Network


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

Hotspot


Dive into the research topics where Kelly R. Bales is active.

Publication


Featured researches published by Kelly R. Bales.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Peripheral anti-Aβ antibody alters CNS and plasma Aβ clearance and decreases brain Aβ burden in a mouse model of Alzheimer's disease

Ronald B. DeMattos; Kelly R. Bales; David J. Cummins; Jean-Cosme Dodart; Steven M. Paul; David M. Holtzman

Active immunization with the amyloid β (Aβ) peptide has been shown to decrease brain Aβ deposition in transgenic mouse models of Alzheimers disease and certain peripherally administered anti-Aβ antibodies were shown to mimic this effect. In exploring factors that alter Aβ metabolism and clearance, we found that a monoclonal antibody (m266) directed against the central domain of Aβ was able to bind and completely sequester plasma Aβ. Peripheral administration of m266 to PDAPP transgenic mice, in which Aβ is generated specifically within the central nervous system (CNS), results in a rapid 1,000-fold increase in plasma Aβ, due, in part, to a change in Aβ equilibrium between the CNS and plasma. Although peripheral administration of m266 to PDAPP mice markedly reduces Aβ deposition, m266 did not bind to Aβ deposits in the brain. Thus, m266 appears to reduce brain Aβ burden by altering CNS and plasma Aβ clearance.


Nature Neuroscience | 2002

Immunization reverses memory deficits without reducing brain Aβ burden in Alzheimer's disease model

Jean-Cosme Dodart; Kelly R. Bales; Kimberley S. Gannon; Stephen J. Greene; Ronald B. DeMattos; Chantal Mathis; Cynthia DeLong; Su Wu; Xin Wu; David M. Holtzman; Steven M. Paul

We have previously shown that chronic treatment with the monoclonal antibody m266, which is specific for amyloid β-peptide (Aβ), increases plasma concentrations of Aβ and reduces Aβ burden in the PDAPP transgenic mouse model of Alzheimers disease (AD). We now report that administration of m266 to PDAPP mice can rapidly reverse memory deficits in both an object recognition task and a holeboard learning and memory task, but without altering brain Aβ burden. We also found that an Aβ/antibody complex was present in both the plasma and the cerebrospinal fluid of m266-treated mice. Our data indicate that passive immunization with this anti-Aβ monoclonal antibody can very rapidly reverse memory impairment in certain learning and memory tasks in the PDAPP mouse model of AD, owing perhaps to enhanced peripheral clearance and (or) sequestration of a soluble brain Aβ species.


Neuron | 2005

Synaptic Activity Regulates Interstitial Fluid Amyloid-β Levels In Vivo

John R. Cirrito; Kelvin A. Yamada; Mary Beth Finn; Robert S. Sloviter; Kelly R. Bales; Patrick May; Darryle D. Schoepp; Steven M. Paul; Steven Mennerick; David M. Holtzman

Summary Aggregation of the amyloid-β (Aβ) peptide in the extracellular space of the brain is central to Alzheimers disease pathogenesis. Aβ aggregation is concentration dependent and brain region specific. Utilizing in vivo microdialysis concurrently with field potential recordings, we demonstrate that Aβ levels in the brain interstitial fluid are dynamically and directly influenced by synaptic activity on a timescale of minutes to hours. Using an acute brain slice model, we show that the rapid effects of synaptic activity on Aβ levels are primarily related to synaptic vesicle exocytosis. These results suggest that synaptic activity may modulate a neurodegenerative disease process, in this case by influencing Aβ metabolism and ultimately region-specific Aβ deposition. The findings also have important implications for treatment development.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Minocycline prevents nigrostriatal dopaminergic neurodegeneration in the MPTP model of Parkinson's disease

Yansheng Du; Zhizhong Ma; Suizhen Lin; Richard Dodel; Feng Gao; Kelly R. Bales; Lazarose C. Triarhou; Eyassu Chernet; Ken W. Perry; David L. Nelson; Susan Luecke; Lee A. Phebus; Frank P. Bymaster; Steven M. Paul

Parkinsons disease is a chronic neurodegenerative disorder characterized by the loss of dopamine neurons in the substantia nigra, decreased striatal dopamine levels, and consequent extrapyramidal motor dysfunction. We now report that minocycline, a semisynthetic tetracycline, recently shown to have neuroprotective effects in animal models of stroke/ischemic injury and Huntingtons disease, prevents nigrostriatal dopaminergic neurodegeneration in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinsons disease. Minocycline treatment also blocked dopamine depletion in the striatum as well as in the nucleus accumbens after MPTP administration. The neuroprotective effect of minocycline is associated with marked reductions in inducible NO synthase (iNOS) and caspase 1 expression. In vitro studies using primary cultures of mesencephalic and cerebellar granule neurons (CGN) and/or glia demonstrate that minocycline inhibits both 1-methyl-4-phenylpyridinium (MPP+)-mediated iNOS expression and NO-induced neurotoxicity, but MPP+-induced neurotoxicity is inhibited only in the presence of glia. Further, minocycline also inhibits NO-induced phosphorylation of p38 mitogen-activated protein kinase (MAPK) in CGN and the p38 MAPK inhibitor, SB203580, blocks NO toxicity of CGN. Our results suggest that minocycline blocks MPTP neurotoxicity in vivo by indirectly inhibiting MPTP/MPP+-induced glial iNOS expression and/or directly inhibiting NO-induced neurotoxicity, most likely by inhibiting the phosphorylation of p38 MAPK. Thus, NO appears to play an important role in MPTP neurotoxicity. Neuroprotective tetracyclines may be effective in preventing or slowing the progression of Parkinsons and other neurodegenerative diseases.


Science Translational Medicine | 2011

Human apoE isoforms differentially regulate brain amyloid-β peptide clearance

Joseph M. Castellano; Jungsu Kim; Floy R. Stewart; Hong Jiang; Ronald B. DeMattos; Bruce W. Patterson; Anne M. Fagan; John C. Morris; Carlos Cruchaga; Alison Goate; Kelly R. Bales; Steven M. Paul; Randall J. Bateman; David M. Holtzman

Human apoE4 increases the concentration of soluble Aβ in the brain by impairing its clearance. Clearing the Debris in Alzheimer’s Disease The strongest risk factor for developing the common sporadic form of Alzheimer’s disease (AD) that occurs in old age is the ε4 allele encoding apolipoprotein E4 (apoE4). Two ε4 alleles can lower the age of onset of AD by 10 to 15 years. In contrast, the ε2 allele decreases the risk of developing this neurodegenerative disorder. APOE is important for lipoprotein metabolism, but how it might be involved in AD has remained unclear. It has been suggested that the apoE4 isoform might somehow help to drive accumulation of the peptide amyloid-β (Aβ), which forms amyloid plaques in the brain that contribute to neuronal death and are the characteristic hallmark of AD. In a tour de force study in humans and mice, Holtzman and his team at Washington University in St. Louis now show that apoE4 contributes to Aβ accumulation in the brain not by affecting Aβ synthesis but by affecting its clearance. First, the authors looked at the Aβ concentration in the cerebrospinal fluid (CSF) of cognitively normal individuals under age 70 carrying different APOE genotypes. They found that those with the ε4/ε4 genotype had a much lower CSF Aβ concentration than did those with the protective ε2/ε3 genotype. A CSF Aβ concentration of less than 500 pg/ml is an indication that Aβ peptide is accumulating in the brain and thus is not moving into the CSF. Next, the researchers analyzed imaging data using a dye called Pittsburgh compound B that binds to amyloid plaques in the brain and showed that those individuals with the ε4/ε4 genotype bound more dye than did those with the other APOE genotypes. They then moved to a mouse model of AD in which the mice expressed one of the three human apoE isoforms. They measured Aβ concentrations in the interstitial fluid of these mice using in vivo microdialysis and then looked at stained hippocampal sections from these mice. They found greater Aβ concentrations in both interstitial fluid and the hippocampus in mice expressing the human apoE4 isoform than in animals expressing either the E3 or E2 isoforms. They discovered that this difference in Aβ concentration between the mice carrying different APOE genotypes was present in young as well as aged mice, suggesting that it predates the appearance of amyloid plaques. They then measured clearance of Aβ from the interstitial fluid of young mice and showed that those with the human apoE4 isoform were less able to clear Aβ than those with the apoE2 or apoE3 isoforms. The researchers showed that processing of the amyloid precursor protein and generation of the Aβ peptide did not vary according to genotype, lending credence to the hypothesis that apoE4 affects clearance of Aβ but not its synthesis. This thorough study sheds new light on how apoE4 is implicated in AD and highlights the Aβ clearance pathway as a new target for developing drugs to slow or even halt the accumulation of amyloid plaques in patients with AD. The apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late-onset, sporadic Alzheimer’s disease (AD). The APOE ε4 allele markedly increases AD risk and decreases age of onset, likely through its strong effect on the accumulation of amyloid-β (Aβ) peptide. In contrast, the APOE ε2 allele appears to decrease AD risk. Most rare, early-onset forms of familial AD are caused by autosomal dominant mutations that often lead to overproduction of Aβ42 peptide. However, the mechanism by which APOE alleles differentially modulate Aβ accumulation in sporadic, late-onset AD is less clear. In a cohort of cognitively normal individuals, we report that reliable molecular and neuroimaging biomarkers of cerebral Aβ deposition vary in an apoE isoform–dependent manner. We hypothesized that human apoE isoforms differentially affect Aβ clearance or synthesis in vivo, resulting in an apoE isoform–dependent pattern of Aβ accumulation later in life. Performing in vivo microdialysis in a mouse model of Aβ-amyloidosis expressing human apoE isoforms (PDAPP/TRE), we find that the concentration and clearance of soluble Aβ in the brain interstitial fluid depends on the isoform of apoE expressed. This pattern parallels the extent of Aβ deposition observed in aged PDAPP/TRE mice. ApoE isoform–dependent differences in soluble Aβ metabolism are observed not only in aged but also in young PDAPP/TRE mice well before the onset of Aβ deposition in amyloid plaques in the brain. Additionally, amyloidogenic processing of amyloid precursor protein and Aβ synthesis, as assessed by in vivo stable isotopic labeling kinetics, do not vary according to apoE isoform in young PDAPP/TRE mice. Our results suggest that APOE alleles contribute to AD risk by differentially regulating clearance of Aβ from the brain, suggesting that Aβ clearance pathways may be useful therapeutic targets for AD prevention.


Journal of Clinical Investigation | 2005

P-glycoprotein deficiency at the blood-brain barrier increases amyloid-β deposition in an Alzheimer disease mouse model

John R. Cirrito; Rashid Deane; Anne M. Fagan; Michael L. Spinner; Maia Parsadanian; Mary Beth Finn; Hong Jiang; Julie L. Prior; Abhay P. Sagare; Kelly R. Bales; Steven M. Paul; Berislav V. Zlokovic; David Piwnica-Worms; David M. Holtzman

Accumulation of amyloid-beta (Abeta) within extracellular spaces of the brain is a hallmark of Alzheimer disease (AD). In sporadic, late-onset AD, there is little evidence for increased Abeta production, suggesting that decreased elimination from the brain may contribute to elevated levels of Abeta and plaque formation. Efflux transport of Abeta across the blood-brain barrier (BBB) contributes to Abeta removal from the brain. P-glycoprotein (Pgp) is highly expressed on the luminal surface of brain capillary endothelial cells and contributes to the BBB. In Pgp-null mice, we show that [I]Abeta40 and [I]Abeta42 microinjected into the CNS clear at half the rate that they do in WT mice. When amyloid precursor protein-transgenic (APP-transgenic) mice were administered a Pgp inhibitor, Abeta levels within the brain interstitial fluid significantly increased within hours of treatment. Furthermore, APP-transgenic, Pgp-null mice had increased levels of brain Abeta and enhanced Abeta deposition compared with APP-transgenic, Pgp WT mice. These data establish a direct link between Pgp and Abeta metabolism in vivo and suggest that Pgp activity at the BBB could affect risk for developing AD as well as provide a novel diagnostic and therapeutic target.


Nature Medicine | 2004

Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-β peptides

Milla Koistinaho; Suizhen Lin; Xin Wu; Michail A. Esterman; Deanna Koger; Jeffrey C. Hanson; Richard E. Higgs; Feng Liu; Seema Malkani; Kelly R. Bales; Steven M. Paul

We have previously shown that apolipoprotein E (Apoe) promotes the formation of amyloid in brain and that astrocyte-specific expression of APOE markedly affects the deposition of amyloid-β peptides (Aβ) in a mouse model of Alzheimer disease. Given the capacity of astrocytes to degrade Aβ, we investigated the potential role of Apoe in this astrocyte-mediated degradation. In contrast to cultured adult wild-type mouse astrocytes, adult Apoe−/− astrocytes do not degrade Aβ present in Aβ plaque–bearing brain sections in vitro. Coincubation with antibodies to either Apoe or Aβ, or with RAP, an antagonist of the low-density lipoprotein receptor family, effectively blocks Aβ degradation by astrocytes. Phase-contrast and confocal microscopy show that Apoe−/− astrocytes do not respond to or internalize Aβ deposits to the same extent as do wild-type astrocytes. Thus, Apoe seems to be important in the degradation and clearance of deposited Aβ species by astrocytes, a process that may be impaired in Alzheimer disease.


Neuron | 2004

ApoE and Clusterin Cooperatively Suppress Aβ Levels and Deposition: Evidence that ApoE Regulates Extracellular Aβ Metabolism In Vivo

Ronald B. DeMattos; John R. Cirrito; Maia Parsadanian; Patrick May; Mark A. O'Dell; Jennie W Taylor; Judith A. K. Harmony; Bruce J. Aronow; Kelly R. Bales; Steven M. Paul; David M. Holtzman

Apolipoprotein E (apoE) and clusterin can influence structure, toxicity, and accumulation of the amyloid-beta (Abeta) peptide in brain. Both molecules may also be involved in Abeta metabolism prior to its deposition. To assess this possibility, we compared PDAPP transgenic mice that develop age-dependent Abeta accumulation in the absence of apoE or clusterin as well as in the absence of both proteins. apoE(-/-) and clusterin(-/-) mice accumulated similar Abeta levels but much less fibrillar Abeta. In contrast, apoE(-/-)/clusterin(-/-) mice had both earlier onset and markedly increased Abeta and amyloid deposition. Both apoE(-/-) and apoE(-/-)/clusterin(-/-) mice had elevated CSF and brain interstitial fluid Abeta, as well as significant differences in the elimination half-life of interstitial fluid Abeta measured by in vivo microdialysis. These findings demonstrate additive effects of apoE and clusterin on influencing Abeta deposition and that apoE plays an important role in regulating extracellular CNS Abeta metabolism independent of Abeta synthesis.


The Journal of Neuroscience | 2005

Exacerbation of Cerebral Amyloid Angiopathy-Associated Microhemorrhage in Amyloid Precursor Protein Transgenic Mice by Immunotherapy Is Dependent on Antibody Recognition of Deposited Forms of Amyloid β

Margaret M. Racke; Laura I. Boone; Deena L. Hepburn; Maia Parsadainian; Matthew T. Bryan; Daniel K. Ness; Kathy S. Piroozi; William H. Jordan; Donna D. Brown; Wherly P. Hoffman; David M. Holtzman; Kelly R. Bales; Bruce D. Gitter; Patrick May; Steven M. Paul; Ronald B. DeMattos

Passive immunization with an antibody directed against the N terminus of amyloid β (Aβ) has recently been reported to exacerbate cerebral amyloid angiopathy (CAA)-related microhemorrhage in a transgenic animal model. Although the mechanism responsible for the deleterious interaction is unclear, a direct binding event may be required. We characterized the binding properties of several monoclonal anti-Aβ antibodies to deposited Aβ in brain parenchyma and CAA. Biochemical analyses demonstrated that the 3D6 and 10D5, two N-terminally directed antibodies, bound with high affinity to deposited forms of Aβ, whereas 266, a central domain antibody, lacked affinity for deposited Aβ. To determine whether 266 or 3D6 would exacerbate CAA-associated microhemorrhage, we treated aged PDAPP mice with either antibody for 6 weeks. We observed an increase in both the incidence and severity of CAA-associated microhemorrhage when PDAPP transgenic mice were treated with the N-terminally directed 3D6 antibody, whereas mice treated with 266 were unaffected. These results may have important implications for future immune-based therapeutic strategies for Alzheimers disease.


Journal of Clinical Investigation | 1999

Expression of human apolipoprotein E reduces amyloid-β deposition in a mouse model of Alzheimer's disease

David M. Holtzman; Kelly R. Bales; Shan Wu; Priyanka Bhat; Maia Parsadanian; Anne M. Fagan; Louis K. Chang; Yuling Sun; Steven M. Paul

The epsilon4 allele of apolipoprotein E (apo E) is associated with an increased risk for developing Alzheimers disease (AD). This may be due to interactions between apo E and the amyloid-beta protein (Abeta). To assess the effects of human apo E isoforms on Abeta deposition in vivo, we bred apo E3 and apo E4 hemizygous (+/-) transgenic mice expressing apo E by astrocytes to mice homozygous (+/+) for a mutant amyloid precursor protein (APPV717F) transgene that develop age-dependent AD neuropathology. All mice were on a mouse apo E null (-/-) background. By nine months of age, APPV717F+/-, apo E-/- mice had developed Abeta deposition, and, as reported previously, the quantity of Abeta deposits was significantly less than that seen in APPV717F+/- mice expressing mouse apo E. In contrast to effects of mouse apo E, similar levels of human apo E3 and apo E4 markedly suppressed early Abeta deposition at nine months of age in APPV717F+/- transgenic mice, even when compared with mice lacking apo E. These findings suggest that human apo E isoforms decrease Abeta aggregation or increase Abeta clearance relative to an environment in which mouse apo E or no apo E is present. The results may have important implications for understanding mechanisms underlying the link between apo E and AD.

Collaboration


Dive into the Kelly R. Bales's collaboration.

Top Co-Authors

Avatar

Steven M. Paul

University of California

View shared research outputs
Top Co-Authors

Avatar

David M. Holtzman

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Ronald B. DeMattos

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maia Parsadanian

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anne M. Fagan

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jirong Lu

Eli Lilly and Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge