Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Matthew J. Mazzella is active.

Publication


Featured researches published by Matthew J. Mazzella.


Journal of Neurochemistry | 2009

Age-dependent dysregulation of brain amyloid precursor protein in the Ts65Dn Down syndrome mouse model

Jennifer H.K. Choi; Jason D. Berger; Matthew J. Mazzella; Jose Morales-Corraliza; Anne M. Cataldo; Ralph A. Nixon; Stephen D. Ginsberg; Efrat Levy; Paul M. Mathews

Individuals with Down syndrome develop β‐amyloid deposition characteristic of early‐onset Alzheimer’s disease (AD) in mid‐life, presumably because of an extra copy of the chromosome 21‐located amyloid precursor protein (App) gene. App mRNA and APP metabolite levels were assessed in the brains of Ts65Dn mice, a mouse model of Down syndrome, using quantitative PCR, western blot analysis, immunoprecipitation, and ELISAs. In spite of the additional App gene copy, App mRNA, APP holoprotein, and all APP metabolite levels in the brains of 4‐month‐old trisomic mice were not increased compared with the levels seen in diploid littermate controls. However starting at 10 months of age, brain APP levels were increased proportional to the App gene dosage imbalance reflecting increased App message levels in Ts65Dn mice. Similar to APP levels, soluble amino‐terminal fragments of APP (sAPPα and sAPPβ) were increased in Ts65Dn mice compared with diploid mice at 12 months but not at 4 months of age. Brain levels of both Aβ40 and Aβ42 were not increased in Ts65Dn mice compared with diploid mice at all ages examined. Therefore, multiple mechanisms contribute to the regulation towards diploid levels of APP metabolites in the Ts65Dn mouse brain.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Modeling familial Danish dementia in mice supports the concept of the amyloid hypothesis of Alzheimer's disease

Janaky Coomaraswamy; Ellen Kilger; Heidrun Wölfing; Claudia Schäfer; Stephan A. Kaeser; Bettina M. Wegenast-Braun; Jasmin K. Hefendehl; Hartwig Wolburg; Matthew J. Mazzella; Jorge Ghiso; Michel Goedert; Haruhiko Akiyama; Francisco García-Sierra; David P. Wolfer; Paul M. Mathews; Mathias Jucker

Familial Danish dementia (FDD) is a progressive neurodegenerative disease with cerebral deposition of Dan-amyloid (ADan), neuroinflammation, and neurofibrillary tangles, hallmark characteristics remarkably similar to those in Alzheimers disease (AD). We have generated transgenic (tg) mouse models of familial Danish dementia that exhibit the age-dependent deposition of ADan throughout the brain with associated amyloid angiopathy, microhemorrhage, neuritic dystrophy, and neuroinflammation. Tg mice are impaired in the Morris water maze and exhibit increased anxiety in the open field. When crossed with TauP301S tg mice, ADan accumulation promotes neurofibrillary lesions, in all aspects similar to the Tau lesions observed in crosses between β-amyloid (Aβ)-depositing tg mice and TauP301S tg mice. Although these observations argue for shared mechanisms of downstream pathophysiology for the sequence-unrelated ADan and Aβ peptides, the lack of codeposition of the two peptides in crosses between ADan- and Aβ-depositing mice points also to distinguishing properties of the peptides. Our results support the concept of the amyloid hypothesis for AD and related dementias, and suggest that different proteins prone to amyloid formation can drive strikingly similar pathogenic pathways in the brain.


PLOS ONE | 2009

In Vivo Turnover of Tau and APP Metabolites in the Brains of Wild-Type and Tg2576 Mice: Greater Stability of sAPP in the β-Amyloid Depositing Mice

Jose Morales-Corraliza; Matthew J. Mazzella; Jason D. Berger; Nicole S. Diaz; Jennifer H.K. Choi; Efrat Levy; Yasuji Matsuoka; Emmanuel Planel; Paul M. Mathews

The metabolism of the amyloid precursor protein (APP) and tau are central to the pathobiology of Alzheimers disease (AD). We have examined the in vivo turnover of APP, secreted APP (sAPP), Aβ and tau in the wild-type and Tg2576 mouse brain using cycloheximide to block protein synthesis. In spite of overexpression of APP in the Tg2576 mouse, APP is rapidly degraded, similar to the rapid turnover of the endogenous protein in the wild-type mouse. sAPP is cleared from the brain more slowly, particularly in the Tg2576 model where the half-life of both the endogenous murine and transgene-derived human sAPP is nearly doubled compared to wild-type mice. The important Aβ degrading enzymes neprilysin and IDE were found to be highly stable in the brain, and soluble Aβ40 and Aβ42 levels in both wild-type and Tg2576 mice rapidly declined following the depletion of APP. The cytoskeletal-associated protein tau was found to be highly stable in both wild-type and Tg2576 mice. Our findings unexpectedly show that of these various AD-relevant protein metabolites, sAPP turnover in the brain is the most different when comparing a wild-type mouse and a β-amyloid depositing, APP overexpressing transgenic model. Given the neurotrophic roles attributed to sAPP, the enhanced stability of sAPP in the β-amyloid depositing Tg2576 mice may represent a neuroprotective response.


Neurobiology of Aging | 2013

Immunization targeting a minor plaque constituent clears β-amyloid and rescues behavioral deficits in an Alzheimer's disease mouse model

Jose Morales-Corraliza; Stephen D. Schmidt; Matthew J. Mazzella; Jason D. Berger; Donald A. Wilson; Daniel W. Wesson; Mathias Jucker; Efrat Levy; Ralph A. Nixon; Paul M. Mathews

Although anti-human β-amyloid (Aβ) immunotherapy clears brain β-amyloid plaques in Alzheimers disease (AD), targeting additional brain plaque constituents to promote clearance has not been attempted. Endogenous murine Aβ is a minor Aβ plaque component in amyloid precursor protein (APP) transgenic AD models, which we show is ∼3%-8% of the total accumulated Aβ in various human APP transgenic mice. Murine Aβ codeposits and colocalizes with human Aβ in amyloid plaques, and the two Aβ species coimmunoprecipitate together from brain extracts. In the human APP transgenic mouse model Tg2576, passive immunization for 8 weeks with a murine-Aβ-specific antibody reduced β-amyloid plaque pathology, robustly decreasing both murine and human Aβ levels. The immunized mice additionally showed improvements in two behavioral assays, odor habituation and nesting behavior. We conclude that passive anti-murine Aβ immunization clears Aβ plaque pathology--including the major human Aβ component--and decreases behavioral deficits, arguing that targeting minor endogenous brain plaque constituents can be beneficial, broadening the range of plaque-associated targets for AD therapeutics.


Methods of Molecular Biology | 2012

Aβ Measurement by Enzyme-Linked Immunosorbent Assay

Stephen D. Schmidt; Matthew J. Mazzella; Ralph A. Nixon; Paul M. Mathews

The neuritic plaque in the brain of Alzheimers disease patients consists of an amyloid composed primarily of Aβ, an approximately 4-kDa peptide derived from the amyloid precursor protein. Multiple lines of evidence suggest that Aβ plays a key role in the pathogenesis of the disease, and potential treatments that target Aβ production and/or Aβ accumulation in the brain as β-amyloid are being aggressively pursued. Methods to quantitate the Aβ peptide are, therefore, invaluable to most studies aimed at a better understanding of the molecular etiology of the disease and in assessing potential therapeutics. Although other techniques have been used to measure Aβ in the brains of AD patients and β-amyloid-depositing transgenic mice, the enzyme-linked immunosorbent assay (ELISA) is one of the most commonly used, reliable, and sensitive methods for quantitating the Aβ peptide. Here we describe methods for the recovery of both soluble and deposited Aβ from brain tissue and the subsequent quantitation of the peptide by sandwich ELISA.


The Journal of Neuroscience | 2016

Brain-Wide Insulin Resistance, Tau Phosphorylation Changes, and Hippocampal Neprilysin and Amyloid-β Alterations in a Monkey Model of Type 1 Diabetes

Jose Morales-Corraliza; Harrison Wong; Matthew J. Mazzella; Shaoli Che; Sang Han Lee; Eva Petkova; Janice D. Wagner; Scott E. Hemby; Stephen D. Ginsberg; Paul M. Mathews

Epidemiological findings suggest that diabetic individuals are at a greater risk for developing Alzheimers disease (AD). To examine the mechanisms by which diabetes mellitus (DM) may contribute to AD pathology in humans, we examined brain tissue from streptozotocin-treated type 1 diabetic adult male vervet monkeys receiving twice-daily exogenous insulin injections for 8–20 weeks. We found greater inhibitory phosphorylation of insulin receptor substrate 1 in each brain region examined of the diabetic monkeys when compared with controls, consistent with a pattern of brain insulin resistance that is similar to that reported in the human AD brain. Additionally, a widespread increase in phosphorylated tau was seen, including brain areas vulnerable in AD, as well as relatively spared structures, such as the cerebellum. An increase in active ERK1/2 was also detected, consistent with DM leading to changes in tau-kinase activity broadly within the brain. In contrast to these widespread changes, we found an increase in soluble amyloid-β (Aβ) levels that was restricted to the temporal lobe, with the greatest increase seen in the hippocampus. Consistent with this localized Aβ increase, a hippocampus-restricted decrease in the protein and mRNA for the Aβ-degrading enzyme neprilysin (NEP) was found, whereas various Aβ-clearing and -degrading proteins were unchanged. Thus, we document multiple biochemical changes in the insulin-controlled DM monkey brain that can link DM with the risk of developing AD, including dysregulation of the insulin-signaling pathway, changes in tau phosphorylation, and a decrease in NEP expression in the hippocampus that is coupled with a localized increase in Aβ. SIGNIFICANCE STATEMENT Given that diabetes mellitus (DM) appears to increase the risk of developing Alzheimers disease (AD), understanding the mechanisms by which DM promotes AD is important. We report that DM in a nonhuman primate brain leads to changes in the levels or posttranslational processing of proteins central to AD pathobiology, including tau, amyloid-β (Aβ), and the Aβ-degrading protease neprilysin. Additional evidence from this model suggests that alterations in brain insulin signaling occurred that are reminiscent of insulin signaling pathway changes seen in human AD. Thus, in an in vivo model highly relevant to humans, we show multiple alterations in the brain resulting from DM that are mechanistically linked to AD risk.


Journal of Alzheimer's Disease | 2013

Early Endosomal Abnormalities and Cholinergic Neuron Degeneration in Amyloid-β Protein Precursor Transgenic Mice

Jennifer H.K. Choi; Gurjinder Kaur; Matthew J. Mazzella; Jose Morales-Corraliza; Efrat Levy; Paul M. Mathews

Early endosomal changes, a prominent pathology in neurons early in Alzheimers disease, also occur in neurons and peripheral tissues in Down syndrome. While in Down syndrome models increased amyloid-β protein precursor (AβPP) expression is known to be a necessary contributor on the trisomic background to this early endosomal pathology, increased AβPP alone has yet to be shown to be sufficient to drive early endosomal alterations in neurons. Comparing two AβPP transgenic mouse models, one that contains the AβPP Swedish K670N/M671L double mutation at the β-cleavage site (APP23) and one that has the AβPP London V717I mutation near the γ-cleavage site (APPLd2), we show significantly altered early endosome morphology in fronto-parietal neurons as well as enlargement of early endosomes in basal forebrain cholinergic neurons of the medial septal nucleus in the APP23 model, which has the higher levels of AβPP β-C-terminal fragment (βCTF) accumulation. Early endosomal changes correlate with a marked loss of the cholinergic population, which is consistent with the known dependence of the large projection cholinergic cells on endosome-mediated retrograde neurotrophic transport. Our findings support the idea that increased expression of AβPP and AβPP metabolites in neurons is sufficient to drive early endosomal abnormalities in vivo, and that disruption of the endocytic system is likely to contribute to basal forebrain cholinergic vulnerability.


Behavioural Brain Research | 2013

Chronic anti-murine Aβ immunization preserves odor guided behaviors in an Alzheimer's β-amyloidosis model.

Daniel W. Wesson; Jose Morales-Corraliza; Matthew J. Mazzella; Donald A. Wilson; Paul M. Mathews

Olfaction is often impaired in Alzheimers disease (AD) and is also dysfunctional in mouse models of the disease. We recently demonstrated that short-term passive anti-murine-Aβ immunization can rescue olfactory behavior in the Tg2576 mouse model overexpressing a human mutation of the amyloid precursor protein (APP) after β-amyloid deposition. Here we tested the ability to preserve normal olfactory behaviors by means of long-term passive anti-murine-Aβ immunization. Seven-month-old Tg2576 and non-transgenic littermate (NTg) mice were IP-injected biweekly with the m3.2 murine-Aβ-specific antibody until 16 mo of age when mice were tested in the odor habituation test. While Tg2576 mice treated with a control antibody showed elevations in odor investigation times and impaired odor habituation compared to NTg, olfactory behavior was preserved to NTg levels in m3.2-immunized Tg2576 mice. Immunized Tg2576 mice had significantly less β-amyloid immunolabeling in the olfactory bulb and entorhinal cortex, yet showed elevations in Thioflavin-S labeled plaques in the piriform cortex. No detectable changes in APP metabolite levels other than Aβ were found following m3.2 immunization. These results demonstrate efficacy of chronic, long-term anti-murine-Aβ m3.2 immunization in preserving normal odor-guided behaviors in a human APP Tg model. Further, these results provide mechanistic insights into olfactory dysfunction as a biomarker for AD by yielding evidence that focal reductions of Aβ may be sufficient to preserve olfaction.


PLOS ONE | 2014

Genetic deletion of transglutaminase 2 does not rescue the phenotypic deficits observed in R6/2 and zQ175 mouse models of Huntington's disease.

Liliana Menalled; Andrea E. Kudwa; Steve Oakeshott; Andrew K. Farrar; Neil G. Paterson; Igor Filippov; Sam Miller; Mei Kwan; Michael Hecht Olsen; Jose Manuel Beltran; Justin Torello; Jon Fitzpatrick; Richard Mushlin; Kimberly H. Cox; Kristi McConnell; Matthew J. Mazzella; Dansha He; Georgina F. Osborne; Rand Al-Nackkash; Gill P. Bates; Pasi Tuunanen; Kimmo Lehtimäki; Dani Brunner; Afshin Ghavami; Sylvie Ramboz; Larry Park; Douglas Macdonald; Ignacio Munoz-Sanjuan; David Howland

Huntingtons disease (HD) is an autosomal dominant, progressive neurodegenerative disorder caused by expansion of CAG repeats in the huntingtin gene. Tissue transglutaminase 2 (TG2), a multi-functional enzyme, was found to be increased both in HD patients and in mouse models of the disease. Furthermore, beneficial effects have been reported from the genetic ablation of TG2 in R6/2 and R6/1 mouse lines. To further evaluate the validity of this target for the treatment of HD, we examined the effects of TG2 deletion in two genetic mouse models of HD: R6/2 CAG 240 and zQ175 knock in (KI). Contrary to previous reports, under rigorous experimental conditions we found that TG2 ablation had no effect on either motor or cognitive deficits, or on the weight loss. In addition, under optimal husbandry conditions, TG2 ablation did not extend R6/2 lifespan. Moreover, TG2 deletion did not change the huntingtin aggregate load in cortex or striatum and did not decrease the brain atrophy observed in either mouse line. Finally, no amelioration of the dysregulation of striatal and cortical gene markers was detected. We conclude that TG2 is not a valid therapeutic target for the treatment of HD.


Frontiers in Behavioral Neuroscience | 2016

Cognitive Training at a Young Age Attenuates Deficits in the zQ175 Mouse Model of HD.

Paul C.P. Curtin; Andrew M. Farrar; Stephen Oakeshott; Jane Sutphen; Jason D. Berger; Matthew J. Mazzella; Kimberly H. Cox; Dansha He; William Alosio; Larry C. Park; David Howland; Daniela Brunner

Huntingtons Disease (HD) is a progressive neurodegenerative disorder that causes motor, cognitive, and psychiatric symptoms. In these experiments, we tested if operant training at an early age affected adult cognitive deficits in the zQ175 KI Het (zQ175) mouse model of HD. In Experiment 1 we trained zQ175 mice in a fixed-ratio/progressive ratio (FR/PR) task to assay learning and motivational deficits. We found pronounced deficits in response rates and task engagement in naïve adult zQ175 mice (32–33 weeks age), while deficits in zQ175 mice trained from 6–7 weeks age were either absent or less severe. When those mice were re-tested as adults, FR/PR performance deficits were absent or otherwise less severe than deficits observed in naïve adult zQ175 relative to wild type (WT) mice. In Experiment 2, we used a Go/No-go operant task to assess the effects of early cognitive testing on response inhibition deficits in zQ175 mice. We found that zQ175 mice that began testing at 7–8 weeks did not exhibit deficits in Go/No-go testing, but when re-tested at 28–29 weeks age exhibited an initial impairment that diminished with training. These transient deficits were nonetheless mild relative to deficits observed among adult zQ175 mice without prior testing experience. In Experiment 3 we trained mice in a two-choice visual discrimination test to evaluate cognitive flexibility. As in prior experiments, we found performance deficits were mild or absent in mice that started training at 6–9 weeks of age, while deficits in naive mice exposed to training at 28–29 weeks were severe. Re-testing mice at 28–29 weeks age, were previously trained starting at 6–9 weeks, revealed that deficits in learning and cognitive flexibility were absent or reduced relative to effects observed in naive adults. In Experiment 4, we tested working memory deficits with a delayed non-match to position (DNMTP) test. Mice with prior experience exhibited mild working memory deficits, with males zQ175 exhibiting no deficits, and females performing significantly worse than WT mice at a single delay interval, whereas naive zQ175 exhibited severe delay-dependent deficits at all intervals exceeding 1 s. In sum, these experiments indicate that CAG-dependent impairments in motivation, motor control, cognitive flexibility, and working memory are sensitive to the environmental enrichment and experience. These findings are of clinical relevance, as HD carrier status can potentially be detected at an early age.

Collaboration


Dive into the Matthew J. Mazzella's collaboration.

Top Co-Authors

Avatar

Paul M. Mathews

Nathan Kline Institute for Psychiatric Research

View shared research outputs
Top Co-Authors

Avatar

Jose Morales-Corraliza

Nathan Kline Institute for Psychiatric Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jason D. Berger

Nathan Kline Institute for Psychiatric Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jennifer H.K. Choi

Nathan Kline Institute for Psychiatric Research

View shared research outputs
Top Co-Authors

Avatar

Mathias Jucker

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stephen D. Schmidt

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Daniel W. Wesson

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge