Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
1.
Nature ; 615(7953): 668-677, 2023 03.
Article in English | MEDLINE | ID: mdl-36890231

ABSTRACT

Extracellular deposition of amyloid-ß as neuritic plaques and intracellular accumulation of hyperphosphorylated, aggregated tau as neurofibrillary tangles are two of the characteristic hallmarks of Alzheimer's disease1,2. The regional progression of brain atrophy in Alzheimer's disease highly correlates with tau accumulation but not amyloid deposition3-5, and the mechanisms of tau-mediated neurodegeneration remain elusive. Innate immune responses represent a common pathway for the initiation and progression of some neurodegenerative diseases. So far, little is known about the extent or role of the adaptive immune response and its interaction with the innate immune response in the presence of amyloid-ß or tau pathology6. Here we systematically compared the immunological milieux in the brain of mice with amyloid deposition or tau aggregation and neurodegeneration. We found that mice with tauopathy but not those with amyloid deposition developed a unique innate and adaptive immune response and that depletion of microglia or T cells blocked tau-mediated neurodegeneration. Numbers of T cells, especially those of cytotoxic T cells, were markedly increased in areas with tau pathology in mice with tauopathy and in the Alzheimer's disease brain. T cell numbers correlated with the extent of neuronal loss, and the cells dynamically transformed their cellular characteristics from activated to exhausted states along with unique TCR clonal expansion. Inhibition of interferon-γ and PDCD1 signalling both significantly ameliorated brain atrophy. Our results thus reveal a tauopathy- and neurodegeneration-related immune hub involving activated microglia and T cell responses, which could serve as therapeutic targets for preventing neurodegeneration in Alzheimer's disease and primary tauopathies.


Subject(s)
Brain , Microglia , Neurofibrillary Tangles , T-Lymphocytes , Tauopathies , Animals , Mice , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Brain/immunology , Brain/metabolism , Brain/pathology , Microglia/immunology , Microglia/metabolism , Neurofibrillary Tangles/immunology , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , tau Proteins/immunology , tau Proteins/metabolism , Tauopathies/immunology , Tauopathies/metabolism , Tauopathies/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Clone Cells/immunology , Clone Cells/metabolism , Clone Cells/pathology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Immunity, Innate
2.
Neurobiol Dis ; 154: 105365, 2021 07.
Article in English | MEDLINE | ID: mdl-33848635

ABSTRACT

The imbalance between production and clearance of amyloid ß (Aß) peptides and their resulting accumulation in the brain is an early and crucial step in the pathogenesis of Alzheimer's disease (AD). Therefore, Aß is strongly positioned as a promising and extensively validated therapeutic target for AD. Investigational disease-modifying approaches aiming at reducing cerebral Aß concentrations include prevention of de novo production of Aß through inhibition of ß-site amyloid precursor protein cleaving enzyme 1 (BACE1), and clearance of Aß deposits via passive Aß immunotherapy. We have developed a novel, high affinity antibody against Aß peptides bearing a pyroglutamate residue at amino acid position 3 (3pE), an Aß species abundantly present in plaque deposits in AD brains. Here, we describe the preclinical characterization of this antibody, and demonstrate a significant reduction in amyloid burden in the absence of microhemorrhages in different mouse models with established plaque deposition. Moreover, we combined antibody treatment with chronic BACE1 inhibitor treatment and demonstrate significant clearance of pre-existing amyloid deposits in transgenic mouse brain, without induction of microhemorrhages and other histopathological findings. Together, these data confirm significant potential for the 3pE-specific antibody to be developed as a passive immunotherapy approach that balances efficacy and safety. Moreover, our studies suggest further enhanced treatment efficacy and favorable safety after combination of the 3pE-specific antibody with BACE1 inhibitor treatment.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Antibodies, Monoclonal/administration & dosage , Aspartic Acid Endopeptidases/antagonists & inhibitors , Immunization, Passive/methods , Peptide Fragments/antagonists & inhibitors , Plaque, Amyloid/drug therapy , Amyloid Precursor Protein Secretases/immunology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Monoclonal/immunology , Aspartic Acid Endopeptidases/immunology , Aspartic Acid Endopeptidases/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Peptide Fragments/immunology , Peptide Fragments/metabolism , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism , Treatment Outcome
3.
Neurosci Lett ; 754: 135869, 2021 05 29.
Article in English | MEDLINE | ID: mdl-33857550

ABSTRACT

Intracellular Aß (iAß) expression, extracellular Aß (eAß) plaque formation and microglial reactivity are characteristic neuropathological events of Alzheimer's disease (AD) and have been detected in several transgenic mouse models of this disease. In this work we decided to investigate the early (2-7 months of age) development of these phenomena at both regional and cellular levels in 5XFAD mice, a severe transgenic mouse model of AD. We demonstrated that 1) Aß pathology develops in many but not all brain regions, 2) iAß is transient and almost always followed by eAß in grey matter regions, and the respective levels are roughly proportional, and 3) in about 1/3 of the grey matter regions with Aß pathology and in several white matter regions, eAß plaques can appear where no iAß-positive structures were detected. We also showed that male and female mice share a similar regional and cellular pattern of Aß pathology development that is more prominent in females. Early iAß is associated to the activation of microglia, while subsequent formation of eAß plaques is associated with markedly increased density of microglial cells that acquire a characteristic clustered phenotype. Present analysis is relevant to set a reference for pathophysiological studies and to define specific targets for the test of therapeutic interventions in this widely used AD transgenic model.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cerebral Cortex/pathology , Plaque, Amyloid/pathology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Amyloid beta-Peptides/analysis , Animals , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Plaque, Amyloid/genetics , Plaque, Amyloid/immunology , Sex Factors
4.
STAR Protoc ; 2(1): 100265, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33490981

ABSTRACT

Alzheimer's disease is characterized by the deposition of extracellular amyloid-beta (Aß) plaques. While microglial phagocytosis is a major mechanism through which Aß is cleared, there is no method for quantitatively assessing Aß phagocytic capacity of microglia in vivo. Here, we present a flow cytometry-based method for investigating the Aß phagocytic capacity of microglia in vivo. This method enables the direct comparison of Aß phagocytic capacity between different microglial subpopulations as well as the direct isolation of Aß phagocytic microglia for downstream applications. For complete details on the use and execution of this protocol, please refer to Lau et al. (2020).


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Microglia/immunology , Phagocytosis , Plaque, Amyloid/immunology , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Plaque, Amyloid/genetics
5.
J Biol Chem ; 296: 100334, 2021.
Article in English | MEDLINE | ID: mdl-33508322

ABSTRACT

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.


Subject(s)
Amyloid/ultrastructure , Complementarity Determining Regions/genetics , Immunoglobulin Light-chain Amyloidosis/genetics , Plaque, Amyloid/genetics , Amino Acid Sequence/genetics , Amyloid/genetics , Amyloid/immunology , Amyloidogenic Proteins/genetics , Amyloidogenic Proteins/immunology , Amyloidogenic Proteins/ultrastructure , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/ultrastructure , Humans , Immunoglobulin Light-chain Amyloidosis/immunology , Immunoglobulin Light-chain Amyloidosis/metabolism , Mutation/genetics , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/pathology , Protein Conformation , Protein Folding
6.
Curr Alzheimer Res ; 17(8): 722-734, 2020.
Article in English | MEDLINE | ID: mdl-33167834

ABSTRACT

BACKGROUND: The pathogenesis of Alzheimer's disease (AD) is not directly caused by the presence of senile plaques but rather by the detrimental effects exerted on neuronal cells by toxic soluble oligomers. Such species are formed early during the aggregation process of the Aß1-42 peptide or can be released from mature fibrils. Nowadays, efficient tools for an early diagnosis, as well as pharmaceutical treatments targeting the harmful agents in samples of AD patients, are still missing. OBJECTIVE: By integrating in vitro immunochemical assay with in vivo neuronal models of toxicity, we aim to understand and target the principles that drive toxicity in AD. METHODS: We evaluated the specificity and sensitivity of A11 and OC conformational antibodies to target a range of pathologically relevant amyloid conformers and rescue their cytotoxic effects in neuronal culture models using a number of cellular readouts. RESULTS: We demonstrated the peculiar ability of conformational antibodies to label pathologically relevant Aß1-42 oligomers and fibrils and to prevent their detrimental effects on neuronal cells. CONCLUSION: Our results substantially improve our knowledge on the role of toxic assemblies in neurodegenerative diseases, thus suggesting new and more effective diagnostic and therapeutic tools for AD.


Subject(s)
Antibodies/therapeutic use , Plaque, Amyloid/immunology , Alzheimer Disease/immunology , Alzheimer Disease/therapy , Amyloid/immunology , Amyloid beta-Peptides/immunology , Animals , Antibodies/immunology , Caspase 3/metabolism , Humans , In Vitro Techniques , Microscopy, Confocal , Neurons/immunology , Peptide Fragments/immunology , Plaque, Amyloid/therapy , Protein Conformation , Rats
7.
J Neuropathol Exp Neurol ; 79(11): 1147-1162, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33011810

ABSTRACT

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.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Macrophages/pathology , Plaque, Amyloid/pathology , Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Brain/immunology , Disease Models, Animal , Female , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Transgenic , Plaque, Amyloid/immunology
8.
Front Immunol ; 11: 1029, 2020.
Article in English | MEDLINE | ID: mdl-32582162

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Antigen Presentation , Peptide Fragments/chemistry , Peptide Fragments/immunology , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloidosis/genetics , Amyloidosis/immunology , Amyloidosis/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Brain/immunology , Brain/pathology , Disease Models, Animal , Gene Expression , Histocompatibility Antigens Class II/metabolism , Humans , Immunomodulation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/genetics , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
9.
Hypertension ; 75(6): 1464-1474, 2020 06.
Article in English | MEDLINE | ID: mdl-32362228

ABSTRACT

Antihypertensive medications targeting the renin-angiotensin system have lowered the incidence and progression of Alzheimer disease. Understanding how these medications function could lead to novel therapeutic strategies. AT4Rs (angiotensin IV receptors) have been associated with angiotensin receptor blockers' cognitive, cerebrovascular, and neuroinflammatory rescue in Alzheimer disease models. Yet, whether AT4Rs act alone or with AT2Rs remains unknown. Here, we investigated whether AT2Rs contribute to losartan's benefits and whether chronic AT2R activation could mimic angiotensin receptor blocker benefits in transgenic mice overexpressing familial Alzheimer disease mutations of the human APP (amyloid precursor protein). Losartan-treated mice (10 mg/kg per day, drinking water, 7 months) received intracerebroventricular (1 month) administration of vehicle or AT2R antagonist PD123319 (1.6 nmol/day). PD123319 countered losartan's benefits on spatial learning and memory, neurovascular coupling, and hampered those on oxidative stress and nitric oxide bioavailability. PD123319 did not oppose losartan's benefits on short-term memory and vasodilatory function and had no benefit on neuroinflammation or Aß (amyloid ß) pathology. Mice receiving either vehicle or selective AT2R agonist compound 21 (intracerebroventricular: 1 nmol/day, 1 month or drinking water: 10 mg/kg per day, 7 months), showed no improvement in memory, vasodilatory function, or nitric oxide bioavailability. Compound 21 treatment normalized neurovascular coupling, reduced astrogliosis independent of persisting microgliosis, and exacerbated oxidative stress in APP mice. Compound 21 reduced dense core Aß plaques, but not diffuse plaques or Aß species. Our findings suggest that targeting AT2Rs is not an ideal strategy for restoring Aß-related cognitive and cerebrovascular deficits.


Subject(s)
Alzheimer Disease , Imidazoles/pharmacology , Neurovascular Coupling/drug effects , Plaque, Amyloid , Pyridines/pharmacology , Receptor, Angiotensin, Type 2/metabolism , Receptors, Angiotensin/metabolism , Renin-Angiotensin System/drug effects , Sulfonamides/pharmacology , Thiophenes/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Cognition/drug effects , Disease Models, Animal , Losartan/pharmacology , Mice , Neuroimmunomodulation , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Vasodilation/drug effects
10.
Proc Natl Acad Sci U S A ; 117(12): 6844-6854, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32144141

ABSTRACT

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.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/pathology , Inflammation/pathology , Neurons/immunology , Plaque, Amyloid/pathology , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloidosis , Animals , Brain/immunology , Brain/metabolism , Disease Models, Animal , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Male , Neurons/metabolism , Neurons/pathology , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism , Rats , Rats, Transgenic
11.
J Neuroimmunol ; 341: 577185, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32045774

ABSTRACT

Alzheimer's disease (AD) is the most common dementia type affecting nearly 44 million people worldwide. Recent findings point to microglia as a significant contributor to neural development, neuroinflammation, and degeneration. Dysregulated immunoactivity in AD has been broadly studied, and current research on animal models enabled us to identify a new cluster of microglia (disease-associated microglia) alongside previously detected glial populations (e.g., plaque-associated microglia, dark microglia, Human Alzheimer's microglia) associated with neuroinflammation and with macrophagic activity. These distinct populations of glia show a spatial distribution within plaques with unique imaging features and distinct gene expression profile. Novel genetic approaches using single-nuclei RNA sequencing (sn-RNA seq) allowed researchers to identify gene expression profiles from fixed human samples. Recent studies, exposing transcriptomic clusters of disease-related cells and analyzing sequenced RNA from sorted myeloid cells, seem to confirm the hypothesis of the central role of glia in the pathogenesis of Alzheimer's disease. These discoveries may shed light on the effects of microglial activation and differences in gene expression profiles, furthering research towards the development of a cell-specific therapy. In this review, we examine recent studies that guide us towards recognizing the role of diverse populations of glial cells and their possible heterogeneous functional states in the pathogenesis of AD in humans.


Subject(s)
Alzheimer Disease/immunology , Microglia/immunology , Nerve Degeneration/immunology , Adaptor Proteins, Signal Transducing/deficiency , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Eye Proteins/physiology , Gene Expression Profiling , Humans , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Microglia/classification , Microglia/metabolism , Microglia/pathology , Nerve Degeneration/pathology , Nerve Growth Factors/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Immunologic/physiology , Serpins/physiology , Transcriptome
12.
Biomacromolecules ; 20(11): 4208-4217, 2019 11 11.
Article in English | MEDLINE | ID: mdl-31600059

ABSTRACT

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.


Subject(s)
Alzheimer Disease/immunology , Amyloid/immunology , Peptide Fragments/chemistry , Plaque, Amyloid/immunology , Protein Corona/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid/chemistry , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Brain/immunology , Brain/pathology , Cytokines/biosynthesis , Cytokines/chemistry , Humans , Microglia/immunology , Neurons/immunology , Neurons/pathology , Peptide Fragments/immunology , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Protein Corona/immunology , Protein Transport/immunology
13.
Acta Neuropathol Commun ; 7(1): 141, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31477180

ABSTRACT

Neuropathological follow-up of patients with Alzheimer's disease (AD) who participated in the first clinical trial of Amyloid-ß 42 (Aß42) immunization (AN1792, Elan Pharmaceuticals) has shown that immunization can induce removal of Aß42 and Aß40 from plaques, whereas analysis of the cerebral vessels has shown increased levels of these Aß peptides in cerebral amyloid angiopathy (CAA). Aß43 has been less frequently studied in AD, but its aggregation propensity and neurotoxic properties suggest it may have an important pathogenic role. In the current study we show by using immunohistochemistry that in unimmunized AD patients Aß43 is a frequent constituent of plaques (6.0% immunostained area), similar to Aß42 (3.9% immunostained area). Aß43 immunostained area was significantly higher than that of Aß40 (2.3%, p = 0.006). In addition, we show that Aß43 is only a minor component of CAA in both parenchymal vessels (1.5 Aß43-positive vessels per cm2 cortex vs. 5.3 Aß42-positive vessels, p = 0.03, and 6.2 Aß40-positive vessels, p = 0.045) and leptomeningeal vessels (5.6% Aß43-positive vessels vs. 17.3% Aß42-positive vessels, p = 0.007, and 27.4% Aß40-positive vessels, p = 0.003). Furthermore, we have shown that Aß43 is cleared from plaques after Aß immunotherapy, similar to Aß42 and Aß40. Cerebrovascular Aß43 levels did not change after immunotherapy.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Brain/immunology , Immunization , Peptide Fragments/immunology , Plaque, Amyloid/immunology , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/pathology , Cerebral Amyloid Angiopathy/immunology , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Female , Humans , Male , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism
15.
Sci Rep ; 9(1): 9870, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31285517

ABSTRACT

Advances in the understanding of Alzheimer's disease (AD) suggest that pathogenesis is not directly related to plaque burden, but rather to soluble toxic amyloid-beta oligomers (AßO). Therapeutic antibodies targeting Aß monomers and/or plaque have shown limited efficacy and dose-limiting adverse events in clinical trials. These findings suggest that antibodies capable of selectively neutralizing toxic AßO may achieve improved efficacy and safety. To this end, we generated monoclonal antibodies against a conformational Aß epitope predicted by computational modeling to be presented on toxic AßO but not monomers or fibrils. The resulting lead antibody, PMN310, showed the desired AßO-selective binding profile. In vitro, PMN310 inhibited AßO propagation and toxicity. In vivo, PMN310 prevented AßO-induced loss of memory formation and reduced synaptic loss and inflammation. A humanized version (huPMN310) compared favorably to other Aß-directed antibodies showing a lack of adverse event-associated binding to Aß deposits in AD brains, and greater selective binding to AßO-enriched AD brain fractions that contain synaptotoxic Aß species. Systemic administration of huPMN310 in mice resulted in brain exposure and kinetics comparable to those of other therapeutic human monoclonal antibodies. Greater selectivity for AßO and the potential to safely administer high doses of huPMN310 are expected to result in enhanced safety and therapeutic potency.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/immunology , Adult , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Animals , Brain/immunology , Child , Cognition/physiology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Plaque, Amyloid/immunology , Young Adult
16.
Acta Neuropathol ; 138(4): 613-630, 2019 10.
Article in English | MEDLINE | ID: mdl-31350575

ABSTRACT

Identification of multiple immune-related genetic risk factors for sporadic AD (sAD) have put the immune system center stage in mechanisms underlying this disorder. Comprehensive analysis of microglia in different stages of AD in human brains revealed microglia activation to follow the progression of AD neuropathological changes and requiring the co-occurrence of beta-Amyloid (Aß) and tau pathology. Carriers of AD-associated risk variants in TREM2 (Triggering receptor expressed on myeloid cells 2) showed a reduction of plaque-associated microglia and a substantial increase in dystrophic neurites and overall pathological tau compared with age and disease stage matched AD patients without TREM2 risk variants. These findings were substantiated by digital spatial profiling of the plaque microenvironment and targeted gene expression profiling on the NanoString nCounter system, which revealed striking brain region dependent differences in immune response patterns within individual cases. The demonstration of profound brain region and risk-variant specific differences in immune activation in human AD brains impacts the applicability of immune-therapeutic approaches for sAD and related neurodegenerative diseases.


Subject(s)
Alzheimer Disease/genetics , Brain/pathology , Membrane Glycoproteins/genetics , Microglia/pathology , Plaque, Amyloid/pathology , Receptors, Immunologic/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/immunology , Disease Progression , Humans , Male , Microglia/immunology , Neurites/immunology , Neurites/pathology , Plaque, Amyloid/immunology , tau Proteins/metabolism
17.
Nat Med ; 25(3): 496-506, 2019 03.
Article in English | MEDLINE | ID: mdl-30692699

ABSTRACT

Apolipoprotein-E (ApoE) has been implicated in Alzheimer's disease, atherosclerosis, and other unresolvable inflammatory conditions but a common mechanism of action remains elusive. We found in ApoE-deficient mice that oxidized lipids activated the classical complement cascade (CCC), resulting in leukocyte infiltration of the choroid plexus (ChP). All human ApoE isoforms attenuated CCC activity via high-affinity binding to the activated CCC-initiating C1q protein (KD~140-580 pM) in vitro, and C1q-ApoE complexes emerged as markers for ongoing complement activity of diseased ChPs, Aß plaques, and atherosclerosis in vivo. C1q-ApoE complexes in human ChPs, Aß plaques, and arteries correlated with cognitive decline and atherosclerosis, respectively. Treatment with small interfering RNA (siRNA) against C5, which is formed by all complement pathways, attenuated murine ChP inflammation, Aß-associated microglia accumulation, and atherosclerosis. Thus, ApoE is a direct checkpoint inhibitor of unresolvable inflammation, and reducing C5 attenuates disease burden.


Subject(s)
Antigen-Antibody Complex/immunology , Apolipoproteins E/immunology , Carotid Artery Diseases/immunology , Choroid Plexus/immunology , Cognitive Dysfunction/immunology , Complement C1q/immunology , Complement Pathway, Classical/immunology , Aged , Aged, 80 and over , Amyloid beta-Peptides/immunology , Animals , Aorta/immunology , Aorta/pathology , Atherosclerosis/immunology , Atherosclerosis/pathology , Brain/immunology , Brain/pathology , Carotid Arteries/immunology , Carotid Arteries/pathology , Carotid Artery Diseases/pathology , Choroid Plexus/pathology , Cognitive Dysfunction/pathology , Complement C5 , Female , Humans , Leukocytes , Male , Mice, Knockout, ApoE , Microscopy, Fluorescence , Middle Aged , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Protein Isoforms/immunology , RNA, Small Interfering
18.
EMBO J ; 38(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30617086

ABSTRACT

Alzheimer's disease is characterized not only by extracellular amyloid plaques and neurofibrillary tangles, but also by microglia-mediated neuroinflammation. Recently, autophagy has been linked to the regulation of the inflammatory response. Thus, we investigated how an impairment of autophagy mediated by BECN1/Beclin1 reduction, as described in Alzheimer's disease patients, would influence cytokine production of microglia. Acutely stimulated microglia from Becn1+/- mice exhibited increased expression of IL-1beta and IL-18 compared to wild-type microglia. Becn1+/-APPPS1 mice also contained enhanced IL-1beta levels. The investigation of the IL-1beta/IL-18 processing pathway showed an elevated number of cells with inflammasomes and increased levels of NLRP3 and cleaved CASP1/Caspase1 in Becn1+/- microglia. Super-resolation microscopy revealed a very close association of NLRP3 aggregates and LC3-positive vesicles. Interestingly, CALCOCO2 colocalized with NLRP3 and its downregulation increased IL-1beta release. These data support the notion that selective autophagy can impact microglia activation by modulating IL-1beta and IL-18 production via NLRP3 degradation and thus present a mechanism how impaired autophagy could contribute to neuroinflammation in Alzheimer's disease.


Subject(s)
Autophagy , Beclin-1/physiology , Inflammation/immunology , Microglia/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Plaque, Amyloid/immunology , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/physiology , Animals , Autophagosomes , Cytokines/metabolism , Female , Inflammasomes , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presenilin-1/physiology
19.
J Neuroimmunol ; 322: 15-25, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29958693

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/immunology , Electroporation/methods , Immunization/methods , Peptide Fragments/immunology , Vaccines, DNA/immunology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/genetics , Animals , Antibody Formation , Biolistics , Dose-Response Relationship, Immunologic , Electromagnetic Phenomena , Forkhead Transcription Factors/genetics , Genes, Reporter , Humans , Immunogenicity, Vaccine , Injections, Intradermal , Mice , Mice, Inbred BALB C , Mice, Transgenic , Peptide Fragments/genetics , Plaque, Amyloid/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Vaccines, DNA/administration & dosage
20.
Sci Rep ; 8(1): 9813, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29955078

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Neuroendocrine Secretory Protein 7B2/deficiency , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Animals , Benzothiazoles/metabolism , Brain/metabolism , Brain/pathology , Brain/physiopathology , Clusterin/metabolism , Disease Models, Animal , Female , Genotype , Heterozygote , Humans , Memory , Mice, Inbred C57BL , Mice, Knockout , Neuroendocrine Secretory Protein 7B2/genetics , Neuroendocrine Secretory Protein 7B2/metabolism , Plaque, Amyloid/immunology , Plaque, Amyloid/physiopathology , Solubility , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...