Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmaceutics ; 12(10)2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33023109

ABSTRACT

Clinical symptoms of Alzheimer's Disease (AD) include behavioral alterations and cognitive impairment. These functional phenotypes early occur in triple-transgenic (3xTg-AD) mice. Specifically, behavioral alterations are first detected when mice are at around 2.5 months old and cognitive impairment in between 3- and 5-month-old mice. In this work, the effect of chronic Aß-immunotherapy on behavioral and cognitive abilities was tested by monthly administering the antibody fragment scFv-h3D6 to 3xTg-AD female mice from 5 to 9 months of age. An untreated group was used as a reference, as well as to attain some information on the effect of training during the longitudinal study. Behavioral and psychological symptoms of dementia (BPSD)-like symptoms were already evident in 5-month-old mice, in the form of neophobia and anxious-like behavior. The exploratory activity decreased over the longitudinal study, not only for 3xTgAD mice but also for the corresponding non-transgenic mice (NTg). Learning abilities of 3xTg-AD mice were not seriously compromised but an impairment in long-term spatial memory was evident at 5 months of age. Interestingly, scFv-h3D6-treatment affected the cognitive impairment displayed by 5-month-old 3xTg-AD mice. It is worth noting that training also reduced cognitive impairment of 3xTg-AD mice over the longitudinal study, suggesting that to properly quantify the isolated therapeutic potential of any drug on cognition using this model it is convenient to perform a prompt, age-matched study rather than a longitudinal study. In addition, a combination of both training and Aß-immunotherapy could constitute a possible approach to treat Alzheimer's disease.

2.
J Alzheimers Dis ; 70(4): 1069-1091, 2019.
Article in English | MEDLINE | ID: mdl-31306135

ABSTRACT

The intracellular deposition of amyloid-ß (Aß) peptides has been described in the brains of both Alzheimer's disease (AD) patients and animal models. A correlation between the intracellular amyloid burden and neurodegeneration has recently been reported in a triple-transgenic AD (3xTg-AD) murine model. In the present study, we assessed the effect of scFv-h3D6, an anti-Aß single-chain variable fragment (scFv) derived from the antibody bapineuzumab, on amyloid pathology in 5-month-old 3xTg-AD female mice, focusing on intracellular Aß clearance, neuronal survival, and functional abilities. We also examined neuroinflammation and the histology of peripheral organ samples to detect any adverse effects. A single intraperitoneal injection of scFv-h3D6 dramatically reduced intracellular Aß burden in the deep layers of the cerebral cortex, pyramidal cells layer of the hippocampus, and basolateral amygdalar nucleus. The treatment prevented neuronal loss in the hippocampus and amygdala, while neither astrogliosis nor microgliosis was induced. Instead, an increase in the size of the white pulp after the treatment indicated that the spleen could be involved in the clearance mechanism. Although the treatment did not ameliorate behavioral and psychological symptoms of dementia-like symptoms, the results of cognitive testing pointed to a noticeable improvement in spatial memory. These findings indicated that the mechanism underlying the therapeutic effect of scFv-h3D6 was the clearance of intracellular Aß, with subsequent prevention of neuronal loss and amelioration of cognitive disabilities. The treatment was safe in terms of neuroinflammation and kidney and liver function, whereas some effects on the spleen were observed.


Subject(s)
Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized/administration & dosage , Intracellular Fluid/metabolism , Neurons/metabolism , Single-Chain Antibodies/administration & dosage , Spatial Memory/physiology , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Female , Intracellular Fluid/drug effects , Mice , Mice, Transgenic , Neurons/drug effects , Spatial Memory/drug effects
3.
PLoS One ; 14(5): e0217793, 2019.
Article in English | MEDLINE | ID: mdl-31150495

ABSTRACT

The success of the targeting of amyloid-ß (Aß) oligomers through immunotherapy in Alzheimer's disease (AD) mouse models has not been translated into the clinics. The use of single-chain variable fragments (scFvs) has been proposed to prevent the potential severe effects of full-length mAbs by precluding crystallizable fraction-mediated microglia activation. The efficacy of scFv-h3D6, a bapineuzumab-derived anti-Aß scFv, has been extensively proven. In this work, we compared scFv-h3D6-EL, an elongated variant of the scFv-h3D6, with its original version to assess whether its characteristic higher thermodynamic stability improved its pharmacokinetic parameters. Although scFv-h3D6-EL had a longer half-life than its original version, its absorption from the peritoneal cavity into the systemic compartment was lower than that of the original version. Moreover, we attempted to determine the mechanism underlying the protective effect of scFv-h3D6. We found that scFv-h3D6 showed compartmental distribution and more interestingly crossed the blood-brain barrier. In the brain, scFv-h3D6 was engulfed by glial cells or internalized by Aß peptide-containing neurons in the early phase post-injection, and was colocalized with the Aß peptide almost exclusively in glial cells in the late phase post-injection. Aß peptide levels in the brain decreased simultaneously with an increase in scFv-h3D6 levels. This observation in addition to the increased tumor necrosis factor-α levels in the late phase post-injection suggested that the engulfment of Aß peptide/scFv-h3D6 complex extruded from large neurons by phagocytic cells was the mechanism underlying Aß peptide withdrawal. The mechanism of action of scFv-h3D6 demonstrates the effectivity of Aß-immunotherapy and lays the background for other studies focused on the finding of a treatment for AD.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Immunoglobulin Fragments/immunology , Single-Chain Antibodies/immunology , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacokinetics , Amyloid beta-Peptides/pharmacology , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Disease Models, Animal , Humans , Immunoglobulin Fragments/pharmacology , Mice , Neurons/metabolism , Protein Sorting Signals/genetics , Protein Stability , Single-Chain Antibodies/pharmacokinetics , Single-Chain Antibodies/pharmacology , Thermodynamics
4.
Biochem Pharmacol ; 155: 380-392, 2018 09.
Article in English | MEDLINE | ID: mdl-30026023

ABSTRACT

Anti-Aß immunotherapy has emerged as a promising approach to treat Alzheimer's disease (AD). The single-chain variable fragment scFv-h3D6 is an anti-Aß antibody fragment that lacks the Fc region, which is associated with the induction of microglial reactivity by the full-length monoclonal antibody bapineuzumab. ScFv-h3D6 was previously shown to restore the levels of apolipoprotein E (apoE) and apolipoprotein J (apoJ) in a triple-transgenic-AD (3xTg-AD) mouse model. Since apoE and apoJ play an important role in the development of AD, we aimed to study the in vivo effect of the combined therapy of scFv-h3D6 with apoE and apoJ mimetic peptides (MPs). Four-and-a-half-month-old 3xTg-AD mice were treated for six weeks with scFv-h3D6, apoE-MP, apoJ-MP, or a combination of scFv-h3D6 with each of the MPs, or a vehicle, and then the results were compared to non-transgenic mice. Magnetic Resonance Imaging showed a general tendency of the different treatments to protect against the reduction in brain volume. Aß burden decreased after treatment with scFv-h3D6, apoE-MP, or apoJ-MP, but the effect was not as evident with the combined therapies. In terms of glial reactivity, apoE-MP showed a potent anti-inflammatory effect that was eased by the presence of scFv-h3D6, whereas the combination of apoJ-MP and scFv-h3D6 was not detrimental. ScFv-h3D6 alone did not induce microglial reactivity, as full-length antibodies do; rather, it reduced it. Endogenous apoE and apoJ levels were decreased by scFv-h3D6, but the MPs lead to a simultaneous increase of both apolipoproteins. While apoE-MP and apoJ-MP demonstrated different effects in the combined therapies with scFv-h3D6, they did not improve the overall protective effect of scFv-h3D6 in reducing the Aß burden, apolipoproteins levels or microglial reactivity.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoproteins E/administration & dosage , Biomimetic Materials/administration & dosage , Clusterin/administration & dosage , Single-Chain Antibodies/administration & dosage , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amino Acid Sequence , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/genetics , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Female , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Transgenic , Random Allocation
5.
PLoS One ; 12(8): e0181480, 2017.
Article in English | MEDLINE | ID: mdl-28771492

ABSTRACT

ScFv-h3D6 has been shown as an efficient therapy in the 3xTg-AD mouse model of Alzheimer's Disease. Because one of the major bottlenecks for the therapeutic uses of proteins produced in Escherichia coli is their potential contamination with endotoxins, LPS were extensively removed by a rather low-efficient, expensive, and time-consuming purification step. In addition, disulfide scrambling is favored in the reducing bacterial cytoplasm albeit the use of reductase deficient strains. To overcome these hurdles, as well as to improve the yield, the yeast Pichia pastoris, an endotoxin-free host system for recombinant protein production, has been used to produce scFv-h3D6, both in flask and in a fed-batch bioreactor. Comparison of the thermal stability of the obtained protein with that from E. coli showed no differences. Opposite to the case of the protein obtained from E. coli, no disulfide scrambled conformations or LPS traces were detected in that produced in P. pastoris. Cytotoxicity assays in SH-SY5Y neuroblastoma cell-cultures demonstrated that proteins from both expression systems were similarly efficient in precluding Aß-induced toxicity. Finally, the 3xTg-AD mouse model was used to test the therapeutic effect of both proteins. Quantification of Aß levels from cortex and hippocampus protein extracts by ELISA, and Aß-immunohistochemistry, showed that both proteins reduced Aß burden. This work demonstrates that scFv-h3D6 obtained from P. pastoris shows the same benefits as those already known for that obtained from E. coli, with multiple advantages in terms of recombinant production and safety.


Subject(s)
Amyloid beta-Peptides/immunology , Genetic Engineering , Peptide Fragments/immunology , Pichia/genetics , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/immunology , Amino Acid Sequence , Animals , Cell Line, Tumor , Humans , Mice , Reproducibility of Results , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/therapeutic use
6.
J Alzheimers Dis ; 59(3): 1079-1096, 2017.
Article in English | MEDLINE | ID: mdl-28697564

ABSTRACT

The main histopathological hallmarks of Alzheimer's disease (AD) are the extracellular deposition of neuritic amyloid plaques, composed of amyloid-ß (Aß) peptide, and the intracellular accumulation of neurofibrillary tangles, composed of hyperphosphorylated tau. Both traits are emulated in the 3xTg-AD mouse model. Because the relevance of this model in the bibliography and the main role of Aß in neuronal impairment, here we have detailed the brain Aß/AßPP distribution to subsequently quantify cellular density and intracellular burden for specific neuronal populations in the early stages of the disease. 6E10 immunoreactivity was evident in the deep layers of the cerebral cortex, in the pyramidal cell layer of the hippocampus, in the basolateral amygdala nucleus, and in the deep cerebellar nuclei macroneurons; whereas the specific neuronal populations with decreased cellular density were the large pyramidal neurons from the layers V-VI in the cerebral cortex, the pyramidal neurons from the CA2-3 region in the hippocampus, and the large neurons from the basolateral nucleus in the amygdala, apart from the already reported deep cerebellar nuclei. Interestingly, we found a strong correlation between intracellular Aß/AßPP burden and cellular density in all these populations. In addition, behavioral testing showed the functional consequences of such a neuronal depletion. Concretely, anxious-like behavior is manifested in the corner and open-field tests, as well as cognitive functions shown to be impaired in the novel object recognition test and Morris water maze paradigm. To our knowledge, this is the first deep characterization of the specific neuronal populations affected in the 3xTg-AD mouse model.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor/metabolism , Brain/pathology , Extracellular Fluid/metabolism , Neurons/pathology , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Calcium-Binding Proteins/metabolism , Epitopes , Exploratory Behavior , Female , Gene Expression Regulation/genetics , Humans , Male , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Mutation/genetics , Nerve Tissue Proteins/metabolism , Peptide Fragments/metabolism , Phosphorylation , Presenilin-1/genetics , Recognition, Psychology , Space Perception , tau Proteins/genetics
7.
J Alzheimers Dis ; 57(4): 1171-1183, 2017.
Article in English | MEDLINE | ID: mdl-28304309

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder that nowadays affects more than 40 million people worldwide and it is predicted to exponentially increase in the coming decades. Because no curative treatment exists, research on the pathophysiology of the disease, as well as the testing of new drugs, are mandatory. For these purposes, animal models constitute a valuable, although perfectible tool. This review takes a tour through several aspects of mouse models of AD, such as the generation of transgenic models, the relevance of the promoter driving the expression of the transgenes, and the concrete transgenes used to simulate AD pathophysiology. Then, transgenic mouse lines harboring mutated human genes at several loci such as APP, PSEN1, APOEɛ4, and ob (leptin) are reviewed. Therefore, not only the accumulation of the Aß peptide is emulated but also cholesterol and insulin metabolism. Further novel information about the disease will allow for the development of more accurate animal models, which in turn will undoubtedly be helpful for bringing preclinical research closer to clinical trials in humans.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Mice, Transgenic , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Humans
9.
MAbs ; 5(5): 660-4, 2013.
Article in English | MEDLINE | ID: mdl-23884149

ABSTRACT

The therapeutic potential of scFv-h3D6 has recently been shown in the 3xTg-AD mice. A clear effect on amyloid ß (Aß) oligomers and certain apolipoproteins in the brain was found, but no effect was seen in the cerebellum. Here, cellular vulnerability of the 3xTg-AD cerebellum is described for the first time, together with its protection by scFv-h3D6. Neuron depletion in the DCN was regionally variable and followed a mediolateral axis of involvement that was greatest in the fastigial nucleus, lesser in the interpositus and negligible in the dentate nucleus. A sole and low intraperitoneal dose of scFv-h3D6 protected 3xTg-AD DCN neurons from death. Further studies might provide interesting information about both the potential of scFv-h3D6 as a therapeutic agent and the role of the cerebellum in AD.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/immunology , Cerebellar Nuclei/drug effects , Neurons/drug effects , Single-Chain Antibodies/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Count , Cell Survival/drug effects , Cerebellar Nuclei/metabolism , Cerebellar Nuclei/pathology , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic , Mutation , Neurons/metabolism , Neurons/pathology , Presenilin-1/genetics , Presenilin-1/metabolism , Single-Chain Antibodies/immunology , tau Proteins/genetics , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...