) treatment. OBJECTIVE: In the present pilot study, the effect of (neuro)inflammation on amyloidopathy was investigated in the marmoset monkey, which has potential as an AD animal model due to its natural cerebral amyloidosis similar to humans. METHODS: Six adult/aged marmosets (Callithrix jacchus) were intracranial injected with amyloid-beta (Aß) fibrils at three cortical locations in the right hemisphere. Additionally, in half of the monkeys, lipopolysaccharide (LPS) was co-injected with the Aß fibrils and injected in the other hemisphere without Aß fibrils. The other three monkeys received phosphate buffered saline instead of LPS, as a control for the inflammatory state. The effect of inflammation on amyloidopathy was also investigated in an additional monkey that suffered from chronic inflammatory wasting syndrome. Mirror histology sections were analyzed to assess amyloidopathy and immune reaction, and peripheral blood for AD biomarker expression. RESULTS: All LPS-injected monkeys showed an early AD immune blood cell expression profile on CD95 and CD45RA. Two out of three monkeys injected with Aß and LPS and the additional monkey, suffering from chronic inflammation, developed plaques. None of the controls, injected with Aß only, developed any plaques. CONCLUSION: This study shows the importance of immune modulation on the susceptibility for amyloidosis, a hallmark of AD, which offers new perspectives for disease modifying approaches in AD.


Assuntos
Amiloidose/imunologia , Córtex Cerebral/imunologia , Inflamação/fisiopatologia , Doença de Alzheimer , Peptídeos beta-Amiloides , Amiloidose/sangue , Amiloidose/diagnóstico por imagem , Amiloidose/patologia , Animais , Biomarcadores/sangue , Callithrix , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/patologia , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/diagnóstico por imagem , Inflamação/patologia , Antígenos Comuns de Leucócito/sangue , Lipopolissacarídeos , Masculino , Microglia/imunologia , Microglia/patologia , Projetos Piloto , Placa Amiloide/diagnóstico por imagem , Placa Amiloide/imunologia , Placa Amiloide/patologia , Doença de Emaciação Crônica/sangue , Doença de Emaciação Crônica/diagnóstico por imagem , Doença de Emaciação Crônica/imunologia , Doença de Emaciação Crônica/patologia , Receptor fas/sangue
14.
Curr Alzheimer Res ; 14(3): 295-302, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27829339

RESUMO

BACKGROUND: The ever-increasing number of people living with Alzheimer's disease urges to develop more effective therapies. Despite considerable success, anti-Alzheimer immunotherapy still faces the challenge of intracerebral and intracellular delivery. This work introduces in situ production of anti-amyloid beta (Aß) antibody after intracerebral injection of PEG-PAsp(DET)/mRNA polyplexes as a novel immunotherapy approach and a safer alternative compared to high systemic antibodies doses or administration of adenovirus encoding anti- Aß antibodies. METHODS: We used mRNA encoding three different Aß-specific scFV with a secretion signal for passive immunotherapy. scFv contained a 6xHis-tag for immuno-detection. The secretion signal from IL2 (IL2ss) was added to allow extracellular engagement of senile plaques. Aß affinity of scFv was measured by surface plasmon resonance. To allow intracellular delivery, scFv were administered as polyplexes formed with our smart copolymer polyethylene glycol-poly[N'-[N-(2-aminoethyl)-2-aminoethyl] aspartamide] [PEG-PAsp (DET)]. We evaluated scFv expression in cellulo by Western blot and ELISA, their ability to disaggregate amyloid aggregates by thioflavine T assay. Moreover, in vivo expression and therapeutic activity were evaluated in a murine amyloidosis model, by anti-6xHis-tag ELISA and anti- Aß ELISA, respectively. RESULTS: The selected anti-amyloid beta scFv showed affinity towards Aß and disaggregated Aß fibers in vitro. Whereas both DNA and mRNA transfection led to scFV expression in cancer cells, only mRNA led to detectable scFv expression in primary neurons. In addition, the use of IL2ss increased by 3.4-fold scFv secretion by primary neurons over mRNA polyplexes devoid of secretion signal. In vivo, a 3 to 11- fold of intracranial scFv levels was measured for mRNA compared to DNA polyplexes and higher in vivo scFv levels were obtained with mRNA containing IL2ss over non-secreted mRNA. Intracranial injection of anti-Aß mRNA polyplexes with IL2ss resulted in 40 % Aß decrease in an acute amyloidosis model; with no decrease detected with control scFv mRNA nor DNA polyplexes. However, no Aß decrease was detected in a more challenging transgenic model of Alzheimer's disease. CONCLUSION: Our results introduce a concerted approach not only for Alzheimer's disease treatment but also for immunotherapy against neurological diseases. The effectivity of our platform required the intracranial delivery of anti-Aß scFv as mRNA not DNA, as mRNA with an IL2ss secretion sequence to favor engagement of Aß in the amyloidosis model, complexation with a smart copolymer for efficient transfection of primary neurons and to achieve detectable mRNA expression in the brain during 48h. Amyloid burden decrease in an acute amyloidosis model was only achieved when these three factors (mRNA coding scFv, smart copolymer, IL2ss) were integrated into a single formulation.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Encéfalo/imunologia , Imunização Passiva , RNA Mensageiro/administração & dosagem , Anticorpos de Cadeia Única/biossíntese , Doença de Alzheimer/imunologia , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/imunologia , Placa Amiloide/imunologia , Placa Amiloide/terapia , Polietilenoglicóis , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia
15.
J Alzheimers Dis ; 52(1): 243-69, 2016 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-27031470

RESUMO

Alzheimer's disease (AD) is a degenerative and irreversible disorder whose progressiveness is dependent on age. It is histopathologically characterized by the massive accumulation of insoluble forms of tau and amyloid-ß (Aß) asneurofibrillary tangles and neuritic plaques, respectively. Many studies have documented that these two polypeptides suffer several posttranslational modifications employing postmortem tissue sections from brains of patients with AD. In order to elucidate the molecular mechanisms underlying the posttranslational modifications of key players in this disease, including Aß and tau, several transgenic mouse models have been developed. One of these models is the 3×Tg-AD transgenic mouse, carrying three transgenes encoding APPSWE, S1M146V, and TauP301L proteins. To further characterize this transgenicmouse, we determined the accumulation of fibrillar Aß as a function of age in relation to the hyperphosphorylation patterns of TauP301L at both its N- and C-terminus in the hippocampal formation by immunofluorescence and confocal microscopy. Moreover, we searched for the expression of activated protein kinases and mediators of inflammation by western blot of wholeprotein extracts from hippocampal tissue sections since 3 to 28 months as well. Our results indicate that the presence of fibrillar Aß deposits correlates with a significant activation of astrocytes and microglia in subiculum and CA1 regions of hippocampus. Accordingly, we also observed a significant increase in the expression of TNF-α associated to neuritic plaques and glial cells. Importantly, there is an overexpression of the stress activated protein kinases SAPK/JNK and Cdk-5 in pyramidal neurons, which might phosphorylate several residues at the C-terminus of TauP301L. Therefore, the accumulation of Aß oligomers results in an inflammatory environment that upregulates kinases involved in hyperphosphorylation of TauP301L polypeptide.


Assuntos
Envelhecimento/imunologia , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/metabolismo , Hipocampo/imunologia , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Modelos Animais de Doenças , Feminino , Hipocampo/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuroglia/imunologia , Neuroglia/patologia , Fosforilação/imunologia , Placa Amiloide/imunologia , Placa Amiloide/patologia , Presenilina-1/genética , Presenilina-1/metabolismo , Células Piramidais/imunologia , Células Piramidais/patologia , Proteínas tau/genética
16.
Prog Histochem Cytochem ; 51(1): 1-8, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26851150

RESUMO

This review discussed the importance of mutated tau, amyloid and neuroinflammatory factors and microglia in Alzheimer disease. In particular tau, CD4 and TNF alpha were included in the review and the colocalizations of these factors were highlighted. It is important to realize the Alzheimer disease may result from the interactions of these factors. Some of these factors may coexist at the same region and at the same time e.g. mutated tau and amyloid in plaques. A summary scheme of etiology leading to the disease was included.


Assuntos
Doença de Alzheimer/genética , Proteínas Amiloidogênicas/genética , Mutação , Placa Amiloide/genética , Proteínas tau/genética , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Proteínas Amiloidogênicas/imunologia , Animais , Encéfalo/imunologia , Encéfalo/patologia , Antígenos CD4/genética , Antígenos CD4/imunologia , Morte Celular , Expressão Gênica , Humanos , Inflamação , Microglia/imunologia , Microglia/patologia , Placa Amiloide/diagnóstico , Placa Amiloide/imunologia , Placa Amiloide/patologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Proteínas tau/imunologia
19.
Nat Med ; 22(2): 135-7, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26779813

RESUMO

Systemic immune suppression may curtail the ability to mount the protective, cell-mediated immune responses that are needed for brain repair. By using mouse models of Alzheimer's disease (AD), we show that immune checkpoint blockade directed against the programmed death-1 (PD-1) pathway evokes an interferon (IFN)-γ-dependent systemic immune response, which is followed by the recruitment of monocyte-derived macrophages to the brain. When induced in mice with established pathology, this immunological response leads to clearance of cerebral amyloid-ß (Aß) plaques and improved cognitive performance. Repeated treatment sessions were required to maintain a long-lasting beneficial effect on disease pathology. These findings suggest that immune checkpoints may be targeted therapeutically in AD.


Assuntos
Doença de Alzheimer/imunologia , Anticorpos/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Memória/efeitos dos fármacos , Placa Amiloide/imunologia , Receptor de Morte Celular Programada 1/imunologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/efeitos dos fármacos , Peptídeos beta-Amiloides/imunologia , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/imunologia , Encéfalo/patologia , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Interferon gama/efeitos dos fármacos , Interferon gama/imunologia , Macrófagos/imunologia , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Placa Amiloide/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Análise de Sequência de RNA , Transdução de Sinais/efeitos dos fármacos
20.
Acta Neuropathol ; 130(5): 699-711, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26433971

RESUMO

Immunotherapy is a promising strategy for the treatment of Alzheimer's disease (AD). Antibodies directed against Amyloid Beta (Aß) are able to successfully clear plaques and reverse cognitive deficits in mouse models. Excitement towards this approach has been tempered by high profile failures in the clinic, one key issue has been the development of inflammatory side effects in the brain (ARIAs). New antibodies are entering the clinic for Alzheimer's disease; therefore, it is important to learn all we can from the current generation. In this study, we directly compared 3 clinical candidates in the same pre-clinical model, with the same effector function, for their ability to clear plaques and induce inflammation in the brain. We produced murine versions of the antibodies: Bapineuzumab (3D6), Crenezumab (mC2) and Gantenerumab (chGantenerumab) with an IgG2a constant region. 18-month transgenic APP mice (Tg2576) were injected bilaterally into the hippocampus with 2 µg of each antibody or control. After 7 days, the mice tissue was analysed for clearance of plaques and neuroinflammation by histology and biochemical analysis. 3D6 was the best binder to plaques and in vitro, whilst mC2 bound the least strongly. This translated into 3D6 effectively clearing plaques and reducing the levels of insoluble Aß, whilst chGantenerumab and mC2 did not. 3D6 caused a significant increase in the levels of pro-inflammatory cytokines IL-1ß and TNFα, and an associated increase in microglial expression of CD11B and CD68. chGantenerumab increased pro-inflammatory cytokines and microglial activation, but minimal changes in CD68, as an indicator of phagocytosis. Injection of mC2 did not cause any significant inflammatory changes. Our results demonstrate that the ability of an antibody to clear plaques and induce inflammation is dependent on the epitope and affinity of the antibody.


Assuntos
Peptídeos beta-Amiloides/imunologia , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais/farmacologia , Fatores Imunológicos/farmacologia , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/genética , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Antígeno CD11b/metabolismo , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/imunologia , Hipocampo/patologia , Humanos , Interleucina-1beta/metabolismo , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/patologia , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , Placa Amiloide/tratamento farmacológico , Placa Amiloide/imunologia , Placa Amiloide/patologia , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA