171 results on '"Cynthia A. Lemere"'
Search Results
2. Microbiota in neuroinflammation and synaptic dysfunction: a focus on Alzheimer’s disease
- Author
-
Diane Bairamian, Sha Sha, Nathalie Rolhion, Harry Sokol, Guillaume Dorothée, Cynthia A. Lemere, and Slavica Krantic
- Subjects
Gut microbiota ,Synaptic dysfunction ,Alzheimer’s disease ,Peripheral immunomodulation ,Neuroinflammation ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background The implication of gut microbiota in the control of brain functions in health and disease is a novel, currently emerging concept. Accumulating data suggest that the gut microbiota exert its action at least in part by modulating neuroinflammation. Given the link between neuroinflammatory changes and neuronal activity, it is plausible that gut microbiota may affect neuronal functions indirectly by impacting microglia, a key player in neuroinflammation. Indeed, increasing evidence suggests that interplay between microglia and synaptic dysfunction may involve microbiota, among other factors. In addition to these indirect microglia-dependent actions of microbiota on neuronal activity, it has been recently recognized that microbiota could also affect neuronal activity directly by stimulation of the vagus nerve. Main messages The putative mechanisms of the indirect and direct impact of microbiota on neuronal activity are discussed by focusing on Alzheimer’s disease, one of the most studied neurodegenerative disorders and the prime cause of dementia worldwide. More specifically, the mechanisms of microbiota-mediated microglial alterations are discussed in the context of the peripheral and central inflammation cross-talk. Next, we highlight the role of microbiota in the regulation of humoral mediators of peripheral immunity and their impact on vagus nerve stimulation. Finally, we address whether and how microbiota perturbations could affect synaptic neurotransmission and downstream cognitive dysfunction. Conclusions There is strong increasing evidence supporting a role for the gut microbiome in the pathogenesis of Alzheimer’s disease, including effects on synaptic dysfunction and neuroinflammation, which contribute to cognitive decline. Putative early intervention strategies based on microbiota modulation appear therapeutically promising for Alzheimer’s disease but still require further investigation.
- Published
- 2022
- Full Text
- View/download PDF
3. Phosphorylated Aβ peptides in human Down syndrome brain and different Alzheimer’s-like mouse models
- Author
-
Sathish Kumar, Cynthia A. Lemere, and Jochen Walter
- Subjects
Alzheimer’s disease ,Amyloid β peptide ,Cerebral amyloid angiopathy ,Down syndrome ,Modified Aβ ,Mouse models ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract The deposition of neurotoxic amyloid-β (Aβ) peptides in extracellular plaques in the brain parenchyma is one of the most prominent neuropathological features of Alzheimer’s disease (AD), and considered to be closely related to the pathogenesis of this disease. A number of recent studies demonstrate the heterogeneity in the composition of Aβ deposits in AD brains, due to the occurrence of elongated, truncated and post-translationally modified Aβ peptides that have peculiar characteristics in aggregation behavior and biostability. Importantly, the detection of modified Aβ species has been explored to characterize distinct stages of AD, with phosphorylated Aβ being present in the clinical phase of AD. People with Down syndrome (DS) develop AD pathology by 40 years of age likely due to the overproduction of Aβ caused by the additional copy of the gene encoding the amyloid precursor protein on chromosome 21. In the current study, we analysed the deposition of phosphorylated and non-phosphorylated Aβ species in human DS, AD, and control brains. In addition, deposition of these Aβ species was analysed in brains of a series of established transgenic AD mouse models using phosphorylation-state specific Aβ antibodies. Significant amounts of Aβ phosphorylated at serine residue 8 (pSer8Aβ) and unmodified Aβ were detected in the brains of DS and AD cases. The brains of different transgenic mouse models with either only human mutant amyloid precursor protein (APP), or combinations of human mutant APP, Presenilin (PS), and tau transgenes showed distinct age-dependent and spatiotemporal deposition of pSer8Aβ in extracellular plaques and within the vasculature. Together, these results demonstrate the deposition of phosphorylated Aβ species in DS brains, further supporting the similarity of Aβ deposition in AD and DS. Thus, the detection of phosphorylated and other modified Aβ species could contribute to the understanding and dissection of the complexity in the age-related and spatiotemporal deposition of Aβ variants in AD and DS as well as in distinct mouse models.
- Published
- 2020
- Full Text
- View/download PDF
4. Development of the clinical candidate PBD-C06, a humanized pGlu3-Aβ-specific antibody against Alzheimer’s disease with reduced complement activation
- Author
-
Thore Hettmann, Stephen D. Gillies, Martin Kleinschmidt, Anke Piechotta, Koki Makioka, Cynthia A. Lemere, Stephan Schilling, Jens-Ulrich Rahfeld, and Inge Lues
- Subjects
Medicine ,Science - Abstract
Abstract In clinical trials with early Alzheimer’s patients, administration of anti-amyloid antibodies reduced amyloid deposits, suggesting that immunotherapies may be promising disease-modifying interventions against Alzheimer’s disease (AD). Specific forms of amyloid beta (Aβ) peptides, for example post-translationally modified Aβ peptides with a pyroglutamate at the N-terminus (pGlu3, pE3), are attractive antibody targets, due to pGlu3-Aβ’s neo-epitope character and its propensity to form neurotoxic oligomeric aggregates. We have generated a novel anti-pGlu3-Aβ antibody, PBD-C06, which is based on a murine precursor antibody that binds with high specificity to pGlu3-Aβ monomers, oligomers and fibrils, including mixed aggregates of unmodified Aβ and pGlu3-Aβ peptides. PBD-C06 was generated by first grafting the murine antigen binding sequences onto suitable human variable light and heavy chains. Subsequently, the humanized antibody was de-immunized and site-specific mutations were introduced to restore original target binding, to eliminate complement activation and to improve protein stability. PBD-C06 binds with the same specificity and avidity as its murine precursor antibody and elimination of C1q binding did not compromise Fcγ-receptor binding or in vitro phagocytosis. Thus, PBD-C06 was specifically designed to target neurotoxic aggregates and to avoid complement-mediated inflammatory responses, in order to lower the risk for vasogenic edemas in the clinic.
- Published
- 2020
- Full Text
- View/download PDF
5. Effector function of anti-pyroglutamate-3 Aβ antibodies affects cognitive benefit, glial activation and amyloid clearance in Alzheimer’s-like mice
- Author
-
Helen Crehan, Bin Liu, Martin Kleinschmidt, Jens-Ulrich Rahfeld, Kevin X. Le, Barbara J. Caldarone, Jeffrey L. Frost, Thore Hettmann, Birgit Hutter-Paier, Brian O’Nuallain, Mi-Ae Park, Marcelo F. DiCarli, Inge Lues, Stephan Schilling, and Cynthia A. Lemere
- Subjects
Pyroglutamate-3 amyloid-β ,Immunotherapy ,APPSWE/PS1ΔE9 ,Phagocytosis ,Microhemorrhage ,microPET ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Pyroglutamate-3 Aβ (pGlu-3 Aβ) is an N-terminally truncated and post-translationally modified Aβ species found in Alzheimer’s disease (AD) brain. Its increased peptide aggregation propensity and toxicity make it an attractive emerging treatment strategy for AD. We address the question of how the effector function of an anti-pGlu-3 Aβ antibody influences the efficacy of immunotherapy in mouse models with AD-like pathology. Methods We compared two different immunoglobulin (Ig) isotypes of the same murine anti-pGlu-3 Aβ mAb (07/1 IgG1 and 07/2a IgG2a) and a general N-terminal Aβ mAb (3A1 IgG1) for their ability to clear Aβ and protect cognition in a therapeutic passive immunotherapy study in aged, plaque-rich APPSWE/PS1ΔE9 transgenic (Tg) mice. We also compared the ability of these antibodies and a CDC-mutant form of 07/2a (07/2a-k), engineered to avoid complement activation, to clear Aβ in an ex vivo phagocytosis assay and following treatment in APPSLxhQC double Tg mice, and to activate microglia using longitudinal microPET imaging with TSPO-specific 18F-GE180 tracer following a single bolus antibody injection in young and old Tg mice. Results We demonstrated significant cognitive improvement, better plaque clearance, and more plaque-associated microglia in the absence of microhemorrhage in aged APPSWE/PS1ΔE9 Tg mice treated with 07/2a, but not 07/1 or 3A1, compared to PBS in our first in vivo study. All mAbs cleared plaques in an ex vivo assay, although 07/2a promoted the highest phagocytic activity. Compared with 07/2a, 07/2a-k showed slightly reduced affinity to Fcγ receptors CD32 and CD64, although the two antibodies had similar binding affinities to pGlu-3 Aβ. Treatment of APPSLxhQC mice with 07/2a and 07/2a-k mAbs in our second in vivo study showed significant plaque-lowering with both mAbs. Longitudinal 18F-GE180 microPET imaging revealed different temporal patterns of microglial activation for 3A1, 07/1, and 07/2a mAbs and no difference between 07/2a-k and PBS-treated Tg mice. Conclusion Our results suggest that attenuation of behavioral deficits and clearance of amyloid is associated with strong effector function of the anti-pGlu-3 Aβ mAb in a therapeutic treatment paradigm. We present evidence that antibody engineering to reduce CDC-mediated complement binding facilitates phagocytosis of plaques without inducing neuroinflammation in vivo. Hence, the results provide implications for tailoring effector function of humanized antibodies for clinical development.
- Published
- 2020
- Full Text
- View/download PDF
6. BrightFocus Alzheimer’s Fast Track 2019
- Author
-
Keith W. Whitaker, Frank M. LaFerla, Harry W. M. Steinbusch, Cynthia A. Lemere, and Diane E. Bovenkamp
- Subjects
Dementia ,Alzheimer’s disease ,Neurodegeneration ,Workshop ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract The 3 day workshop “Alzheimer’s Fast Track” is a unique opportunity for graduate students, postdoctoral fellows, or other early-career scientists, focused on Alzheimer’s disease research, to gain new knowledge and become an expert in where this emerging scientific field is moving. In addition, it is not only about receiving a good overview, but also learning to write and defend a successful application for securing funding for Alzheimer’s disease research projects.
- Published
- 2019
- Full Text
- View/download PDF
7. Acute Effects of Focused Ultrasound-Induced Blood-Brain Barrier Opening on Anti-Pyroglu3 Abeta Antibody Delivery and Immune Responses
- Author
-
Praveen Bathini, Tao Sun, Mathias Schenk, Stephan Schilling, Nathan J. McDannold, and Cynthia A. Lemere
- Subjects
focused ultrasound ,pyroglutamate-3 Aβ ,microglia ,Microbiology ,QR1-502 - Abstract
Alzheimer’s Disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid plaques and hyperphosphorylated tau in the brain. Currently, therapeutic agents targeting amyloid appear promising for AD, however, delivery to the CNS is limited due to the blood-brain-barrier (BBB). Focused ultrasound (FUS) is a method to induce a temporary opening of the BBB to enhance the delivery of therapeutic agents to the CNS. In this study, we evaluated the acute effects of FUS and whether the use of FUS-induced BBB opening enhances the delivery of 07/2a mAb, an anti-pyroglutamate-3 Aβ antibody, in aged 24 mo-old APP/PS1dE9 transgenic mice. FUS was performed either unilaterally or bilaterally with mAb infusion and the short-term effect was analyzed 4 h and 72 h post-treatment. Quantitative analysis by ELISA showed a 5–6-fold increase in 07/2a mAb levels in the brain at both time points and an increased brain-to-blood ratio of the antibody. Immunohistochemistry demonstrated an increase in IgG2a mAb detection particularly in the cortex, enhanced immunoreactivity of resident Iba1+ and phagocytic CD68+ microglial cells, and a transient increase in the infiltration of Ly6G+ immune cells. Cerebral microbleeds were not altered in the unilaterally or bilaterally sonicated hemispheres. Overall, this study shows the potential of FUS therapy for the enhanced delivery of CNS therapeutics.
- Published
- 2022
- Full Text
- View/download PDF
8. Anti-Aβ antibodies incapable of reducing cerebral Aβ oligomers fail to attenuate spatial reference memory deficits in J20 mice
- Author
-
Alexandra J. Mably, Wen Liu, Jessica M. Mc Donald, Jean-Cosme Dodart, Frédérique Bard, Cynthia A. Lemere, Brian O'Nuallain, and Dominic M. Walsh
- Subjects
Alzheimer's disease ,Amyloid β-protein ,Immunotherapy ,Oligomers ,APP transgenic mice ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Compelling genetic evidence links the amyloid precursor protein (APP) to Alzheimer's disease (AD). A leading hypothesis proposes that a small amphipathic fragment of APP, the amyloid β-protein (Aβ), self-associates to form soluble assemblies loosely referred to as “oligomers” and that these are primary mediators of synaptic dysfunction. As such, Aβ, and specifically Aβ oligomers, are targets for disease modifying therapies. Currently, the most advanced experimental treatment for AD relies on the use of anti-Aβ antibodies. In this study, we tested the ability of the monomer-preferring antibody, m266 and a novel aggregate-preferring antibody, 1C22, to attenuate spatial reference memory impairments in J20 mice. Chronic treatment with m266 resulted in a ~70-fold increase in Aβ detected in the bloodstream, and a ~50% increase in water-soluble brain Aβ — and in both cases Aβ was bound to m266. In contrast, 1C22 increased the levels of free Aβ in the bloodstream, and bound to amyloid deposits in J20 brain. However, neither 1C22 nor m266 attenuated the cognitive deficits evident in 12 month old J20 mice. Moreover, both antibodies failed to alter the levels of soluble Aβ oligomers in J20 brain. These results suggest that Aβ oligomers may mediate the behavioral deficits seen in J20 mice and highlight the need for the development of aggregate-preferring antibodies that can reach the brain in sufficient levels to neutralize bioactive Aβ oligomers.Aside from the lack of positive effect of m266 and 1C22 on cognition, a substantial number of deaths occurred in m266- and 1C22-immunized J20 mice. These fatalities were specific to anti-Aβ antibodies and to the J20 mouse line since treatment of wild type or PDAPP mice with these antibodies did not cause any deaths. These and other recent results indicate that J20 mice are particularly susceptible to targeting of the APP/Aβ/tau axis. Notwithstanding the specificity of fatalities for J20 mice, it is worrying that the murine precursor (m266) of a lead experimental therapeutic, Solanezumab, did not engage with putatively pathogenic Aβ oligomers.
- Published
- 2015
- Full Text
- View/download PDF
9. Passive Aβ Immunotherapy: Current Achievements and Future Perspectives
- Author
-
Stephan Schilling, Jens-Ulrich Rahfeld, Inge Lues, and Cynthia A. Lemere
- Subjects
amyloid-β ,monoclonal antibodies ,posttranslational modifications ,drug development ,Organic chemistry ,QD241-441 - Abstract
Passive immunotherapy has emerged as a very promising approach for the treatment of Alzheimer’s disease and other neurodegenerative disorders, which are characterized by the misfolding and deposition of amyloid peptides. On the basis of the amyloid hypothesis, the majority of antibodies in clinical development are directed against amyloid β (Aβ), the primary amyloid component in extracellular plaques. This review focuses on the current status of Aβ antibodies in clinical development, including their characteristics and challenges that came up in clinical trials with these new biological entities (NBEs). Emphasis is placed on the current view of common side effects observed with passive immunotherapy, so-called amyloid-related imaging abnormalities (ARIAs), and potential ways to overcome this issue. Among these new ideas, a special focus is placed on molecules that are directed against post-translationally modified variants of the Aβ peptide, an emerging approach for development of new antibody molecules.
- Published
- 2018
- Full Text
- View/download PDF
10. Biochemical and immunohistochemical analysis of an Alzheimer's disease mouse model reveals the presence of multiple cerebral Aβ assembly forms throughout life
- Author
-
Ganesh M. Shankar, Malcolm A. Leissring, Anthony Adame, Xiaoyan Sun, Edward Spooner, Eliezer Masliah, Dennis J. Selkoe, Cynthia A. Lemere, and Dominic M. Walsh
- Subjects
Amyloid β-protein ,Aggregation ,Oligomers ,Amyloid precursor protein ,J20 mice ,Synaptophysin ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The amyloid β-protein (Aβ) is believed to play a causal role in Alzheimer's disease, however, the mechanism by which Aβ mediates its effect and the assembly form(s) of Aβ responsible remain unclear. Several APP transgenic mice have been shown to accumulate Aβ and to develop cognitive deficits. We have studied one such model, the J20 mouse. Using an immunoprecipitation/Western blotting technique we find an age-dependent increase in Aβ monomer and SDS-stable dimer. But prior to the earliest detection of Aβ dimers, immunohistochemical analysis revealed an increase in oligomer immunoreactivity that was coincident with reduced hippocampal MAP2 and synaptophysin staining. Moreover, biochemical fractionation and ELISA analysis revealed evidence of TBS and triton-insoluble sedimentable Aβ aggregates at the earliest ages studied. These data demonstrate the presence of multiple assembly forms of Aβ throughout the life of J20 mice and highlight the difficulty in attributing synaptotoxicity to a single Aβ species.
- Published
- 2009
- Full Text
- View/download PDF
11. Cytosolic Phospholipase A2(cPLA2) Immunoreactivity Is Elevated in Alzheimer's Disease Brain
- Author
-
Diane T. Stephenson, Cynthia A. Lemere, Dennis J. Selkoe, and James A. Clemens
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Phospholipase A2(PLA2) is the key enzyme that initiates the arachidonic acid cascade, which leads to the generation of multiple eicosanoid products. Many of these products are believed to play an important role in the inflammatory process. Activation of PLA2is observed under pathological conditions where inflammation is present. Cytosolic PLA2(cPLA2) is activated by very low levels of calcium and is thought to control receptor-mediated eicosanoid production and to participate in intracellular signal transduction processes. In view of the presence of numerous inflammatory mediators and acute phase proteins in the Alzheimer's disease (AD) brain, localization of cPLA2in AD brain was evaluated and compared to that observed in nonneurologically diseased controls. In this study, a monoclonal antibody raised against cPLA2was used to immunostain tissue sections of human cerebral cortex. Five AD cases and six neurologically normal cases were evaluated in the occipital cortex and the cerebellum. Two of the AD cases were also examined in other cortical regions. Granular-like staining with anti-cPLA2was found to be associated with astrocytes in the cortex of both control and AD cases. Colocalization with GFAP confirmed that cPLA2immunoreactivity is associated almost exclusively with protoplasmic astrocytes. Staining was abolished when sections were labeled with antibody that had been preadsorbed with purified cPLA2. In AD brain, cPLA2immunoreactive astrocytes were greater in number and more intensely stained than those in control cases. cPLA2immunoreactivity was virtually absent in the cerebellum of AD and control cases, despite the presence in this region of diffuse amyloid in two AD cases and amyloid angiopathy in a third case. In the cortex, cPLA2immunoreactive astrocytes were detected in regions that contained numerous Aβ deposits. The finding of elevated levels of cPLA2immunoreactivity in AD brain supports the hypothesis that there is an active inflammatory process occurring in AD.
- Published
- 1996
- Full Text
- View/download PDF
12. APOE and immunity:Research highlights
- Author
-
Courtney M. Kloske, Christopher J. Barnum, Andre F. Batista, Elizabeth M. Bradshaw, Adam M. Brickman, Guojun Bu, Jessica Dennison, Mary D. Gearon, Alison M. Goate, Christian Haass, Michael T. Heneka, William T. Hu, Lenique K. L. Huggins, Nahdia S. Jones, Radosveta Koldamova, Cynthia A. Lemere, Shane A. Liddelow, Edoardo Marcora, Samuel E. Marsh, Henrietta M. Nielsen, Kellen K. Petersen, Melissa Petersen, Stefanie D. Piña‐Escudero, Wei Qiao Qiu, Yakeel T. Quiroz, Eric Reiman, Claire Sexton, Malú Gámez Tansey, Julia TCW, Charlotte E. Teunissen, Betty M. Tijms, Rik van der Kant, Rebecca Wallings, Stacie C. Weninger, Whitney Wharton, Donna M. Wilcock, Tyler James Wishard, Susan L Worley, Henrik Zetterberg, and Maria C. Carrillo
- Subjects
Epidemiology ,microglia ,neuroinflammation ,pathology [Alzheimer Disease] ,Cellular and Molecular Neuroscience ,Apolipoproteins E ,Developmental Neuroscience ,TREM2 ,Humans ,ddc:610 ,tau ,Inflammation ,Health Policy ,astrocytes ,neurodegeneration ,pathology [Microglia] ,biomarkers ,Alzheimer's disease ,neuroglia ,amyloid beta ,Psychiatry and Mental health ,genetics [Apolipoproteins E] ,Neurology (clinical) ,Geriatrics and Gerontology ,APOE ,dementia - Abstract
INTRODUCTION: At the Alzheimer's Association's APOE and Immunity virtual conference, held in October 2021, leading neuroscience experts shared recent research advances on and inspiring insights into the various roles that both the apolipoprotein E gene (APOE) and facets of immunity play in neurodegenerative diseases, including Alzheimer's disease and other dementias. METHODS: The meeting brought together more than 1200 registered attendees from 62 different countries, representing the realms of academia and industry. RESULTS: During the 4-day meeting, presenters illuminated aspects of the cross-talk between APOE and immunity, with a focus on the roles of microglia, triggering receptor expressed on myeloid cells 2 (TREM2), and components of inflammation (e.g., tumor necrosis factor α [TNFα]). DISCUSSION: This manuscript emphasizes the importance of diversity in current and future research and presents an integrated view of innate immune functions in Alzheimer's disease as well as related promising directions in drug development.
- Published
- 2023
- Full Text
- View/download PDF
13. High-Energy, Whole-Body Proton Irradiation Differentially Alters Long-Term Brain Pathology and Behavior Dependent on Sex and Alzheimer’s Disease Mutations
- Author
-
Robert G. Hinshaw, Maren K. Schroeder, Jason Ciola, Curran Varma, Brianna Colletti, Bin Liu, Grace Geyu Liu, Qiaoqiao Shi, Jacqueline P. Williams, M. Kerry O’Banion, Barbara J. Caldarone, and Cynthia A. Lemere
- Subjects
space radiation ,Organic Chemistry ,neurodegeneration ,General Medicine ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,radiation ,Physical and Theoretical Chemistry ,CNS ,Molecular Biology ,Alzheimer’s disease ,Spectroscopy ,proton - Abstract
Whole-body exposure to high-energy particle radiation remains an unmitigated hazard to human health in space. Ongoing experiments at the NASA Space Radiation Laboratory and elsewhere repeatedly show persistent changes in brain function long after exposure to simulations of this unique radiation environment, although, as is also the case with proton radiotherapy sequelae, how this occurs and especially how it interacts with common comorbidities is not well-understood. Here, we report modest differential changes in behavior and brain pathology between male and female Alzheimer’s-like and wildtype littermate mice 7–8 months after exposure to 0, 0.5, or 2 Gy of 1 GeV proton radiation. The mice were examined with a battery of behavior tests and assayed for amyloid beta pathology, synaptic markers, microbleeds, microglial reactivity, and plasma cytokines. In general, the Alzheimer’s model mice were more prone than their wildtype littermates to radiation-induced behavior changes, and hippocampal staining for amyloid beta pathology and microglial activation in these mice revealed a dose-dependent reduction in males but not in females. In summary, radiation-induced, long-term changes in behavior and pathology, although modest, appear specific to both sex and the underlying disease state.
- Published
- 2023
- Full Text
- View/download PDF
14. Space-like 56Fe irradiation manifests mild, early sex-specific behavioral and neuropathological changes in wildtype and Alzheimer’s-like transgenic mice
- Author
-
Bin Liu, Robert G. Hinshaw, Kevin X. Le, Mi-Ae Park, Shuyan Wang, Anthony P. Belanger, Shipra Dubey, Jeffrey L. Frost, Qiaoqiao Shi, Peter Holton, Lee Trojanczyk, Vladimir Reiser, Paul A. Jones, William Trigg, Marcelo F. Di Carli, Paul Lorello, Barbara J. Caldarone, Jacqueline P. Williams, M. Kerry O’Banion, and Cynthia A. Lemere
- Published
- 2019
- Full Text
- View/download PDF
15. Complement C3 Lowering in Adult Inducible Conditional Knockout Mice: Long‐Lasting Effects
- Author
-
Andre Felipe Batista, Maren K Schroeder, Khyrul Khan, Esra Yalcin, Michelle Mei, Jessy Presumey, Shaomin A Li, Michael Carroll, and Cynthia A. Lemere
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
- Full Text
- View/download PDF
16. Temporal Characterization of the Amyloidogenic APP/PS1dE9;hAPOE4 Mouse Model of Alzheimer’s Disease
- Author
-
Martine Grenon, Praveen Bathini, Maria‐Giusy Papavergi, Maren K Schroeder, and Cynthia A. Lemere
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
- Full Text
- View/download PDF
17. Therapeutic Efficacy of the Murine Precursor to PBD‐C06: a Novel CDC‐Mutant Anti‐pGlu3Aβ mAb
- Author
-
Praveen Bathini, Martine Grenon, Maria‐Giusy Papavergi, Brittani R. Price, Maren K. Schroeder, Martin Kleinschmidt, Jens‐Ulrich Rahfeld, Stephan Schilling, and Cynthia A. Lemere
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
- Full Text
- View/download PDF
18. Microglia Do Not Take Up Soluble Amyloid-beta Peptides, But Partially Degrade Them by Secreting Insulin-degrading Enzyme
- Author
-
Liangping Li, Cynthia A. Lemere, Hongjun Fu, and Bin Liu
- Subjects
0301 basic medicine ,Amyloid beta ,Phagocytosis ,Pharmacology ,Insulysin ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Alzheimer Disease ,In vivo ,Insulin-degrading enzyme ,medicine ,Animals ,Humans ,Cytochalasin B ,Amyloid beta-Peptides ,biology ,Microglia ,Chemistry ,General Neuroscience ,Pinocytosis ,Brain ,Peptide Fragments ,030104 developmental biology ,medicine.anatomical_structure ,Enzyme inhibitor ,biology.protein ,030217 neurology & neurosurgery - Abstract
Microglia play important roles in the pathogenesis of Alzheimer’s disease (AD), in part, by affecting the clearance of amyloid-β (Aβ) peptides. Most studies, however, used synthetic soluble Aβ (sAβ) at higher concentrations. The exact mechanisms underlying microglia-mediated clearance of physiological sAβ at very low concentrations remain unclear. Here we reported that there were much more Iba-1- and CD68-positive microglia and significantly less sAβ left in the brain of adult mice 5 days after the surgery of sAβ microinjection compared to 2 h after the surgery (p < 0.05). However, very few Iba-1- and CD68-positive microglia co-localized with microinjected fluorescently labeled sAβ (FLsAβ(42)) 5 days after the surgery. Also, there was no co-localization of FLsAβ(42) with a lysosomal marker (LAMP-1) 5 days after the surgery. There was no significant difference in the percentage of Aβ(+)/PE-CD11b(+)/APC-CD45(low) microglia between the control group and the group microinjected with TBS-soluble Aβ extracted from the brains of AD patients (p > 0.05). The degradation of physiological sAβ was prevented by a highly selective insulin-degrading enzyme inhibitor (Ii1) but not by a phagocytosis inhibitor (polyinosinic acid) or pinocytosis inhibitor (cytochalasin B) in vitro. Furthermore, the reduction of synthetic and physiological sAβ in the brain was partially prevented by the co-injection of Ii1 in vivo (p < 0.05). Our results demonstrate that microglia do not take up synthetic or physiological sAβ, but partially degrade it via the secretion of insulin-degrading enzyme, which will be beneficial for understanding how sAβ is removed from the brain by microglia.
- Published
- 2020
- Full Text
- View/download PDF
19. Development of the clinical candidate PBD-C06, a humanized pGlu3-Aβ-specific antibody against Alzheimer’s disease with reduced complement activation
- Author
-
Anke Piechotta, Cynthia A. Lemere, Stephan Schilling, Inge Lues, Thore Hettmann, Koki Makioka, Jens-Ulrich Rahfeld, Stephen D. Gillies, and Martin Kleinschmidt
- Subjects
lcsh:Medicine ,Humanized antibody ,Epitopes ,Mice ,0302 clinical medicine ,Edema ,lcsh:Science ,Complement Activation ,0303 health sciences ,Multidisciplinary ,biology ,Chemistry ,Drug discovery ,Endocytosis ,3. Good health ,Cell biology ,Pyrrolidonecarboxylic Acid ,Medicine ,Immunotherapy ,Antibody ,Protein Binding ,Amyloid ,Amyloid beta ,Phagocytosis ,Science ,Antibodies, Monoclonal, Humanized ,Article ,03 medical and health sciences ,Alzheimer Disease ,Animals ,Humans ,Avidity ,Alleles ,030304 developmental biology ,Inflammation ,Amyloid beta-Peptides ,Complement C1q ,lcsh:R ,Complementarity Determining Regions ,In vitro ,Complement system ,Mutation ,biology.protein ,lcsh:Q ,Antibody therapy ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery - Abstract
In clinical trials with early Alzheimer’s patients, administration of anti-amyloid antibodies reduced amyloid deposits, suggesting that immunotherapies may be promising disease-modifying interventions against Alzheimer’s disease (AD). Specific forms of amyloid beta (Aβ) peptides, for example post-translationally modified Aβ peptides with a pyroglutamate at the N-terminus (pGlu3, pE3), are attractive antibody targets, due to pGlu3-Aβ’s neo-epitope character and its propensity to form neurotoxic oligomeric aggregates. We have generated a novel anti-pGlu3-Aβ antibody, PBD-C06, which is based on a murine precursor antibody that binds with high specificity to pGlu3-Aβ monomers, oligomers and fibrils, including mixed aggregates of unmodified Aβ and pGlu3-Aβ peptides. PBD-C06 was generated by first grafting the murine antigen binding sequences onto suitable human variable light and heavy chains. Subsequently, the humanized antibody was de-immunized and site-specific mutations were introduced to restore original target binding, to eliminate complement activation and to improve protein stability. PBD-C06 binds with the same specificity and avidity as its murine precursor antibody and elimination of C1q binding did not compromise Fcγ-receptor binding or in vitro phagocytosis. Thus, PBD-C06 was specifically designed to target neurotoxic aggregates and to avoid complement-mediated inflammatory responses, in order to lower the risk for vasogenic edemas in the clinic.
- Published
- 2020
20. Effector function of anti-pyroglutamate-3 Aβ antibodies affects cognitive benefit, glial activation and amyloid clearance in Alzheimer’s-like mice
- Author
-
Stephan Schilling, Brian O'Nuallain, Marcelo F. DiCarli, Mi-Ae Park, Helen Crehan, Kevin X. Le, Bin Liu, Birgit Hutter-Paier, Martin Kleinschmidt, Thore Hettmann, Cynthia A. Lemere, Jens Ulrich Rahfeld, Barbara J. Caldarone, Jeffrey L. Frost, and Inge Lues
- Subjects
0301 basic medicine ,Alzheimer Vaccines ,Amyloid ,medicine.drug_class ,Cognitive Neuroscience ,Mice, Transgenic ,Pharmacology ,Monoclonal antibody ,lcsh:RC346-429 ,lcsh:RC321-571 ,Mice ,03 medical and health sciences ,Microhemorrhage ,Cognition ,0302 clinical medicine ,Phagocytosis ,Alzheimer Disease ,In vivo ,medicine ,Animals ,Pyroglutamate-3 amyloid-β ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Neuroinflammation ,lcsh:Neurology. Diseases of the nervous system ,APPSWE/PS1ΔE9 ,Amyloid beta-Peptides ,biology ,Microglia ,Chemistry ,Research ,Immunization, Passive ,Antibodies, Monoclonal ,microPET ,Pyrrolidonecarboxylic Acid ,3. Good health ,Complement system ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Immunoglobulin G ,biology.protein ,Neurology (clinical) ,Immunotherapy ,Antibody ,Neuroglia ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery ,Ex vivo - Abstract
Background Pyroglutamate-3 Aβ (pGlu-3 Aβ) is an N-terminally truncated and post-translationally modified Aβ species found in Alzheimer’s disease (AD) brain. Its increased peptide aggregation propensity and toxicity make it an attractive emerging treatment strategy for AD. We address the question of how the effector function of an anti-pGlu-3 Aβ antibody influences the efficacy of immunotherapy in mouse models with AD-like pathology. Methods We compared two different immunoglobulin (Ig) isotypes of the same murine anti-pGlu-3 Aβ mAb (07/1 IgG1 and 07/2a IgG2a) and a general N-terminal Aβ mAb (3A1 IgG1) for their ability to clear Aβ and protect cognition in a therapeutic passive immunotherapy study in aged, plaque-rich APPSWE/PS1ΔE9 transgenic (Tg) mice. We also compared the ability of these antibodies and a CDC-mutant form of 07/2a (07/2a-k), engineered to avoid complement activation, to clear Aβ in an ex vivo phagocytosis assay and following treatment in APPSLxhQC double Tg mice, and to activate microglia using longitudinal microPET imaging with TSPO-specific 18F-GE180 tracer following a single bolus antibody injection in young and old Tg mice. Results We demonstrated significant cognitive improvement, better plaque clearance, and more plaque-associated microglia in the absence of microhemorrhage in aged APPSWE/PS1ΔE9 Tg mice treated with 07/2a, but not 07/1 or 3A1, compared to PBS in our first in vivo study. All mAbs cleared plaques in an ex vivo assay, although 07/2a promoted the highest phagocytic activity. Compared with 07/2a, 07/2a-k showed slightly reduced affinity to Fcγ receptors CD32 and CD64, although the two antibodies had similar binding affinities to pGlu-3 Aβ. Treatment of APPSLxhQC mice with 07/2a and 07/2a-k mAbs in our second in vivo study showed significant plaque-lowering with both mAbs. Longitudinal 18F-GE180 microPET imaging revealed different temporal patterns of microglial activation for 3A1, 07/1, and 07/2a mAbs and no difference between 07/2a-k and PBS-treated Tg mice. Conclusion Our results suggest that attenuation of behavioral deficits and clearance of amyloid is associated with strong effector function of the anti-pGlu-3 Aβ mAb in a therapeutic treatment paradigm. We present evidence that antibody engineering to reduce CDC-mediated complement binding facilitates phagocytosis of plaques without inducing neuroinflammation in vivo. Hence, the results provide implications for tailoring effector function of humanized antibodies for clinical development.
- Published
- 2020
21. Focus Ultrasound‐Induced Blood‐Brain Barrier opening enhances anti‐pGlu3 Aβ mAb delivery and amyloid‐beta plaque clearance
- Author
-
Praveen Bathini, Tao Sun, Qiaoqiao Shi, Yongzhi Zhang, Nadine Taudte, Mathias Schenk, Thore Hettmann, Stephan Schilling, Nathan McDannold, and Cynthia A Lemere
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2021
- Full Text
- View/download PDF
22. Paving the Way for Therapy
- Author
-
Roger H. Reeves, Alain D. Dekker, Marie-Claude Potier, Cynthia A. Lemere, Pablo Caviedes, Jean M. Delabar, Jorge Busciglio, Elizabeth Head, Mara Dierssen, Anita Bhattacharyya, Peter Paul De Deyn, and Molecular Neuroscience and Ageing Research (MOLAR)
- Subjects
0301 basic medicine ,Down syndrome ,Medical education ,Intellectual and Developmental Disabilities (IDD) ,Clinical Sciences ,Attendance ,MOUSE MODEL ,medicine.disease ,Brain Disorders ,Syndrome Conferences, Symposia or Workshops ,03 medical and health sciences ,Important research ,030104 developmental biology ,Trisomy 21 Research Society ,Multidisciplinary approach ,Intellectual disability ,medicine ,Acquired Cognitive Impairment ,Genetics ,Alzheimer disease ,Trisomy ,Psychology ,Genetics (clinical) - Abstract
In the last decade, a number of important research advances in different fields have allowed Down syndrome (DS) research to flourish, creating a time of both unparalleled opportunity and considerable challenge. Building a scientific framework that distills mechanisms involved in the developmental intellectual disability of DS as well as the early-onset component of Alzheimer disease and the several other comorbidities associated with the condition is a challenge that scientists are now tackling using novel technologies and multidisciplinary approaches. The Trisomy 21 Research Society (T21RS) was founded in 2014 to address these evolving needs and challenges. In June of 2017, the T21RS held its 2nd International Conference in Chicago, USA. With more than 200 scientists, advocates, people with DS, and family members in attendance, the meeting served as a forum for the discussion of the latest research and clinical advances as well as the most compelling needs of people with DS and their families.
- Published
- 2019
- Full Text
- View/download PDF
23. Aducanumab produced a clinically meaningful benefit in association with amyloid lowering
- Author
-
Alireza Atri, Jeffrey L. Cummings, Stephen Salloway, Cynthia A. Lemere, Paul S. Aisen, and Marwan N. Sabbagh
- Subjects
medicine.medical_specialty ,Amyloid ,Neurology ,medicine.drug_class ,Donanemab ,Cognitive Neuroscience ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Amyloidogenic Proteins ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Lecanemab ,Viewpoint ,Clinical trials ,Alzheimer Disease ,Internal medicine ,medicine ,Humans ,Aducanumab ,RC346-429 ,Amyloid beta-Peptides ,Geriatrics gerontology ,business.industry ,Clinical trial ,Gantenerumab ,Monoclonal antibodies ,Neurology. Diseases of the nervous system ,Neurology (clinical) ,business ,Geriatric psychiatry ,FDA ,RC321-571 ,medicine.drug - Published
- 2021
24. Ultrasound-mediated blood-brain barrier disruption improves anti-pyroglutamate3 Aβ antibody efficacy and enhances phagocyte infiltration into brain in aged Alzheimer’s disease-like mice
- Author
-
Qiaoqiao Shi, Tao Sun, Yongzhi Zhang, Chanikarn Power, Camilla Hoesch, Shawna Antonelli, Maren K. Schroeder, Barbara J. Caldarone, Nadine Taudte, Mathias Schenk, Thore Hettmann, Stephan Schilling, Nathan J. McDannold, and Cynthia A. Lemere
- Subjects
Microglia ,biology ,medicine.drug_class ,business.industry ,medicine.medical_treatment ,Monocyte ,Immunotherapy ,Pharmacology ,T-maze ,Hippocampal formation ,Monoclonal antibody ,Blood–brain barrier ,medicine.anatomical_structure ,mental disorders ,medicine ,biology.protein ,Antibody ,business - Abstract
Pyroglutamate-3 amyloid-β (pGlu3 Aβ) is an N-terminally modified, toxic form of amyloid-β that is present in cerebral amyloid plaques and vascular deposits. Using the Fc-competent murine anti-pGlu3 Aβ monoclonal antibody (mAb), 07/2a, we present here a nonpharmacological approach using focused ultrasound (FUS) with intravenous (i.v.) injection of microbubbles (MB) to facilitate i.v. delivery of the 07/2a mAb across the blood brain barrier (BBB) in order to improve Aβ removal and restore memory in aged APP/PS1 mice, an Alzheimer’s disease (AD)-like model of amyloidogenesis.Compared to sham-treated controls, aged APP/PS1 mice treated with 07/2a immediately prior to FUS-mediated BBB disruption (mAb + FUS-BBBD combination treatment) showed significantly better spatial learning and memory in the Water T Maze. FUS-BBBD treatment alone improved contextual fear learning and memory in aged WT and APP/PS1 mice, respectively. APP/PS1 mice given the combination treatment had reduced Aβ42 and pGlu3 Aβ hippocampal plaque burden compared to PBS-treated APP/PS1 mice.Hippocampal synaptic puncta density and synaptosomal synaptic protein levels were also higher in APP/PS1 mice treated with 07/2a just prior to BBB disruption. Increased Iba-1+ microglia were observed in the hippocampi of AD mice treated with 07/2a with and without FUS-BBBD, and APP/PS1 mice that received hippocampal BBB disruption and 07/2a showed increased Ly6G+ monocytes in hippocampal CA3. FUS-induced BBB disruption did not increase the incidence of microhemorrhage in mice with or without 07/2a mAb treatment.Our findings suggest that FUS is useful tool that may enhance delivery of an anti-pGlu3 Aβ mAb for immunotherapy. FUS-mediated BBB disruption in combination with the 07/2a mAb also appears to facilitate monocyte infiltration in this AD model. Overall, these effects resulted in greater sparing of synapses and improved cognitive function without causing overt damage, suggesting the possibility of FUS as a noninvasive method to increase the therapeutic efficacy in AD patients.
- Published
- 2021
- Full Text
- View/download PDF
25. Focused ultrasound with anti-pGlu3 Aβ enhances efficacy in Alzheimer's disease-like mice via recruitment of peripheral immune cells
- Author
-
Chanikarn Power, Qiaoqiao Shi, Yongzhi Zhang, Camilla Hoesch, Shawna Antonelli, Stephan Schilling, Tao Sun, Cynthia A. Lemere, Maren K. Schroeder, Thore Hettmann, Barbara J. Caldarone, Nadine Taudte, Nathan McDannold, and Mathias Schenk
- Subjects
medicine.drug_class ,medicine.medical_treatment ,Pharmaceutical Science ,Mice, Transgenic ,Plaque, Amyloid ,02 engineering and technology ,Pharmacology ,Hippocampal formation ,Blood–brain barrier ,Monoclonal antibody ,Article ,03 medical and health sciences ,Mice ,Immune system ,Alzheimer Disease ,medicine ,Animals ,Humans ,030304 developmental biology ,Aged ,0303 health sciences ,Amyloid beta-Peptides ,Microglia ,business.industry ,Monocyte ,Brain ,Immunotherapy ,021001 nanoscience & nanotechnology ,Pyrrolidonecarboxylic Acid ,Disease Models, Animal ,medicine.anatomical_structure ,Drug delivery ,0210 nano-technology ,business - Abstract
Pyroglutamate-3 amyloid-β (pGlu3 Aβ) is an N-terminally modified, pathogenic form of amyloid-β that is present in cerebral amyloid plaques and vascular deposits. Here, for the first time, wee used focused ultrasound (FUS) with microbubbles to enhance the intravenous delivery of an Fc-competent anti-pGlu3 Aβ monoclonal antibody, 07/2a mAb, across the blood brain barrier (BBB) in an attempt to improve Aβ removal and memory in aged APP/PS1dE9 mice, an Alzheimer’s disease-like model of amyloidogenesis. First, we demonstrated that bilateral hippocampal FUS-BBBD led to a 5.5-fold increase of 07/2a mAb in the brains of sonicated aged APP/PS1dE9 mice compared to non-sonicated mice 72 hours following a single treatment. Then, we determined that three weekly treatments with 07/2a mAb alone improved spatial learning and memory in aged, plaque-rich APP/PS1dE9 mice, and that this improvement occurred faster and in a higher percentage of animals when combined with FUS-BBBD. APP/PS1dE9 mice given the combination treatment had reduced hippocampal plaque burden compared to PBS-treated APP/PS1dE9 mice. Furthermore, synaptic protein levels were higher in hippocampal synaptosomes from APP/PS1 mice given the combination treatment compared to sham controls, and there were more CA3 synaptic puncta labeled in the APP/PS1dE9 mice given the combination treatment compared to those given mAb alone. Plaque-associated microglia were present in the hippocampi of APP/PS1dE9 mice treated with 07/2a mAb with and without FUS-BBBD. However, we discovered that plaque-associated Ly6G+ monocytes were only present in the hippocampi of APP/PS1dE9 mice that were given FUS-BBBD alone or even more so, the combination treatment. Lastly, FUS- BBBD did not increase the incidence of microhemorrhage in mice with or without 07/2a mAb treatment. Our findings suggest that FUS is a useful tool to enhance delivery and efficacy of an anti-pGlu3 Aβ mAb for immunotherapy either via an additive effect or an independent mechanism. We revealed a potential novel mechanism wherein the combination of 07/2a mAb with FUS-BBBD led to greater monocyte infiltration and recruitment to plaques in this AD-like model. Overall, these effects resulted in greater plaque removal, sparing of synapses and improved cognitive function without causing overt damage, suggesting the possibility of FUS-BBBD as a noninvasive method to increase the therapeutic efficacy of drugs or biologics in AD patients.
- Published
- 2021
26. Combination of the glutaminyl cyclase inhibitor PQ912 (Varoglutamstat) and the murine monoclonal antibody PBD-C06 (m6) shows additive effects on brain Aβ pathology in transgenic mice
- Author
-
Stephan Schilling, Jens-Ulrich Rahfeld, Birgit Hutter-Paier, Cynthia A. Lemere, Koki Makioka, Torsten Hoffmann, Falk Ponath, Mathias Schenk, Inge Lues, and Publica
- Subjects
Genetically modified mouse ,Combination therapy ,medicine.drug_class ,QH301-705.5 ,medicine.medical_treatment ,drug combination ,hAPPsl×hQC mice ,Mice, Transgenic ,Pharmacology ,Monoclonal antibody ,Catalysis ,Inorganic Chemistry ,Antibodies, Monoclonal, Murine-Derived ,Mice ,Alzheimer Disease ,medicine ,Animals ,Humans ,Transgenic mice ,Physical and Theoretical Chemistry ,Biology (General) ,Imidazolines ,Molecular Biology ,QD1-999 ,Spectroscopy ,Amyloid beta-Peptides ,biology ,Chemistry ,Communication ,Organic Chemistry ,Therapeutic effect ,Glutaminyl cyclase ,General Medicine ,Immunotherapy ,Alzheimer's disease ,Aminoacyltransferases ,Small molecule ,Peptide Fragments ,Anti-pyroglutamyl β-amyloid antibody ,Computer Science Applications ,Immuntherapie ,glutaminyl cyclase inhibitor ,biology.protein ,Benzimidazoles ,immunotherapy ,Antibody ,Alzheimer’s disease - Abstract
Compelling evidence suggests that pyroglutamate-modified Aβ (pGlu3-Aβ; AβN3pG) peptides play a pivotal role in the development and progression of Alzheimer’s disease (AD). Approaches targeting pGlu3-Aβ by glutaminyl cyclase (QC) inhibition (Varoglutamstat) or monoclonal antibodies (Donanemab) are currently in clinical development. Here, we aimed at an assessment of combination therapy of Varoglutamstat (PQ912) and a pGlu3-Aβ-specific antibody (m6) in transgenic mice. Whereas the single treatments at subtherapeutic doses show moderate (16–41%) but statistically insignificant reduction of Aβ42 and pGlu-Aβ42 in mice brain, the combination of both treatments resulted in significant reductions of Aβ by 45–65%. Evaluation of these data using the Bliss independence model revealed a combination index of ≈1, which is indicative for an additive effect of the compounds. The data are interpreted in terms of different pathways, in which the two drugs act. While PQ912 prevents the formation of pGlu3-Aβ in different compartments, the antibody is able to clear existing pGlu3-Aβ deposits. The results suggest that combination of the small molecule Varoglutamstat and a pE3Aβ-directed monoclonal antibody may allow a reduction of the individual compound doses while maintaining the therapeutic effect.
- Published
- 2021
27. Further understanding the connection between Alzheimer's disease and Down syndrome
- Author
-
Michael S. Rafii, H. Diana Rosas, Heather M. Snyder, Huntington Potter, Florence Lai, James Hendrix, Andre Strydom, Anna J. Esbensen, Ann-Charlotte Granholm, Fabian Fernandez, Priya S. Kishnani, Michelle Sie Whitten, Lisa J. Bain, Elliott J. Mufson, William C. Mobley, Shahid Zaman, Patrick J. Lao, Cynthia A. Lemere, Elizabeth Head, Adam M. Brickman, Sigan L. Hartley, Joaquín M. Espinosa, Maria C. Carrillo, and Juan Fortea
- Subjects
0301 basic medicine ,Gerontology ,Down syndrome ,Aging ,Future studies ,Epidemiology ,media_common.quotation_subject ,Intellectual and Developmental Disabilities (IDD) ,Clinical Sciences ,Disease ,Neurodegenerative ,Article ,Unmet needs ,vascular contributions ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Presentation ,0302 clinical medicine ,Developmental Neuroscience ,Alzheimer Disease ,medicine ,Acquired Cognitive Impairment ,Dementia ,Humans ,2.1 Biological and endogenous factors ,Aetiology ,media_common ,trisomy ,Amyloid beta-Peptides ,Health Policy ,Neurosciences ,Foundation (evidence) ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Alzheimer's disease ,medicine.disease ,Brain Disorders ,Psychiatry and Mental health ,030104 developmental biology ,Good Health and Well Being ,Geriatrics ,Neurological ,Life expectancy ,Neurology (clinical) ,Geriatrics and Gerontology ,Down Syndrome ,Psychology ,030217 neurology & neurosurgery - Abstract
Improved medical care of individuals with Down syndrome (DS) has led to an increase in life expectancy to over the age of 60 years. In conjunction, there has been an increase in age-related co-occurring conditions including Alzheimer's disease (AD). Understanding the factors that underlie symptom and age of clinical presentation of dementia in people with DS may provide insights into the mechanisms of sporadic and DS-associated AD (DS-AD). In March 2019, the Alzheimer's Association, Global Down Syndrome Foundation and the LuMind IDSC Foundation partnered to convene a workshop to explore the state of the research on the intersection of AD and DS research; to identify research gaps and unmet needs; and to consider how best to advance the field. This article provides a summary of discussions, including noting areas of emerging science and discovery, considerations for future studies, and identifying open gaps in our understanding for future focus.
- Published
- 2020
28. Phosphorylated Aβ peptides in human Down syndrome brain and different Alzheimer's-like mouse models
- Author
-
Jochen Walter, Cynthia A. Lemere, and Sathish Kumar
- Subjects
Genetically modified mouse ,Transgene ,Down syndrome ,Mutant ,Plaque, Amyloid ,Mouse models ,Presenilin ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,Serine ,Cellular and Molecular Neuroscience ,Mice ,Alzheimer Disease ,Extracellular ,medicine ,Amyloid precursor protein ,Animals ,Humans ,Phosphorylation ,Cerebral amyloid angiopathy ,lcsh:Neurology. Diseases of the nervous system ,Amyloid beta-Peptides ,biology ,Chemistry ,Research ,Brain ,Modified Aβ ,medicine.disease ,Cell biology ,Disease Models, Animal ,biology.protein ,Neurology (clinical) ,Post-translational modification ,Amyloid β peptide ,Alzheimer’s disease - Abstract
The deposition of neurotoxic amyloid-β (Aβ) peptides in extracellular plaques in the brain parenchyma is one of the most prominent neuropathological features of Alzheimer’s disease (AD), and considered to be closely related to the pathogenesis of this disease. A number of recent studies demonstrate the heterogeneity in the composition of Aβ deposits in AD brains, due to the occurrence of elongated, truncated and post-translationally modified Aβ peptides that have peculiar characteristics in aggregation behavior and biostability. Importantly, the detection of modified Aβ species has been explored to characterize distinct stages of AD, with phosphorylated Aβ being present in the clinical phase of AD. People with Down syndrome (DS) develop AD pathology by 40 years of age likely due to the overproduction of Aβ caused by the additional copy of the gene encoding the amyloid precursor protein on chromosome 21. In the current study, we analysed the deposition of phosphorylated and non-phosphorylated Aβ species in human DS, AD, and control brains. In addition, deposition of these Aβ species was analysed in brains of a series of established transgenic AD mouse models using phosphorylation-state specific Aβ antibodies. Significant amounts of Aβ phosphorylated at serine residue 8 (pSer8Aβ) and unmodified Aβ were detected in the brains of DS and AD cases. The brains of different transgenic mouse models with either only human mutant amyloid precursor protein (APP), or combinations of human mutant APP, Presenilin (PS), and tau transgenes showed distinct age-dependent and spatiotemporal deposition of pSer8Aβ in extracellular plaques and within the vasculature. Together, these results demonstrate the deposition of phosphorylated Aβ species in DS brains, further supporting the similarity of Aβ deposition in AD and DS. Thus, the detection of phosphorylated and other modified Aβ species could contribute to the understanding and dissection of the complexity in the age-related and spatiotemporal deposition of Aβ variants in AD and DS as well as in distinct mouse models.
- Published
- 2020
29. Deposition of phosphorylated amyloid-β in brains of aged nonhuman primates and canines
- Author
-
Sathish Kumar, Carl W. Cotman, Roberta M. Palmour, Cynthia A. Lemere, Jeffrey L. Frost, Elizabeth Head, and Jochen Walter
- Subjects
0301 basic medicine ,Amyloid β ,Chemistry ,General Neuroscience ,Molecular biology ,Cercopithecus aethiops ,Pathology and Forensic Medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Phosphorylation ,Neurology (clinical) ,Deposition (chemistry) ,030217 neurology & neurosurgery - Published
- 2018
- Full Text
- View/download PDF
30. Age-related epigenetic changes in hippocampal subregions of four animal models of Alzheimer's disease
- Author
-
Daniel L.A. van den Hove, Bart P. F. Rutten, Roberta M. Palmour, Monique Havermans, Roy Lardenoije, Kevin X. Le, Anne van Casteren, Cynthia A. Lemere, Psychiatrie & Neuropsychologie, RS: MHeNs - R3 - Neuroscience, and Promovendi MHN
- Subjects
0301 basic medicine ,Aging ,DNA methyltransferase ,Hippocampal formation ,Hippocampus ,CALORIC RESTRICTION ,DNA Methyltransferase 3A ,Epigenesis, Genetic ,Amyloid beta-Protein Precursor ,Mice ,0302 clinical medicine ,DNA hydroxymethylation ,Chlorocebus aethiops ,Genetics ,TRANSGENIC MICE ,DNA methylation ,Alzheimer's disease ,AMYLOID-BETA ,HUMAN BRAIN ,Animal models ,HISTONE ACETYLATION ,APP/PS1 MOUSE MODEL ,Genetically modified mouse ,DNA Hydroxymethylation ,medicine.medical_specialty ,Amyloid beta ,S-ADENOSYLMETHIONINE ,Mice, Transgenic ,Biology ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Species Specificity ,Alzheimer Disease ,Internal medicine ,medicine ,Animals ,Humans ,Epigenetics ,Molecular Biology ,A-BETA LEVELS ,Dentate gyrus ,PLAQUE-FORMATION ,GLOBAL DNA METHYLATION ,Cell Biology ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,biology.protein ,030217 neurology & neurosurgery - Abstract
Both aging and Alzheimer's disease (AD) are associated with widespread epigenetic changes, with most evidence suggesting global hypomethylation in AD. It is, however, unclear how these age-related epigenetic changes are linked to molecular aberrations as expressed in animal models of AD. Here, we investigated age-related changes of epigenetic markers of DNA methylation and hydroxymethylation in a range of animal models of AD, and their correlations with amyloid plaque load. Three transgenic mouse models, including the J20, APP/PS1dE9 and 3xTg-AD models, as well as Caribbean vervets (a non-transgenic non-human primate model of AD) were investigated. In the J20 mouse model, an age-related decrease in DNA methylation was found in the dentate gyrus (DG) and a decrease in the ratio between DNA methylation and hydroxymethylation was found in the DG and cornu ammonis (CA) 3. In the 3xTg-AD mice, an age-related increase in DNA methylation was found in the DG and CA1-2. No significant age-related alterations were found in the APP/PS1dE9 mice and non-human primate model. In the J20 model, hippocampal plaque load showed a significant negative correlation with DNA methylation in the DG, and with the ratio a negative correlation in the DG and CA3. For the APP/PS1dE9 model a negative correlation between the ratio and plaque load was observed in the CA3, as well as a negative correlation between DNA methyltransferase 3A (DNMT3A) levels and plaque load in the DG and CA3. Thus, only the J20 model showed an age-related reduction in global DNA methylation, while DNA hypermethylation was observed in the 3xTg-AD model. Given these differences between animal models, future studies are needed to further elucidate the contribution of different AD-related genetic variation to age-related epigenetic changes.
- Published
- 2018
- Full Text
- View/download PDF
31. Long-Term Sex- and Genotype-Specific Effects of 56Fe Irradiation on Wild-Type and APPswe/PS1dE9 Transgenic Mice
- Author
-
Maren K. Schroeder, Bin Liu, Robert G. Hinshaw, Mi-Ae Park, Shuyan Wang, Shipra Dubey, Grace Geyu Liu, Qiaoqiao Shi, Peter Holton, Vladimir Reiser, Paul A. Jones, William Trigg, Marcelo F. Di Carli, Barbara J. Caldarone, Jacqueline P. Williams, M. Kerry O’Banion, and Cynthia A. Lemere
- Subjects
sex differences ,Male ,Time Factors ,Genotype ,QH301-705.5 ,Mice, Transgenic ,Article ,Catalysis ,Inorganic Chemistry ,Amyloid beta-Protein Precursor ,Mice ,56Fe radiation ,Alzheimer Disease ,Presenilin-1 ,Animals ,Biology (General) ,Physical and Theoretical Chemistry ,QD1-999 ,Molecular Biology ,Spectroscopy ,Spatial Memory ,Iron Radioisotopes ,Sex Characteristics ,Behavior, Animal ,Organic Chemistry ,General Medicine ,Computer Science Applications ,Chemistry ,Gamma Rays ,Female ,Alzheimer’s disease - Abstract
Space radiation presents a substantial threat to travel beyond Earth. Relatively low doses of high-energy particle radiation cause physiological and behavioral impairments in rodents and may pose risks to human spaceflight. There is evidence that 56Fe irradiation, a significant component of space radiation, may be more harmful to males than to females and worsen Alzheimer’s disease pathology in genetically vulnerable models. Yet, research on the long-term, sex- and genotype-specific effects of 56Fe irradiation is lacking. Here, we irradiated 4-month-old male and female, wild-type and Alzheimer’s-like APP/PS1 mice with 0, 0.10, or 0.50 Gy of 56Fe ions (1GeV/u). Mice underwent microPET scans before and 7.5 months after irradiation, a battery of behavioral tests at 11 months of age and were sacrificed for pathological and biochemical analyses at 12 months of age. 56Fe irradiation worsened amyloid-beta (Aβ) pathology, gliosis, neuroinflammation and spatial memory, but improved motor coordination, in male transgenic mice and worsened fear memory in wild-type males. Although sham-irradiated female APP/PS1 mice had more cerebral Aβ and gliosis than sham-irradiated male transgenics, female mice of both genotypes were relatively spared from radiation effects 8 months later. These results provide evidence for sex-specific, long-term CNS effects of space radiation.
- Published
- 2021
- Full Text
- View/download PDF
32. APOE ε4 Association With Cognition and Alzheimer Disease Biomarkers in Down Syndrome—Implications for Clinical Trials and Treatments for All
- Author
-
Cynthia A. Lemere, Elizabeth Head, and David M. Holtzman
- Subjects
Apolipoprotein E ,Oncology ,medicine.medical_specialty ,Down syndrome ,business.industry ,MEDLINE ,Cognition ,medicine.disease ,Clinical trial ,Internal medicine ,medicine ,Neurology (clinical) ,Alzheimer's disease ,business ,Association (psychology) - Published
- 2021
- Full Text
- View/download PDF
33. mTORC2 (Rictor) in Alzheimer’s Disease and Reversal of Amyloid-β Expression-Induced Insulin Resistance and Toxicity in Rat Primary Cortical Neurons
- Author
-
Bumsup Kwon, Austin Y. Ha, Kyohei Itamura, Henry W. Querfurth, Suzanne M. de la Monte, Han-Kyu Lee, and Cynthia A. Lemere
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Cell Survival ,medicine.medical_treatment ,Mice, Transgenic ,Mechanistic Target of Rapamycin Complex 2 ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,mTORC2 ,Article ,Rats, Sprague-Dawley ,03 medical and health sciences ,Insulin resistance ,Alzheimer Disease ,Internal medicine ,Autophagy ,medicine ,Animals ,Humans ,Protein kinase B ,Cells, Cultured ,PI3K/AKT/mTOR pathway ,Aged ,Aged, 80 and over ,Cerebral Cortex ,Neurons ,Amyloid beta-Peptides ,Chemistry ,Cell growth ,General Neuroscience ,Insulin ,General Medicine ,Middle Aged ,medicine.disease ,Peptide Fragments ,Psychiatry and Mental health ,Clinical Psychology ,030104 developmental biology ,Endocrinology ,Cell culture ,Female ,Insulin Resistance ,biological phenomena, cell phenomena, and immunity ,Geriatrics and Gerontology ,Proto-Oncogene Proteins c-akt - Abstract
Mammalian target of rapamycin complex 1 (mTORC1), a nutrient sensor and central controller of cell growth and proliferation, is altered in various models of Alzheimer's disease (AD). Even less studied or understood in AD is mammalian target of rapamycin complex 2 (mTORC2) that influences cellular metabolism, in part through the regulations of Akt/PKB and SGK. Dysregulation of insulin/PI3K/Akt signaling is another important feature of AD pathogenesis. We found that both total mTORC1 and C2 protein levels and individual C1 and C2 enzymatic activities were decreased in human AD brain samples. In two rodent AD models, mTORC1 and C2 activities were also decreased. In a neuronal culture model of AD characterized by accumulation of cellular amyloid-β (Aβ)42, mTORC1 activity was reduced. Autophagic vesicles and markers were correspondingly increased and new protein synthesis was inhibited, consistent with mTORC1 hypofunction. Interestingly, mTORC2 activity in neural culture seemed resistant to the effects of intracellular amyloid. In various cell lines, Aβ expression provoked insulin resistance, characterized by inhibition of stimulated Akt phosphorylation, and an increase in negative mTORC1 regular, p-AMPK, itself a nutrient sensor. Rapamycin decreased phospho-mTOR and to lesser degree p-Rictor. This further suppression of mTORC1 activity protected cells from Aβ-induced toxicity and insulin resistance. More striking, Rictor over-expression fully reversed the Aβ-effects on primary neuronal cultures. Finally, using in vitro assay, Rictor protein addition completely overcame oligomeric Aβ-induced inhibition of the PDK-Akt activation step. We conclude that striking a new balance by restoring mTORC2 abundance and/or inhibition of mTORC1 has therapeutic potential in AD.
- Published
- 2017
- Full Text
- View/download PDF
34. BrightFocus Alzheimer's Fast Track 2019
- Author
-
Frank M. LaFerla, Diane E. Bovenkamp, Cynthia A. Lemere, Harry W.M. Steinbusch, Keith W. Whitaker, and Academic Affairs
- Subjects
0301 basic medicine ,Scientific field ,lcsh:Geriatrics ,Meeting Report ,lcsh:RC346-429 ,Education ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Alzheimer Disease ,medicine ,Humans ,Dementia ,Neurodegeneration ,Molecular Biology ,Workshop ,lcsh:Neurology. Diseases of the nervous system ,Medical education ,Alzheimer's disease ,medicine.disease ,Research Personnel ,lcsh:RC952-954.6 ,030104 developmental biology ,ComputingMethodologies_PATTERNRECOGNITION ,Graduate students ,ComputingMilieux_COMPUTERSANDSOCIETY ,Neurology (clinical) ,Fast track ,Psychology ,Alzheimer’s disease ,030217 neurology & neurosurgery - Abstract
The 3 day workshop “Alzheimer’s Fast Track” is a unique opportunity for graduate students, postdoctoral fellows, or other early-career scientists, focused on Alzheimer’s disease research, to gain new knowledge and become an expert in where this emerging scientific field is moving. In addition, it is not only about receiving a good overview, but also learning to write and defend a successful application for securing funding for Alzheimer’s disease research projects.
- Published
- 2019
35. Scientific Advising and Reviewing: On strengthening the bond between the Alzheimer's Association and the scientific community
- Author
-
David S. Knopman, Todd E. Golde, Bruce T. Lamb, Zaven S. Khachaturian, Mary Sano, Hui Zheng, Suzanne Craft, Gerard D. Schellenberg, Laura N. Gitlin, Margaret A. Pericak-Vance, Heather M. Snyder, Cynthia A. Lemere, Julie A. Schneider, and Maria C. Carrillo
- Subjects
Epidemiology ,business.industry ,Health Policy ,Bond ,Association (object-oriented programming) ,Research ,education ,MEDLINE ,Public relations ,Caregiver support ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Alzheimer Disease ,Political science ,Humans ,Neurology (clinical) ,Geriatrics and Gerontology ,Public education ,business ,Societies ,Intersectoral Collaboration ,health care economics and organizations - Abstract
From its inception in 1980, advancement of research was one of the primary missions of the Alzheimer's Association (also known as Alzheimer's Disease and Related Disorders Association) in addition to leading in family caregiver support, better care, public education, and awareness. Over the past 30 years, the Association has grown and expanded its engagement with the scientific community. In the past 10 years, its research budget has more than doubled, greatly increasing the number of research grants funded and the number of strategic projects supported. The leadership and members of the Medical and Scientific Advisory Council recognized that the growth of the Alzheimer's Association and the expanded mission of Medical & Scientific Relations Division necessitated a change in the mission and charge of the external scientific advisory function to the Association.
- Published
- 2019
36. Space-like 56Fe irradiation manifests mild, early sex-specific behavioral and neuropathological changes in wildtype and Alzheimer’s-like transgenic mice
- Author
-
Lee A. Trojanczyk, M. Kerry O'Banion, Qiaoqiao Shi, Paul A. Jones, Shuyan Wang, Bin Liu, Marcelo F. Di Carli, Robert G. Hinshaw, Paul Lorello, Jacqueline P. Williams, Jeffrey L. Frost, Vladimir Reiser, Kevin X. Le, Mi-Ae Park, William Trigg, Anthony P. Belanger, Cynthia A. Lemere, Shipra Dubey, Barbara J. Caldarone, and Peter Holton
- Subjects
Male ,0301 basic medicine ,lcsh:Medicine ,Behavioural methods ,0302 clinical medicine ,lcsh:Science ,Iron Radioisotopes ,Multidisciplinary ,Behavior, Animal ,Microglia ,biology ,Brain ,Alzheimer's disease ,Experimental models of disease ,medicine.anatomical_structure ,Cerebral blood flow ,Female ,Genetically modified mouse ,medicine.medical_specialty ,Transgene ,Mice, Transgenic ,Neuropathology ,Motor Activity ,Article ,03 medical and health sciences ,Sex Factors ,Alzheimer Disease ,Internal medicine ,Presenilin-1 ,medicine ,Translocator protein ,Animals ,Humans ,Learning ,Neuroinflammation ,Inflammation ,Amyloid beta-Peptides ,business.industry ,lcsh:R ,Wild type ,Dose-Response Relationship, Radiation ,Space Flight ,Astrobiology ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,biology.protein ,lcsh:Q ,business ,030217 neurology & neurosurgery - Abstract
Space travel will expose people to high-energy, heavy particle radiation, and the cognitive deficits induced by this exposure are not well understood. To investigate the short-term effects of space radiation, we irradiated 4-month-old Alzheimer’s disease (AD)-like transgenic (Tg) mice and wildtype (WT) littermates with a single, whole-body dose of 10 or 50 cGy 56Fe ions (1 GeV/u) at Brookhaven National Laboratory. At ~1.5 months post irradiation, behavioural testing showed sex-, genotype-, and dose-dependent changes in locomotor activity, contextual fear conditioning, grip strength, and motor learning, mainly in Tg but not WT mice. There was little change in general health, depression, or anxiety. Two months post irradiation, microPET imaging of the stable binding of a translocator protein ligand suggested no radiation-specific change in neuroinflammation, although initial uptake was reduced in female mice independently of cerebral blood flow. Biochemical and immunohistochemical analyses revealed that radiation reduced cerebral amyloid-β levels and microglia activation in female Tg mice, modestly increased microhemorrhages in 50 cGy irradiated male WT mice, and did not affect synaptic marker levels compared to sham controls. Taken together, we show specific short-term changes in neuropathology and behaviour induced by 56Fe irradiation, possibly having implications for long-term space travel.
- Published
- 2019
- Full Text
- View/download PDF
37. Active Amyloid-β Vaccination Results in Epigenetic Changes in the Hippocampus of an Alzheimer's Disease-Like Mouse Model
- Author
-
Sophie E Jung, Bart P. F. Rutten, Bin Liu, Daniel L.A. van den Hove, Peter Blackburn, Monique Havermans, Roy Lardenoije, Cynthia A. Lemere, Psychiatrie & Neuropsychologie, RS: MHeNs - R2 - Mental Health, RS: MHeNs - R3 - Neuroscience, and MUMC+: MA Psychiatrie (3)
- Subjects
0301 basic medicine ,Male ,Plaque, Amyloid ,Active immunotherapy ,Active immunization ,Hippocampus ,DNA Methyltransferase 3A ,Epigenesis, Genetic ,chemistry.chemical_compound ,Amyloid beta-Protein Precursor ,Random Allocation ,0302 clinical medicine ,DNA methylation ,biology ,Vaccination ,Alzheimer's disease ,IMMUNIZATION ,amyloid-beta ,Neurology ,IMPROVES COGNITION ,Mice, Inbred DBA ,5-HYDROXYMETHYLCYTOSINE ,Female ,PLAQUES ,DNA Hydroxymethylation ,Amyloid beta ,mouse model ,HYDROXYMETHYLATION ,DNA-METHYLATION ,Mice, Transgenic ,AN1792 ,03 medical and health sciences ,Alzheimer Disease ,Presenilin-1 ,Animals ,Humans ,Epigenetics ,IMMUNOTHERAPY ,5-Hydroxymethylcytosine ,Amyloid beta-Peptides ,epigenetics ,Epigenome ,GLOBAL DNA ,Mice, Inbred C57BL ,PATHOLOGY ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Immunology ,biology.protein ,Neurology (clinical) ,active vaccine ,030217 neurology & neurosurgery - Abstract
Background: While evidence accumulates for a role of epigenetic modifications in the pathophysiological cascade of Alzheimer’s disease (AD), amyloid-β (Aβ)-targeted active immunotherapy approaches are under investigation to prevent or slow the progression of AD. The impact of Aβ active vaccines on epigenetic markers has not been studied thus far. Objective: The current study aims to establish the relationship between active immunotherapy with a MER5101-based vaccine (consisting of Aβ1-15 copies conjugated with a 7 aa spacer to the diphtheria toxoid carrier protein, formulated in a Th2-biased adjuvant) and epigenetic DNA modifications in the hippocampus of APPswe/PS1dE9 mice. Methods: As we previously reported, immunotherapy started when the mice were 10 months of age and behavioral testing occurred at 14 months of age, after which the mice were sacrificed for further analysis of their brains. In this add-on study, global levels of DNA methylation and hydroxymethylation, and DNA methyltransferase 3A (DNMT3A) were determined using quantitative immunohistochemistry, and compared to our previously analyzed immunization-induced changes in AD-related neuropathology and cognition. Results: Active immunization did not affect global DNA methylation levels but instead, resulted in decreased DNA hydroxymethylation and DNMT3A levels. Independent of immunization, inverse correlations with behavioral performance were observed for levels of DNA methylation and hydroxymethylation, as well as DNMT3A, while Aβ pathology and synaptic markers did not correlate with DNA methylation levels but did positively correlate with DNA hydroxymethylation and levels of DNMT3A. Conclusion: Our results indicate that active Aβ vaccination has significant effects on the epigenome in the hippocampus of APPswe/PS1dE9 mice, and suggest that DNA methylation and hydroxymethylation may be involved in cognitive functioning.
- Published
- 2019
38. Antibody Engineering & Therapeutics 2016: The Antibody Society's annual meeting, December 11–15, 2016, San Diego, CA
- Author
-
Kerry A. Chester, Dennis R. Burton, Anne Messer, Trudi Veldman, James S. Huston, Mark R Alfenito, Gregory P. Adams, Andrew Bradbury, Jamie K. Scott, Janice M. Reichert, Paul W. H. I. Parren, Paul Carter, Janine Schuurman, James W Larrick, and Cynthia A. Lemere
- Subjects
0301 basic medicine ,Bispecific antibody ,Immunology ,Antibody engineering ,antibody-drug conjugates ,international non-proprietary names ,Library science ,Meeting Preview ,03 medical and health sciences ,0302 clinical medicine ,Antibody Repertoire ,Antibodies monoclonal ,antibody repertoire ,Immunology and Allergy ,Medicine ,Computational design ,Effector functions ,reproducibility ,biology ,business.industry ,Medical school ,030104 developmental biology ,biology.protein ,next-generation sequencing ,bispecific antibodies ,Antibody ,business ,030217 neurology & neurosurgery - Abstract
Antibody Engineering & Therapeutics, the largest meeting devoted to antibody science and technology and the annual meeting of The Antibody Society, will be held in San Diego, CA on December 11-15, 2016. Each of 14 sessions will include six presentations by leading industry and academic experts. In this meeting preview, the session chairs discuss the relevance of their topics to current and future antibody therapeutics development. Session topics include bispecifics and designer polyclonal antibodies; antibodies for neurodegenerative diseases; the interface between passive and active immunotherapy; antibodies for non-cancer indications; novel antibody display, selection and screening technologies; novel checkpoint modulators / immuno-oncology; engineering antibodies for T-cell therapy; novel engineering strategies to enhance antibody functions; and the biological Impact of Fc receptor engagement. The meeting will open with keynote speakers Dennis R. Burton (The Scripps Research Institute), who will review progress toward a neutralizing antibody-based HIV vaccine; Olivera J. Finn, (University of Pittsburgh School of Medicine), who will discuss prophylactic cancer vaccines as a source of therapeutic antibodies; and Paul Richardson (Dana-Farber Cancer Institute), who will provide a clinical update on daratumumab for multiple myeloma. In a featured presentation, a representative of the World Health Organization's INN expert group will provide a perspective on antibody naming. “Antibodies to watch in 2017” and progress on The Antibody Society's 2016 initiatives will be presented during the Society's special session. In addition, two pre-conference workshops covering ways to accelerate antibody drugs to the clinic and the applications of next-generation sequencing in antibody discovery and engineering will be held on Sunday December 11, 2016.
- Published
- 2016
- Full Text
- View/download PDF
39. Traumatic Brain Injury in Aged Mice Induces Chronic Microglia Activation, Synapse Loss, and Complement-Dependent Memory Deficits
- Author
-
Cynthia A. Lemere, Maria-Serena Paladini, Xi Feng, Susanna Rosi, Karen Krukowski, Lara-Kirstie Riparip, Austin Chou, Myriam M. Chaumeil, and Brice Tiret
- Subjects
0301 basic medicine ,Traumatic ,Male ,Aging ,Hippocampus ,microglia ,Cell Count ,Neurodegenerative ,Inbred C57BL ,Alzheimer's Disease ,Synapse ,lcsh:Chemistry ,Mice ,0302 clinical medicine ,Injury - Trauma - (Head and Spine) ,Models ,synapse ,Brain Injuries, Traumatic ,2.1 Biological and endogenous factors ,complement ,Aetiology ,lcsh:QH301-705.5 ,Spectroscopy ,education.field_of_study ,Microglia ,traumatic brain injury ,Brain ,Cognition ,General Medicine ,Magnetic Resonance Imaging ,3. Good health ,Computer Science Applications ,medicine.anatomical_structure ,Blood-Brain Barrier ,Neurological ,Disease Progression ,Female ,Traumatic brain injury ,Contusions ,Population ,Models, Biological ,Catalysis ,Article ,Inorganic Chemistry ,03 medical and health sciences ,Immune system ,Phagocytosis ,Behavioral and Social Science ,medicine ,Acquired Cognitive Impairment ,Genetics ,Animals ,Physical and Theoretical Chemistry ,education ,Molecular Biology ,C1q ,Memory Disorders ,Chemical Physics ,business.industry ,Organic Chemistry ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Complement System Proteins ,medicine.disease ,Biological ,nervous system diseases ,Complement system ,Brain Disorders ,Mice, Inbred C57BL ,030104 developmental biology ,nervous system ,lcsh:Biology (General) ,lcsh:QD1-999 ,Brain Injuries ,Chronic Disease ,Synapses ,Injury (total) Accidents/Adverse Effects ,Dementia ,Other Biological Sciences ,business ,Injury - Traumatic brain injury ,Other Chemical Sciences ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Traumatic brain injury (TBI) is of particular concern for the aging community since there is both increased incidence of TBI and decreased functional recovery in this population. In addition, TBI is the strongest environmental risk factor for development of Alzheimer&rsquo, s disease and other dementia-related neurodegenerative disorders. Critical changes that affect cognition take place over time following the initial insult. Our previous work identified immune system activation as a key contributor to cognitive deficits observed in aged animals. Using a focal contusion model in the current study, we demonstrate a brain lesion and cavitation formation, as well as prolonged blood&ndash, brain barrier breakdown. These changes were associated with a prolonged inflammatory response, characterized by increased microglial cell number and phagocytic activity 30 days post injury, corresponding to significant memory deficits. We next aimed to identify the injury-induced cellular and molecular changes that lead to chronic cognitive deficits in aged animals, and measured increases in complement initiation components C1q, C3, and CR3, which are known to regulate microglial&ndash, synapse interactions. Specifically, we found significant accumulation of C1q on synapses within the hippocampus, which was paralleled by synapse loss 30 days post injury. We used genetic and pharmacological approaches to determine the mechanistic role of complement initiation on cognitive loss in aging animals after TBI. Notably, both genetic and pharmacological blockade of the complement pathway prevented memory deficits in aged injured animals. Thus, therapeutically targeting early components of the complement cascade represents a significant avenue for possible clinical intervention following TBI in the aging population.
- Published
- 2018
40. P1‐099: COMBINATION OF A GLUTAMINYL CYCLASE INHIBITOR (PQ912) AND A PYROGLUTAMATE‐Aβ SPECIFIC ANTIBODY (PBD‐M06) SHOWS ADDITIVE EFFECTS IN A MOUSE MODEL WITH ALZHEIMER'S DISEASE‐LIKE PATHOLOGY
- Author
-
Birgit Hutter-Paier, Stephan Schilling, Cynthia A. Lemere, Martina Farcher, Torsten Hoffmann, and Inge Lues
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Specific antibody ,Developmental Neuroscience ,Epidemiology ,Chemistry ,Health Policy ,Glutaminyl cyclase ,Neurology (clinical) ,Disease ,Geriatrics and Gerontology ,Pharmacology - Published
- 2018
- Full Text
- View/download PDF
41. Passive Aβ Immunotherapy: Current Achievements and Future Perspectives
- Author
-
Cynthia A. Lemere, Inge Lues, Stephan Schilling, and Jens-Ulrich Rahfeld
- Subjects
Cytotoxicity, Immunologic ,Diagnostic Imaging ,0301 basic medicine ,Amyloid ,Amyloid β ,medicine.drug_class ,medicine.medical_treatment ,Pharmaceutical Science ,Plaque, Amyloid ,Review ,amyloid-β ,Monoclonal antibody ,Analytical Chemistry ,lcsh:QD241-441 ,03 medical and health sciences ,0302 clinical medicine ,lcsh:Organic chemistry ,Alzheimer Disease ,Drug Discovery ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,Clinical Trials as Topic ,Amyloid beta-Peptides ,business.industry ,Passive Immunotherapy ,Organic Chemistry ,Aβ peptide ,Antibodies, Monoclonal ,Immunotherapy ,drug development ,Biochemistry of Alzheimer's disease ,Disease Models, Animal ,030104 developmental biology ,Drug development ,Blood-Brain Barrier ,Chemistry (miscellaneous) ,Molecular Medicine ,posttranslational modifications ,monoclonal antibodies ,business ,Protein Processing, Post-Translational ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Passive immunotherapy has emerged as a very promising approach for the treatment of Alzheimer’s disease and other neurodegenerative disorders, which are characterized by the misfolding and deposition of amyloid peptides. On the basis of the amyloid hypothesis, the majority of antibodies in clinical development are directed against amyloid β (Aβ), the primary amyloid component in extracellular plaques. This review focuses on the current status of Aβ antibodies in clinical development, including their characteristics and challenges that came up in clinical trials with these new biological entities (NBEs). Emphasis is placed on the current view of common side effects observed with passive immunotherapy, so-called amyloid-related imaging abnormalities (ARIAs), and potential ways to overcome this issue. Among these new ideas, a special focus is placed on molecules that are directed against post-translationally modified variants of the Aβ peptide, an emerging approach for development of new antibody molecules.
- Published
- 2018
42. In VivoDetection of Age- and Disease-Related Increases in Neuroinflammation by18F-GE180 TSPO MicroPET Imaging in Wild-Type and Alzheimer's Transgenic Mice
- Author
-
Shuyan Wang, Mi-Ae Park, Peter Holton, Bin Liu, Paul A. Jones, William Trigg, Jeffrey L. Frost, Shipra Dubey, Marcelo F. Di Carli, Kevin X. Le, Vladimir Reiser, Cynthia A. Lemere, and Anthony P. Belanger
- Subjects
Male ,Genetically modified mouse ,Aging ,Fluorine Radioisotopes ,medicine.medical_specialty ,Pathology ,Carbazoles ,Hippocampus ,Mice, Transgenic ,Mice ,Receptors, GABA ,Alzheimer Disease ,In vivo ,Internal medicine ,medicine ,Translocator protein ,Animals ,Humans ,Neuroinflammation ,Inflammation ,biology ,Microglia ,Chemistry ,General Neuroscience ,Articles ,medicine.disease ,Mice, Inbred C57BL ,Endocrinology ,medicine.anatomical_structure ,Positron-Emission Tomography ,Disease Progression ,biology.protein ,Alzheimer's disease ,Ex vivo - Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Neuroinflammation appears to play an important role in AD pathogenesis. Ligands of the 18 kDa translocator protein (TSPO), a marker for activated microglia, have been used as positron emission tomography (PET) tracers to reflect neuroinflammation in humans and mouse models. Here, we used the novel TSPO-targeted PET tracer18F-GE180 (flutriciclamide) to investigate differences in neuroinflammation between young and old WT and APP/PS1dE9 transgenic (Tg) mice.In vivoPET scans revealed an overt age-dependent elevation in whole-brain uptake of18F-GE180 in both WT and Tg mice, and a significant increase in whole-brain uptake of18F-GE180 (peak-uptake and retention) in old Tg mice compared with young Tg mice and all WT mice. Similarly, the18F-GE180 binding potential in hippocampus was highest to lowest in old Tg > old WT > young Tg > young WT mice using MRI coregistration.Ex vivoPET and autoradiography analysis further confirmed ourin vivoPET results: enhanced uptake and specific binding (SUV75%) of18F-GE180 in hippocampus and cortex was highest in old Tg mice followed by old WT, young Tg, and finally young WT mice.18F-GE180 specificity was confirmed by anin vivocold tracer competition study. We also examined18F-GE180 metabolites in 4-month-old WT mice and found that, although total radioactivity declined over 2 h, of the remaining radioactivity, ∼90% was due to parent18F-GE180. In conclusion,18F-GE180 PET scans may be useful for longitudinal monitoring of neuroinflammation during AD progression and treatment.SIGNIFICANCE STATEMENTMicroglial activation, a player in Alzheimer's disease (AD) pathogenesis, is thought to reflect neuroinflammation. Usingin vivomicroPET imaging with a novel TSPO radioligand,18F-GE180, we detected significantly enhanced neuroinflammation during normal aging in WT mice and in response to AD-associated pathology in APP/PS1dE9 Tg mice, an AD mouse model. Increased uptake and specific binding of18F-GE180 in whole brain and hippocampus were confirmed byex vivoPET and autoradiography. The binding specificity and stability of18F-GE180 was further confirmed by a cold tracer competition study and a metabolite study, respectively. Therefore,18F-GE180 PET imaging may be useful for longitudinal monitoring of neuroinflammation during AD progression and treatment and may also be useful for other neurodegenerative diseases.
- Published
- 2015
- Full Text
- View/download PDF
43. ComplementC3-Deficient Mice Fail to Display Age-Related Hippocampal Decline
- Author
-
Katherine Merry, Jean-Cosme Dodart, Cynthia A. Lemere, Sarah B. Matousek, Jessica E. Kenison, Jeffrey L. Frost, Kenneth J. Colodner, Qiaoqiao Shi, Shaomin Li, Soyon Hong, Kevin X. Le, Beth Stevens, and Barbara J. Caldarone
- Subjects
Male ,Aging ,medicine.medical_specialty ,Synaptophysin ,Hippocampus ,Biology ,Hippocampal formation ,Synapse ,Mice ,Internal medicine ,Conditioning, Psychological ,medicine ,Animals ,Aging brain ,Maze Learning ,Mice, Knockout ,Neuronal Plasticity ,Innate immune system ,General Neuroscience ,Age Factors ,Excitatory Postsynaptic Potentials ,Long-term potentiation ,Articles ,Complement C3 ,Fear ,Synapsins ,Adaptation, Physiological ,Complement system ,Mice, Inbred C57BL ,Electrophysiology ,Endocrinology ,Phosphopyruvate Hydratase ,Synapses ,Exploratory Behavior ,Neuroscience ,Synaptosomes - Abstract
The complement system is part of the innate immune response responsible for removing pathogens and cellular debris, in addition to helping to refine CNS neuronal connections via microglia-mediated pruning of inappropriate synapses during brain development. However, less is known about the role of complement during normal aging. Here, we studied the role of the central complement component, C3, in synaptic health and aging. We examined behavior as well as electrophysiological, synaptic, and neuronal changes in the brains ofC3-deficient male mice (C3KO) compared with age-, strain-, and gender-matched C57BL/6J (wild-type, WT) control mice at postnatal day 30, 4 months, and 16 months of age. We found the following: (1) region-specific and age-dependent synapse loss in aged WT mice that was not observed inC3KO mice; (2) age-dependent neuron loss in hippocampal CA3 (but not in CA1) that followed synapse loss in aged WT mice, neither of which were observed in agedC3KO mice; and (3) significantly enhanced LTP and cognition and less anxiety in agedC3KO mice compared with aged WT mice. Importantly, CA3 synaptic puncta were similar between WT andC3KO mice at P30. Together, our results suggest a novel and prominent role for complement protein C3 in mediating aged-related and region-specific changes in synaptic function and plasticity in the aging brain.SIGNIFICANCE STATEMENTThe complement cascade, part of the innate immune response to remove pathogens, also plays a role in synaptic refinement during brain development by the removal of weak synapses. We investigated whether complement C3, a central component, affects synapse loss during aging. Wild-type (WT) andC3knock-out (C3KO) mice were examined at different ages. The mice were similar at 1 month of age. However, with aging, WT mice lost synapses in specific brain regions, especially in hippocampus, an area important for memory, whereasC3KO mice were protected. AgedC3KO mice also performed better on learning and memory tests than aged WT mice. Our results suggest that complement C3, or its downstream signaling, is detrimental to synapses during aging.
- Published
- 2015
- Full Text
- View/download PDF
44. Glio-vascular changes during ageing in wild-type and Alzheimer׳s disease-like APP/PS1 mice
- Author
-
Maria Alexandra Brito, Dora Brites, Cynthia A. Lemere, and Cátia S. Janota
- Subjects
Male ,Aging ,Pathology ,medicine.medical_specialty ,Mice, Transgenic ,Hippocampal formation ,Biology ,Microgliosis ,Hippocampus ,Article ,Pathogenesis ,Amyloid beta-Protein Precursor ,Alzheimer Disease ,Cortex (anatomy) ,Parenchyma ,Presenilin-1 ,medicine ,Animals ,Humans ,Gliosis ,Senile plaques ,Molecular Biology ,Cerebral Cortex ,General Neuroscience ,medicine.disease ,Capillaries ,Astrogliosis ,Mice, Inbred C57BL ,medicine.anatomical_structure ,nervous system ,Blood-Brain Barrier ,Ageing ,Neurology (clinical) ,Pericytes ,Neuroglia ,Neuroscience ,Developmental Biology - Abstract
Vascular and glial involvement in the development of neurodegenerative disorders, such as Alzheimer's disease (AD), and age-related brain vulnerabilities have been suggested. Therefore, we sought to: (i) investigate which vascular and glial events are evident in ageing and/or AD, (ii) to establish the temporal evolution of vascular and glial changes in AD-like and wild-type (WT) mice and (iii) to relate them to amyloid-β (Aβ) peptide accumulation. We examined immunohistochemically hippocampi and cortex from APP/PS1dE9 and WT C57BL/6 mice along ageing and disease progression (young-adulthood, middle- and old-age). Ageing resulted in the increase in receptor for advanced glycation endproducts expression, as well as the entrance of thrombin and albumin in hippocampal parenchyma. In contrast, the loss of platelet-derived growth factor receptor-β (PDGFR-β) positive cells, in both regions, was only related to AD pathogenesis. Hypovascularization was affected by both ageing and AD in the hippocampus, but resulted from the interaction between both factors in the cortex. Astrogliosis was a result of AD in hippocampus and of both factors in cortex, while microgliosis was associated with fibrillar amyloid plaques in AD-like mice and with the interaction between both factors in each of the studied regions. In sum, these data show that senile plaques precede vascular and glial alterations only in hippocampus, whereas in cortex, vascular and glial alterations, namely the loss of PDGFR-β-positive cells and astrogliosis, accompanied the first senile plaques. Hence, this study points to vascular and glial events that co-exist in AD pathogenesis and age-related brain vulnerabilities.
- Published
- 2015
- Full Text
- View/download PDF
45. The epigenetics of aging and neurodegeneration
- Author
-
Gunter Kenis, Cynthia A. Lemere, Paul D. Coleman, Patrick R. Hof, Roy Lardenoije, Diego Mastroeni, Konstantinos Kompotis, Bart P. F. Rutten, Daniel L.A. van den Hove, Artemis Iatrou, Harry W.M. Steinbusch, Promovendi MHN, MUMC+: MA Niet Med Staf Psychiatrie (9), Psychiatrie & Neuropsychologie, and RS: MHeNs - R3 - Neuroscience
- Subjects
Aging ,Epigenetic regulation of neurogenesis ,RNA, Untranslated ,Parkinson's disease ,BACE1-AS ,Epigenetics of schizophrenia ,Biology ,Chromatin remodeling ,Article ,Epigenesis, Genetic ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Epigenetics ,Neurodegeneration ,General Neuroscience ,Neurodegenerative Diseases ,Huntington's disease ,DNA Methylation ,Alzheimer's disease ,medicine.disease ,DNA demethylation ,DNA methylation ,Neuroscience - Abstract
Epigenetics is a quickly growing field encompassing mechanisms regulating gene expression that do not involve changes in the genotype. Epigenetics is of increasing relevance to neuroscience, with epigenetic mechanisms being implicated in brain development and neuronal differentiation, as well as in more dynamic processes related to cognition. Epigenetic regulation covers multiple levels of gene expression; from direct modifications of the DNA and histone tails, regulating the level of transcription, to interactions with messenger RNAs, regulating the level of translation. Importantly, epigenetic dysregulation currently garners much attention as a pivotal player in aging and age-related neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, where it may mediate interactions between genetic and environmental risk factors, or directly interact with disease-specific pathological factors. We review current knowledge about the major epigenetic mechanisms, including DNA methylation and DNA demethylation, chromatin remodeling and non-coding RNAs, as well as the involvement of these mechanisms in normal aging and in the pathophysiology of the most common neurodegenerative diseases. Additionally, we examine the current state of epigenetics-based therapeutic strategies for these diseases, which either aim to restore the epigenetic homeostasis or skew it to a favorable direction to counter disease pathology. Finally, methodological challenges of epigenetic investigations and future perspectives are discussed.
- Published
- 2015
46. Dissecting the Contribution of Vascular Alterations and Aging to Alzheimer’s Disease
- Author
-
Cynthia A. Lemere, Cátia S. Janota, and Maria Alexandra Brito
- Subjects
0301 basic medicine ,Aging ,medicine.medical_specialty ,Neurology ,Neuroscience (miscellaneous) ,Disease ,Biology ,Blood–brain barrier ,Models, Biological ,Pathogenesis ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Alzheimer Disease ,medicine ,Animals ,Humans ,Dementia ,Cognitive decline ,Amyloid beta-Peptides ,Neurodegeneration ,Neurotoxicity ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Blood-Brain Barrier ,Blood Vessels ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by cognitive decline that afflicts as many as 45 % of individuals who survive past the age of 85. AD has been associated with neurovascular dysfunction and brain accumulation of amyloid-β peptide, as well as tau phosphorylation and neurodegeneration, but the pathogenesis of the disease is still somewhat unclear. According to the amyloid cascade hypothesis of AD, accumulation of amyloid-β peptide (Aβ) aggregates initiates a sequence of events leading to neuronal injury and loss, and dementia. Alternatively, the vascular hypothesis of AD incorporates the vascular contribution to the disease, stating that a primary insult to brain microcirculation (e.g., stroke) not only contributes to amyloidopathy but initiates a non-amyloidogenic pathway of vascular-mediated neuronal dysfunction and injury, which involves blood-brain barrier compromise, with increased permeability of blood vessels, leakage of blood-borne components into the brain, and, consequently, neurotoxicity. Vascular dysfunction also includes a diminished brain capillary flow, causing multiple focal ischemic or hypoxic microinjuries, diminished amyloid-β clearance, and formation of neurotoxic oligomers, which lead to neuronal dysfunction. Here we present and discuss relevant findings on the contribution of vascular alterations during aging to AD, with the hope that a better understanding of the players in the "orchestra" of neurodegeneration will be useful in developing therapies to modulate the "symphony".
- Published
- 2015
- Full Text
- View/download PDF
47. [F4–06–02]: UPDATE OF THE AMYLOID HYPOTHESIS: COMPLEMENT MODULATES THE GLIAL RESPONSE TO Aβ PLAQUES AND MEDIATES SYNAPSE LOSS
- Author
-
Cynthia A. Lemere
- Subjects
Synapse ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology ,Biology ,Neuroscience ,Complement (complexity) ,Biochemistry of Alzheimer's disease - Published
- 2017
- Full Text
- View/download PDF
48. The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases
- Author
-
Asaf Madi, Elaine O’Loughlin, Anna Worthmann, Oleg Butovsky, Lien Beckers, Tsuneya Ikezu, Frank L. Heppner, Kristin Hartmann, Vladimir Litvak, Mar Gacias, Markus Glatzel, Zain Fanek, Zachary Humulock, Howard L. Weiner, Susanne Krasemann, Joerg Heeren, Jordi Ochando, Zhiping Weng, Ron Cialic, Christian Haass, Yang Xu, Narghes Calcagno, David M. Holtzman, Tobias Zrzavy, Bogdan Budnik, Cynthia A. Lemere, George Tweet, Hans Lassmann, Scott T. Smith, Hao Chen, Caroline Baufeld, Rachid El Fatimy, David J. Greco, Charlotte Madore, Fargol Mazaheri, Emily C. Tjon, Patricia Conde-Sanroman, and Jason D. Ulrich
- Subjects
0301 basic medicine ,Apolipoprotein E ,Apoptosis ,Plaque, Amyloid ,genetics [Alzheimer Disease] ,immunology [Phagocytosis] ,metabolism [Microglia] ,metabolism [Neurodegenerative Diseases] ,Monocytes ,metabolism [Apolipoproteins E] ,pathology [Alzheimer Disease] ,Mice ,Amyloid beta-Protein Precursor ,0302 clinical medicine ,Superoxide Dismutase-1 ,Transforming Growth Factor beta ,metabolism [Amyloid beta-Protein Precursor] ,Immunology and Allergy ,Cluster Analysis ,Amyotrophic lateral sclerosis ,Receptors, Immunologic ,Cerebral Cortex ,Neurons ,Mice, Knockout ,metabolism [Superoxide Dismutase-1] ,Membrane Glycoproteins ,Microglia ,metabolism [Transforming Growth Factor beta] ,Neurodegeneration ,metabolism [Receptors, Immunologic] ,Neurodegenerative Diseases ,deficiency [Apolipoproteins E] ,immunology [Apoptosis] ,immunology [Microglia] ,genetics [Superoxide Dismutase-1] ,Infectious Diseases ,medicine.anatomical_structure ,Phenotype ,metabolism [Neurons] ,Gene Targeting ,Female ,Alzheimer's disease ,metabolism [Alzheimer Disease] ,Signal Transduction ,Encephalomyelitis, Autoimmune, Experimental ,Immunology ,metabolism [Amyloid beta-Peptides] ,Mice, Transgenic ,Biology ,03 medical and health sciences ,immunology [Monocytes] ,Apolipoproteins E ,Trem2 protein, mouse ,Phagocytosis ,Alzheimer Disease ,medicine ,Immune Tolerance ,Animals ,Humans ,genetics [Phagocytosis] ,ddc:610 ,pathology [Plaque, Amyloid] ,genetics [Apoptosis] ,immunology [Neurodegenerative Diseases] ,Amyloid beta-Peptides ,TREM2 ,Multiple sclerosis ,metabolism [Cerebral Cortex] ,Gene Expression Profiling ,Transforming growth factor beta ,medicine.disease ,metabolism [Plaque, Amyloid] ,Disease Models, Animal ,030104 developmental biology ,nervous system ,Gene Expression Regulation ,genetics [Neurodegenerative Diseases] ,biology.protein ,genetics [Apolipoproteins E] ,pathology [Cerebral Cortex] ,Transcriptome ,Neuroscience ,metabolism [Membrane Glycoproteins] ,030217 neurology & neurosurgery ,metabolism [Monocytes] - Abstract
Microglia play a pivotal role in the maintenance of brain homeostasis but lose homeostatic function during neurodegenerative disorders. We identified a specific apolipoprotein E (APOE)-dependent molecular signature in microglia from models of amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and Alzheimer's disease (AD) and in microglia surrounding neuritic β-amyloid (Aβ)-plaques in the brains of people with AD. The APOE pathway mediated a switch from a homeostatic to a neurodegenerative microglia phenotype after phagocytosis of apoptotic neurons. TREM2 (triggering receptor expressed on myeloid cells 2) induced APOE signaling, and targeting the TREM2-APOE pathway restored the homeostatic signature of microglia in ALS and AD mouse models and prevented neuronal loss in an acute model of neurodegeneration. APOE-mediated neurodegenerative microglia had lost their tolerogenic function. Our work identifies the TREM2-APOE pathway as a major regulator of microglial functional phenotype in neurodegenerative diseases and serves as a novel target that could aid in the restoration of homeostatic microglia.
- Published
- 2017
- Full Text
- View/download PDF
49. MER5101, a Novel Aβ1-15:DT Conjugate Vaccine, Generates a Robust Anti-Aβ Antibody Response and Attenuates Aβ Pathology and Cognitive Deficits in APPswe/PS1ΔE9 Transgenic Mice
- Author
-
Peter Blackburn, Jing Sun, Jeffrey L. Frost, Bin Liu, Stephen Grimes, Cynthia A. Lemere, and Hongjun Fu
- Subjects
Pathology ,Diphtheria Toxoid ,T-Lymphocytes ,medicine.medical_treatment ,Conditioning, Classical ,Morris water navigation task ,Epitope ,Immunoglobulin G ,Amyloid beta-Protein Precursor ,Mice ,Cricetinae ,Gliosis ,Cerebral Cortex ,biology ,General Neuroscience ,Fear ,Cytokines ,Antibody ,Adjuvant ,medicine.medical_specialty ,Dose-Response Relationship, Immunologic ,Enzyme-Linked Immunosorbent Assay ,Mice, Transgenic ,CHO Cells ,Transfection ,Statistics, Nonparametric ,Article ,Immune system ,Alzheimer Disease ,Conjugate vaccine ,Glial Fibrillary Acidic Protein ,Presenilin-1 ,medicine ,Animals ,Humans ,Immunoprecipitation ,Maze Learning ,Cell Proliferation ,Amyloid beta-Peptides ,Immunotherapy ,Peptide Fragments ,Disease Models, Animal ,Culture Media, Conditioned ,Antibody Formation ,Mutation ,Immunology ,biology.protein ,Immunization ,Cognition Disorders ,Epitope Mapping ,Spleen - Abstract
Active amyloid-β (Aβ) immunotherapy is under investigation to prevent or treat early Alzheimer's disease (AD). In 2002, a Phase II clinical trial (AN1792) was halted due to meningoencephalitis in ∼6% of the AD patients, possibly caused by a T-cell-mediated immunological response. Thus, generating a vaccine that safely generates high anti-Aβ antibody levels in the elderly is required. In this study, MER5101, a novel conjugate of Aβ1-15 peptide (a B-cell epitope fragment) conjugated to an immunogenic carrier protein, diphtheria toxoid (DT), and formulated in a nanoparticular emulsion-based adjuvant, was administered to 10-month-old APPswe/PS1ΔE9 transgenic (Tg) and wild-type (Wt) mice. High anti-Aβ antibody levels were observed in both vaccinated APPswe/PS1ΔE9 Tg and Wt mice. Antibody isotypes were mainly IgG1 and IgG2b, suggesting a Th2-biased response. Restimulation of splenocytes with the Aβ1-15:DT conjugate resulted in a strong proliferative response, whereas proliferation was absent after restimulation with Aβ1-15 or Aβ1-40/42 peptides, indicating a cellular immune response against DT while avoiding an Aβ-specific T-cell response. Moreover, significant reductions in cerebral Aβ plaque burden, accompanied by attenuated microglial activation and increased synaptic density, were observed in MER5101-vaccinated APPswe/PS1ΔE9 Tg mice compared with Tg adjuvant controls. Last, MER5101-immunized APPswe/PS1ΔE9 Tg mice showed improvement of cognitive deficits in both contextual fear conditioning and the Morris water maze. Our novel, highly immunogenic Aβ conjugate vaccine, MER5101, shows promise for improving Aβ vaccine safety and efficacy and therefore, may be useful for preventing and/or treating early AD.
- Published
- 2013
- Full Text
- View/download PDF
50. De-repression of FOXO3a death axis by microRNA-132 and -212 causes neuronal apoptosis in Alzheimer's disease
- Author
-
Charles R. Vanderburg, Dominic M. Walsh, Christine Esau, Tatiana Veremeyko, Cynthia A. Lemere, Anna M. Krichevsky, Hon-Kit Andus Wong, and Nehal Patel
- Subjects
Down-Regulation ,Apoptosis ,Bioinformatics ,Models, Biological ,Neuroprotection ,Mice ,Alzheimer Disease ,RNA interference ,microRNA ,Genetics ,medicine ,Animals ,Humans ,Gene silencing ,PTEN ,p300-CBP Transcription Factors ,Molecular Biology ,Genetics (clinical) ,Neurons ,Regulation of gene expression ,biology ,Akt/PKB signaling pathway ,Forkhead Box Protein O3 ,Neurodegeneration ,PTEN Phosphohydrolase ,Brain ,Forkhead Transcription Factors ,Hydrogen Peroxide ,General Medicine ,medicine.disease ,Rats ,Cell biology ,MicroRNAs ,Gene Expression Regulation ,biology.protein ,RNA Interference ,Signal Transduction - Abstract
Alzheimer's disease (AD) is a multifactorial and fatal neurodegenerative disorder for which the mechanisms leading to profound neuronal loss are incompletely recognized. MicroRNAs (miRNAs) are recently discovered small regulatory RNA molecules that repress gene expression and are increasingly acknowledged as prime regulators involved in human brain pathologies. Here we identified two homologous miRNAs, miR-132 and miR-212, downregulated in temporal cortical areas and CA1 hippocampal neurons of human AD brains. Sequence-specific inhibition of miR-132 and miR-212 induces apoptosis in cultured primary neurons, whereas their overexpression is neuroprotective against oxidative stress. Using primary neurons and PC12 cells, we demonstrate that miR-132/212 controls cell survival by direct regulation of PTEN, FOXO3a and P300, which are all key elements of AKT signaling pathway. Silencing of these three target genes by RNAi abrogates apoptosis caused by the miR-132/212 inhibition. We further demonstrate that mRNA and protein levels of PTEN, FOXO3a, P300 and most of the direct pro-apoptotic transcriptional targets of FOXO3a are significantly elevated in human AD brains. These results indicate that the miR-132/miR-212/PTEN/FOXO3a signaling pathway contributes to AD neurodegeneration.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.