Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Cell Host Microbe ; 29(9): 1437-1453.e8, 2021 09 08.
Article En | MEDLINE | ID: mdl-34428428

The SARS-CoV-2 pandemic has affected more than 185 million people worldwide resulting in over 4 million deaths. To contain the pandemic, there is a continued need for safe vaccines that provide durable protection at low and scalable doses and can be deployed easily. Here, AAVCOVID-1, an adeno-associated viral (AAV), spike-gene-based vaccine candidate demonstrates potent immunogenicity in mouse and non-human primates following a single injection and confers complete protection from SARS-CoV-2 challenge in macaques. Peak neutralizing antibody titers are sustained at 1 year and complemented by functional memory T cell responses. The AAVCOVID vector has no relevant pre-existing immunity in humans and does not elicit cross-reactivity to common AAVs used in gene therapy. Vector genome persistence and expression wanes following injection. The single low-dose requirement, high-yield manufacturability, and 1-month stability for storage at room temperature may make this technology well suited to support effective immunization campaigns for emerging pathogens on a global scale.


Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19 Vaccines/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/genetics , Dependovirus/genetics , Dependovirus/metabolism , Female , Humans , Immunogenicity, Vaccine/immunology , Immunologic Memory/immunology , Macaca fascicularis , Macaca mulatta , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology , Transgenes/genetics , Vaccination/methods , Viral Load/immunology
2.
bioRxiv ; 2021 Jan 19.
Article En | MEDLINE | ID: mdl-33442684

The SARS-CoV-2 pandemic has affected more than 70 million people worldwide and resulted in over 1.5 million deaths. A broad deployment of effective immunization campaigns to achieve population immunity at global scale will depend on the biological and logistical attributes of the vaccine. Here, two adeno-associated viral (AAV)-based vaccine candidates demonstrate potent immunogenicity in mouse and nonhuman primates following a single injection. Peak neutralizing antibody titers remain sustained at 5 months and are complemented by functional memory T-cells responses. The AAVrh32.33 capsid of the AAVCOVID vaccine is an engineered AAV to which no relevant pre-existing immunity exists in humans. Moreover, the vaccine is stable at room temperature for at least one month and is produced at high yields using established commercial manufacturing processes in the gene therapy industry. Thus, this methodology holds as a very promising single dose, thermostable vaccine platform well-suited to address emerging pathogens on a global scale.

3.
J Med Chem ; 61(24): 11021-11036, 2018 12 27.
Article En | MEDLINE | ID: mdl-30407821

Spinal muscular atrophy (SMA), a rare neuromuscular disorder, is the leading genetic cause of death in infants and toddlers. SMA is caused by the deletion or a loss of function mutation of the survival motor neuron 1 (SMN1) gene. In humans, a second closely related gene SMN2 exists; however it codes for a less stable SMN protein. In recent years, significant progress has been made toward disease modifying treatments for SMA by modulating SMN2 pre-mRNA splicing. Herein, we describe the discovery of LMI070/branaplam, a small molecule that stabilizes the interaction between the spliceosome and SMN2 pre-mRNA. Branaplam (1) originated from a high-throughput phenotypic screening hit, pyridazine 2, and evolved via multiparameter lead optimization. In a severe mouse SMA model, branaplam treatment increased full-length SMN RNA and protein levels, and extended survival. Currently, branaplam is in clinical studies for SMA.


Brain/drug effects , ERG1 Potassium Channel/metabolism , Muscular Atrophy, Spinal/drug therapy , Pyridazines/chemistry , Administration, Oral , Animals , Brain/metabolism , Cell Line , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , ERG1 Potassium Channel/antagonists & inhibitors , Humans , Mice, Inbred C57BL , Motor Neurons/drug effects , Muscular Atrophy, Spinal/genetics , Pyridazines/pharmacology , Quantitative Structure-Activity Relationship , RNA Splicing , Rats, Sprague-Dawley , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/genetics
4.
PLoS One ; 11(12): e0167077, 2016.
Article En | MEDLINE | ID: mdl-27907033

INTRODUCTION AND OBJECTIVE: Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disorder. SMA is caused by homozygous loss of the SMN1 gene and retention of the SMN2 gene resulting in reduced levels of full length SMN protein that are insufficient for motor neuron function. Various treatments that restore levels of SMN are currently in clinical trials and biomarkers are needed to determine the response to treatment. Here, we sought to investigate in SMA mice a set of plasma analytes, previously identified in patients with SMA to correlate with motor function. The goal was to determine whether levels of plasma markers were altered in the SMNΔ7 mouse model of SMA and whether postnatal SMN restoration resulted in normalization of the biomarkers. METHODS: SMNΔ7 and control mice were treated with antisense oligonucleotides (ASO) targeting ISS-N1 to increase SMN protein from SMN2 or scramble ASO (sham treatment) via intracerebroventricular injection on postnatal day 1 (P1). Brain, spinal cord, quadriceps muscle, and liver were analyzed for SMN protein levels at P12 and P90. Ten plasma biomarkers (a subset of biomarkers in the SMA-MAP panel available for analysis in mice) were analyzed in plasma obtained at P12, P30, and P90. RESULTS: Of the eight plasma biomarkers assessed, 5 were significantly changed in sham treated SMNΔ7 mice compared to control mice and were normalized in SMNΔ7 mice treated with ASO. CONCLUSION: This study defines a subset of the SMA-MAP plasma biomarker panel that is abnormal in the most commonly used mouse model of SMA. Furthermore, some of these markers are responsive to postnatal SMN restoration. These findings support continued clinical development of these potential prognostic and pharmacodynamic biomarkers.


Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Oligonucleotides, Antisense/genetics , Spinal Cord/metabolism , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Animals , Animals, Newborn , Biomarkers/metabolism , Brain/metabolism , Brain/pathology , Clinical Trials as Topic , Disease Models, Animal , Gene Expression Regulation , Genetic Complementation Test , Humans , Injections, Intraventricular , Liver/metabolism , Liver/pathology , Mice , Mice, Transgenic , Motor Neurons/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Oligonucleotides, Antisense/metabolism , Spinal Cord/pathology , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/metabolism
5.
Assay Drug Dev Technol ; 12(6): 315-41, 2014 Aug.
Article En | MEDLINE | ID: mdl-25147906

Spinal muscular atrophy (SMA) is an autosomal recessive genetic disorder resulting in degeneration of α-motor neurons of the anterior horn and proximal muscle weakness. It is the leading cause of genetic mortality in children younger than 2 years. It affects ∼1 in 11,000 live births. In 95% of cases, SMA is caused by homozygous deletion of the SMN1 gene. In addition, all patients possess at least one copy of an almost identical gene called SMN2. A single point mutation in exon 7 of the SMN2 gene results in the production of low levels of full-length survival of motor neuron (SMN) protein at amounts insufficient to compensate for the loss of the SMN1 gene. Although no drug treatments are available for SMA, a number of drug discovery and development programs are ongoing, with several currently in clinical trials. This review describes the assays used to identify candidate drugs for SMA that modulate SMN2 gene expression by various means. Specifically, it discusses the use of high-throughput screening to identify candidate molecules from primary screens, as well as the technical aspects of a number of widely used secondary assays to assess SMN messenger ribonucleic acid (mRNA) and protein expression, localization, and function. Finally, it describes the process of iterative drug optimization utilized during preclinical SMA drug development to identify clinical candidates for testing in human clinical trials.


Drug Discovery/methods , Genetic Therapy/methods , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Animals , Drug Discovery/trends , Genetic Therapy/trends , High-Throughput Screening Assays/methods , High-Throughput Screening Assays/trends , Humans , Muscular Atrophy, Spinal/diagnosis , Survival of Motor Neuron 2 Protein/genetics
6.
PLoS One ; 8(4): e60113, 2013.
Article En | MEDLINE | ID: mdl-23565191

OBJECTIVES: Spinal Muscular Atrophy (SMA) presents challenges in (i) monitoring disease activity and predicting progression, (ii) designing trials that allow rapid assessment of candidate therapies, and (iii) understanding molecular causes and consequences of the disease. Validated biomarkers of SMA motor and non-motor function would offer utility in addressing these challenges. Our objectives were (i) to discover additional markers from the Biomarkers for SMA (BforSMA) study using an immunoassay platform, and (ii) to validate the putative biomarkers in an independent cohort of SMA patients collected from a multi-site natural history study (NHS). METHODS: BforSMA study plasma samples (N = 129) were analyzed by immunoassay to identify new analytes correlating to SMA motor function. These immunoassays included the strongest candidate biomarkers identified previously by chromatography. We selected 35 biomarkers to validate in an independent cohort SMA type 1, 2, and 3 samples (N = 158) from an SMA NHS. The putative biomarkers were tested for association to multiple motor scales and to pulmonary function, neurophysiology, strength, and quality of life measures. We implemented a Tobit model to predict SMA motor function scores. RESULTS: 12 of the 35 putative SMA biomarkers were significantly associated (p<0.05) with motor function, with a 13(th) analyte being nearly significant. Several other analytes associated with non-motor SMA outcome measures. From these 35 biomarkers, 27 analytes were selected for inclusion in a commercial panel (SMA-MAP) for association with motor and other functional measures. CONCLUSIONS: Discovery and validation using independent cohorts yielded a set of SMA biomarkers significantly associated with motor function and other measures of SMA disease activity. A commercial SMA-MAP biomarker panel was generated for further testing in other SMA collections and interventional trials. Future work includes evaluating the panel in other neuromuscular diseases, for pharmacodynamic responsiveness to experimental SMA therapies, and for predicting functional changes over time in SMA patients.


Blood Proteins , Muscular Atrophy, Spinal/blood , Adolescent , Adult , Biomarkers/blood , Child , Child, Preschool , Humans , Infant , Mass Spectrometry , Middle Aged , Motor Activity , Muscular Atrophy, Spinal/diagnosis , Prognosis , Proteomics/methods , ROC Curve , Reagent Kits, Diagnostic , Reproducibility of Results , Young Adult
7.
PLoS One ; 7(11): e50763, 2012.
Article En | MEDLINE | ID: mdl-23226377

OBJECTIVES: Survival Motor Neuron (SMN) protein levels may become key pharmacodynamic (PD) markers in spinal muscular atrophy (SMA) clinical trials. SMN protein in peripheral blood mononuclear cells (PBMCs) can be quantified for trials using an enzyme-linked immunosorbent assay (ELISA). We developed protocols to collect, process, store and analyze these samples in a standardized manner for SMA clinical studies, and to understand the impact of age and intraindividual variability over time on PBMC SMN signal. METHODS: Several variables affecting SMN protein signal were evaluated using an ELISA. Samples were from healthy adults, adult with respiratory infections, SMA patients, and adult SMA carriers. RESULTS: Delaying PBMCs processing by 45 min, 2 hr or 24 hr after collection or isolation allows sensitive detection of SMN levels and high cell viability (>90%). SMN levels from PBMCs isolated by EDTA tubes/Lymphoprep gradient are stable with processing delays and have greater signal compared to CPT-collected samples. SMN signal in healthy individuals varies up to 8x when collected at intervals up to 1 month. SMN signals from individuals with respiratory infections show 3-5x changes, driven largely by the CD14 fraction. SMN signal in PBMC frozen lysates are relatively stable for up to 6 months. Cross-sectional analysis of PBMCs from SMA patients and carriers suggest SMN protein levels decline with age. CONCLUSIONS: The sources of SMN signal variability in PBMCs need to be considered in the design and of SMA clinical trials, and interpreted in light of recent medical history. Improved normalization to DNA or PBMC subcellular fractions may mitigate signal variability and should be explored in SMA patients.


Enzyme-Linked Immunosorbent Assay/methods , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , SMN Complex Proteins/metabolism , Adolescent , Adult , Carrier State/blood , Cell Count , Cell Separation , Child , Child, Preschool , Female , Humans , Infant , Leukocytes, Mononuclear/pathology , Male , Middle Aged , Muscular Atrophy, Spinal/blood , Respiratory Tract Infections/blood , Young Adult
8.
Hum Mol Genet ; 21(20): 4431-47, 2012 Oct 15.
Article En | MEDLINE | ID: mdl-22802075

A number of mouse models for spinal muscular atrophy (SMA) have been genetically engineered to recapitulate the severity of human SMA by using a targeted null mutation at the mouse Smn1 locus coupled with the transgenic addition of varying copy numbers of human SMN2 genes. Although this approach has been useful in modeling severe SMA and very mild SMA, a mouse model of the intermediate form of the disease would provide an additional research tool amenable for drug discovery. In addition, many of the previously engineered SMA strains are multi-allelic by design, containing a combination of transgenes and targeted mutations in the homozygous state, making further genetic manipulation difficult. A new genetic engineering approach was developed whereby variable numbers of SMN2 sequences were incorporated directly into the murine Smn1 locus. Using combinations of these alleles, we generated an allelic series of SMA mouse strains harboring no, one, two, three, four, five, six or eight copies of SMN2. We report here the characterization of SMA mutants in this series that displayed a range in disease severity from embryonic lethal to viable with mild neuromuscular deficits.


Muscular Atrophy, Spinal/genetics , Neuromuscular Junction/genetics , Alleles , Animals , Behavior, Animal , Disease Models, Animal , Genotype , Humans , Mice , Mice, Inbred Strains , Neuromuscular Junction/metabolism , Phenotype , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism
9.
PLoS One ; 7(4): e33572, 2012.
Article En | MEDLINE | ID: mdl-22558076

BACKGROUND: The universal presence of a gene (SMN2) nearly identical to the mutated SMN1 gene responsible for Spinal Muscular Atrophy (SMA) has proved an enticing incentive to therapeutics development. Early disappointments from putative SMN-enhancing agent clinical trials have increased interest in improving the assessment of SMN expression in blood as an early "biomarker" of treatment effect. METHODS: A cross-sectional, single visit, multi-center design assessed SMN transcript and protein in 108 SMA and 22 age and gender-matched healthy control subjects, while motor function was assessed by the Modified Hammersmith Functional Motor Scale (MHFMS). Enrollment selectively targeted a broad range of SMA subjects that would permit maximum power to distinguish the relative influence of SMN2 copy number, SMA type, present motor function, and age. RESULTS: SMN2 copy number and levels of full-length SMN2 transcripts correlated with SMA type, and like SMN protein levels, were lower in SMA subjects compared to controls. No measure of SMN expression correlated strongly with MHFMS. A key finding is that SMN2 copy number, levels of transcript and protein showed no correlation with each other. CONCLUSION: This is a prospective study that uses the most advanced techniques of SMN transcript and protein measurement in a large selectively-recruited cohort of individuals with SMA. There is a relationship between measures of SMN expression in blood and SMA type, but not a strong correlation to motor function as measured by the MHFMS. Low SMN transcript and protein levels in the SMA subjects relative to controls suggest that these measures of SMN in accessible tissues may be amenable to an "early look" for target engagement in clinical trials of putative SMN-enhancing agents. Full length SMN transcript abundance may provide insight into the molecular mechanism of phenotypic variation as a function of SMN2 copy number. TRIAL REGISTRY: Clinicaltrials.gov NCT00756821.


Biomarkers/metabolism , DNA Copy Number Variations/physiology , Muscular Atrophy, Spinal/metabolism , Age Factors , Analysis of Variance , Case-Control Studies , Cross-Sectional Studies , DNA Copy Number Variations/genetics , DNA Primers/genetics , Female , Humans , Male , Motor Activity/physiology , Muscular Atrophy, Spinal/genetics , Prospective Studies , Real-Time Polymerase Chain Reaction , Survival of Motor Neuron 2 Protein/metabolism
10.
PLoS One ; 7(4): e35462, 2012.
Article En | MEDLINE | ID: mdl-22558154

BACKGROUND: Spinal Muscular Atrophy (SMA) is a neurodegenerative motor neuron disorder resulting from a homozygous mutation of the survival of motor neuron 1 (SMN1) gene. The gene product, SMN protein, functions in RNA biosynthesis in all tissues. In humans, a nearly identical gene, SMN2, rescues an otherwise lethal phenotype by producing a small amount of full-length SMN protein. SMN2 copy number inversely correlates with disease severity. Identifying other novel biomarkers could inform clinical trial design and identify novel therapeutic targets. OBJECTIVE: To identify novel candidate biomarkers associated with disease severity in SMA using unbiased proteomic, metabolomic and transcriptomic approaches. MATERIALS AND METHODS: A cross-sectional single evaluation was performed in 108 children with genetically confirmed SMA, aged 2-12 years, manifesting a broad range of disease severity and selected to distinguish factors associated with SMA type and present functional ability independent of age. Blood and urine specimens from these and 22 age-matched healthy controls were interrogated using proteomic, metabolomic and transcriptomic discovery platforms. Analyte associations were evaluated against a primary measure of disease severity, the Modified Hammersmith Functional Motor Scale (MHFMS) and to a number of secondary clinical measures. RESULTS: A total of 200 candidate biomarkers correlate with MHFMS scores: 97 plasma proteins, 59 plasma metabolites (9 amino acids, 10 free fatty acids, 12 lipids and 28 GC/MS metabolites) and 44 urine metabolites. No transcripts correlated with MHFMS. DISCUSSION: In this cross-sectional study, "BforSMA" (Biomarkers for SMA), candidate protein and metabolite markers were identified. No transcript biomarker candidates were identified. Additional mining of this rich dataset may yield important insights into relevant SMA-related pathophysiology and biological network associations. Additional prospective studies are needed to confirm these findings, demonstrate sensitivity to change with disease progression, and assess potential impact on clinical trial design. TRIAL REGISTRY: Clinicaltrials.gov NCT00756821.


Muscular Atrophy, Spinal/genetics , Phenotype , Severity of Illness Index , Amino Acids/metabolism , Analysis of Variance , Biomarkers/blood , Biomarkers/urine , Case-Control Studies , Child , Child, Preschool , Chromatography, Liquid , Cross-Sectional Studies , Fatty Acids, Nonesterified/metabolism , Gas Chromatography-Mass Spectrometry , Gene Dosage/genetics , Gene Expression Profiling/methods , Humans , Metabolomics/methods , Motor Activity/physiology , Pilot Projects , Proteomics/methods , Survival of Motor Neuron 2 Protein/genetics , Tandem Mass Spectrometry
11.
J Mol Biol ; 421(4-5): 525-36, 2012 Aug 24.
Article En | MEDLINE | ID: mdl-22197375

Alzheimer's disease, the most common cause of dementia in the elderly and characterized by the deposition and accumulation of plaques, is composed in part of ß-amyloid (Aß) peptides, loss of neurons, and the accumulation of neurofibrillary tangles. Here, we describe ponezumab, a humanized monoclonal antibody, and show how it binds specifically to the carboxyl (C)-terminus of Aß40. Ponezumab can label Aß that is deposited in brain parenchyma found in sections from Alzheimer's disease casualties and in transgenic mouse models that overexpress Aß. Importantly, ponezumab does not label full-length, non-cleaved amyloid precursor protein on the cell surface. The C-terminal epitope of the soluble Aß present in the circulation appears to be available for ponezumab binding because systemic administration of ponezumab greatly elevates plasma Aß40 levels in a dose-dependent fashion after administration to a mouse model that overexpress human Aß. Administration of ponezumab to transgenic mice also led to a dose-dependent reduction in hippocampal amyloid load. To further explore the nature of ponezumab binding to Aß40, we determined the X-ray crystal structure of ponezumab in complex with Aß40 and found that the Aß40 carboxyl moiety makes extensive contacts with ponezumab. Furthermore, the structure-function analysis supported this critical requirement for carboxy group of AßV40 in the Aß-ponezumab interaction. These findings provide novel structural insights into the in vivo conformation of the C-terminus of Aß40 and the brain Aß-lowering efficacy that we observed following administration of ponezumab in transgenic mouse models.


Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amino Acid Sequence , Amyloid beta-Peptides/blood , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Brain/pathology , Crystallography, X-Ray , Disease Models, Animal , Humans , Injections, Intravenous , Mice , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Neuroprotective Agents/administration & dosage , Plasma/chemistry , Protein Binding , Protein Conformation
12.
PLoS One ; 6(8): e24269, 2011.
Article En | MEDLINE | ID: mdl-21904622

OBJECTIVES: Genetic defects leading to the reduction of the survival motor neuron protein (SMN) are a causal factor for Spinal Muscular Atrophy (SMA). While there are a number of therapies under evaluation as potential treatments for SMA, there is a critical lack of a biomarker method for assessing efficacy of therapeutic interventions, particularly those targeting upregulation of SMN protein levels. Towards this end we have engaged in developing an immunoassay capable of accurately measuring SMN protein levels in blood, specifically in peripheral blood mononuclear cells (PBMCs), as a tool for validating SMN protein as a biomarker in SMA. METHODS: A sandwich enzyme-linked immunosorbent assay (ELISA) was developed and validated for measuring SMN protein in human PBMCs and other cell lysates. Protocols for detection and extraction of SMN from transgenic SMA mouse tissues were also developed. RESULTS: The assay sensitivity for human SMN is 50 pg/mL. Initial analysis reveals that PBMCs yield enough SMN to analyze from blood volumes of less than 1 mL, and SMA Type I patients' PBMCs show ∼90% reduction of SMN protein compared to normal adults. The ELISA can reliably quantify SMN protein in human and mouse PBMCs and muscle, as well as brain, and spinal cord from a mouse model of severe SMA. CONCLUSIONS: This SMN ELISA assay enables the reliable, quantitative and rapid measurement of SMN in healthy human and SMA patient PBMCs, muscle and fibroblasts. SMN was also detected in several tissues in a mouse model of SMA, as well as in wildtype mouse tissues. This SMN ELISA has general translational applicability to both preclinical and clinical research efforts.


Enzyme-Linked Immunosorbent Assay/methods , Muscular Atrophy, Spinal/metabolism , SMN Complex Proteins/analysis , Adult , Aged , Aged, 80 and over , Animals , Cells, Cultured , Female , Humans , In Vitro Techniques , Male , Mice , Reproducibility of Results , SMN Complex Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 108(28): E279-87, 2011 Jul 12.
Article En | MEDLINE | ID: mdl-21690377

Age-related macular degeneration (AMD) is a leading cause of visual dysfunction worldwide. Amyloid ß (Aß) peptides, Aß1-40 (Aß40) and Aß1-42 (Aß42), have been implicated previously in the AMD disease process. Consistent with a pathogenic role for Aß, we show here that a mouse model of AMD that invokes multiple factors that are known to modify AMD risk (aged human apolipoprotein E 4 targeted replacement mice on a high-fat, cholesterol-enriched diet) presents with Aß-containing deposits basal to the retinal pigmented epithelium (RPE), histopathologic changes in the RPE, and a deficit in scotopic electroretinographic response, which is reflective of impaired visual function. Strikingly, these electroretinographic deficits are abrogated in a dose-dependent manner by systemic administration of an antibody targeting the C termini of Aß40 and Aß42. Concomitant reduction in the levels of Aß and activated complement components in sub-RPE deposits and structural preservation of the RPE are associated with anti-Aß40/42 antibody immunotherapy and visual protection. These observations are consistent with the reduction in amyloid plaques and improvement of cognitive function in mouse models of Alzheimer's disease treated with anti-Aß antibodies. They also implicate Aß in the pathogenesis of AMD and identify Aß as a viable therapeutic target for its treatment.


Amyloid beta-Peptides/antagonists & inhibitors , Macular Degeneration/therapy , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/therapeutic use , Apolipoprotein E4/genetics , Complement System Proteins/metabolism , Dietary Fats/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Immunologic , Female , Humans , Immunotherapy , Macular Degeneration/etiology , Macular Degeneration/pathology , Macular Degeneration/physiopathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/immunology , Vision, Low/physiopathology , Vision, Low/prevention & control
14.
J Neurosci ; 29(15): 4964-71, 2009 Apr 15.
Article En | MEDLINE | ID: mdl-19369565

In amyloid precursor protein (APP) models of amyloid deposition, the amount of amyloid deposits increase with mouse age. At a first approximation, the extent of amyloid accumulation may either reflect small excesses of production over clearance that accumulate over time or, alternatively, indicate a steady-state equilibrium at that age, reflecting the instantaneous excess of production over clearance, which increases as the organism ages. To discriminate between these options, we reversibly suppressed amyloid deposition in Tg2576 mice with the anti-Abeta antibody 2H6, starting at 8 months, just before the first histological deposits can be discerned. Six months later, we stopped the suppression and monitored the progression of amyloid accumulation in control APP mice and suppressed APP mice over the next 3 months. The accumulation hypothesis would predict that the rate of amyloid from 14 to 17 months would be similar in the suppressed and control mice, while the equilibrium hypothesis would predict that the increase would be faster in the suppressed group, possibly catching up completely with the control mice. The results strongly support the accumulation hypothesis, with no evidence of the suppressed mice catching up with the control mice as predicted by equilibrium models. If anything, there was a slower rate of increase in the suppressed APP mice than the control mice, suggesting that a slow seeding mechanism likely precedes a rapid fibrillogenesis in determining the extent of amyloid deposition.


Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease/prevention & control , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/immunology , Animals , Antibodies/administration & dosage , Brain/metabolism , Brain/pathology , Drosophila Proteins/administration & dosage , Mice , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Time Factors
15.
Learn Mem ; 15(9): 625-32, 2008 Sep.
Article En | MEDLINE | ID: mdl-18772249

Two experiments were conducted to investigate the possibility of faster forgetting by PDAPP mice (a well-established model of Alzheimer's disease as reported by Games and colleagues in an earlier paper). Experiment 1, using mice aged 13-16 mo, confirmed the presence of a deficit in a spatial reference memory task in the water maze by hemizygous PDAPP mice relative to littermate controls. However, after overtraining to a criterion of equivalent navigational performance, a series of memory retention tests revealed faster forgetting in the PDAPP group. Very limited retraining was sufficient to reinstate good memory in both groups, indicating that their faster forgetting may be due to retrieval failure rather than trace decay. In Experiment 2, 6-mo-old PDAPP and controls were required to learn each of a series of spatial locations to criterion with their memory assessed 10 min after learning each location. No memory deficit was apparent in the PDAPP mice initially, but a deficit built up through the series of locations suggestive of increased sensitivity to interference. Faster forgetting and increased interference may each reflect a difficulty in accessing memory traces. This interpretation of one aspect of the cognitive deficit in human mutant APP mice has parallels to deficits observed in patients with Alzheimer's disease, further supporting the validity of transgenic models of the disease.


Alzheimer Disease/physiopathology , Memory/physiology , Spatial Behavior/physiology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Disease Models, Animal , Humans , Immunohistochemistry , Maze Learning/physiology , Mice , Mice, Transgenic
16.
Neurobiol Aging ; 29(6): 861-73, 2008 Jun.
Article En | MEDLINE | ID: mdl-17331621

Accumulation of cerebral amyloid-beta (Abeta) has been implicated as a putative causal factor in the development of Alzheimer's disease (AD). Transgenic mice like the PDAPP line overexpress human mutant Amyloid Precursor Protein (hAPP) and recapitulate many features of AD, including amyloid neuropathology and cognitive deficits. Inhibition of the beta-site aspartyl cleaving enzyme (BACE1) enzyme responsible for the first proteolytic cleavage that ultimately generates Abeta has been proposed as a strategy for AD therapy. To assess the theoretical repercussions of beta-secretase activity reduction in an in vivo model of AD, BACE1(-/-) mice bred to the PDAPP line were examined in a series of behavioral tasks. Although BACE1 gene ablation abolished hAbeta accumulation, BACE1(-/-) mice had unexpected sensorimotor impairments, spatial memory deficits, and displayed seizures, phenotypes which were severe on the PDAPP background. These results suggest that while excess Abeta is functionally pathological, BACE1-mediated processing of APP and other substrates play a role in "normal" learning, memory and sensorimotor processes.


Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/genetics , Aspartic Acid Endopeptidases/genetics , Behavior, Animal , Mental Disorders/physiopathology , Alzheimer Disease/complications , Animals , Gene Deletion , Mental Disorders/complications , Mice , Mice, Knockout
17.
J Neurosci ; 27(10): 2654-62, 2007 Mar 07.
Article En | MEDLINE | ID: mdl-17344403

The behavioral and biochemical impact of active immunization against human beta-amyloid (Abeta) was assessed using male transgenic (Tg) mice overexpressing a human mutant amyloid precursor protein (heterozygous PDAPP mice) and littermate controls. Administration of aggregated Abeta42 occurred at monthly intervals from 7 months ("prevention") or 11 months ("reversal"), followed by double-blind behavioral training at 16 months on a cued task, then serial spatial learning in a water maze. Using a 2 x 2 design, with Abeta42 adjuvanted with MPL-AF (adjuvant formulation of monophosphoryl lipid A) or MPL-AF alone, PDAPP mice were impaired compared with non-Tg littermates on two separate measures of serial spatial learning. Immunization caused no overall rescue of learning but limited the accumulation of total Abeta and Abeta42 levels in cortex and hippocampus by up to 60%. In immunized PDAPP mice, significant negative correlations were observed between hippocampal and cortical Abeta levels and learning capacity, particularly in the prevention study, and correlations between learning capacity and antibody titer. Moreover, a subset of PDAPP mice with very low Abeta levels (hippocampal Abeta levels of <6000 ng/g or cortical Abeta levels of <1000 ng/g) was indistinguishable from non-Tg controls. Mice in the prevention study were also rescued from cognitive impairment more effectively than those in the reversal study. The combination of variability in antibody response and differential levels of Abeta accumulation across the population of immunized PDAPP mice may be responsible for success in cognitive protection with only a subset of these animals, but the similarity to the findings of certain human vaccination trials is noteworthy.


Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Immunization , Maze Learning/physiology , Mutation , Peptide Fragments/immunology , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Animals , Antibodies/blood , Behavior, Animal/physiology , Cerebral Cortex/metabolism , Cues , Hippocampus/metabolism , Humans , Male , Mice , Mice, Transgenic , Peptide Fragments/metabolism , Swimming , Task Performance and Analysis
18.
J Alzheimers Dis ; 9(3 Suppl): 133-49, 2006.
Article En | MEDLINE | ID: mdl-16914852

Progress in understanding and treating Alzheimer's disease (AD) has been tremendously bolstered by the era of transgenic models of AD. The identification of disease-causing mutations in proteins such as amyloid-beta precursor protein (betaAPP) and presenilin1 (PS1), together with the discovery of other high risk factors (e.g., Apolipoprotein E4), as well as pathogenic mutations in the tau protein has led to the creation of several transgenic mice, including those expressing bi- and tri-genic constructs. Each model has unique pathologies that provide insights into disease mechanisms and interactive features of neuropathologic cascades. More importantly, therapeutic hypotheses are now testable in a manner unheard of less than 15 years ago. The wealth of new approaches currently in clinical and preclinical evaluations can be directly attributed to the impact of these animals on our ability to model relevant aspects of the disease. As a result, we may see containment or even the elimination of AD in the near future as a direct consequence of these advances.


Alzheimer Disease/pathology , Brain/pathology , Disease Models, Animal , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/immunology , Brain/metabolism , Immunization/methods , Mice , Mice, Transgenic , Nerve Degeneration/pathology , Plaque, Amyloid/pathology , Vaccines/therapeutic use
...