Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Mol Oncol ; 17(11): 2337-2355, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37609678

ABSTRACT

Targeted therapies for prostate, breast, and ovarian cancers are based on their activity against primary tumors rather than their anti-metastatic activity. Consequently, there is an urgent need for new agents targeting the metastatic process. Emerging evidence correlates in vitro and in vivo cancer invasion and metastasis with increased activity of the proteases mesotrypsin (prostate and breast cancer) and kallikrein 6 (KLK6; ovarian cancer). Thus, mesotrypsin and KLK6 are attractive putative targets for therapeutic intervention. As potential therapeutics for advanced metastatic prostate, breast, and ovarian cancers, we report novel mesotrypsin- and KLK6-based therapies, based on our previously developed mutants of the human amyloid ß-protein precursor Kunitz protease inhibitor domain (APPI). These mutants, designated APPI-3M (prostate and breast cancer) and APPI-4M (ovarian cancer), demonstrated significant accumulation in tumors and therapeutic efficacy in orthotopic preclinical models, with the advantages of long retention times in vivo, high affinity and favorable pharmacokinetic properties. The applicability of the APPIs, as a novel therapy and for imaging purposes, is supported by their good safety profile and their controlled and scalable manufacturability in bioreactors.


Subject(s)
Breast Neoplasms , Ovarian Neoplasms , Male , Humans , Female , Serine Proteinase Inhibitors/therapeutic use , Amyloid beta-Peptides/therapeutic use , Prostate/pathology , Amyloid beta-Protein Precursor/pharmacology , Amyloid beta-Protein Precursor/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Kallikreins/genetics
2.
Geroscience ; 45(3): 1343-1381, 2023 06.
Article in English | MEDLINE | ID: mdl-37022634

ABSTRACT

Alzheimer's disease (AD) is an age-associated neurodegenerative disease. As the population ages, the increasing prevalence of AD threatens massive healthcare costs in the coming decades. Unfortunately, traditional drug development efforts for AD have proven largely unsuccessful. A geroscience approach to AD suggests that since aging is the main driver of AD, targeting aging itself may be an effective way to prevent or treat AD. Here, we discuss the effectiveness of geroprotective interventions on AD pathology and cognition in the widely utilized triple-transgenic mouse model of AD (3xTg-AD) which develops both ß-amyloid and tau pathologies characteristic of human AD, as well as cognitive deficits. We discuss the beneficial impacts of calorie restriction (CR), the gold standard for geroprotective interventions, and the effects of other dietary interventions including protein restriction. We also discuss the promising preclinical results of geroprotective pharmaceuticals, including rapamycin and medications for type 2 diabetes. Though these interventions and treatments have beneficial effects in the 3xTg-AD model, there is no guarantee that they will be as effective in humans, and we discuss the need to examine these interventions in additional animal models as well as the urgent need to test if some of these approaches can be translated from the lab to the bedside for the treatment of humans with AD.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Neurodegenerative Diseases , Mice , Humans , Animals , Alzheimer Disease/prevention & control , Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/therapeutic use , tau Proteins/metabolism , Mice, Transgenic , Disease Models, Animal
3.
Carbohydr Res ; 525: 108747, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36773398

ABSTRACT

The clinically important anticoagulant heparin, a member of the glycosaminoglycan family of carbohydrates that is extracted predominantly from porcine and bovine tissue sources, has previously been shown to inhibit the ß-site amyloid precursor protein cleaving enzyme 1 (BACE-1), a key drug target in Alzheimer's Disease. In addition, heparin has been shown to exert favourable bioactivities through a number of pathophysiological pathways involved in the disease processes of Alzheimer's Disease including inflammation, oxidative stress, tau phosphorylation and amyloid peptide generation. Despite the multi-target potential of heparin as a therapeutic option for Alzheimer's disease, the repurposing of this medically important biomolecule has to-date been precluded by its high anticoagulant potential. An alternative source to mammalian-derived glycosaminoglycans are those extracted from marine environments and these have been shown to display an expanded repertoire of sequence-space and heterogeneity compared to their mammalian counterparts. Furthermore, many marine-derived glycosaminoglycans appear to retain favourable bioactivities, whilst lacking the high anticoagulant potential of their mammalian counterparts. Here we describe a sulphated, marine-derived glycosaminoglycan extract from the Atlantic Sea Scallop, Placopecten magellanicus that displays high inhibitory potential against BACE-1 (IC50 = 4.8 µg.mL-1) combined with low anticoagulant activity; 25-fold less than that of heparin. This extract possesses a more favourable therapeutic profile compared to pharmaceutical heparin of mammalian provenance and is composed of a mixture of heparan sulphate (HS), with a high content of 6-sulphated N-acetyl glucosamine (64%), and chondroitin sulphate.


Subject(s)
Alzheimer Disease , Pectinidae , Animals , Cattle , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/therapeutic use , Anticoagulants/chemistry , Glycosaminoglycans/pharmacology , Heparin/pharmacology , Mammals/metabolism , Pectinidae/metabolism , Swine , Amyloid Precursor Protein Secretases
4.
Trends Mol Med ; 29(2): 99-111, 2023 02.
Article in English | MEDLINE | ID: mdl-36509631

ABSTRACT

The beta-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) has long been considered a conventional target for Alzheimer's disease (AD). Unfortunately, AD clinical trials of most BACE1 inhibitors were discontinued due to ineffective cognitive improvement or safety challenges. Recent studies investigating the involvement of BACE1 in metabolic, vascular, and immune functions have indicated a role in aging, diabetes, hypertension, and cancer. These novel BACE1 functions have helped to identify new 'druggable' targets for BACE1 against aging comorbidities. In this review, we discuss BACE1 regulation during aging, and then provide recent insights into its enzymatic and nonenzymatic involvement in aging and age-related diseases. Our study not only proposes the perspective of BACE1's actions in various systems, but also provides new directions for using BACE1 inhibitors and modulators to delay aging and to treat age-related diseases.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Humans , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Aging/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/therapeutic use
5.
Mini Rev Med Chem ; 23(4): 380-398, 2023.
Article in English | MEDLINE | ID: mdl-35786334

ABSTRACT

Alzheimer's Disease (AD) is a common neurodegenerative disorder that is almost incurable with the existing therapeutic interventions. Due to the high-risk factors associated with this disease, there is a global pursuit of new anti-AD agents. Herein, we explore the biochemical pathways which are responsible for the initiation/propagation of the disease. It is observed that out of the two isoforms of ß-secretase, ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) and ß-site amyloid precursor protein cleaving enzyme 2 (BACE2) present in the brain, BACE1 plays the predominant role in the commencement of AD. Moreover, the catalytic activities of acetylcholinesterase and butyrylcholinesterase regulate the concentration of neurotransmitters, and they are needed to be kept under control during the signs of AD. Hence, these two enzymes also serve as potential targets for the treatment of AD patients. Keeping in view the multifactorial nature of the disease, we also reviewed the multitarget approach for the treatment of AD. It is tried to identify the common structural features of those molecules which act on different cellular targets during AD therapy.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Aspartic Acid Endopeptidases/therapeutic use , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/therapeutic use , Acetylcholinesterase , Butyrylcholinesterase
6.
Bioorg Chem ; 129: 106137, 2022 12.
Article in English | MEDLINE | ID: mdl-36108590

ABSTRACT

Pomegranate (Punica granatum L.) extract has been reported to inhibit cholinesterase and the ß-site amyloid precursor protein cleaving enzyme 1 (BACE1); however, most of its constituents' potential inhibition of these enzymes remains unknown. Thus, we investigated the anti-Alzheimer's disease (anti-AD) potential of 16 ellagitannin and gallotannin, and nine anthocyanin derivatives' inhibition of BACE1, AChE, and BChE, and gallagic acid inhibited both the best. Further, a kinetic study identified different modes of inhibition, and a molecular docking simulation revealed that active compounds inhibited these three enzymes with low binding energy through hydrophilic and hydrophobic interactions in the active site cavities. Gallagic acid and castalagin decreased Aß peptides secretion from neuroblastoma cells that overexpressed human ß-amyloid precursor protein significantly by 10 µM. Further, treatment with gallagic acid and castalagin reduced BACE1 and APPsß expression levels significantly without affecting amyloid precursor protein (APP) levels in the amyloidogenic pathway. Co-incubation of Aß42 with gallagic acid reduced Aß42-induced intracellular reactive oxygen species (ROS) production significantly. Our results suggest that pomegranate constituents, specifically gallagic acid, may be useful in developing therapeutic treatment modalities for AD.


Subject(s)
Alzheimer Disease , Pomegranate , Humans , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/therapeutic use , Molecular Docking Simulation , Cholinesterases , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism
7.
Exp Mol Med ; 54(4): 433-446, 2022 04.
Article in English | MEDLINE | ID: mdl-35396575

ABSTRACT

Alzheimer's disease (AD) is caused by synaptic and neuronal loss in the brain. One of the characteristic hallmarks of AD is senile plaques containing amyloid ß-peptide (Aß). Aß is produced from amyloid precursor protein (APP) by sequential proteolytic cleavages by ß-secretase and γ-secretase, and the polymerization of Aß into amyloid plaques is thought to be a key pathogenic event in AD. Since γ-secretase mediates the final cleavage that liberates Aß, γ-secretase has been widely studied as a potential drug target for the treatment of AD. γ-Secretase is a transmembrane protein complex containing presenilin, nicastrin, Aph-1, and Pen-2, which are sufficient for γ-secretase activity. γ-Secretase cleaves >140 substrates, including APP and Notch. Previously, γ-secretase inhibitors (GSIs) were shown to cause side effects in clinical trials due to the inhibition of Notch signaling. Therefore, more specific regulation or modulation of γ-secretase is needed. In recent years, γ-secretase modulators (GSMs) have been developed. To modulate γ-secretase and to understand its complex biology, finding the binding sites of GSIs and GSMs on γ-secretase as well as identifying transiently binding γ-secretase modulatory proteins have been of great interest. In this review, decades of findings on γ-secretase in AD are discussed.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/therapeutic use , Humans , Proteolysis , Signal Transduction/physiology
8.
Curr Stem Cell Res Ther ; 17(4): 370-388, 2022.
Article in English | MEDLINE | ID: mdl-35236271

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by progressive cognitive deterioration. All recent therapeutic strategies tend to inhibit the generation of the Aß peptide. These approaches tend to mediate both α - and γ -secretases to undergo the nonamyloidogenic pathway. ADAM10 is the main α-secretase that cleaves APP, and it is regulated by the metabolic product of vitamin A (retinoic acid), which is being widely used recently in AD research as a target for treatment. Mesenchymal stem cells (MSCs) are also used recently as a promising regenerative therapy for AD. OBJECTIVES: The present study aimed to: (1) study the effect of MSCs with/without acitretin on the regulation of Adam10 gene expression in AlCl3-induced AD rat model, and (2) validate the hypothesis that AD is a time-dependent progressive disease that spreads spontaneously even after the stopping of exposure to AlCl3. METHODS: The experimental work has been designed to include three successive phases; AlCl3 induction phase (I), AlCl3 withdrawal phase (W), and therapeutic phase (T). Forty-five male albino Wistar rats were randomly divided into 2 main groups: the control (C) group (15 rats) and AD group (30 rats). The therapeutic potential of MSCs with/without acitretin has been evaluated at behavioral, physiological, molecular, and histopathological levels. RESULTS: Among the three therapeutic groups, combined administration of both MSC and acitretin showed the best compensatory effects on most of the measured parameters. CONCLUSION: The present study approved that AD is a time-dependent progressive disease which spreads spontaneously without more AlCl3 exposure.


Subject(s)
Alzheimer Disease , Mesenchymal Stem Cells , Neurodegenerative Diseases , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , ADAM10 Protein/therapeutic use , Acitretin/metabolism , Acitretin/pharmacology , Acitretin/therapeutic use , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/therapeutic use , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/therapeutic use , Animals , Male , Mesenchymal Stem Cells/metabolism , Rats
9.
Drug Deliv Transl Res ; 12(11): 2667-2677, 2022 11.
Article in English | MEDLINE | ID: mdl-35015254

ABSTRACT

Antibody drugs that target amyloid ß (Aß) are considered possible treatments for Alzheimer's disease; however, most have been dropped from clinical trials. We hypothesized that administration route for antiAß antibody (AntiAß) might affect its therapeutic potential and thus compared delivery of antibodies to the brain and their effect on cognitive dysfunction and amyloid disposition via intravenous (i.v.) and intranasal routes with and without the cell-penetrating peptide, L-penetratin. We demonstrated that intranasal administration with L-penetratin more efficiently delivered human immunoglobulin G (IgG), a model molecule for AntiAß, to the brain compared with i.v. injection. We found that multiple intranasal treatments with Alexa 594-labeled AntiAß (A594-AntiAß) with L-penetratin significantly improved learning by mice with aged amyloid precursor protein (APP) knock-in (App KI mice). Further, intranasal administration of A594-AntiAß increased the amount of soluble Aß (1-42) in the brain, suggesting suppression of Aß aggregation in insoluble form and involvement of activated microglia in Aß clearance. Thus, administration route may be critical for efficient delivery of AntiAß to the brain, and the nose-to-brain delivery with L-penetratin can maximize its therapeutic efficacy.


Subject(s)
Alzheimer Disease , Cell-Penetrating Peptides , Aged , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/pharmacology , Amyloid beta-Protein Precursor/therapeutic use , Animals , Brain/metabolism , Disease Models, Animal , Humans , Immunoglobulin G/metabolism , Injections, Intravenous , Mice
10.
Curr Opin Neurobiol ; 72: 101-110, 2022 02.
Article in English | MEDLINE | ID: mdl-34689040

ABSTRACT

Secretases are a group of proteases that are major drug targets considered for the prevention and treatment of Alzheimer's disease (AD). Secretases do not only process the AD-linked neuronal amyloid precursor protein (APP) but also the triggering receptor expressed on myeloid cells 2 (TREM2), thereby controlling microglial functions. This review highlights selected recent discoveries for the α-secretases a disintegrin and metalloprotease 10 (ADAM10) and a disintegrin and metalloprotease 17 (ADAM17), the ß-secretase ß-site APP cleaving enzyme 1 (BACE1) and γ-secretase and their link to AD. New genetic evidence strengthens the role of α-secretases in AD through cleavage of APP and TREM2. Novel proteins were linked to AD, which control α- and ß-secretase activity through transcriptional and post-translational mechanisms. Finally, new opportunities but also challenges are discussed for pharmacologically targeting ß- and γ-secretase cleavage of APP and α-secretase cleavage of TREM2 with the aim to prevent or treat AD.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , ADAM10 Protein/metabolism , ADAM10 Protein/therapeutic use , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/therapeutic use , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/therapeutic use , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/therapeutic use , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Aspartic Acid Endopeptidases/therapeutic use , Humans , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/therapeutic use , Proteolysis , Receptors, Immunologic/metabolism , Receptors, Immunologic/therapeutic use
11.
J Nutr ; 152(1): 140-152, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34636875

ABSTRACT

BACKGROUND: There is growing evidence of strong associations between the pathogenesis of Alzheimer's disease (AD) and dysbiotic oral and gut microbiota. Recent studies demonstrated that isoorientin (ISO) is anti-inflammatory and alleviates markers of AD, which were hypothesized to be mediated by the oral and gut microbiota. OBJECTIVES: We studied the effects of oral administration of ISO on AD-related markers and the oral and gut microbiota in mice. METHODS: Eight-month-old amyloid precursor protein/presenilin-1 (AP) transgenic male mice were randomly allocated to 3 groups of 15 mice each: vehicle (AP) alone or with a low dose of ISO (AP + ISO-L; 25 mg/kg) or a high dose of ISO (AP + ISO-H; 50 mg/kg). Age-matched wild-type (WT) C57BL/6 male littermates were used as controls. The 4 groups were treated intragastrically with ISO or sterilized ultrapure water for 2 months. AD-related markers in the brain, serum, colon, and liver were analyzed with immunohistochemical and histochemical staining, Western blotting, and ELISA. Oral and gut microbiotas were analyzed using 16S ribosomal RNA gene sequencing. RESULTS: The high-dose ISO treatment significantly decreased amyloid beta 42-positive deposition by 38.1% and 45.2% in the cortex and hippocampus, respectively, of AP mice (P < 0.05). Compared with the AP group, both ISO treatments reduced brain phospho-Tau, phosphor-p65, phosphor-inhibitor of NF-κB, and brain and serum LPS and TNF-α by 17.9%-72.5% and increased brain and serum IL-4 and IL-10 by 130%-210% in the AP + ISO-L and AP + ISO-H groups (P < 0.05). Abundances of 26, 25, and 23 microbial taxa in oral, fecal and cecal samples, respectively, were increased in both the AP + ISO-L and AP + ISO-H groups relative to the AP group [linear discriminant analysis (LDA) >3.0; P < 0.05]. Gram-negative bacteria, Alteromonas, Campylobacterales, and uncultured Bacteroidales bacterium were positively correlated (rho = 0.28-0.59; P < 0.05) with the LPS levels and responses of inflammatory cytokines. CONCLUSIONS: The microbiota-gut-brain axis is a potential mechanism by which ISO reduces AD-related markers in AP mice.


Subject(s)
Alzheimer Disease , Gastrointestinal Microbiome , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/pharmacology , Amyloid beta-Protein Precursor/therapeutic use , Animals , Disease Models, Animal , Luteolin , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1
12.
Curr Drug Targets ; 23(3): 266-285, 2022.
Article in English | MEDLINE | ID: mdl-34370634

ABSTRACT

INTRODUCTION: Alzheimer's disease (AD) is an intensifying neurodegenerative illness due to its irreversible nature. Identification of ß-site Amyloid Precursor Protein (APP) cleaving en-zyme1 (BACE1) has been a significant medicinal focus towards AD treatment, and this has opened ground for several investigations. Despite the numerous works in this direction, no BACE1 inhibitor has made it to the final approval stage as an anti-AD drug. METHODS: We provide an introductory background of the subject with a general overview of the pathogenesis of AD. The review features BACE1 inhibitor design and development with a focus on some clinical trials and discontinued drugs. Using the topical keywords BACE1, inhibitor design, and computational/theoretical study in the Web of Science and Scopus database, we retrieved over 49 relevant articles. The search years are from 2010 and 2020, with analysis conducted from May 2020 to March 2021. RESULTS AND DISCUSSION: Researchers have employed computational methodologies to unravel po-tential BACE1 inhibitors with a significant outcome. The most used computer-aided approach in BACE1 inhibitor design and binding/interaction studies are pharmacophore development, quantita-tive structure-activity relationship (QSAR), virtual screening, docking, and molecular dynamics (MD) simulations. These methods, plus more advanced ones including quantum mechan-ics/molecular mechanics (QM/MM) and QM, have proven substantial in the computational frame-work for BACE1 inhibitor design. Computational chemists have embraced the incorporation of in vitro assay to provide insight into the inhibition performance of identified molecules with potential inhibition towards BACE1. Significant IC50 values up to 50 nM, better than clinical trial com-pounds, are available in the literature. CONCLUSION: The continuous failure of potent BACE1 inhibitors at clinical trials is attracting many queries prompting researchers to investigate newer concepts necessary for effective inhibitor de-sign. The considered properties for efficient BACE1 inhibitor design seem enormous and require thorough scrutiny. Lately, researchers noticed that besides appreciable binding affinity and Blood-Brain Barrier (BBB) permeation, BACE1 inhibitor must show low or no affinity for permeability-glycoprotein. Computational modeling methods have profound applications in drug discovery strat-egies. With the volume of recent in silico studies on BACE1 inhibition, the prospect of identifying potent molecules that would reach the approved level is feasible. Investigators should try pushing many of the identified BACE1 compounds with significant anti-AD properties to preclinical and clinical trial stages. We also advise computational research on allosteric inhibitor design, exosite modeling, and multisite inhibition of BACE1. These alternatives might be a solution to BACE1 drug discovery in AD therapy.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/therapeutic use , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Humans , Molecular Dynamics Simulation
13.
Mol Brain ; 11(1): 7, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426354

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease driven in large part by accumulated deposits in the brain of the amyloid precursor protein (APP) cleavage product amyloid-ß peptide (Aß). However, AD is also characterised by reductions in secreted amyloid precursor protein-alpha (sAPPα), an alternative cleavage product of APP. In contrast to the neurotoxicity of accumulated Αß, sAPPα has many neuroprotective and neurotrophic properties. Increasing sAPPα levels has the potential to serve as a therapeutic treatment that mitigates the effects of Aß and rescue cognitive function. Here we tested the hypothesis that lentivirus-mediated expression of a human sAPPα construct in a mouse model of AD (APPswe/PS1dE9), begun before the onset of plaque pathology, could prevent later behavioural and electrophysiological deficits. Male mice were given bilateral intra-hippocampal injections at 4 months of age and tested 8-10 months later. Transgenic mice expressing sAPPα performed significantly better than untreated littermates in all aspects of the spatial water maze task. Expression of sAPPα also resulted in partial rescue of long-term potentiation (LTP), tested in vitro. These improvements occurred in the absence of changes in amyloid pathology. Supporting these findings on LTP, lentiviral-mediated expression of sAPPα for 3 months from 10 months of age, or acute sAPPα treatment in hippocampal slices from 18 to 20 months old transgenic mice, completely reversed the deficits in LTP. Together these findings suggest that sAPPα has wide potential to act as either a preventative or restorative therapeutic treatment in AD by mitigating the effects of Aß toxicity and enhancing cognitive reserve.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/therapeutic use , Lentivirus/metabolism , Memory Disorders/drug therapy , Memory Disorders/physiopathology , Neuronal Plasticity , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Amyloid/drug effects , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/administration & dosage , Amyloid beta-Protein Precursor/pharmacology , Animals , Behavior, Animal , Biomarkers/metabolism , Disease Models, Animal , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Memory Disorders/pathology , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology , Synaptic Transmission/drug effects , Transduction, Genetic
15.
Bioconjug Chem ; 28(4): 1102-1114, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28151644

ABSTRACT

Antibody drug conjugates (ADCs) provide an efficacious and relatively safe means by which chemotherapeutic agents can be specifically targeted to cancer cells. In addition to the selection of antibody targets, ADCs offer a modular design that allows selection of ADC characteristics through the choice of linker chemistries, toxins, and conjugation sites. Many studies have indicated that release of toxins bound to antibodies via noncleavable linker chemistries relies on the internalization and intracellular trafficking of the ADC. While this can make noncleavable ADCs more stable in the serum, it can also result in lower efficacy when their respective targets are not internalized efficiently or are recycled back to the cell surface following internalization. Here, we show that a lysosomally targeted ADC against the protein APLP2 mediates cell killing, both in vitro and in vivo, more effectively than an ADC against Trop2, a protein with less efficient lysosomal targeting. We also engineered a bispecific ADC with one arm targeting HER2 for the purpose of directing the ADC to tumors, and the other arm targeting APLP2, whose purpose is to direct the ADC to lysosomes for toxin release. This proof-of-concept bispecific ADC demonstrates that this technology can be used to shift the intracellular trafficking of a constitutively recycled target by directing one arm of the antibody against a lysosomally delivered protein. Our data also show limitations of this approach and potential future directions for development.


Subject(s)
Drug Delivery Systems , Immunoconjugates/pharmacology , Lysosomes/metabolism , Transcytosis , Amyloid beta-Protein Precursor/immunology , Amyloid beta-Protein Precursor/therapeutic use , Animals , Antibodies, Bispecific/therapeutic use , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Immunoconjugates/metabolism , Mice, Nude , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/therapeutic use , Receptor, ErbB-2/immunology , Receptor, ErbB-2/therapeutic use
16.
Neurol Sci ; 36(4): 593-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25380623

ABSTRACT

This study was performed to understand whether P165 improves learning and memory by restoring insulin action using a diabetes mellitus (DM) rat model. A total of 34 male Sprague-Dawley rats were randomly divided into four groups: control group (n = 8), DM group (n = 8), DM group treated with a low dose of P165 (n = 9), and DM group treated with a high dose of P165 (n = 9). After 8 weeks of treatment, the animals were killed and the expression of insulin signaling-related proteins was examined in the hippocampus by Western blot and immunohistochemical staining. Administration of P165 in diabetic rats did not induce a significant effect on the fasting blood glucose level. The expression of IR, IRS-1, AKT, p-CREB, and Bcl-2 proteins was significantly enhanced in the hippocampus in diabetic rats. Treatment of diabetic rats with P165 at both low and high doses significantly attenuated the expression levels of these proteins. Moreover, immunohistochemistry staining showed that IR, IRS-1, AKT, p-CREB, and Bcl-2 were abundantly expressed in the CA1 region of the hippocampus. The number of cells positively stained for the above proteins was significantly higher in diabetic tissues compared to control tissues, whereas P165 treatments induced a significant reduction in the expression of these proteins. The expression of IR, IRS-1, AKT, p-CREB, and Bcl-2 was enhanced in DM rats, and administration of P165 normalized the expression of these molecules, suggesting that P165 can improve impaired insulin signal transduction.


Subject(s)
Amyloid beta-Protein Precursor/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Peptide Fragments/therapeutic use , Signal Transduction/drug effects , Amyloid beta-Protein Precursor/pharmacology , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , CREB-Binding Protein/metabolism , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Male , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Insulin/metabolism , Streptozocin/toxicity
17.
J Neurochem ; 128(1): 196-204, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23919582

ABSTRACT

We have previously shown that following traumatic brain injury (TBI) the presence of the amyloid precursor protein (APP) may be neuroprotective. APP knockout mice have increased neuronal death and worse cognitive and motor outcomes following TBI, which is rescued by treatment with exogenous sAPPα (the secreted ectodomain of APP generated by α-secretase cleavage). Two neuroprotective regions were identified in sAPPα, the N and C-terminal domains D1 and D6a/E2 respectively. As both D1 and D6a/E2 contain heparin binding activity it was hypothesized that this is responsible for the neuroprotective activity. In this study, we focused on the heparin binding site, encompassed by residues 96-110 in D1, which has previously been shown to have neurotrophic properties. We found that treatment with APP96-110 rescued motor and cognitive deficits in APP-/- mice following focal TBI. APP96-110 also provided neuroprotection in Sprague-Dawley rats following diffuse TBI. Treatment with APP96-110 significantly improved functional outcome as well as preserve histological cellular morphology in APP-/- mice following focal controlled cortical impact injury. Furthermore, following administration of APP96-110 in rats after diffuse impact acceleration TBI, motor and cognitive outcomes were significantly improved and axonal injury reduced. These data define the heparin binding site in the D1 domain of sAPPα, represented by the sequence APP96-110, as the neuroprotective site to confer neuroprotection following TBI. The product of α-secretase cleavage of the amyloid precursor protein, sAPPα is neuroprotective following traumatic brain injury (TBI). Of interest was whether this neuroprotective activity could be further delineated to a heparin binding region within sAPPα, corresponding to the region APP96-110 (see diagram demonstrating the domain structure of sAPPα). Indeed treatment with APP96-110 improved functional outcome following TBI, an effect that was not seen with a mutated version of the peptide that had reduced heparin binding affinity.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain Injuries/metabolism , Brain Injuries/prevention & control , Heparin/metabolism , Neuroprotective Agents/metabolism , Amino Acid Sequence , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/therapeutic use , Animals , Binding Sites/physiology , Heparin/chemistry , Heparin/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Neuroprotective Agents/therapeutic use , Protein Structure, Tertiary , Random Allocation , Rats , Rats, Sprague-Dawley
18.
Biomed Res Int ; 2013: 709145, 2013.
Article in English | MEDLINE | ID: mdl-24089686

ABSTRACT

APP/PS1 double-transgenic mouse models of Alzheimer's disease (AD), which overexpress mutated forms of the gene for human amyloid precursor protein (APP) and presenilin 1 (PS1), have provided robust neuropathological hallmarks of AD-like pattern at early ages. This study characterizes immunocytochemical patterns of AD mouse brain as a model for human AD treated with the EB101 vaccine. In this novel vaccine, a new approach has been taken to circumvent past failures by judiciously selecting an adjuvant consisting of a physiological matrix embedded in liposomes, composed of naturally occurring phospholipids (phosphatidylcholine, phosphatidylglycerol, and cholesterol). Our findings showed that administration of amyloid-ß1₋42 (Aß) and sphingosine-1-phosphate emulsified in liposome complex (EB101) to APP/PS1 mice before onset of Aß deposition (7 weeks of age) and/or at an older age (35 weeks of age) is effective in halting the progression and clearing the AD-like neuropathological hallmarks. Passive immunization with EB101 did not activate inflammatory responses from the immune system and astrocytes. Consistent with a decreased inflammatory background, the basal immunological interaction between the T cells and the affected areas (hippocampus) in the brain of treated mice was notably reduced. These results demonstrate that immunization with EB101 vaccine prevents and attenuates AD neuropathology in this type of double-transgenic mice.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/immunology , Amyloid beta-Peptides/genetics , Presenilin-1/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/immunology , Amyloid beta-Protein Precursor/therapeutic use , Animals , Astrocytes/drug effects , Astrocytes/immunology , Disease Models, Animal , Humans , Immunotherapy, Active , Mice , Mice, Transgenic , Presenilin-1/immunology
19.
J Diabetes Res ; 2013: 689841, 2013.
Article in English | MEDLINE | ID: mdl-23844373

ABSTRACT

Researchers have proposed that amyloid precursor protein 17 peptide (APP17 peptide), an active fragment of amyloid precursor protein (APP) in the nervous system, has therapeutic effects on neurodegeneration. Diabetic encephalopathy (DE) is a neurological disease caused by diabetes. Here we use multiple experimental approaches to investigate the effect of APP17 peptide on changes in learning behavior and glycol metabolism in rats. It was found that rats with DE treated by APP17 peptide showed reversed behavioral alternation. The [(18)F]-FDG-PET images and other results all showed that the APP17 peptide could promote glucose metabolism in the brain of the DE rat model. Meanwhile, the insulin signaling was markedly increased as shown by increased phosphorylation of Akt and enhanced GLUT4 activation. Compared with the DE group, the activities of SOD, GSH-Px, and CAT in the rat hippocampal gyrus were increased, while MDA decreased markedly in the DE + APP17 peptide group. No amyloid plaques in the cortex and the hippocampus were detected in either group, indicating that the experimental animals in the current study were not suffering from Alzheimer's disease. These results indicate that APP17 peptide could be used to treat DE effectively.


Subject(s)
Amyloid beta-Protein Precursor/therapeutic use , Brain/drug effects , Diabetic Neuropathies/prevention & control , Glycols/metabolism , Nerve Degeneration/prevention & control , Peptide Fragments/therapeutic use , Amyloid beta-Protein Precursor/pharmacology , Animals , Behavior, Animal/drug effects , Blood Glucose/metabolism , Brain/metabolism , Cell Proliferation/drug effects , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , Learning/drug effects , Male , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects
20.
PLoS One ; 7(4): e34209, 2012.
Article in English | MEDLINE | ID: mdl-22545081

ABSTRACT

Amyloid precursor protein (APP) plays a pivotal role in Alzheimer's disease (AD) pathogenesis, but its normal physiological functions are less clear. Combined deletion of the APP and APP-like protein 2 (APLP2) genes in mice results in post-natal lethality, suggesting that APP performs an essential, if redundant, function during embryogenesis. We previously showed that injection of antisense morpholino to reduce APP levels in zebrafish embryos caused convergent-extension defects. Here we report that a reduction in APP levels causes defective axonal outgrowth of facial branchiomotor and spinal motor neurons, which involves disorganized axonal cytoskeletal elements. The defective outgrowth is caused in a cell-autonomous manner and both extracellular and intracellular domains of human APP are required to rescue the defective phenotype. Interestingly, wild-type human APP rescues the defective phenotype but APPswe mutation, which causes familial AD, does not. Our results show that the zebrafish model provides a powerful system to delineate APP functions in vivo and to study the biological effects of APP mutations.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Axons/pathology , Gene Knockdown Techniques , Motor Neurons/pathology , Zebrafish/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/therapeutic use , Animals , Axons/metabolism , Cells, Cultured , Gene Expression Regulation, Developmental , Humans , Motor Neurons/metabolism , Mutation , Phenotype , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...