279 results on '"Serrano-Pozo A"'
Search Results
2. Astrocyte tau deposition in progressive supranuclear palsy is associated with dysregulation of MAPT transcription
- Author
-
Jackson, Rosemary J., Melloni, Alexandra, Fykstra, Dustin P., Serrano-Pozo, Alberto, Shinobu, Leslie, and Hyman, Bradley T.
- Published
- 2024
- Full Text
- View/download PDF
3. Interneuron diversity in the human dorsal striatum
- Author
-
Garma, Leonardo D., Harder, Lisbeth, Barba-Reyes, Juan M., Marco Salas, Sergio, Díez-Salguero, Mónica, Nilsson, Mats, Serrano-Pozo, Alberto, Hyman, Bradley T., and Muñoz-Manchado, Ana B.
- Published
- 2024
- Full Text
- View/download PDF
4. Real-time imaging of mitochondrial redox reveals increased mitochondrial oxidative stress associated with amyloid β aggregates in vivo in a mouse model of Alzheimer’s disease
- Author
-
Calvo-Rodriguez, Maria, Kharitonova, Elizabeth K., Snyder, Austin C., Hou, Steven S., Sanchez-Mico, Maria Virtudes, Das, Sudeshna, Fan, Zhanyun, Shirani, Hamid, Nilsson, K. Peter R., Serrano-Pozo, Alberto, and Bacskai, Brian J.
- Published
- 2024
- Full Text
- View/download PDF
5. Astrocyte tau deposition in progressive supranuclear palsy is associated with dysregulation of MAPT transcription
- Author
-
Rosemary J. Jackson, Alexandra Melloni, Dustin P. Fykstra, Alberto Serrano-Pozo, Leslie Shinobu, and Bradley T. Hyman
- Subjects
Progressive supranuclear palsy ,Astrocytes ,MAPT ,Tufted astrocytes ,Tau ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Progressive supranuclear palsy (PSP) is a neurodegenerative disease characterized by 4R tau deposition in neurons as well as in astrocytes and oligodendrocytes. While astrocytic tau deposits are rarely observed in normal aging (so-called aging-related tau astrogliopathy, ARTAG) and Alzheimer’s disease (AD), astrocytic tau in the form of tufted astrocytes is a pathognomonic hallmark of PSP. Classical biochemical experiments emphasized tau synthesis in neurons in the central nervous system, suggesting that astrocytic tau inclusions might be derived from uptake of extracellular neuronal-derived tau. However, recent single-nucleus RNAseq experiments highlight the fact that MAPT, the gene encoding tau, is also expressed by astrocytes, albeit in lower amounts. We, therefore, revisited the question of whether astrocyte-driven expression of tau might contribute to astrocytic tau aggregates in PSP by performing fluorescent in situ hybridization/immunohistochemical co-localization in human postmortem brain specimens from individuals with PSP and AD with ARTAG as well as normal controls. We find that, in PSP but not in AD, tau-immunoreactive astrocytes have higher levels of MAPT mRNA compared to astrocytes that do not have tau aggregates. These results suggest that astrocytic responses in PSP are unique to this tauopathy and support the possibility that fundamental changes in PSP astrocyte-endogenous mRNA biology contribute to increased synthesis of tau protein and underlies the formation of the astrocytic tau deposits characteristic of PSP.
- Published
- 2024
- Full Text
- View/download PDF
6. Interneuron diversity in the human dorsal striatum
- Author
-
Leonardo D. Garma, Lisbeth Harder, Juan M. Barba-Reyes, Sergio Marco Salas, Mónica Díez-Salguero, Mats Nilsson, Alberto Serrano-Pozo, Bradley T. Hyman, and Ana B. Muñoz-Manchado
- Subjects
Science - Abstract
Abstract Deciphering the striatal interneuron diversity is key to understanding the basal ganglia circuit and to untangling the complex neurological and psychiatric diseases affecting this brain structure. We performed snRNA-seq and spatial transcriptomics of postmortem human caudate nucleus and putamen samples to elucidate the diversity and abundance of interneuron populations and their inherent transcriptional structure in the human dorsal striatum. We propose a comprehensive taxonomy of striatal interneurons with eight main classes and fourteen subclasses, providing their full transcriptomic identity and spatial expression profile as well as additional quantitative FISH validation for specific populations. We have also delineated the correspondence of our taxonomy with previous standardized classifications and shown the main transcriptomic and class abundance differences between caudate nucleus and putamen. Notably, based on key functional genes such as ion channels and synaptic receptors, we found matching known mouse interneuron populations for the most abundant populations, the recently described PTHLH and TAC3 interneurons. Finally, we were able to integrate other published datasets with ours, supporting the generalizability of this harmonized taxonomy.
- Published
- 2024
- Full Text
- View/download PDF
7. Real-time imaging of mitochondrial redox reveals increased mitochondrial oxidative stress associated with amyloid β aggregates in vivo in a mouse model of Alzheimer’s disease
- Author
-
Maria Calvo-Rodriguez, Elizabeth K. Kharitonova, Austin C. Snyder, Steven S. Hou, Maria Virtudes Sanchez-Mico, Sudeshna Das, Zhanyun Fan, Hamid Shirani, K. Peter R. Nilsson, Alberto Serrano-Pozo, and Brian J. Bacskai
- Subjects
Oxidative stress ,ROS ,Alzheimer’s disease ,Mitochondria ,SS31 ,Neurodegeneration ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Reactive oxidative stress is a critical player in the amyloid beta (Aβ) toxicity that contributes to neurodegeneration in Alzheimer’s disease (AD). Damaged mitochondria are one of the main sources of reactive oxygen species and accumulate in Aβ plaque-associated dystrophic neurites in the AD brain. Although Aβ causes neuronal mitochondria reactive oxidative stress in vitro, this has never been directly observed in vivo in the living mouse brain. Here, we tested for the first time whether Aβ plaques and soluble Aβ oligomers induce mitochondrial oxidative stress in surrounding neurons in vivo, and whether this neurotoxic effect can be abrogated using mitochondrial-targeted antioxidants. Methods We expressed a genetically encoded fluorescent ratiometric mitochondria-targeted reporter of oxidative stress in mouse models of the disease and performed intravital multiphoton microscopy of neuronal mitochondria and Aβ plaques. Results For the first time, we demonstrated by direct observation in the living mouse brain exacerbated mitochondrial oxidative stress in neurons after both Aβ plaque deposition and direct application of soluble oligomeric Aβ onto the brain, and determined the most likely pathological sequence of events leading to oxidative stress in vivo. Oxidative stress could be inhibited by both blocking calcium influx into mitochondria and treating with the mitochondria-targeted antioxidant SS31. Remarkably, the latter ameliorated plaque-associated dystrophic neurites without impacting Aβ plaque burden. Conclusions Considering these results, combination of mitochondria-targeted compounds with other anti-amyloid beta or anti-tau therapies hold promise as neuroprotective drugs for the prevention and/or treatment of AD.
- Published
- 2024
- Full Text
- View/download PDF
8. NeuraHealth: An Automated Screening Pipeline to Detect Undiagnosed Cognitive Impairment in Electronic Health Records with Deep Learning and Natural Language Processing
- Author
-
Tyagi, Tanish, Magdamo, Colin G., Noori, Ayush, Li, Zhaozhi, Liu, Xiao, Deodhar, Mayuresh, Hong, Zhuoqiao, Ge, Wendong, Ye, Elissa M., Sheu, Yi-han, Alabsi, Haitham, Brenner, Laura, Robbins, Gregory K., Zafar, Sahar, Benson, Nicole, Moura, Lidia, Hsu, John, Serrano-Pozo, Alberto, Prokopenko, Dimitry, Tanzi, Rudolph E., Hyman, Bradley T., Blacker, Deborah, Mukerji, Shibani S., Westover, M. Brandon, and Das, Sudeshna
- Subjects
Computer Science - Computation and Language - Abstract
Dementia related cognitive impairment (CI) is a neurodegenerative disorder, affecting over 55 million people worldwide and growing rapidly at the rate of one new case every 3 seconds. 75% cases go undiagnosed globally with up to 90% in low-and-middle-income countries, leading to an estimated annual worldwide cost of USD 1.3 trillion, forecasted to reach 2.8 trillion by 2030. With no cure, a recurring failure of clinical trials, and a lack of early diagnosis, the mortality rate is 100%. Information in electronic health records (EHR) can provide vital clues for early detection of CI, but a manual review by experts is tedious and error prone. Several computational methods have been proposed, however, they lack an enhanced understanding of the linguistic context in complex language structures of EHR. Therefore, I propose a novel and more accurate framework, NeuraHealth, to identify patients who had no earlier diagnosis. In NeuraHealth, using patient EHR from Mass General Brigham BioBank, I fine-tuned a bi-directional attention-based deep learning natural language processing model to classify sequences. The sequence predictions were used to generate structured features as input for a patient level regularized logistic regression model. This two-step framework creates high dimensionality, outperforming all existing state-of-the-art computational methods as well as clinical methods. Further, I integrate the models into a real-world product, a web app, to create an automated EHR screening pipeline for scalable and high-speed discovery of undetected CI in EHR, making early diagnosis viable in medical facilities and in regions with scarce health services.
- Published
- 2022
9. Using Deep Learning to Identify Patients with Cognitive Impairment in Electronic Health Records
- Author
-
Tyagi, Tanish, Magdamo, Colin G., Noori, Ayush, Li, Zhaozhi, Liu, Xiao, Deodhar, Mayuresh, Hong, Zhuoqiao, Ge, Wendong, Ye, Elissa M., Sheu, Yi-han, Alabsi, Haitham, Brenner, Laura, Robbins, Gregory K., Zafar, Sahar, Benson, Nicole, Moura, Lidia, Hsu, John, Serrano-Pozo, Alberto, Prokopenko, Dimitry, Tanzi, Rudolph E., Hyman, Bradley T., Blacker, Deborah, Mukerji, Shibani S., Westover, M. Brandon, and Das, Sudeshna
- Subjects
Computer Science - Computation and Language ,Computer Science - Machine Learning - Abstract
Dementia is a neurodegenerative disorder that causes cognitive decline and affects more than 50 million people worldwide. Dementia is under-diagnosed by healthcare professionals - only one in four people who suffer from dementia are diagnosed. Even when a diagnosis is made, it may not be entered as a structured International Classification of Diseases (ICD) diagnosis code in a patient's charts. Information relevant to cognitive impairment (CI) is often found within electronic health records (EHR), but manual review of clinician notes by experts is both time consuming and often prone to errors. Automated mining of these notes presents an opportunity to label patients with cognitive impairment in EHR data. We developed natural language processing (NLP) tools to identify patients with cognitive impairment and demonstrate that linguistic context enhances performance for the cognitive impairment classification task. We fine-tuned our attention based deep learning model, which can learn from complex language structures, and substantially improved accuracy (0.93) relative to a baseline NLP model (0.84). Further, we show that deep learning NLP can successfully identify dementia patients without dementia-related ICD codes or medications., Comment: Machine Learning for Health (ML4H) - Extended Abstract
- Published
- 2021
10. Reactive astrocyte nomenclature, definitions, and future directions
- Author
-
Escartin, Carole, Galea, Elena, Lakatos, András, O’Callaghan, James P, Petzold, Gabor C, Serrano-Pozo, Alberto, Steinhäuser, Christian, Volterra, Andrea, Carmignoto, Giorgio, Agarwal, Amit, Allen, Nicola J, Araque, Alfonso, Barbeito, Luis, Barzilai, Ari, Bergles, Dwight E, Bonvento, Gilles, Butt, Arthur M, Chen, Wei-Ting, Cohen-Salmon, Martine, Cunningham, Colm, Deneen, Benjamin, De Strooper, Bart, Díaz-Castro, Blanca, Farina, Cinthia, Freeman, Marc, Gallo, Vittorio, Goldman, James E, Goldman, Steven A, Götz, Magdalena, Gutiérrez, Antonia, Haydon, Philip G, Heiland, Dieter H, Hol, Elly M, Holt, Matthew G, Iino, Masamitsu, Kastanenka, Ksenia V, Kettenmann, Helmut, Khakh, Baljit S, Koizumi, Schuichi, Lee, C Justin, Liddelow, Shane A, MacVicar, Brian A, Magistretti, Pierre, Messing, Albee, Mishra, Anusha, Molofsky, Anna V, Murai, Keith K, Norris, Christopher M, Okada, Seiji, Oliet, Stéphane HR, Oliveira, João F, Panatier, Aude, Parpura, Vladimir, Pekna, Marcela, Pekny, Milos, Pellerin, Luc, Perea, Gertrudis, Pérez-Nievas, Beatriz G, Pfrieger, Frank W, Poskanzer, Kira E, Quintana, Francisco J, Ransohoff, Richard M, Riquelme-Perez, Miriam, Robel, Stefanie, Rose, Christine R, Rothstein, Jeffrey D, Rouach, Nathalie, Rowitch, David H, Semyanov, Alexey, Sirko, Swetlana, Sontheimer, Harald, Swanson, Raymond A, Vitorica, Javier, Wanner, Ina-Beate, Wood, Levi B, Wu, Jiaqian, Zheng, Binhai, Zimmer, Eduardo R, Zorec, Robert, Sofroniew, Michael V, and Verkhratsky, Alexei
- Subjects
Neurosciences ,Brain Disorders ,Neurological ,Aging ,Animals ,Astrocytes ,Brain ,Brain Diseases ,Brain Injuries ,Humans ,Spinal Cord ,Spinal Cord Injuries ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery - Abstract
Reactive astrocytes are astrocytes undergoing morphological, molecular, and functional remodeling in response to injury, disease, or infection of the CNS. Although this remodeling was first described over a century ago, uncertainties and controversies remain regarding the contribution of reactive astrocytes to CNS diseases, repair, and aging. It is also unclear whether fixed categories of reactive astrocytes exist and, if so, how to identify them. We point out the shortcomings of binary divisions of reactive astrocytes into good-vs-bad, neurotoxic-vs-neuroprotective or A1-vs-A2. We advocate, instead, that research on reactive astrocytes include assessment of multiple molecular and functional parameters-preferably in vivo-plus multivariate statistics and determination of impact on pathological hallmarks in relevant models. These guidelines may spur the discovery of astrocyte-based biomarkers as well as astrocyte-targeting therapies that abrogate detrimental actions of reactive astrocytes, potentiate their neuro- and glioprotective actions, and restore or augment their homeostatic, modulatory, and defensive functions.
- Published
- 2021
11. DECIPHERING THE INTERNEURON DIVERSITY IN THE HUMAN CAUDATE NUCLEUS AND PUTAMEN USING SNRNA-SEQ.
- Author
-
Ana Muñoz Manchado, Leonardo Garma, Lisbeth Harder, Juan Manuel Barba Reyes, Mónica Díez Salguero, Alberto Serrano Pozo, and Bradley Hyman
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2023
- Full Text
- View/download PDF
12. OLIGODENDROCYTES AS A CRITICAL CELL-TYPE IN PARKINSON'S DISEASE
- Author
-
Juan Manuel Barba Reyes, Lisbeth Harder, Mónica Díez Salguero, Leonardo Garma, Alberto Serrano Pozo, Bradley Hyman, and Ana Muñoz Manchado
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2023
- Full Text
- View/download PDF
13. Cyclic multiplex fluorescent immunohistochemistry and machine learning reveal distinct states of astrocytes and microglia in normal aging and Alzheimer’s disease
- Author
-
Muñoz-Castro, Clara, Noori, Ayush, Magdamo, Colin G., Li, Zhaozhi, Marks, Jordan D., Frosch, Matthew P., Das, Sudeshna, Hyman, Bradley T., and Serrano-Pozo, Alberto
- Published
- 2022
- Full Text
- View/download PDF
14. Cyclic multiplex fluorescent immunohistochemistry and machine learning reveal distinct states of astrocytes and microglia in normal aging and Alzheimer’s disease
- Author
-
Clara Muñoz-Castro, Ayush Noori, Colin G. Magdamo, Zhaozhi Li, Jordan D. Marks, Matthew P. Frosch, Sudeshna Das, Bradley T. Hyman, and Alberto Serrano-Pozo
- Subjects
Alzheimer’s disease ,Amyloid plaques ,Astrocytes ,Immunohistochemistry ,Microglia ,Neurofibrillary tangles ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Astrocytes and microglia react to Aβ plaques, neurofibrillary tangles, and neurodegeneration in the Alzheimer’s disease (AD) brain. Single-nuclei and single-cell RNA-seq have revealed multiple states or subpopulations of these glial cells but lack spatial information. We have developed a methodology of cyclic multiplex fluorescent immunohistochemistry on human postmortem brains and image analysis that enables a comprehensive morphological quantitative characterization of astrocytes and microglia in the context of their spatial relationships with plaques and tangles. Methods Single FFPE sections from the temporal association cortex of control and AD subjects were subjected to 8 cycles of multiplex fluorescent immunohistochemistry, including 7 astroglial, 6 microglial, 1 neuronal, Aβ, and phospho-tau markers. Our analysis pipeline consisted of: (1) image alignment across cycles; (2) background subtraction; (3) manual annotation of 5172 ALDH1L1+ astrocytic and 6226 IBA1+ microglial profiles; (4) local thresholding and segmentation of profiles; (5) machine learning on marker intensity data; and (6) deep learning on image features. Results Spectral clustering identified three phenotypes of astrocytes and microglia, which we termed “homeostatic,” “intermediate,” and “reactive.” Reactive and, to a lesser extent, intermediate astrocytes and microglia were closely associated with AD pathology (≤ 50 µm). Compared to homeostatic, reactive astrocytes contained substantially higher GFAP and YKL-40, modestly elevated vimentin and TSPO as well as EAAT1, and reduced GS. Intermediate astrocytes had markedly increased EAAT2, moderately increased GS, and intermediate GFAP and YKL-40 levels. Relative to homeostatic, reactive microglia showed increased expression of all markers (CD68, ferritin, MHC2, TMEM119, TSPO), whereas intermediate microglia exhibited increased ferritin and TMEM119 as well as intermediate CD68 levels. Machine learning models applied on either high-plex signal intensity data (gradient boosting machines) or directly on image features (convolutional neural networks) accurately discriminated control vs. AD diagnoses at the single-cell level. Conclusions Cyclic multiplex fluorescent immunohistochemistry combined with machine learning models holds promise to advance our understanding of the complexity and heterogeneity of glial responses as well as inform transcriptomics studies. Three distinct phenotypes emerged with our combination of markers, thus expanding the classic binary “homeostatic vs. reactive” classification to a third state, which could represent “transitional” or “resilient” glia.
- Published
- 2022
- Full Text
- View/download PDF
15. Real-time imaging of mitochondrial redox reveals increased mitochondrial oxidative stress associated with amyloid ß aggregates in vivo in a mouse model of Alzheimer's disease
- Author
-
Calvo-Rodriguez, Maria, Kharitonova, Elizabeth K., Snyder, Austin C., Hou, Steven S., Sanchez-Mico, Maria Virtudes, Das, Sudeshna, Fan, Zhanyun, Shirani, Hamid, Nilsson, Peter, Serrano-Pozo, Alberto, Bacskai, Brian J., Calvo-Rodriguez, Maria, Kharitonova, Elizabeth K., Snyder, Austin C., Hou, Steven S., Sanchez-Mico, Maria Virtudes, Das, Sudeshna, Fan, Zhanyun, Shirani, Hamid, Nilsson, Peter, Serrano-Pozo, Alberto, and Bacskai, Brian J.
- Abstract
BackgroundReactive oxidative stress is a critical player in the amyloid beta (A beta) toxicity that contributes to neurodegeneration in Alzheimer's disease (AD). Damaged mitochondria are one of the main sources of reactive oxygen species and accumulate in A beta plaque-associated dystrophic neurites in the AD brain. Although A beta causes neuronal mitochondria reactive oxidative stress in vitro, this has never been directly observed in vivo in the living mouse brain. Here, we tested for the first time whether A beta plaques and soluble A beta oligomers induce mitochondrial oxidative stress in surrounding neurons in vivo, and whether this neurotoxic effect can be abrogated using mitochondrial-targeted antioxidants.MethodsWe expressed a genetically encoded fluorescent ratiometric mitochondria-targeted reporter of oxidative stress in mouse models of the disease and performed intravital multiphoton microscopy of neuronal mitochondria and A beta plaques.ResultsFor the first time, we demonstrated by direct observation in the living mouse brain exacerbated mitochondrial oxidative stress in neurons after both A beta plaque deposition and direct application of soluble oligomeric A beta onto the brain, and determined the most likely pathological sequence of events leading to oxidative stress in vivo. Oxidative stress could be inhibited by both blocking calcium influx into mitochondria and treating with the mitochondria-targeted antioxidant SS31. Remarkably, the latter ameliorated plaque-associated dystrophic neurites without impacting A beta plaque burden.ConclusionsConsidering these results, combination of mitochondria-targeted compounds with other anti-amyloid beta or anti-tau therapies hold promise as neuroprotective drugs for the prevention and/or treatment of AD., Funding Agencies|National Institute on Aging; Schepens Eye Research Institute Gene Transfer Vector Core
- Published
- 2024
- Full Text
- View/download PDF
16. Update on modifiable risk factors for Alzheimer’s disease and related dementias
- Author
-
Universidad de Sevilla. Departamento de Bioquímica y Biología Molecular, National Institutes on Aging. United States, Jaisa Aad, Methasit, Muñoz Castro, Clara, Serrano Pozo, Alberto, Universidad de Sevilla. Departamento de Bioquímica y Biología Molecular, National Institutes on Aging. United States, Jaisa Aad, Methasit, Muñoz Castro, Clara, and Serrano Pozo, Alberto
- Abstract
Purpose of review. All human beings undergo a lifelong cumulative exposure to potentially preventable adverse factors such as toxins, infections, traumatisms, and cardiovascular risk factors, collectively termed exposome. The interplay between the individual’s genetics and exposome is thought to have a large impact in health outcomes such as cancer and cardiovascular disease. Likewise, a growing body of evidence is supporting the idea that preventable factors explain a sizable proportion of Alzheimer’s disease and related dementia (ADRD) cases. Recent findings. Here, we will review the most recent epidemiological, experimental preclinical, and interventional clinical studies examining some of these potentially modifiable risk factors for ADRD. We will focus on new evidence regarding cardiovascular risk factors, air pollution, viral and other infectious agents, traumatic brain injury, and hearing loss. Summary. While greater and higher quality epidemiological and experimental evidence is needed to unequivocally confirm their causal link with ADRD and/or unravel the underlying mechanisms, these modifiable risk factors may represent a window of opportunity to reduce ADRD incidence and prevalence at the population level via health screenings, and education and health policies.
- Published
- 2024
17. Development and Evaluation of a Natural Language Processing Annotation Tool to Facilitate Phenotyping of Cognitive Status in Electronic Health Records: Diagnostic Study
- Author
-
Ayush Noori, Colin Magdamo, Xiao Liu, Tanish Tyagi, Zhaozhi Li, Akhil Kondepudi, Haitham Alabsi, Emily Rudmann, Douglas Wilcox, Laura Brenner, Gregory K Robbins, Lidia Moura, Sahar Zafar, Nicole M Benson, John Hsu, John R Dickson, Alberto Serrano-Pozo, Bradley T Hyman, Deborah Blacker, M Brandon Westover, Shibani S Mukerji, and Sudeshna Das
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 ,Public aspects of medicine ,RA1-1270 - Abstract
BackgroundElectronic health records (EHRs) with large sample sizes and rich information offer great potential for dementia research, but current methods of phenotyping cognitive status are not scalable. ObjectiveThe aim of this study was to evaluate whether natural language processing (NLP)–powered semiautomated annotation can improve the speed and interrater reliability of chart reviews for phenotyping cognitive status. MethodsIn this diagnostic study, we developed and evaluated a semiautomated NLP-powered annotation tool (NAT) to facilitate phenotyping of cognitive status. Clinical experts adjudicated the cognitive status of 627 patients at Mass General Brigham (MGB) health care, using NAT or traditional chart reviews. Patient charts contained EHR data from two data sets: (1) records from January 1, 2017, to December 31, 2018, for 100 Medicare beneficiaries from the MGB Accountable Care Organization and (2) records from 2 years prior to COVID-19 diagnosis to the date of COVID-19 diagnosis for 527 MGB patients. All EHR data from the relevant period were extracted; diagnosis codes, medications, and laboratory test values were processed and summarized; clinical notes were processed through an NLP pipeline; and a web tool was developed to present an integrated view of all data. Cognitive status was rated as cognitively normal, cognitively impaired, or undetermined. Assessment time and interrater agreement of NAT compared to manual chart reviews for cognitive status phenotyping was evaluated. ResultsNAT adjudication provided higher interrater agreement (Cohen κ=0.89 vs κ=0.80) and significant speed up (time difference mean 1.4, SD 1.3 minutes; P
- Published
- 2022
- Full Text
- View/download PDF
18. Meta-analysis of mouse transcriptomic studies supports a context-dependent astrocyte reaction in acute CNS injury versus neurodegeneration
- Author
-
Sudeshna Das, Zhaozhi Li, Ayush Noori, Bradley T. Hyman, and Alberto Serrano-Pozo
- Subjects
Acute CNS injury ,Astrocyte reaction ,Meta-analysis ,Neurodegenerative diseases ,Transcriptomics ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Neuronal damage in acute CNS injuries and chronic neurodegenerative diseases is invariably accompanied by an astrocyte reaction in both mice and humans. However, whether and how the nature of the CNS insult—acute versus chronic—influences the astrocyte response, and whether astrocyte transcriptomic changes in these mouse models faithfully recapitulate the astrocyte reaction in human diseases remains to be elucidated. We hypothesized that astrocytes set off different transcriptomic programs in response to acute versus chronic insults, besides a shared “pan-injury” signature common to both types of conditions, and investigated the presence of these mouse astrocyte signatures in transcriptomic studies from human neurodegenerative diseases. Methods We performed a meta-analysis of 15 published astrocyte transcriptomic datasets from mouse models of acute injury (n = 6) and chronic neurodegeneration (n = 9) and identified pan-injury, acute, and chronic signatures, with both upregulated (UP) and downregulated (DOWN) genes. Next, we investigated these signatures in 7 transcriptomic datasets from various human neurodegenerative diseases. Results In mouse models, the number of UP/DOWN genes per signature was 64/21 for pan-injury and 109/79 for acute injury, whereas only 13/27 for chronic neurodegeneration. The pan-injury-UP signature was represented by the classic cytoskeletal hallmarks of astrocyte reaction (Gfap and Vim), plus extracellular matrix (i.e., Cd44, Lgals1, Lgals3, Timp1), and immune response (i.e., C3, Serping1, Fas, Stat1, Stat2, Stat3). The acute injury-UP signature was enriched in protein synthesis and degradation (both ubiquitin-proteasome and autophagy systems), intracellular trafficking, and anti-oxidant defense genes, whereas the acute injury-DOWN signature included genes that regulate chromatin structure and transcriptional activity, many of which are transcriptional repressors. The chronic neurodegeneration-UP signature was further enriched in astrocyte-secreted extracellular matrix proteins (Lama4, Cyr61, Thbs4), while the DOWN signature included relevant genes such as Agl (glycogenolysis), S1pr1 (immune modulation), and Sod2 (anti-oxidant). Only the pan-injury-UP mouse signature was clearly present in some human neurodegenerative transcriptomic datasets. Conclusions Acute and chronic CNS injuries lead to distinct astrocyte gene expression programs beyond their common astrocyte reaction signature. However, caution should be taken when extrapolating astrocyte transcriptomic findings from mouse models to human diseases.
- Published
- 2020
- Full Text
- View/download PDF
19. Increased mitochondrial calcium levels associated with neuronal death in a mouse model of Alzheimer’s disease
- Author
-
Maria Calvo-Rodriguez, Steven S. Hou, Austin C. Snyder, Elizabeth K. Kharitonova, Alyssa N. Russ, Sudeshna Das, Zhanyun Fan, Alona Muzikansky, Monica Garcia-Alloza, Alberto Serrano-Pozo, Eloise Hudry, and Brian J. Bacskai
- Subjects
Science - Abstract
Calvo-Rodriguez et al. show elevated calcium levels in neuronal mitochondria in a mouse model of cerebral β-amyloidosis after plaque deposition, which precede rare neuron death events in this model. The mechanism involves toxic extracellular Aβ oligomers and the mitochondrial calcium uniporter.
- Published
- 2020
- Full Text
- View/download PDF
20. Tau molecular diversity contributes to clinical heterogeneity in Alzheimer’s disease
- Author
-
Dujardin, Simon, Commins, Caitlin, Lathuiliere, Aurelien, Beerepoot, Pieter, Fernandes, Analiese R., Kamath, Tarun V., De Los Santos, Mark B., Klickstein, Naomi, Corjuc, Diana L., Corjuc, Bianca T., Dooley, Patrick M., Viode, Arthur, Oakley, Derek H., Moore, Benjamin D., Mullin, Kristina, Jean-Gilles, Dinorah, Clark, Ryan, Atchison, Kevin, Moore, Renee, Chibnik, Lori B., Tanzi, Rudolph E., Frosch, Matthew P., Serrano-Pozo, Alberto, Elwood, Fiona, Steen, Judith A., Kennedy, Matthew E., and Hyman, Bradley T.
- Published
- 2020
- Full Text
- View/download PDF
21. Primary age-related tauopathy (PART): a common pathology associated with human aging
- Author
-
Crary, John F, Trojanowski, John Q, Schneider, Julie A, Abisambra, Jose F, Abner, Erin L, Alafuzoff, Irina, Arnold, Steven E, Attems, Johannes, Beach, Thomas G, Bigio, Eileen H, Cairns, Nigel J, Dickson, Dennis W, Gearing, Marla, Grinberg, Lea T, Hof, Patrick R, Hyman, Bradley T, Jellinger, Kurt, Jicha, Gregory A, Kovacs, Gabor G, Knopman, David S, Kofler, Julia, Kukull, Walter A, Mackenzie, Ian R, Masliah, Eliezer, McKee, Ann, Montine, Thomas J, Murray, Melissa E, Neltner, Janna H, Santa-Maria, Ismael, Seeley, William W, Serrano-Pozo, Alberto, Shelanski, Michael L, Stein, Thor, Takao, Masaki, Thal, Dietmar R, Toledo, Jonathan B, Troncoso, Juan C, Vonsattel, Jean Paul, White, Charles L, Wisniewski, Thomas, Woltjer, Randall L, Yamada, Masahito, and Nelson, Peter T
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Neurodegenerative ,Brain Disorders ,Aging ,Alzheimer's Disease ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Dementia ,Alzheimer's Disease Related Dementias (ADRD) ,Acquired Cognitive Impairment ,Aetiology ,Detection ,screening and diagnosis ,4.2 Evaluation of markers and technologies ,2.1 Biological and endogenous factors ,4.1 Discovery and preclinical testing of markers and technologies ,Neurological ,Brain ,Diagnosis ,Differential ,Humans ,Plaque ,Amyloid ,Tauopathies ,Terminology as Topic ,TPSD ,TOD ,Braak ,Neuropathology ,Consensus ,Clinical Sciences ,Neurology & Neurosurgery - Abstract
We recommend a new term, "primary age-related tauopathy" (PART), to describe a pathology that is commonly observed in the brains of aged individuals. Many autopsy studies have reported brains with neurofibrillary tangles (NFTs) that are indistinguishable from those of Alzheimer's disease (AD), in the absence of amyloid (Aβ) plaques. For these "NFT+/Aβ-" brains, for which formal criteria for AD neuropathologic changes are not met, the NFTs are mostly restricted to structures in the medial temporal lobe, basal forebrain, brainstem, and olfactory areas (bulb and cortex). Symptoms in persons with PART usually range from normal to amnestic cognitive changes, with only a minority exhibiting profound impairment. Because cognitive impairment is often mild, existing clinicopathologic designations, such as "tangle-only dementia" and "tangle-predominant senile dementia", are imprecise and not appropriate for most subjects. PART is almost universally detectable at autopsy among elderly individuals, yet this pathological process cannot be specifically identified pre-mortem at the present time. Improved biomarkers and tau imaging may enable diagnosis of PART in clinical settings in the future. Indeed, recent studies have identified a common biomarker profile consisting of temporal lobe atrophy and tauopathy without evidence of Aβ accumulation. For both researchers and clinicians, a revised nomenclature will raise awareness of this extremely common pathologic change while providing a conceptual foundation for future studies. Prior reports that have elucidated features of the pathologic entity we refer to as PART are discussed, and working neuropathological diagnostic criteria are proposed.
- Published
- 2014
22. Differential gene expression data from the human central nervous system across Alzheimer's disease, Lewy body diseases, and the amyotrophic lateral sclerosis and frontotemporal dementia spectrum
- Author
-
Ayush Noori, Aziz M. Mezlini, Bradley T. Hyman, Alberto Serrano-Pozo, and Sudeshna Das
- Subjects
Alzheimer's disease ,Amyotrophic lateral sclerosis ,Differential expression ,Frontotemporal dementia ,Lewy body diseases ,Meta-analysis ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Science (General) ,Q1-390 - Abstract
In Noori et al. [1], we hypothesized that there is a shared gene expression signature underlying neurodegenerative proteinopathies including Alzheimer's disease (AD), Lewy body diseases (LBD), and the amyotrophic lateral sclerosis and frontotemporal dementia (ALS-FTD) spectrum. To test this hypothesis, we performed a systematic review and meta-analysis of 60 human central nervous system transcriptomic datasets in the public Gene Expression Omnibus and ArrayExpress repositories, comprising a total of 2,600 AD, LBD, and ALS-FTD patients and age-matched controls which passed our stringent quality control pipeline. Here, we provide the results of differential expression analyses with data quality reports for each of these 60 datasets. This atlas of differential expression across AD, LBD, and ALS-FTD may guide future work to elucidate the pathophysiological drivers of these individual diseases as well as the common substrate of neurodegeneration.
- Published
- 2021
- Full Text
- View/download PDF
23. Systematic review and meta-analysis of human transcriptomics reveals neuroinflammation, deficient energy metabolism, and proteostasis failure across neurodegeneration
- Author
-
Ayush Noori, Aziz M. Mezlini, Bradley T. Hyman, Alberto Serrano-Pozo, and Sudeshna Das
- Subjects
Alzheimer's disease ,Amyotrophic lateral sclerosis ,Frontotemporal dementia ,Lewy body diseases ,Meta-analysis ,Mitochondrial energy metabolism ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD), Lewy body diseases (LBD), and the amyotrophic lateral sclerosis and frontotemporal dementia (ALS-FTD) spectrum are defined by the accumulation of specific misfolded protein aggregates. However, the mechanisms by which each proteinopathy leads to neurodegeneration remain elusive. We hypothesized that there is a common “pan-neurodegenerative” gene expression signature driving pathophysiology across these clinically and pathologically diverse proteinopathies. To test this hypothesis, we performed a systematic review of human CNS transcriptomics datasets from AD, LBD, and ALS-FTD patients and age-matched controls in the Gene Expression Omnibus (GEO) and ArrayExpress databases, followed by consistent processing of each dataset, meta-analysis, pathway enrichment, and overlap analyses. After applying pre-specified eligibility criteria and stringent data pre-processing, a total of 2600 samples from 26 AD, 21 LBD, and 13 ALS-FTD datasets were included in the meta-analysis. The pan-neurodegenerative gene signature is characterized by an upregulation of innate immunity, cytoskeleton, and transcription and RNA processing genes, and a downregulation of the mitochondrial electron transport chain. Pathway enrichment analyses also revealed the upregulation of neuroinflammation (including Toll-like receptor, TNF, and NFκB signaling) and phagocytosis, and the downregulation of mitochondrial oxidative phosphorylation, lysosomal acidification, and ubiquitin-proteasome pathways. Our findings suggest that neuroinflammation and a failure in both neuronal energy metabolism and protein degradation systems are consistent features underlying neurodegenerative diseases, despite differences in the extent of neuronal loss and brain regions involved.
- Published
- 2021
- Full Text
- View/download PDF
24. Meta-analysis of mouse transcriptomic studies supports a context-dependent astrocyte reaction in acute CNS injury versus neurodegeneration
- Author
-
Das, Sudeshna, Li, Zhaozhi, Noori, Ayush, Hyman, Bradley T., and Serrano-Pozo, Alberto
- Published
- 2020
- Full Text
- View/download PDF
25. Increased mitochondrial calcium levels associated with neuronal death in a mouse model of Alzheimer’s disease
- Author
-
Calvo-Rodriguez, Maria, Hou, Steven S., Snyder, Austin C., Kharitonova, Elizabeth K., Russ, Alyssa N., Das, Sudeshna, Fan, Zhanyun, Muzikansky, Alona, Garcia-Alloza, Monica, Serrano-Pozo, Alberto, Hudry, Eloise, and Bacskai, Brian J.
- Published
- 2020
- Full Text
- View/download PDF
26. Distinct transcriptomic responses to Aβ plaques, neurofibrillary tangles, and APOE in Alzheimer's disease.
- Author
-
Das, Sudeshna, Li, Zhaozhi, Wachter, Astrid, Alla, Srinija, Noori, Ayush, Abdourahman, Aicha, Tamm, Joseph A., Woodbury, Maya E., Talanian, Robert V., Biber, Knut, Karran, Eric H., Hyman, Bradley T., and Serrano‐Pozo, Alberto
- Abstract
INTRODUCTION: Omics studies have revealed that various brain cell types undergo profound molecular changes in Alzheimer's disease (AD) but the spatial relationships with plaques and tangles and APOE‐linked differences remain unclear. METHODS: We performed laser capture microdissection of amyloid beta (Aβ) plaques, the 50 μm halo around them, tangles with the 50 μm halo around them, and areas distant (> 50 μm) from plaques and tangles in the temporal cortex of AD and control donors, followed by RNA‐sequencing. RESULTS: Aβ plaques exhibited upregulated microglial (neuroinflammation/phagocytosis) and downregulated neuronal (neurotransmission/energy metabolism) genes, whereas tangles had mostly downregulated neuronal genes. Aβ plaques had more differentially expressed genes than tangles. We identified a gradient Aβ plaque > peri‐plaque > tangle > distant for these changes. AD APOE ε4 homozygotes had greater changes than APOE ε3 across locations, especially within Aβ plaques. DISCUSSION: Transcriptomic changes in AD consist primarily of neuroinflammation and neuronal dysfunction, are spatially associated mainly with Aβ plaques, and are exacerbated by the APOE ε4 allele. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Endothelial Cells are Heterogeneous in Different Brain Regions and are Dramatically Altered in Alzheimer’s Disease
- Author
-
Bryant, Annie, primary, Li, Zhaozhi, additional, Jayakumar, Rojashree, additional, Serrano-Pozo, Alberto, additional, Woost, Benjamin, additional, Hu, Miwei, additional, Woodbury, Maya E., additional, Wachter, Astrid, additional, Lin, Gen, additional, Kwon, Taekyung, additional, Talanian, Robert V., additional, Biber, Knut, additional, Karran, Eric H., additional, Hyman, Bradley T., additional, Das, Sudeshna, additional, and Bennett, Rachel, additional
- Published
- 2023
- Full Text
- View/download PDF
28. Editorial: The Role of Glia in Alzheimer's Disease
- Author
-
Beatriz G. Pérez-Nievas and Alberto Serrano-Pozo
- Subjects
Alzheimer's disease ,amyloid plaques ,astrocytes ,chemokines/chemokine receptors ,glia ,microglia ,Neurology. Diseases of the nervous system ,RC346-429 - Published
- 2019
- Full Text
- View/download PDF
29. Plaque-Associated Local Toxicity Increases over the Clinical Course of Alzheimer Disease
- Author
-
Serrano-Pozo, Alberto, Betensky, Rebecca A., Frosch, Matthew P., and Hyman, Bradley T.
- Published
- 2016
- Full Text
- View/download PDF
30. Interneuron diversity in the human dorsal striatum
- Author
-
Leonardo Garma, Lisbeth Harder, Juan Barba-Reyes, Monica Diez-Salguero, Alberto Serrano-Pozo, Bradley Hyman, and Ana Munoz-Manchado
- Abstract
Deciphering the striatal interneuron diversity is key to understanding the basal ganglia circuit and to untangle the complex neurological and psychiatric diseases affecting this brain structure. We performed snRNA-seq of postmortem human caudate nucleus and putamen samples to elucidate the diversity and abundance of interneuron populations and their transcriptional structure in the human dorsal striatum. We propose a new taxonomy of striatal interneurons with eight main classes and fourteen subclasses and provide their specific markers and some quantitative FISH validation, particularly for a novel PTHLH-expressing population. For the most abundant populations, PTHLH and TAC3, we found matching known mouse interneuron populations based on key functional genes such as ion channels and synaptic receptors. Remarkably, human TAC3 and mouse Th populations share important similarities including the expression of the neuropeptide tachykinin 3. Finally, we were able to integrate other published datasets supporting the generalizability of this new harmonized taxonomy.
- Published
- 2023
31. Distinct Transcriptomic Responses to Aβ plaques, Neurofibrillary Tangles, and APOE in Alzheimer’s Disease
- Author
-
Sudeshna Das, Zhaozhi Li, Astrid Wachter, Srinija Alla, Ayush Noori, Aicha Abdourahman, Joseph A. Tamm, Maya E. Woodbury, Robert V. Talanian, Knut Biber, Eric H. Karran, Bradley T. Hyman, and Alberto Serrano-Pozo
- Subjects
Article - Abstract
INTRODUCTIONOmics studies have revealed that various brain cell types undergo profound molecular changes in Alzheimer’s disease (AD) but the spatial relationships with plaques and tangles andAPOE-linked differences remain unclear.METHODSWe performed laser capture microdissection of Aβ plaques, the 50μm halo around them, tangles with the 50μm halo around them, and areas distant (>50μm) from plaques and tangles in the temporal cortex of AD and control donors, followed by RNA-sequencing.RESULTSAβ plaques exhibited upregulated microglial (neuroinflammation/phagocytosis) and downregulated neuronal (neurotransmission/energy metabolism) genes, whereas tangles had mostly downregulated neuronal genes. Aβ plaques had more differentially expressed genes than tangles. We identified a gradient Aβ plaque>peri-plaque>tangle>distant for these changes. ADAPOEε4 homozygotes had greater changes thanAPOEε3 across locations, especially within Aβ plaques.DISCUSSIONTranscriptomic changes in AD consist primarily of neuroinflammation and neuronal dysfunction, are spatially associated mainly with Aβ plaques, and are exacerbated by theAPOEε4 allele.
- Published
- 2023
32. Neuropathology-Independent Association BetweenAPOEGenotype and Cognitive Decline Rate in the Normal Aging-Early Alzheimer Continuum
- Author
-
Qian, Jing, primary, Zhang, Yiding, additional, Betensky, Rebecca A., additional, Hyman, Bradley T., additional, and Serrano-Pozo, Alberto, additional
- Published
- 2023
- Full Text
- View/download PDF
33. Author Correction: Tau molecular diversity contributes to clinical heterogeneity in Alzheimer’s disease
- Author
-
Dujardin, Simon, Commins, Caitlin, Lathuiliere, Aurelien, Beerepoot, Pieter, Fernandes, Analiese R., Kamath, Tarun V., De Los Santos, Mark B., Klickstein, Naomi, Corjuc, Diana L., Corjuc, Bianca T., Dooley, Patrick M., Viode, Arthur, Oakley, Derek H., Moore, Benjamin D., Mullin, Kristina, Jean-Gilles, Dinorah, Clark, Ryan, Atchison, Kevin, Moore, Renee, Chibnik, Lori B., Tanzi, Rudolph E., Frosch, Matthew P., Serrano-Pozo, Alberto, Elwood, Fiona, Steen, Judith A., Kennedy, Matthew E., and Hyman, Bradley T.
- Published
- 2021
- Full Text
- View/download PDF
34. Endothelial Cells are Heterogeneous in Different Brain Regions and are Dramatically Altered in Alzheimer’s Disease
- Author
-
Bryant, Annie, Li, Zhaozhi, Jayakumar, Rojashree, Serrano-Pozo, Alberto, Woost, Benjamin, Hu, Miwei, Woodbury, Maya E., Wachter, Astrid, Lin, Gen, Kwon, Taekyung, Talanian, Robert V., Biber, Knut, Karran, Eric H., Hyman, Bradley T., Das, Sudeshna, and Bennett, Rachel
- Subjects
General Neuroscience ,Article - Abstract
Vascular endothelial cells play an important role in maintaining brain health, but their contribution to Alzheimer’s disease (AD) is obscured by limited understanding of the cellular heterogeneity in normal aged brain and in disease. To address this, we performed single nucleus RNAseq on tissue from 32 AD and non-AD donors each with five cortical regions: entorhinal cortex, inferior temporal gyrus, prefrontal cortex, visual association cortex and primary visual cortex. Analysis of 51,586 endothelial cells revealed unique gene expression patterns across the five regions in non-AD donors. Alzheimer’s brain endothelial cells were characterized by upregulated protein folding genes and distinct transcriptomic differences in response to amyloid beta plaques and cerebral amyloid angiopathy (CAA). This dataset demonstrates previously unrecognized regional heterogeneity in the endothelial cell transcriptome in both aged non-AD and AD brain.Significance StatementIn this work, we show that vascular endothelial cells collected from five different brain regions display surprising variability in gene expression. In the presence of Alzheimer’s disease pathology, endothelial cell gene expression is dramatically altered with clear differences in regional and temporal changes. These findings help explain why certain brain regions appear to differ in susceptibility to disease-related vascular remodeling events that may impact blood flow.
- Published
- 2023
35. Astrocyte transcriptomic changes along the spatiotemporal progression of Alzheimer’s disease
- Author
-
ALBERTO SERRANO-POZO, Ayush Noori, and Clara Muñoz-Castro
- Abstract
Astrocytes play a critical role in brain homeostasis and normal functions but their changes along the spatiotemporal progression of Alzheimer’s disease (AD) neuropathology remain largely unknown. Here we performed single-nucleus RNA-sequencing on brain regions along the stereotypical progression of AD pathology from donors ranging the entire normal aging-AD continuum comprising 628,943 astrocyte nuclei from 32 donors across 5 brain regions. We discovered temporal gene-expression-trajectories with gene sets differentially activated at various disease stages. Surprisingly, a gene set enriched in proteostasis and energy metabolism, was upregulated in late-stage but unexpectedly returned to baseline levels in end-stage, suggesting exhaustion of response in “burnt-out” astrocytes. The spatial gene-expression-trajectories revealed that astrocytic genes of tripartite synapses are dysregulated in parallel to the stereotypical progression of tangle pathology across regions. We identified astrocyte heterogeneity across brain regions with a continuum from homeostatic to reactive cells through “intermediate” transitional states. These findings suggest complex astrocytic dysfunction in AD neurodegeneration.
- Published
- 2022
36. Deciphering the Astrocyte Reaction in Alzheimer’s Disease
- Author
-
Beatriz G. Perez-Nievas and Alberto Serrano-Pozo
- Subjects
Alzheimer’s disease ,amyloid plaques ,astrocytes ,glia ,microglia ,neurofibrillary tangles ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Reactive astrocytes were identified as a component of senile amyloid plaques in the cortex of Alzheimer’s disease (AD) patients several decades ago. However, their role in AD pathophysiology has remained elusive ever since, in part owing to the extrapolation of the literature from primary astrocyte cultures and acute brain injury models to a chronic neurodegenerative scenario. Recent accumulating evidence supports the idea that reactive astrocytes in AD acquire neurotoxic properties, likely due to both a gain of toxic function and a loss of their neurotrophic effects. However, the diversity and complexity of this glial cell is only beginning to be unveiled, anticipating that astrocyte reaction might be heterogeneous as well. Herein we review the evidence from mouse models of AD and human neuropathological studies and attempt to decipher the main conundrums that astrocytes pose to our understanding of AD development and progression. We discuss the morphological features that characterize astrocyte reaction in the AD brain, the consequences of astrocyte reaction for both astrocyte biology and AD pathological hallmarks, and the molecular pathways that have been implicated in this reaction.
- Published
- 2018
- Full Text
- View/download PDF
37. Effect of
- Author
-
Alberto, Serrano-Pozo, Zhaozhi, Li, Ayush, Noori, Huong N, Nguyen, Aziz, Mezlini, Liang, Li, Eloise, Hudry, Rosemary J, Jackson, Bradley T, Hyman, and Sudeshna, Das
- Subjects
Article - Abstract
The roles of APOEε4 and APOEε2—the strongest genetic risk and protective factors for Alzheimer’s disease—in glial responses remain elusive. We tested the hypothesis that APOE alleles differentially impact glial responses by investigating their effects on the glial transcriptome from elderly control brains with no neuritic amyloid plaques. We identified a cluster of microglial genes that are upregulated in APOEε4 and downregulated in APOEε2 carriers relative to APOEε3 homozygotes. This microglia-APOE cluster is enriched in phagocytosis—including TREM2 and TYROBP—and proinflammatory genes, and is also detectable in brains with frequent neuritic plaques. Next, we tested these findings in APOE knock-in mice exposed to acute (lipopolysaccharide challenge) and chronic (cerebral β-amyloidosis) insults and found that these mice partially recapitulate human APOE-linked expression patterns. Thus, the APOEε4 allele might prime microglia towards a phagocytic and proinflammatory state through an APOE–TREM2–TYROBP axis in normal aging as well as in Alzheimer’s disease.
- Published
- 2022
38. Acute and Chronic Sustained Hypoxia Do Not Substantially Regulate Amyloid-β Peptide Generation In Vivo.
- Author
-
Alberto Serrano-Pozo, Manuel A Sánchez-García, Antonio Heras-Garvín, Rosana March-Díaz, Victoria Navarro, Marisa Vizuete, José López-Barneo, Javier Vitorica, and Alberto Pascual
- Subjects
Medicine ,Science - Abstract
Recent epidemiological evidence has linked hypoxia with the development of Alzheimer disease (AD). A number of in vitro and in vivo studies have reported that hypoxia can induce amyloid-β peptide accumulation through various molecular mechanisms including the up-regulation of the amyloid-β precursor protein, the β-secretase Bace1, or the γγ-secretase complex components, as well as the down-regulation of Aβ-degrading enzymes.To investigate the effects of acute and chronic sustained hypoxia in Aβ generation in vivo.2-3 month-old C57/Bl6J wild-type mice were exposed to either normoxia (21% O2) or hypoxia (9% O2) for either 4 to 72 h (acute) or 21-30 days (chronic sustained) in a hermetic chamber. Brain mRNA levels of Aβ-related genes were measured by quantitative real-time PCR, whereas levels of Bace1 protein, full length AβPP, and its C-terminal fragments (C99/C88 ratio) were measured by Western blot. In addition, 8 and 14-month-old APP/PS1 transgenic mice were subjected to 9% O2 for 21 days and levels of Aβ40, Aβ42, full length AβPP, and soluble AβPPα (sAβPPα) were measured by ELISA or WB.Hypoxia (either acute or chronic sustained) did not impact the transcription of any of the Aβ-related genes in young wild-type mice. A significant reduction of Bace1 protein level was noted with acute hypoxia for 16 h but did not correlate with an increased level of full length AβPP or a decreased C99/C83 ratio. Chronic sustained hypoxia did not significantly alter the levels of Bace1, full length AβPP or the C99/C83 ratio. Last, chronic sustained hypoxia did not significantly change the levels of Aβ40, Aβ42, full length AβPP, or sAβPPα in either young or aged APP/PS1 mice.Our results argue against a hypoxia-induced shift of AβPP proteolysis from the non-amyloidogenic to the amyloidogenic pathways. We discuss the possible methodological caveats of previous in vivo studies.
- Published
- 2017
- Full Text
- View/download PDF
39. Plasma biomarkers for prognosis of cognitive decline in patients with mild cognitive impairment
- Author
-
Pia Kivisäkk, Colin Magdamo, Bianca A Trombetta, Ayush Noori, Yi kai E Kuo, Lori B Chibnik, Becky C Carlyle, Alberto Serrano-Pozo, Clemens R Scherzer, Bradley T Hyman, Sudeshna Das, and Steven E Arnold
- Subjects
General Engineering - Abstract
Plasma-based biomarkers present a promising approach in the research and clinical practice of Alzheimer's disease as they are inexpensive, accessible and minimally invasive. In particular, prognostic biomarkers of cognitive decline may aid in planning and management of clinical care. Although recent studies have demonstrated the prognostic utility of plasma biomarkers of Alzheimer pathology or neurodegeneration, such as pTau-181 and NF-L, whether other plasma biomarkers can further improve prediction of cognitive decline is undetermined. We conducted an observational cohort study to determine the prognostic utility of plasma biomarkers in predicting progression to dementia for individuals presenting with mild cognitive impairment due to probable Alzheimer's disease. We used the Olink™ Proximity Extension Assay technology to measure the level of 460 circulating proteins in banked plasma samples of all participants. We used a discovery data set comprised 60 individuals with mild cognitive impairment (30 progressors and 30 stable) and a validation data set consisting of 21 stable and 21 progressors. We developed a machine learning model to distinguish progressors from stable and used 44 proteins with significantly different plasma levels in progressors versus stable along with age, sex, education and baseline cognition as candidate features. A model with age, education, APOE genotype, baseline cognition, plasma pTau-181 and 12 plasma Olink protein biomarker levels was able to distinguish progressors from stable with 86.7% accuracy (mean area under the curve = 0.88). In the validation data set, the model accuracy was 78.6%. The Olink proteins selected by the model included those associated with vascular injury and neuroinflammation (e.g. IL-8, IL-17A, TIMP-4, MMP7). In addition, to compare these prognostic biomarkers to those that are altered in Alzheimer's disease or other types of dementia relative to controls, we analyzed samples from 20 individuals with Alzheimer, 30 with non-Alzheimer dementias and 34 with normal cognition. The proteins NF-L and PTP-1B were significantly higher in both Alzheimer and non-Alzheimer dementias compared with cognitively normal individuals. Interestingly, the prognostic markers of decline at the mild cognitive impairment stage did not overlap with those that differed between dementia and control cases. In summary, our findings suggest that plasma biomarkers of inflammation and vascular injury are associated with cognitive decline. Developing a plasma biomarker profile could aid in prognostic deliberations and identify individuals at higher risk of dementia in clinical practice.
- Published
- 2022
40. Hypoxia compromises the mitochondrial metabolism of Alzheimer’s disease microglia via HIF1
- Author
-
Adrian Viehweger, Almudena Gerpe, Manuel A. Sanchez-Garcia, Maria V. Sanchez-Mico, Alberto Pascual, Angela Gomez-Arboledas, José López-Barneo, Jose Carlos Davila, Javier Vitorica, Alicia E. Rosales-Nieves, Marisa Vizuete, Victoria Navarro, Edurne Berra, Antonio Heras-Garvin, Nieves Lara-Ureña, Rosana March-Díaz, Carmen Romero-Molina, Cristina Forja, Elisabeth Sanchez-Mejias, Antonia Gutierrez, Maria I. Alvarez-Vergara, Clara Ortega-de San Luis, Tammie Bishop, Emma J. Hodson, Alberto Serrano-Pozo, Instituto de Salud Carlos III, Ministerio de Educación, Cultura y Deporte (España), Ministerio de Economía, Industria y Competitividad (España), European Commission, Junta de Andalucía, and Fundación Domingo Martínez
- Subjects
Aging ,Microglia ,Cellular respiration ,HIF1 ,Neuroscience (miscellaneous) ,Hippocampus ,Neuropathology ,Mitochondrion ,Biology ,Hypoxia (medical) ,Aerobic respiration ,medicine.anatomical_structure ,Downregulation and upregulation ,medicine ,Cancer research ,Genetic predisposition ,Anaerobic glycolysis ,Geriatrics and Gerontology ,medicine.symptom ,Hypoxia ,Alzheimer’s disease - Abstract
Genetic Alzheimer’s disease (AD) risk factors associate with reduced defensive amyloid β plaque-associated microglia (AβAM), but the contribution of modifiable AD risk factors to microglial dysfunction is unknown. In AD mouse models, we observe concomitant activation of the hypoxia-inducible factor 1 (HIF1) pathway and transcription of mitochondrial-related genes in AβAM, and elongation of mitochondria, a cellular response to maintain aerobic respiration under low nutrient and oxygen conditions. Overactivation of HIF1 induces microglial quiescence in cellulo, with lower mitochondrial respiration and proliferation. In vivo, overstabilization of HIF1, either genetically or by exposure to systemic hypoxia, reduces AβAM clustering and proliferation and increases Aβ neuropathology. In the human AD hippocampus, upregulation of HIF1α and HIF1 target genes correlates with reduced Aβ plaque microglial coverage and an increase of Aβ plaque-associated neuropathology. Thus, hypoxia (a modifiable AD risk factor) hijacks microglial mitochondrial metabolism and converges with genetic susceptibility to cause AD microglial dysfunction., R.M.-D. was the recipient of a Sara Borrell fellowship from Instituto de Salud Carlos III (ISCIII) (CD09/0007). N.L.-U., C.O.-d.S.L., C.R.-M. and M.I.A.-V. were the recipients of FPU fellowships from Spanish Ministry of Education, Culture and Sport (FPU14/02115, AP2010‐1598, FPU16/02050 and FPU15/02898, respectively). A.H.-G. was the recipient of an FPI fellowship from the Spanish Ministry of Education, Culture and Sport (BES-2010-033886). This work was supported by grants from the Spanish MINEICO, ISCIII and FEDER (European Union) (SAF2012‐33816, SAF2015‐64111‐R, SAF2017-90794-REDT and PIE13/0004 to A.P.); by the Regional Government of Andalusia co-funded by CEC and FEDER funds (European Union) (‘Proyectos de Excelencia’; P12‐CTS‐2138 and P12‐CTS‐2232 to A.P.); by the ‘Ayuda de Biomedicina 2018’, Fundación Domingo Martínez (to A.P.) ; by the ISCIII of Spain, co-financed by FEDER funds (European Union) through grants PI18/01556 (to J.V.) and PI18/01557 (to A. Gutierrez); by Junta de Andalucía, co-financed by FEDER funds (grants UMA18-FEDERJA-211 (to A. Gutierrez) and US‐1262734 (to J.V.)); and by Spanish MINEICO (BFU2016-76872-R and BES-2011-047721 to E.B.).
- Published
- 2021
41. Association of APOE Genotype With Heterogeneity of Cognitive Decline Rate in Alzheimer Disease
- Author
-
Bradley T. Hyman, Rebecca A. Betensky, Alberto Serrano-Pozo, and Jing Qian
- Subjects
0301 basic medicine ,Oncology ,Apolipoprotein E ,medicine.medical_specialty ,Clinical Dementia Rating ,business.industry ,Neuropathology ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Internal medicine ,Genotype ,Medicine ,Neurology (clinical) ,Senile plaques ,Allele ,Cognitive decline ,Alzheimer's disease ,business ,030217 neurology & neurosurgery - Abstract
ObjectiveTo test the hypothesis that the APOE genotype is a significant driver of heterogeneity in Alzheimer disease (AD) clinical progression, which could have important implications for clinical trial design and interpretation.MethodsWe applied novel reverse-time longitudinal models to analyze the trajectories of Clinical Dementia Rating Sum of Boxes (CDR-SOB) and Mini-Mental State Examination (MMSE) scores—2 common outcome measures in AD clinical trials—in 1,102 autopsy-proven AD cases (moderate/frequent neuritic plaques and Braak tangle stage III or greater) from the National Alzheimer's Coordinating Center Neuropathology database resembling participants with mild to moderate AD in therapeutic clinical trials.ResultsAPOE ε4 carriers exhibited ≈1.5 times faster CDR-SOB increase than APOE ε3/ε3 carriers (2.12 points per year vs 1.44 points per year) and ≈1.3 times faster increase than APOE ε2 carriers (1.65 points per year), whereas APOE ε2 vs APOE ε3/ε3 difference was not statistically significant. APOE ε4 carriers had ≈1.1 times faster MMSE decline than APOE ε3/ε3 carriers (−3.45 vs −3.03 points per year) and ≈1.4 times faster decline than APOE ε2 carriers (−2.43 points per year), whereas APOE ε2 carriers had ≈1.2 times slower decline than APOE ε3/ε3 carriers (−2.43 vs −3.03 points per year). These findings remained largely unchanged after controlling for the effect of AD neuropathologic changes on the rate of cognitive decline and for the presence and severity of comorbid pathologies.ConclusionCompared to the APOE ε3/ε3 reference genotype, the APOE ε2 and ε4 alleles have opposite (slowing and accelerating, respectively) effects on the rate of cognitive decline, which are clinically relevant and largely independent of the differential APOE allele effects on AD and comorbid pathologies. Thus, APOE genotype contributes to the heterogeneity in rate of clinical progression in AD.
- Published
- 2021
42. Development and Evaluation of a Natural Language Processing Annotation Tool to Facilitate Phenotyping of Cognitive Status in Electronic Health Records: Diagnostic Study
- Author
-
Noori, Ayush, primary, Magdamo, Colin, additional, Liu, Xiao, additional, Tyagi, Tanish, additional, Li, Zhaozhi, additional, Kondepudi, Akhil, additional, Alabsi, Haitham, additional, Rudmann, Emily, additional, Wilcox, Douglas, additional, Brenner, Laura, additional, Robbins, Gregory K, additional, Moura, Lidia, additional, Zafar, Sahar, additional, Benson, Nicole M, additional, Hsu, John, additional, R Dickson, John, additional, Serrano-Pozo, Alberto, additional, Hyman, Bradley T, additional, Blacker, Deborah, additional, Westover, M Brandon, additional, Mukerji, Shibani S, additional, and Das, Sudeshna, additional
- Published
- 2022
- Full Text
- View/download PDF
43. Plasma biomarkers for prognosis of cognitive decline in patients with mild cognitive impairment
- Author
-
Kivisäkk, Pia, primary, Magdamo, Colin, additional, Trombetta, Bianca A, additional, Noori, Ayush, additional, Kuo, Yi kai E, additional, Chibnik, Lori B, additional, Carlyle, Becky C, additional, Serrano-Pozo, Alberto, additional, Scherzer, Clemens R, additional, Hyman, Bradley T, additional, Das, Sudeshna, additional, and Arnold, Steven E, additional
- Published
- 2022
- Full Text
- View/download PDF
44. A Phenotypic Change But Not Proliferation Underlies Glial Responses in Alzheimer Disease
- Author
-
Serrano-Pozo, Alberto, Gómez-Isla, Teresa, Growdon, John H., Frosch, Matthew P., and Hyman, Bradley T.
- Published
- 2013
- Full Text
- View/download PDF
45. Additional file 1 of Cyclic multiplex fluorescent immunohistochemistry and machine learning reveal distinct states of astrocytes and microglia in normal aging and Alzheimer’s disease
- Author
-
Ministerio de Ciencia, Innovación y Universidades (España), National Institute on Aging (US), Alzheimer's Association, Real Colegio Complutense, Muñoz-Castro, Clara, Noori, Ayush, Magdamo, Colin G., Li, Zhaozhi, Marks, Jordan D., Frosch, Matthew P., Das, Sudeshna, Hyman, Bradley T., Serrano-Pozo, Alberto, Ministerio de Ciencia, Innovación y Universidades (España), National Institute on Aging (US), Alzheimer's Association, Real Colegio Complutense, Muñoz-Castro, Clara, Noori, Ayush, Magdamo, Colin G., Li, Zhaozhi, Marks, Jordan D., Frosch, Matthew P., Das, Sudeshna, Hyman, Bradley T., and Serrano-Pozo, Alberto
- Abstract
Additional File 1: Table S1. Demographic and neuropathological characteristics of study subjects. Description: Abbreviations: ADNC = AD neuropathological changes; APOE = Apolipoprotein E genotype; CAA = cerebral amyloid angiopathy; CVD = cerebrovascular disease; F = female; LBD = Lewy body disease; M = male; NA = Not available/applicable; NOS = not otherwise specified; NP Dx = neuropathological diagnosis. Table S2. Primary and secondary antibodies used in this study and sequence of immunohistochemistry cycles. Description: Note: GFAP and DAPI detection are needed in all the cycles to guarantee an adequate alignment of the images. Abbreviations: AF488 = AlexaFluor 488; Cy = cyanine; Dk = donkey; Gt = goat; Ms = mouse; Rb = rabbit. All secondary antibodies were purchased from Jackson ImmunoResearch Labs, West Grove, PA. Table S3. Results of mixed effects regression models. Description: Results of mixed effects regression models with diagnosis (CTRL vs. AD) or state (homeostatic vs. intermediate vs. reactive) as a fixed effect, respectively, and subject ID as random effect in both cases, are reported. Table S4. Model performance statistics for CTRL vs. AD binary classifiers. Description: Model performance statistics for the binary classification task of CTRL vs. AD for both the gradient boosting machine (GBM) and the convolutional neural network (CNN) machine learning models are reported. For all heuristics except for AUC and AUCPR (which are not threshold-dependent), the threshold was chosen by maximizing the accuracy. 95% confidence intervals were estimated by bootstrapping the hold-out test set across 500 iterations. Table S5. Results of Bayesian hyperparameter optimization. Description: The final hyperparameters determined by the Optuna hyperparameter tuning framework are reported. The Optuna optimizer maximized the out-of-sample area under the receiver operating characteristic (ROC) curve (AUC), which in turn was determined by 3-fold cross-validation for each tria
- Published
- 2022
46. Inhibition of amyloid-β plaque formation by α-synuclein
- Author
-
Bachhuber, Teresa, Katzmarski, Natalie, McCarter, Joanna F., Loreth, Desiree, Tahirovic, Sabina, Kamp, Frits, Abou-Ajram, Claudia, Nuscher, Brigitte, Serrano-Pozo, Alberto, Muller, Alexandra, Prinz, Marco, Steiner, Harald, Hyman, Bradley T., Haass, Christian, and Meyer-Luehmann, Melanie
- Subjects
Development and progression -- Genetic aspects -- Research ,Biological sciences ,Health - Abstract
Amyloid-β (Aβ) plaques and α-synuclein (α-syn)-rich Lewy bodies are the major neuropathological hallmarks of Alzheimer's disease (AD) and Parkinson's disease, respectively. An overlap of pathologies is found in most individuals with dementia with Lewy bodies (DLB) (1) and in more than 50% of AD cases (2). Their brains display substantial α-syn accumulation not only in Lewy bodies, but also in dystrophic neurites decorating Aβ plaques (2-4). Several studies report binding and coaggregation of Aβ and α-syn (5-7), yet the precise role of α-syn in amyloid plaque formation remains elusive. Here we performed intracerebral injections of α-syn-containing preparations into amyloid precursor protein (APP) transgenic mice (expressing [APP695.sup.KM670/671NL] and [PSEN1.sup.L166P] under the control of the neuron-specific Thy-1 promoter; referred to here as 'APPPS1'). Unexpectedly, α-syn failed to cross-seed Aβ plaques in vivo, but rather it inhibited plaque formation in APPPS1 mice coexpressing [SNCA.sup.A30P] (referred to here as 'APPPS1 x [A30P]aSYN' double-transgenic mice). This was accompanied by increased Ap levels in cerebrospinal fluid despite unchanged overall Aβ levels. Notably, the seeding activity of Aβ-containing brain homogenates was considerably reduced by α-syn, and Aβ deposition was suppressed in grafted tissue from [A30P]aSYN transgenic mice. Thus, we conclude that an interaction between Aβ and α-syn leads to inhibition of Aβ deposition and to reduced plaque formation., The initial discovery of α-syn within plaques as the precursor of the non-amyloid component (NAC) (8) and the binding of Aβ and α-syn in vitro and in situ (5,6) suggested [...]
- Published
- 2015
47. The Synaptic Accumulation of Hyperphosphorylated Tau Oligomers in Alzheimer Disease Is Associated With Dysfunction of the Ubiquitin-Proteasome System
- Author
-
Tai, Hwan-Ching, Serrano-Pozo, Alberto, Hashimoto, Tadafumi, Frosch, Matthew P., Spires-Jones, Tara L., and Hyman, Bradley T.
- Published
- 2012
- Full Text
- View/download PDF
48. Additional file 2 of Cyclic multiplex fluorescent immunohistochemistry and machine learning reveal distinct states of astrocytes and microglia in normal aging and Alzheimer���s disease
- Author
-
Mu��oz-Castro, Clara, Noori, Ayush, Magdamo, Colin G., Li, Zhaozhi, Marks, Jordan D., Frosch, Matthew P., Das, Sudeshna, Hyman, Bradley T., and Serrano-Pozo, Alberto
- Abstract
Additional File 2: Figure S1. A �� pathology in the temporal pole cortex. Description: Immunohistochemistry for A�� (mouse monoclonal antibody, clone 6F/3D, Agilent, #M0872, 1:600) with peroxidase/DAB was performed in nearly-adjacent sections to those used for cyclic multiplex fluorescent immunohistochemistry in a Leica BOND-III automated stainer. Sections were counterstained with hematoxylin. Scale bars: 5 mm, insets 200 ��m. Figure S2. Phospho-tau pathology in the temporal pole cortex. Description: Immunohistochemsitry for phospho-tauSer202/Thr205(mouse monoclonal antibody, clone AT8, Thermo-Scientific, #MN1020, 1:10,000) with peroxidase/DAB was performed in nearly-adjacent sections to those used for cyclic multiplex fluorescent immunohistochemistry in a Leica BOND-III automated stainer. Sections were counterstained with hematoxylin. Scale bars: 5 mm, insets 200 ��m. Figure S3. Expression levels of selected markers across astrocytic and microglial subclusters from public single-nuclei RNA-seq studies. Description: Bubble plots illustrate the percent of nuclei (bubble size) and the gene expression levels (z-scores, color bar) of the astrocytic and microglial markers used in our cyclic multiplex fluorescent immunohistochemistry protocol across the astrocytic and microglial subclusters rendered by several published single-nuclei RNA-seq data sets. Note that our set of markers discriminates some of these transcriptomic subclusters. Figure S4. Characterization of astrocytes and microglia in AD vs. CTRL by cortical layer. Description: Box and whisker plots illustrate the distribution (box: median and interquartile range [IQR]; whiskers: 1.5 �� IQR) of mean gray intensity (MGI) z-scores for (a) each astrocytic marker and (b) each microglial marker across the CTRL and AD groups by cortical layer. Only layers II to VI were included in this study. Figure S5. Characterization of astrocytic and microglial states by cortical layer. Description: Box and whisker plots show the distribution (box: median and interquartile range [IQR]; whiskers: 1.5 �� IQR) of mean gray intensity (MGI) z-scores for each astrocytic (a) or microglial (b) marker across the three phenotypes by cortical layer. Only layers II to VI were included in this study. Figure S6. Effects of proximity to AD neuropathological changes on astrocytic and microglial phenotypes from two CTRL subjects with abundant A�� plaques. Description: (a) Representative high-plex image of astrocytes from a CTRL subject with abundant A�� plaques; note the differences with AD astrocytes in Fig. 5a. For clarity, only ALDH1L1, EAAT2, and GFAP markers are shown together with A��. Scale bar: 100 ��m, insets a1���a3: 10 ��m. (b) Histograms show the proportion of each astrocyte phenotype in n=2 CTRL subjects with abundant A�� plaques relative to all their astrocytes as a function of their distance (��m, x axis) to the nearest A�� plaque. Note that there are equal numbers of astrocytes within 25 ��m from the nearest A�� plaque classified as homeostatic, intermediate, or reactive. (c) Representative high-plex image of microglia from the same field of the same CTRL with abundant A�� plaques; note the differences when compared to AD microglia in Fig. 5c. For clarity, only IBA1, TMEM119, and CD68 markers are shown together with A��. Scale bar: 100 ��m, insets c1���c3: 10 ��m. (d) Histograms indicate the proportion of each microglial phenotype in n=2 CTRL subjects with abundant A�� plaques relative to all their microglial profiles as a function of their distance (��m, x axis) to the nearest A�� plaque. Note that most microglia in the vicinity of A�� plaques were classified as homeostatic, suggesting that their phenotypic transition to intermediate and reactive had not yet occurred. Figure S7. Differences in neuritic component of A�� plaques from CTRL and AD subjects. Description: Representative images of A�� and phospho-tau (PHF1) immunohistochemistry corresponding to the same fields of the AD and CTRL subjects shown in Fig. 5 and Fig. S6, respectively. Note the differences in the PHF1+ neuritic changes between CTRL and AD A�� plaques. Scale bar: 100 ��m, insets a1 and b1: 10 ��m. Figure S8. Gradient boosting machine models accurately discriminate between glial phenotypes. Description: Receiver operating characteristic (ROC) curves demonstrate the high discriminative power of the gradient boosting machine (GBM) models to discern between states (i.e., homeostatic vs. intermediate vs. reactive) of (a) astrocytes and (b) microglia based on mean gray intensity (MGI) data from thousands of high-plex single-cell profiles. Rankings of the variable importance scores shown in the horizontal bar plots reveal the most relevant markers for each classification task, respectively. Figure S9. Application of deep learning model interpretability functions to astrocytes with extreme classification probabilities. Description: Examples of the convolutional neural network (CNN) model interpretability functions applied to astrocytes with extreme classification probabilities (i.e., confident and correct predictions). Columns 1 and 5 show DAPI and all astrocyte markers of the high-plex image of a single astrocyte cell body from a CTRL and an AD subject, respectively, after performing the CNN normalization steps described (i.e., segmentation, interpolation, channel-level z-score). Hence, the signal intensity is represented by dynamic range rather than by pixel intensity. Columns 2���4 and 6���8 show the saliency (2 and 6), integrated gradient (3 and 7), and GradCAM (4 and 8) maps, which illustrate the pixels of each marker that the CNN considered most important for the classification of these two astrocytes as CTRL or AD. Figure S10. Application of deep learning model interpretability functions to microglia with extreme classification probabilities. Description: Examples of the convolutional neural network (CNN) model interpretability functions applied to microglia with extreme classification probabilities (i.e., confident and correct predictions). Columns 1 and 5 show DAPI and all microglial markers of the high-plex image of a single microglial cell from a CTRL and an AD subject, respectively, after performing the CNN normalization steps described (i.e., segmentation, interpolation, channel-level z-score). Hence, the signal intensity is represented by dynamic range rather than by pixel intensity. Columns 2���4 and 6���8 show the saliency (2 and 6), integrated gradient (3 and 7), and GradCAM (4 and 8) maps, which illustrate the pixels of each marker that the CNN considered most important for the classification of these two microglia as CTRL or AD.
- Published
- 2022
- Full Text
- View/download PDF
49. Neuropathology-Independent Association BetweenAPOEGenotype and Cognitive Decline Rate in the Normal Aging-Early Alzheimer Continuum
- Author
-
Jing Qian, Yiding Zhang, Rebecca A. Betensky, Bradley T. Hyman, and Alberto Serrano-Pozo
- Subjects
Neurology (clinical) ,Genetics (clinical) - Abstract
Background and ObjectivesWe previously found that theAPOEgenotype affects the rate of cognitive decline in mild-to-moderate Alzheimer disease (AD) dementia independently of its effects on AD neuropathologic changes (ADNC) and copathologies. In this study, we tested the hypothesis that theAPOEalleles differentially affect the rate of cognitive decline at the normal aging-early AD continuum and that this association is independent of their effects on classical ADNC and copathologies.MethodsWe analyzedAPOEassociations with the cognitive trajectories (Clinical Dementia Rating scale Sum of Boxes [CDR-SOB] and Mini-Mental State Examination [MMSE]) of more than 1,000 individuals from a national clinicopathologic sample who had either no, mild (sparse neuritic plaques and the Braak neurofibrillary tangle [NFT] stage I/II), or intermediate (moderate neuritic plaques and the Braak NFT stage III/IV) ADNC levels at autopsy via 2 latent classes reverse-time longitudinal modeling.ResultsCarrying theAPOEε4 allele was associated with a faster rate of cognitive decline by both CDR-SOB and MMSE relative toAPOEε3 homozygotes. This association remained statistically significant after adjusting for ADNC severity, comorbid pathologies, and the effects of ADNC on the slope of cognitive decline. Our modeling strategy identified 2 latent classes in whichAPOEε4 carriers declined faster thanAPOEε3 homozygotes, with latent class 1 members representing slow decliners (CDR-SOB: 76.7% of individuals, 0.195 vs 0.146 points/y inAPOEε4 vsAPOEε3/ε3; MMSE: 88.6% of individuals, −0.303 vs −0.153 points/y inAPOEε4 vsAPOEε3/ε3), whereas latent class 2 members were fast decliners (CDR-SOB: 23.3% of participants, 1.536 vs 1.487 points/y inAPOEε4 vsAPOEε3/ε3; MMSE: 11.4% of participants, −2.538 vs −2.387 points/y inAPOEε4 vsAPOEε3/ε3). Compared with slow decliners, fast decliners were more likely to carry theAPOEε4 allele, younger at initial visit and death, more impaired at initial and last visits, and more likely to have intermediate (vs none or mild) ADNC levels, as well as concurrent Lewy bodies and hippocampal sclerosis at autopsy.DiscussionIn a large national sample selected to represent the normal aging-early AD continuum, theAPOEε4 allele is associated with a modest but statistically significant acceleration of the cognitive decline rate even after controlling for its effects on ADNC and comorbid pathologies.
- Published
- 2023
50. Active deep learning to detect cognitive concerns in electronic health records
- Author
-
Colin G. Magdamo, Zhuoqiao Mia Hong, Ayush Noori, Yi‐Han Sheu, Mayuresh Deodhar, Elissa M Ye, Wendong Ge, Haoqi Sun, Laura Brenner, Gregory K. Robbins, Shibani Mukerji, Sahar F. Zafar, Nicole Benson, Lidia Maria V. Moura, John Hsu, Steven E. Arnold, Bradley T. Hyman, Alberto Serrano‐Pozo, M Brandon Westover, Deborah Blacker, and Sudeshna Das
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.