1
|
Merino-Serrais P, Plaza-Alonso S, Tapia-Gonzalez S, León-Espinosa G, DeFelipe J. Parvalbumin interneurons in the hippocampal formation of individuals with Alzheimer's disease: a neuropathological study of abnormal phosphorylated tau in neurons. Front Neuroanat 2025; 19:1571514. [PMID: 40275866 PMCID: PMC12018435 DOI: 10.3389/fnana.2025.1571514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder in the elderly. Recent efforts have centered on understanding early events that trigger AD, aiming to facilitate early diagnosis and intervention for improved patient outcomes. The traditional histopathological features observed in AD encompass the extracellular accumulation of amyloid-beta protein and the intracellular abnormal phosphorylation of Tau protein (pTau). However, elucidating how these pathological hallmarks ultimately contribute to cognitive deficits remains a complex challenge. While AD is commonly conceptualized as a disorder characterized by synaptic failure, substantial knowledge gaps persist regarding the mechanisms underlying the onset and progression of the disease, underscoring the need for novel and more effective therapeutic approaches. In this context, the impairment of GABAergic paravalbumin (PV+) neurons has been proposed as a crucial factor contributing to neuronal network dysfunction and cognitive decline in AD. The presence of pTau in pyramidal neurons is directly linked to their impairment in AD; however, the effect of pTau in PV+ neurons remains unclear. In this present study, we analyzed the existence of PV+ neurons containing pTau using immunocytochemistry in the hippocampal formation and entorhinal cortex of human samples from diagnosed AD cases and individuals without neurological or psychiatric disorders. Two pTau isoforms, pTauAT8 and pTaupS396, corresponding to early and late stages of AD respectively, were examined. Our findings indicate that most PV+ neurons across the hippocampal formation and entorhinal cortex did not contain pTau in either group cases. Interestingly, while AD cases diagnosed with dementia exhibited a higher number of pTau+ neurons, the majority of PV+/pTau+ neurons were found in individuals with no neurological alterations. This suggests that the presence of pTau in PV+ neurons does not directly correlate with the overall abundance of pTau+ neurons. Given that PV+ neuron impairment is a key pathogenic mechanism in AD and is associated with cognitive decline, understanding the changes in PV+ neurons during AD progression could provide critical insights into the alterations of neuronal circuits underlying the disease.
Collapse
Affiliation(s)
- Paula Merino-Serrais
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Sergio Plaza-Alonso
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Silvia Tapia-Gonzalez
- Laboratorio de Neurofisiología Celular, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Gonzalo León-Espinosa
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Madrid, Spain
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| |
Collapse
|
2
|
Rodriguez-Rodriguez P, Wang W, Tsagkogianni C, Feng I, Morello-Megias A, Jain K, Alanko V, Kahvecioglu HA, Mohammadi E, Li X, Flajolet M, Sandebring-Matton A, Maioli S, Vidal N, Milosevic A, Roussarie JP. Cell-type specific profiling of human entorhinal cortex at the onset of Alzheimer's disease neuropathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630881. [PMID: 39803521 PMCID: PMC11722323 DOI: 10.1101/2024.12.31.630881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Neurons located in layer II of the entorhinal cortex (ECII) are the primary site of pathological tau accumulation and neurodegeneration at preclinical stages of Alzheimer's disease (AD). Exploring the alterations that underlie the early degeneration of these cells is essential to develop therapies that curb the disease before symptom onset. Here we performed cell-type specific profiling of human EC at the onset of AD neuropathology. We identify an early response to amyloid pathology by microglia and oligodendrocytes. Importantly, we provide the first insight into neuronal alterations that coincide with incipient tau pathology: the signaling pathway for Reelin, recently shown to be a major AD resilience gene is dysregulated in ECII neurons, while the secreted synaptic organizer molecules NPTX2 and CBLN4, emerging AD biomarkers, are downregulated in surrounding neurons. By uncovering the complex multicellular landscape of EC at these early AD stages, this study paves the way for detailed characterization of the mechanisms governing NFT formation and opens long-needed novel therapeutic avenues.
Collapse
Affiliation(s)
| | - Wei Wang
- Bioinformatics Resource Center, The Rockefeller University. New York, NY, USA
| | - Christina Tsagkogianni
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | - Irena Feng
- Boston University Chobanian & Avedisian School of Medicine. Boston, MA, USA
| | - Ana Morello-Megias
- Boston University Chobanian & Avedisian School of Medicine. Boston, MA, USA
| | - Kaahini Jain
- Boston University Chobanian & Avedisian School of Medicine. Boston, MA, USA
| | - Vilma Alanko
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | | | - Elyas Mohammadi
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | - Xiaofei Li
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | | | - Anna Sandebring-Matton
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | - Silvia Maioli
- Department of Neurobiology Care Sciences and Society, Karolinska Institute. Stockholm, Sweden
| | - Noemi Vidal
- Pathology department. Biobank HUB-ICO-IDIBELL, University Hospital of Bellvitge. Barcelona, Spain
| | - Ana Milosevic
- Laboratory of Developmental Genetics, The Rockefeller University. New York, NY, USA
| | | |
Collapse
|
3
|
Cooper LN, Ansari MY, Capshaw G, Galazyuk A, Lauer AM, Moss CF, Sears KE, Stewart M, Teeling EC, Wilkinson GS, Wilson RC, Zwaka TP, Orman R. Bats as instructive animal models for studying longevity and aging. Ann N Y Acad Sci 2024; 1541:10-23. [PMID: 39365995 PMCID: PMC11580778 DOI: 10.1111/nyas.15233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Bats (order Chiroptera) are emerging as instructive animal models for aging studies. Unlike some common laboratory species, they meet a central criterion for aging studies: they live for a long time in the wild or in captivity, for 20, 30, and even >40 years. Healthy aging (i.e., healthspan) in bats has drawn attention to their potential to improve the lives of aging humans due to bat imperviousness to viral infections, apparent low rate of tumorigenesis, and unique ability to repair DNA. At the same time, bat longevity also permits the accumulation of age-associated systemic pathologies that can be examined in detail and manipulated, especially in captive animals. Research has uncovered additional and critical advantages of bats. In multiple ways, bats are better analogs to humans than are rodents. In this review, we highlight eight diverse areas of bat research with relevance to aging: genome sequencing, telomeres, and DNA repair; immunity and inflammation; hearing; menstruation and menopause; skeletal system and fragility; neurobiology and neurodegeneration; stem cells; and senescence and mortality. These examples demonstrate the broad relevance of the bat as an animal model and point to directions that are particularly important for human aging studies.
Collapse
Affiliation(s)
- Lisa Noelle Cooper
- Department of Anatomy and NeurobiologyNortheast Ohio Medical UniversityRootstownOhioUSA
| | - Mohammad Y. Ansari
- Department of Anatomy and NeurobiologyNortheast Ohio Medical UniversityRootstownOhioUSA
| | - Grace Capshaw
- Department of Psychological and Brain SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Alex Galazyuk
- Department of Anatomy and NeurobiologyNortheast Ohio Medical UniversityRootstownOhioUSA
| | - Amanda M. Lauer
- Department of Otolaryngology – HNSJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Cynthia F. Moss
- Department of Psychological and Brain SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Karen E. Sears
- Department of Ecology and Evolutionary Biology, Department of Molecular, Cellular, and Developmental BiologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Mark Stewart
- Department of Physiology & PharmacologySUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Emma C. Teeling
- School of Biology and Environmental Science, Science Centre EastUniversity College DublinDublinIreland
| | - Gerald S. Wilkinson
- Department of BiologyUniversity of Maryland at College ParkCollege ParkMarylandUSA
| | | | - Thomas P. Zwaka
- Black Family Stem Cell Institute, Huffington Center for Cell‐based Research in Parkinson's Disease, Department of Cell, Developmental and Regenerative BiologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Rena Orman
- Department of Physiology & PharmacologySUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| |
Collapse
|
4
|
Vanessa Becerra-Hernández L, Casanova MF, Buriticá E. Cortical calretinin-positive neurons: Functional and ontogenetic characteristics and their relationship to brain pathologies. Brain Res 2024; 1846:149285. [PMID: 39490954 DOI: 10.1016/j.brainres.2024.149285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 06/02/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
Cortical GABAergic interneurons can be classified according to electrophysiological, biochemical, and/or morphological criteria. In humans, the use of calcium-binding proteins allows us to differentiate three subpopulations of GABAergic interneurons with minimal overlap. Cortical calretinin-positive neurons mainly include bipolar and double-bouquet morphologies, with a largely non-rapid and adaptive firing pattern, originating from the ganglionic eminence and the ventricular and subventricular regions of the developing brain. These cells are distributed from layer I to VI of the neocortex, with predominance in layers II and III. Given their morphology, distribution of processes, and elucidated synaptic contacts, these neurons are considered important in the control of intraminicolumnar processing through vertical inhibition. They have been extensively studied in the context of pathologies characterized by excitation/inhibition imbalance, such as Alzheimer's disease, epilepsy, traumatic brain injury, and autism. In light of the current evidence, this review considers these aspects in depth and discusses the pathophysiological role and selective vulnerability (pathoclisis) vs. the resistance that these interneurons can present against different types of injury.
Collapse
Affiliation(s)
- Lina Vanessa Becerra-Hernández
- Centro de Estudios Cerebrales, Facultad de Salud, Universidad del Valle, Cali, Colombia; Departamento de Ciencias Básicas de la Salud, Pontificia Universidad Javeriana, Cali, Colombia.
| | - Manuel F Casanova
- Department of Biomedical Sciences, School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| | - Efraín Buriticá
- Centro de Estudios Cerebrales, Facultad de Salud, Universidad del Valle, Cali, Colombia
| |
Collapse
|
5
|
Michaud F, Francavilla R, Topolnik D, Iloun P, Tamboli S, Calon F, Topolnik L. Altered firing output of VIP interneurons and early dysfunctions in CA1 hippocampal circuits in the 3xTg mouse model of Alzheimer's disease. eLife 2024; 13:RP95412. [PMID: 39264364 PMCID: PMC11392531 DOI: 10.7554/elife.95412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
Alzheimer's disease (AD) leads to progressive memory decline, and alterations in hippocampal function are among the earliest pathological features observed in human and animal studies. GABAergic interneurons (INs) within the hippocampus coordinate network activity, among which type 3 interneuron-specific (I-S3) cells expressing vasoactive intestinal polypeptide and calretinin play a crucial role. These cells provide primarily disinhibition to principal excitatory cells (PCs) in the hippocampal CA1 region, regulating incoming inputs and memory formation. However, it remains unclear whether AD pathology induces changes in the activity of I-S3 cells, impacting the hippocampal network motifs. Here, using young adult 3xTg-AD mice, we found that while the density and morphology of I-S3 cells remain unaffected, there were significant changes in their firing output. Specifically, I-S3 cells displayed elongated action potentials and decreased firing rates, which was associated with a reduced inhibition of CA1 INs and their higher recruitment during spatial decision-making and object exploration tasks. Furthermore, the activation of CA1 PCs was also impacted, signifying early disruptions in CA1 network functionality. These findings suggest that altered firing patterns of I-S3 cells might initiate early-stage dysfunction in hippocampal CA1 circuits, potentially influencing the progression of AD pathology.
Collapse
Affiliation(s)
- Felix Michaud
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Ruggiero Francavilla
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Dimitry Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Parisa Iloun
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Suhel Tamboli
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| | - Frederic Calon
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Lisa Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, Canada
- Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, Canada
| |
Collapse
|
6
|
Kampmann M. Molecular and cellular mechanisms of selective vulnerability in neurodegenerative diseases. Nat Rev Neurosci 2024; 25:351-371. [PMID: 38575768 DOI: 10.1038/s41583-024-00806-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 04/06/2024]
Abstract
The selective vulnerability of specific neuronal subtypes is a hallmark of neurodegenerative diseases. In this Review, I summarize our current understanding of the brain regions and cell types that are selectively vulnerable in different neurodegenerative diseases and describe the proposed underlying cell-autonomous and non-cell-autonomous mechanisms. I highlight how recent methodological innovations - including single-cell transcriptomics, CRISPR-based screens and human cell-based models of disease - are enabling new breakthroughs in our understanding of selective vulnerability. An understanding of the molecular mechanisms that determine selective vulnerability and resilience would shed light on the key processes that drive neurodegeneration and point to potential therapeutic strategies to protect vulnerable cell populations.
Collapse
Affiliation(s)
- Martin Kampmann
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Olkhova EA, Smith LA, Dennis BH, Ng YS, LeBeau FEN, Gorman GS. Delineating mechanisms underlying parvalbumin neuron impairment in different neurological and neurodegenerative disorders: the emerging role of mitochondrial dysfunction. Biochem Soc Trans 2024; 52:553-565. [PMID: 38563502 PMCID: PMC11088917 DOI: 10.1042/bst20230191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024]
Abstract
Given the current paucity of effective treatments in many neurological disorders, delineating pathophysiological mechanisms among the major psychiatric and neurodegenerative diseases may fuel the development of novel, potent treatments that target shared pathways. Recent evidence suggests that various pathological processes, including bioenergetic failure in mitochondria, can perturb the function of fast-spiking, parvalbumin-positive neurons (PV+). These inhibitory neurons critically influence local circuit regulation, the generation of neuronal network oscillations and complex brain functioning. Here, we survey PV+ cell vulnerability in the major neuropsychiatric, and neurodegenerative diseases and review associated cellular and molecular pathophysiological alterations purported to underlie disease aetiology.
Collapse
Affiliation(s)
- Elizaveta A. Olkhova
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
| | - Laura A. Smith
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
| | - Bethany H. Dennis
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
| | - Yi Shiau Ng
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, U.K
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4HH, U.K
| | - Fiona E. N. LeBeau
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
| | - Gráinne S. Gorman
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, U.K
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4HH, U.K
| |
Collapse
|
8
|
Acharya M, Singh N, Gupta G, Tambuwala MM, Aljabali AAA, Chellappan DK, Dua K, Goyal R. Vitamin D, Calbindin, and calcium signaling: Unraveling the Alzheimer's connection. Cell Signal 2024; 116:111043. [PMID: 38211841 DOI: 10.1016/j.cellsig.2024.111043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
Calcium is a ubiquitous second messenger that is indispensable in regulating neurotransmission and memory formation. A precise intracellular calcium level is achieved through the concerted action of calcium channels, and calcium exerts its effect by binding to an array of calcium-binding proteins, including calmodulin (CAM), calcium-calmodulin complex-dependent protein kinase-II (CAMK-II), calbindin (CAL), and calcineurin (CAN). Calbindin orchestrates a plethora of signaling events that regulate synaptic transmission and depolarizing signals. Vitamin D, an endogenous fat-soluble metabolite, is synthesized in the skin upon exposure to ultraviolet B radiation. It modulates calcium signaling by increasing the expression of the calcium-sensing receptor (CaSR), stimulating phospholipase C activity, and regulating the expression of calcium channels such as TRPV6. Vitamin D also modulates the activity of calcium-binding proteins, including CAM and calbindin, and increases their expression. Calbindin, a high-affinity calcium-binding protein, is involved in calcium buffering and transport in neurons. It has been shown to inhibit apoptosis and caspase-3 activity stimulated by presenilin 1 and 2 in AD. Whereas CAM, another calcium-binding protein, is implicated in regulating neurotransmitter release and memory formation by phosphorylating CAN, CAMK-II, and other calcium-regulated proteins. CAMK-II and CAN regulate actin-induced spine shape changes, which are further modulated by CAM. Low levels of both calbindin and vitamin D are attributed to the pathology of Alzheimer's disease. Further research on vitamin D via calbindin-CAMK-II signaling may provide newer insights, revealing novel therapeutic targets and strategies for treatment.
Collapse
Affiliation(s)
- Manish Acharya
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University, Himachal Pradesh, India
| | - Nicky Singh
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University, Himachal Pradesh, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur 302017, India
| | - Murtaza M Tambuwala
- Lincoln Medical School, Universities of Nottingham and Lincoln College of Science, Brayford Pool Campus, Lincoln LN6 7TS, UK.
| | - Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Yarmouk University, Irbid 21163, Jordan.
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| | - Rohit Goyal
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University, Himachal Pradesh, India.
| |
Collapse
|
9
|
Perez SM, Boley AM, McCoy AM, Lodge DJ. Aberrant Dopamine System Function in the Ferrous Amyloid Buthionine (FAB) Rat Model of Alzheimer's Disease. Int J Mol Sci 2023; 24:7196. [PMID: 37108357 PMCID: PMC10138591 DOI: 10.3390/ijms24087196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Antipsychotics increase the risk of death in elderly patients with Alzheimer's disease (AD). Thus, there is an immediate need for novel therapies to treat comorbid psychosis in AD. Psychosis has been attributed to a dysregulation of the dopamine system and is associated with aberrant regulation by the hippocampus. Given that the hippocampus is a key site of pathology in AD, we posit that aberrant regulation of the dopamine system may contribute to comorbid psychosis in AD. A ferrous amyloid buthionine (FAB) rodent model was used to model a sporadic form of AD. FAB rats displayed functional hippocampal alterations, which were accompanied by decreases in spontaneous, low-frequency oscillations and increases in the firing rates of putative pyramidal neurons. Additionally, FAB rats exhibited increases in dopamine neuron population activity and augmented responses to the locomotor-inducing effects of MK-801, as is consistent with rodent models of psychosis-like symptomatology. Further, working memory deficits in the Y-maze, consistent with an AD-like phenotype, were observed in FAB rats. These data suggest that the aberrant hippocampal activity observed in AD may contribute to dopamine-dependent psychosis, and that the FAB model may be useful for the investigation of comorbid psychosis related to AD. Understanding the pathophysiology that leads to comorbid psychosis in AD will ultimately lead to the discovery of novel targets for the treatment of this disease.
Collapse
Affiliation(s)
- Stephanie M. Perez
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA; (A.M.B.); (D.J.L.)
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| | - Angela M. Boley
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA; (A.M.B.); (D.J.L.)
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| | - Alexandra M. McCoy
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA; (A.M.B.); (D.J.L.)
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| | - Daniel J. Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA; (A.M.B.); (D.J.L.)
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| |
Collapse
|
10
|
Tang Y, Yan Y, Mao J, Ni J, Qing H. The hippocampus associated GABAergic neural network impairment in early-stage of Alzheimer's disease. Ageing Res Rev 2023; 86:101865. [PMID: 36716975 DOI: 10.1016/j.arr.2023.101865] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is the commonest neurodegenerative disease with slow progression. Pieces of evidence suggest that the GABAergic system is impaired in the early stage of AD, leading to hippocampal neuron over-activity and further leading to memory and cognitive impairment in patients with AD. However, the precise impairment mechanism of the GABAergic system on the pathogenesis of AD is still unclear. The impairment of neural networks associated with the GABAergic system is tightly associated with AD. Therefore, we describe the roles played by hippocampus-related GABAergic circuits and their impairments in AD neuropathology. In addition, we give our understand on the process from GABAergic circuit impairment to cognitive and memory impairment, since recent studies on astrocyte in AD plays an important role behind cognition dysfunction caused by GABAergic circuit impairment, which helps better understand the GABAergic system and could open up innovative AD therapy.
Collapse
Affiliation(s)
- Yuanhong Tang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Jian Mao
- Zhengzhou Tobacco Institute of China National Tobacco Company, Zhengzhou 450001, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; Department of Biology, Shenzhen MSU-BIT University, Shenzhen 518172, China.
| |
Collapse
|
11
|
Highet B, Wiseman JA, Mein H, Parker R, Ryan B, Turner CP, Jing Y, Singh-Bains MK, Liu P, Dragunow M, Faull RLM, Murray HC, Curtis MA. PSA-NCAM Regulatory Gene Expression Changes in the Alzheimer's Disease Entorhinal Cortex Revealed with Multiplexed in situ Hybridization. J Alzheimers Dis 2023; 92:371-390. [PMID: 36744342 DOI: 10.3233/jad-220986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common form of dementia and is characterized by a substantial reduction of neuroplasticity. Our previous work demonstrated that neurons involved in memory function may lose plasticity because of decreased protein levels of polysialylated neural cell adhesion molecule (PSA-NCAM) in the entorhinal cortex (EC) of the human AD brain, but the cause of this decrease is unclear. OBJECTIVE To investigate genes involved in PSA-NCAM regulation which may underlie its decrease in the AD EC. METHODS We subjected neurologically normal and AD human EC sections to multiplexed fluorescent in situ hybridization and immunohistochemistry to investigate genes involved in PSA-NCAM regulation. Gene expression changes were sought to be validated in both human tissue and a mouse model of AD. RESULTS In the AD EC, a cell population expressing a high level of CALB2 mRNA and a cell population expressing a high level of PST mRNA were both decreased. CALB2 mRNA and protein were not decreased globally, indicating that the decrease in CALB2 was specific to a sub-population of cells. A significant decrease in PST mRNA expression was observed with single-plex in situ hybridization in middle temporal gyrus tissue microarray cores from AD patients, which negatively correlated with tau pathology, hinting at global loss in PST expression across the AD brain. No significant differences in PSA-NCAM or PST protein expression were observed in the MAPT P301S mouse brain at 9 months of age. CONCLUSION We conclude that PSA-NCAM dysregulation may cause subsequent loss of structural plasticity in AD, and this may result from a loss of PST mRNA expression. Due PSTs involvement in structural plasticity, intervention for AD may be possible by targeting this disrupted plasticity pathway.
Collapse
Affiliation(s)
- Blake Highet
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - James A Wiseman
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Hannah Mein
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Remai Parker
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Brigid Ryan
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Clinton P Turner
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand.,Department of Anatomical Pathology, LabPlus, Auckland City Hospital, New Zealand
| | - Yu Jing
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Malvindar K Singh-Bains
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Ping Liu
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Helen C Murray
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland, New Zealand
| |
Collapse
|
12
|
Ali AB, Islam A, Constanti A. The fate of interneurons, GABA A receptor sub-types and perineuronal nets in Alzheimer's disease. Brain Pathol 2022; 33:e13129. [PMID: 36409151 PMCID: PMC9836378 DOI: 10.1111/bpa.13129] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurological disease, which is associated with gradual memory loss and correlated with synaptic hyperactivity and abnormal oscillatory rhythmic brain activity that precedes phenotypic alterations and is partly responsible for the spread of the disease pathology. Synaptic hyperactivity is thought to be because of alteration in the homeostasis of phasic and tonic synaptic inhibition, which is orchestrated by the GABAA inhibitory system, encompassing subclasses of interneurons and GABAA receptors, which play a vital role in cognitive functions, including learning and memory. Furthermore, the extracellular matrix, the perineuronal nets (PNNs) which often go unnoticed in considerations of AD pathology, encapsulate the inhibitory cells and neurites in critical brain regions and have recently come under the light for their crucial role in synaptic stabilisation and excitatory-inhibitory balance and when disrupted, serve as a potential trigger for AD-associated synaptic imbalance. Therefore, in this review, we summarise the current understanding of the selective vulnerability of distinct interneuron subtypes, their synaptic and extrasynaptic GABAA R subtypes as well as the changes in PNNs in AD, detailing their contribution to the mechanisms of disease development. We aim to highlight how seemingly unique malfunction in each component of the interneuronal GABA inhibitory system can be tied together to result in critical circuit dysfunction, leading to the irreversible symptomatic damage observed in AD.
Collapse
|
13
|
Agrawal H, Mehendale AM. A Review of Proteins Associated With Neuroprotection and Regeneration in Alzheimer's Disease. Cureus 2022; 14:e30412. [DOI: 10.7759/cureus.30412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022] Open
|
14
|
Katsuki F, Gerashchenko D, Brown RE. Alterations of sleep oscillations in Alzheimer's disease: A potential role for GABAergic neurons in the cortex, hippocampus, and thalamus. Brain Res Bull 2022; 187:181-198. [PMID: 35850189 DOI: 10.1016/j.brainresbull.2022.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/01/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023]
Abstract
Sleep abnormalities are widely reported in patients with Alzheimer's disease (AD) and are linked to cognitive impairments. Sleep abnormalities could be potential biomarkers to detect AD since they are often observed at the preclinical stage. Moreover, sleep could be a target for early intervention to prevent or slow AD progression. Thus, here we review changes in brain oscillations observed during sleep, their connection to AD pathophysiology and the role of specific brain circuits. Slow oscillations (0.1-1 Hz), sleep spindles (8-15 Hz) and their coupling during non-REM sleep are consistently reduced in studies of patients and in AD mouse models although the timing and magnitude of these alterations depends on the pathophysiological changes and the animal model studied. Changes in delta (1-4 Hz) activity are more variable. Animal studies suggest that hippocampal sharp-wave ripples (100-250 Hz) are also affected. Reductions in REM sleep amount and slower oscillations during REM are seen in patients but less consistently in animal models. Thus, changes in a variety of sleep oscillations could impact sleep-dependent memory consolidation or restorative functions of sleep. Recent mechanistic studies suggest that alterations in the activity of GABAergic neurons in the cortex, hippocampus and thalamic reticular nucleus mediate sleep oscillatory changes in AD and represent a potential target for intervention. Longitudinal studies of the timing of AD-related sleep abnormalities with respect to pathology and dysfunction of specific neural networks are needed to identify translationally relevant biomarkers and guide early intervention strategies to prevent or delay AD progression.
Collapse
Affiliation(s)
- Fumi Katsuki
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA.
| | - Dmitry Gerashchenko
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA
| | - Ritchie E Brown
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA 02132, USA
| |
Collapse
|
15
|
Sanchez-Varo R, Mejias-Ortega M, Fernandez-Valenzuela JJ, Nuñez-Diaz C, Caceres-Palomo L, Vegas-Gomez L, Sanchez-Mejias E, Trujillo-Estrada L, Garcia-Leon JA, Moreno-Gonzalez I, Vizuete M, Vitorica J, Baglietto-Vargas D, Gutierrez A. Transgenic Mouse Models of Alzheimer's Disease: An Integrative Analysis. Int J Mol Sci 2022; 23:5404. [PMID: 35628216 PMCID: PMC9142061 DOI: 10.3390/ijms23105404] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/10/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) constitutes the most prominent form of dementia among elderly individuals worldwide. Disease modeling using murine transgenic mice was first initiated thanks to the discovery of heritable mutations in amyloid precursor protein (APP) and presenilins (PS) genes. However, due to the repeated failure of translational applications from animal models to human patients, along with the recent advances in genetic susceptibility and our current understanding on disease biology, these models have evolved over time in an attempt to better reproduce the complexity of this devastating disease and improve their applicability. In this review, we provide a comprehensive overview about the major pathological elements of human AD (plaques, tauopathy, synaptic damage, neuronal death, neuroinflammation and glial dysfunction), discussing the knowledge that available mouse models have provided about the mechanisms underlying human disease. Moreover, we highlight the pros and cons of current models, and the revolution offered by the concomitant use of transgenic mice and omics technologies that may lead to a more rapid improvement of the present modeling battery.
Collapse
Affiliation(s)
- Raquel Sanchez-Varo
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| | - Marina Mejias-Ortega
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Juan Jose Fernandez-Valenzuela
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Cristina Nuñez-Diaz
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Laura Caceres-Palomo
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Laura Vegas-Gomez
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Elisabeth Sanchez-Mejias
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Laura Trujillo-Estrada
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Juan Antonio Garcia-Leon
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Ines Moreno-Gonzalez
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Marisa Vizuete
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Departamento Bioquimica y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, Instituto de Biomedicina de Sevilla (IBIS)-Hospital Universitario Virgen del Rocio/CSIC, 41012 Seville, Spain
| | - Javier Vitorica
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Departamento Bioquimica y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, Instituto de Biomedicina de Sevilla (IBIS)-Hospital Universitario Virgen del Rocio/CSIC, 41012 Seville, Spain
| | - David Baglietto-Vargas
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Antonia Gutierrez
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| |
Collapse
|
16
|
Carollia perspicillata: A Small Bat with Tremendous Translational Potential for Studies of Brain Aging and Neurodegeneration. Biomedicines 2021; 9:biomedicines9101454. [PMID: 34680571 PMCID: PMC8533637 DOI: 10.3390/biomedicines9101454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/04/2021] [Accepted: 10/10/2021] [Indexed: 11/30/2022] Open
Abstract
As the average human lifespan lengthens, the impact of neurodegenerative disease increases, both on the individual suffering neurodegeneration and on the community that supports those individuals. Studies aimed at understanding the mechanisms of neurodegeneration have relied heavily on observational studies of humans and experimental studies in animals, such as mice, in which aspects of brain structure and function can be manipulated to target mechanistic steps. An animal model whose brain is structurally closer to the human brain, that lives much longer than rodents, and whose husbandry is practical may be valuable for mechanistic studies that cannot readily be conducted in rodents. To demonstrate that the long-lived Seba’s short-tailed fruit bat, Carollia perspicillata, may fit this role, we used immunohistochemical labeling for NeuN and three calcium-binding proteins, calretinin, parvalbumin, and calbindin, to define hippocampal formation anatomy. Our findings demonstrate patterns of principal neuron organization that resemble primate and human hippocampal formation and patterns of calcium-binding protein distribution that help to define subregional boundaries. Importantly, we present evidence for a clear prosubiculum in the bat brain that resembles primate prosubiculum. Based on the similarities between bat and human hippocampal formation anatomy, we suggest that Carollia has unique advantages for the study of brain aging and neurodegeneration. A captive colony of Carollia allows age tracking, diet and environment control, pharmacological manipulation, and access to behavioral, physiological, anatomical, and molecular evaluation.
Collapse
|
17
|
Giesers NK, Wirths O. Loss of Hippocampal Calretinin and Parvalbumin Interneurons in the 5XFAD Mouse Model of Alzheimer's Disease. ASN Neuro 2021; 12:1759091420925356. [PMID: 32423230 PMCID: PMC7238451 DOI: 10.1177/1759091420925356] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The deposition of amyloid-β peptides in the form of extracellular plaques
and neuronal degeneration belong to the hallmark features of
Alzheimer’s disease (AD). In addition, impaired calcium homeostasis
and altered levels in calcium-binding proteins seem to be associated
with the disease process. In this study, calretinin- (CR) and
parvalbumin- (PV) positive gamma-aminobutyric acid-producing
(GABAergic) interneurons were quantified in different hippocampal
subfields of 12-month-old wild-type mice, as well as in the transgenic
AD mouse models 5XFAD and Tg4-42. While, in comparison with wild-type
mice, CR-positive interneurons were mainly reduced in the CA1 and
CA2/3 regions in plaque-bearing 5XFAD mice, PV-positive interneurons
were reduced in all analyzed subfields including the dentate gyrus. No
reduction in CR- and PV-positive interneuron numbers was detected in
the non-plaque-forming Tg4-42 mouse, although this model has been
previously demonstrated to harbor a massive loss of CA1 pyramidal
neurons. These results provide information about hippocampal
interneuron numbers in two relevant AD mouse models, suggesting that
interneuron loss in this brain region may be related to extracellular
amyloid burden.
Collapse
Affiliation(s)
- Naomi K Giesers
- Department of Psychiatry and Psychotherapy, Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| | - Oliver Wirths
- Department of Psychiatry and Psychotherapy, Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
18
|
Lauterborn JC, Scaduto P, Cox CD, Schulmann A, Lynch G, Gall CM, Keene CD, Limon A. Increased excitatory to inhibitory synaptic ratio in parietal cortex samples from individuals with Alzheimer's disease. Nat Commun 2021; 12:2603. [PMID: 33972518 PMCID: PMC8110554 DOI: 10.1038/s41467-021-22742-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Synaptic disturbances in excitatory to inhibitory (E/I) balance in forebrain circuits are thought to contribute to the progression of Alzheimer's disease (AD) and dementia, although direct evidence for such imbalance in humans is lacking. We assessed anatomical and electrophysiological synaptic E/I ratios in post-mortem parietal cortex samples from middle-aged individuals with AD (early-onset) or Down syndrome (DS) by fluorescence deconvolution tomography and microtransplantation of synaptic membranes. Both approaches revealed significantly elevated E/I ratios for AD, but not DS, versus controls. Gene expression studies in an independent AD cohort also demonstrated elevated E/I ratios in individuals with AD as compared to controls. These findings provide evidence of a marked pro-excitatory perturbation of synaptic E/I balance in AD parietal cortex, a region within the default mode network that is overly active in the disorder, and support the hypothesis that E/I imbalances disrupt cognition-related shifts in cortical activity which contribute to the intellectual decline in AD.
Collapse
Affiliation(s)
- Julie C Lauterborn
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA, USA.
| | - Pietro Scaduto
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases. School of Medicine, University of Texas Medical Branch at Galveston, Galveston, USA
| | - Conor D Cox
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA, USA
| | - Anton Schulmann
- National Institute of Mental Health, Human Genetics Branch, Bethesda, MD, USA
| | - Gary Lynch
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA, USA
- Department of Psychiatry & Human Behavior, University of California at Irvine, Irvine, CA, 92697, USA
| | - Christine M Gall
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Agenor Limon
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases. School of Medicine, University of Texas Medical Branch at Galveston, Galveston, USA.
| |
Collapse
|
19
|
Domínguez-Álvaro M, Montero-Crespo M, Blazquez-Llorca L, Plaza-Alonso S, Cano-Astorga N, DeFelipe J, Alonso-Nanclares L. 3D Analysis of the Synaptic Organization in the Entorhinal Cortex in Alzheimer's Disease. eNeuro 2021; 8:ENEURO.0504-20.2021. [PMID: 34039651 PMCID: PMC8225407 DOI: 10.1523/eneuro.0504-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/26/2021] [Accepted: 04/16/2021] [Indexed: 01/01/2023] Open
Abstract
The entorhinal cortex (EC) is especially vulnerable in the early stages of Alzheimer's disease (AD). In particular, cognitive deficits have been linked to alterations in the upper layers of EC. In the present report, we examined Layers II and III from eight human brain autopsies (four subjects with no recorded neurologic alterations and four AD cases). We used stereological methods to assess cortical atrophy of the EC and possible changes in the volume occupied by different cortical elements (neuronal and glial cell bodies; blood vessels; and neuropil). We performed 3D ultrastructural analyses of synapses using focused ion beam/scanning electron microscopy (FIB/SEM) to examine possible alterations related to AD. At the light microscope level, we found a significantly lower volume fraction occupied by neuronal bodies in Layer III and a higher volume fraction occupied by glial cell bodies in Layer II in AD cases. At the ultrastructural level, we observed that (1) there was a significantly lower synaptic density in both layers in AD cases; (2) synapses were larger and more complex in Layer II in AD cases; and (3) there was a greater proportion of small and simple synapses in Layer III in AD cases than in control individuals. These structural differences may play a role in the anatomic basis for the impairment of cognitive functions in AD.
Collapse
Affiliation(s)
- M Domínguez-Álvaro
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
| | - M Montero-Crespo
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
| | - L Blazquez-Llorca
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
- Sección Departamental de Anatomía y Embriología (Veterinaria), Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - S Plaza-Alonso
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
| | - N Cano-Astorga
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
| | - J DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
| | - L Alonso-Nanclares
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III 28031, Madrid, Spain
| |
Collapse
|
20
|
Leng K, Li E, Eser R, Piergies A, Sit R, Tan M, Neff N, Li SH, Rodriguez RD, Suemoto CK, Leite REP, Ehrenberg AJ, Pasqualucci CA, Seeley WW, Spina S, Heinsen H, Grinberg LT, Kampmann M. Molecular characterization of selectively vulnerable neurons in Alzheimer's disease. Nat Neurosci 2021; 24:276-287. [PMID: 33432193 PMCID: PMC7854528 DOI: 10.1038/s41593-020-00764-7] [Citation(s) in RCA: 278] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 11/20/2020] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is characterized by the selective vulnerability of specific neuronal populations, the molecular signatures of which are largely unknown. To identify and characterize selectively vulnerable neuronal populations, we used single-nucleus RNA sequencing to profile the caudal entorhinal cortex and the superior frontal gyrus-brain regions where neurofibrillary inclusions and neuronal loss occur early and late in AD, respectively-from postmortem brains spanning the progression of AD-type tau neurofibrillary pathology. We identified RORB as a marker of selectively vulnerable excitatory neurons in the entorhinal cortex and subsequently validated their depletion and selective susceptibility to neurofibrillary inclusions during disease progression using quantitative neuropathological methods. We also discovered an astrocyte subpopulation, likely representing reactive astrocytes, characterized by decreased expression of genes involved in homeostatic functions. Our characterization of selectively vulnerable neurons in AD paves the way for future mechanistic studies of selective vulnerability and potential therapeutic strategies for enhancing neuronal resilience.
Collapse
Affiliation(s)
- Kun Leng
- Institute for Neurodegenerative Disease, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA
| | - Emmy Li
- Institute for Neurodegenerative Disease, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Rana Eser
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Antonia Piergies
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Rene Sit
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Norma Neff
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Song Hua Li
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Roberta Diehl Rodriguez
- Department of Neurology, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil
| | - Claudia Kimie Suemoto
- Department of Pathology, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil
- Division of Geriatrics, Department of Clinical Medicine, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil
| | | | - Alexander J Ehrenberg
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Carlos A Pasqualucci
- Department of Pathology, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil
| | - William W Seeley
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Salvatore Spina
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Helmut Heinsen
- Department of Pathology, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil
- Department of Psychiatry, University of Würzburg, Würzburg, Germany
| | - Lea T Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pathology, Universidade de São Paulo, Faculdade de Medicina, São Paulo, Brazil.
- Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Martin Kampmann
- Institute for Neurodegenerative Disease, University of California, San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Niccoli T, Kerr F, Snoeren I, Fabian D, Aleyakpo B, Ivanov D, Sofola-Adesakin O, Cryar A, Adcott J, Thornton J, Partridge L. Activating transcription factor 4-dependent lactate dehydrogenase activation as a protective response to amyloid beta toxicity. Brain Commun 2021; 3:fcab053. [PMID: 33977265 PMCID: PMC8093921 DOI: 10.1093/braincomms/fcab053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 02/07/2022] [Accepted: 02/09/2021] [Indexed: 12/21/2022] Open
Abstract
Accumulation of amyloid beta peptides is thought to initiate the pathogenesis of Alzheimer's disease. However, the precise mechanisms mediating their neurotoxicity are unclear. Our microarray analyses show that, in Drosophila models of amyloid beta 42 toxicity, genes involved in the unfolded protein response and metabolic processes are upregulated in brain. Comparison with the brain transcriptome of early-stage Alzheimer's patients revealed a common transcriptional signature, but with generally opposing directions of gene expression changes between flies and humans. Among these differentially regulated genes, lactate dehydrogenase (Ldh) was up-regulated by the greatest degree in amyloid beta 42 flies and the human orthologues (LDHA and LDHB) were down-regulated in patients. Functional analyses revealed that either over-expression or inhibition of Ldh by RNA interference (RNAi) slightly exacerbated climbing defects in both healthy and amyloid beta 42-induced Drosophila. This suggests that metabolic responses to lactate dehydrogenase must be finely-tuned, and that its observed upregulation following amyloid beta 42 production could potentially represent a compensatory protection to maintain pathway homeostasis in this model, with further manipulation leading to detrimental effects. The increased Ldh expression in amyloid beta 42 flies was regulated partially by unfolded protein response signalling, as ATF4 RNAi diminished the transcriptional response and enhanced amyloid beta 42-induced climbing phenotypes. Further functional studies are required to determine whether Ldh upregulation provides compensatory neuroprotection against amyloid beta 42-induced loss of activating transcription factor 4 activity and endoplasmatic reticulum stress. Our study thus reveals dysregulation of lactate dehydrogenase signalling in Drosophila models and patients with Alzheimer's disease, which may lead to a detrimental loss of metabolic homeostasis. Importantly, we observed that down-regulation of ATF4-dependent endoplasmic reticulum-stress signalling in this context appears to prevent Ldh compensation and to exacerbate amyloid beta 42-dependent neuronal toxicity. Our findings, therefore, suggest caution in the use of therapeutic strategies focussed on down-regulation of this pathway for the treatment of Alzheimer's disease, since its natural response to the toxic peptide may induce beneficial neuroprotective effects.
Collapse
Affiliation(s)
- Teresa Niccoli
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
- Correspondence may also be addressed to: Teresa Niccoli E-mail:
| | - Fiona Kerr
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
- Department of Biological and Biomedical Sciences, School of Health & Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK
- Department of Life Sciences, School of Applied Sciences, Edinburgh Napier University, Edinburgh EH11 4BN, UK
- Correspondence may also be addressed to: Fiona Kerr Department of Life Sciences, School of Applied Sciences, Edinburgh Napier University, Sighthill Campus Edinburgh EH11 4BN, UK E-mail:
| | - Inge Snoeren
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Daniel Fabian
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Benjamin Aleyakpo
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Dobril Ivanov
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
- UK Dementia Research Institute (UKDRI), Cardiff University, Cardiff CF24 4HQ, UK
| | - Oyinkan Sofola-Adesakin
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Adam Cryar
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Jennifer Adcott
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Janet Thornton
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Linda Partridge
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
- Correspondence to: Linda Partridge Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London Darwin Building, Gower Street, London WC1E 6BT, UK E-mail:
| |
Collapse
|
22
|
Kang J, Shen J. Cell-autonomous role of Presenilin in age-dependent survival of cortical interneurons. Mol Neurodegener 2020; 15:72. [PMID: 33302995 PMCID: PMC7731773 DOI: 10.1186/s13024-020-00419-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/01/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Mutations in the PSEN1 and PSEN2 genes are the major cause of familial Alzheimer's disease. Previous studies demonstrated that Presenilin (PS), the catalytic subunit of γ-secretase, is required for survival of excitatory neurons in the cerebral cortex during aging. However, the role of PS in inhibitory interneurons had not been explored. METHODS To determine PS function in GABAergic neurons, we generated inhibitory neuron-specific PS conditional double knockout (IN-PS cDKO) mice, in which PS is selectively inactivated by Cre recombinase expressed under the control of the endogenous GAD2 promoter. We then performed behavioral, biochemical, and histological analyses to evaluate the consequences of selective PS inactivation in inhibitory neurons. RESULTS IN-PS cDKO mice exhibit earlier mortality and lower body weight despite normal food intake and basal activity. Western analysis of protein lysates from various brain sub-regions of IN-PS cDKO mice showed significant reduction of PS1 levels and dramatic accumulation of γ-secretase substrates. Interestingly, IN-PS cDKO mice develop age-dependent loss of GABAergic neurons, as shown by normal number of GAD67-immunoreactive interneurons in the cerebral cortex at 2-3 months of age but reduced number of cortical interneurons at 9 months. Moreover, age-dependent reduction of Parvalbumin- and Somatostatin-immunoreactive interneurons is more pronounced in the neocortex and hippocampus of IN-PS cDKO mice. Consistent with these findings, the number of apoptotic cells is elevated in the cerebral cortex of IN-PS cDKO mice, and the enhanced apoptosis is due to dramatic increases of apoptotic interneurons, whereas the number of apoptotic excitatory neurons is unaffected. Furthermore, progressive loss of interneurons in the cerebral cortex of IN-PS cDKO mice is accompanied with astrogliosis and microgliosis. CONCLUSION Our results together support a cell-autonomous role of PS in the survival of cortical interneurons during aging. Together with earlier studies, these findings demonstrate a universal, essential requirement of PS in the survival of both excitatory and inhibitory neurons during aging.
Collapse
Affiliation(s)
- Jongkyun Kang
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115 USA
| | - Jie Shen
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115 USA
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
23
|
Ali F, Baringer SL, Neal A, Choi EY, Kwan AC. Parvalbumin-Positive Neuron Loss and Amyloid-β Deposits in the Frontal Cortex of Alzheimer's Disease-Related Mice. J Alzheimers Dis 2020; 72:1323-1339. [PMID: 31743995 DOI: 10.3233/jad-181190] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) has several hallmark features including amyloid-β (Aβ) plaque deposits and neuronal loss. Here, we characterized Aβ plaque aggregation and parvalbumin-positive (PV) GABAergic neurons in 6-9-month-old 5xFAD mice harboring mutations associated with familial AD. We used immunofluorescence staining to compare three regions in the frontal cortex-prelimbic (PrL), cingulate (Cg, including Cg1 and Cg2), and secondary motor (M2) cortices-along with primary somatosensory (S1) cortex. We quantified the density of Aβ plaques, which showed significant laminar and regional vulnerability. There were more plaques of larger sizes in deep layers compared to superficial layers. Total plaque burden was higher in frontal regions compared to S1. We also found layer- and region-specific differences across genotype in the density of PV interneurons. PV neuron density was lower in 5xFAD mice than wild-type, particularly in deep layers of frontal regions, with Cg (-50%) and M2 (-39%) exhibiting the largest reduction. Using in vivo two-photon imaging, we longitudinally visualized the loss of frontal cortical PV neurons across four weeks in the AD mouse model. Overall, these results provide information about Aβ deposits and PV neuron density in a widely used mouse model for AD, implicating deep layers of frontal cortical regions as being especially vulnerable.
Collapse
Affiliation(s)
- Farhan Ali
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | | | - Arianna Neal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Esther Y Choi
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Alex C Kwan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
24
|
Neuron Loss in Alzheimer's Disease: Translation in Transgenic Mouse Models. Int J Mol Sci 2020; 21:ijms21218144. [PMID: 33143374 PMCID: PMC7663280 DOI: 10.3390/ijms21218144] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
Transgenic mouse models represent an essential tool for the exploration of Alzheimer’s disease (AD) pathological mechanisms and the development of novel treatments, which at present provide only symptomatic and transient effects. While a variety of mouse models successfully reflects the main neuropathological hallmarks of AD, such as extracellular amyloid-β (Aβ) deposits, intracellular accumulation of Tau protein, the development of micro- and astrogliosis, as well as behavioral deficits, substantial neuron loss, as a key feature of the disease, seems to be more difficult to achieve. In this review, we summarize information on classic and more recent transgenic mouse models for AD, focusing in particular on loss of pyramidal, inter-, and cholinergic neurons. Although the cause of neuron loss in AD is still a matter of scientific debate, it seems to be linked to intraneuronal Aβ accumulation in several transgenic mouse models, especially in pyramidal neurons.
Collapse
|
25
|
Xu Y, Zhao M, Han Y, Zhang H. GABAergic Inhibitory Interneuron Deficits in Alzheimer's Disease: Implications for Treatment. Front Neurosci 2020; 14:660. [PMID: 32714136 PMCID: PMC7344222 DOI: 10.3389/fnins.2020.00660] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized clinically by severe cognitive deficits and pathologically by amyloid plaques, neuronal loss, and neurofibrillary tangles. Abnormal amyloid β-protein (Aβ) deposition in the brain is often thought of as a major initiating factor in AD neuropathology. However, gamma-aminobutyric acid (GABA) inhibitory interneurons are resistant to Aβ deposition, and Aβ decreases synaptic glutamatergic transmission to decrease neural network activity. Furthermore, there is now evidence suggesting that neural network activity is aberrantly increased in AD patients and animal models due to functional deficits in and decreased activity of GABA inhibitory interneurons, contributing to cognitive deficits. Here we describe the roles played by excitatory neurons and GABA inhibitory interneurons in Aβ-induced cognitive deficits and how altered GABA interneurons regulate AD neuropathology. We also comprehensively review recent studies on how GABA interneurons and GABA receptors can be exploited for therapeutic benefit. GABA interneurons are an emerging therapeutic target in AD, with further clinical trials urgently warranted.
Collapse
Affiliation(s)
- Yilan Xu
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Manna Zhao
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Yuying Han
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Heng Zhang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
26
|
Ubeda-Bañon I, Saiz-Sanchez D, Flores-Cuadrado A, Rioja-Corroto E, Gonzalez-Rodriguez M, Villar-Conde S, Astillero-Lopez V, Cabello-de la Rosa JP, Gallardo-Alcañiz MJ, Vaamonde-Gamo J, Relea-Calatayud F, Gonzalez-Lopez L, Mohedano-Moriano A, Rabano A, Martinez-Marcos A. The human olfactory system in two proteinopathies: Alzheimer's and Parkinson's diseases. Transl Neurodegener 2020; 9:22. [PMID: 32493457 PMCID: PMC7271529 DOI: 10.1186/s40035-020-00200-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/20/2020] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's and Parkinson's diseases are the most prevalent neurodegenerative disorders. Their etiologies are idiopathic, and treatments are symptomatic and orientated towards cognitive or motor deficits. Neuropathologically, both are proteinopathies with pathological aggregates (plaques of amyloid-β peptide and neurofibrillary tangles of tau protein in Alzheimer's disease, and Lewy bodies mostly composed of α-synuclein in Parkinson's disease). These deposits appear in the nervous system in a predictable and accumulative sequence with six neuropathological stages. Both disorders present a long prodromal period, characterized by preclinical signs including hyposmia. Interestingly, the olfactory system, particularly the anterior olfactory nucleus, is initially and preferentially affected by the pathology. Cerebral atrophy revealed by magnetic resonance imaging must be complemented by histological analyses to ascertain whether neuronal and/or glial loss or neuropil remodeling are responsible for volumetric changes. It has been proposed that these proteinopathies could act in a prion-like manner in which a misfolded protein would be able to force native proteins into pathogenic folding (seeding), which then propagates through neurons and glia (spreading). Existing data have been examined to establish why some neuronal populations are vulnerable while others are resistant to pathology and to what extent glia prevent and/or facilitate proteinopathy spreading. Connectomic approaches reveal a number of hubs in the olfactory system (anterior olfactory nucleus, olfactory entorhinal cortex and cortical amygdala) that are key interconnectors with the main hubs (the entorhinal-hippocampal-cortical and amygdala-dorsal motor vagal nucleus) of network dysfunction in Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Isabel Ubeda-Bañon
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Daniel Saiz-Sanchez
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Alicia Flores-Cuadrado
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Ernesto Rioja-Corroto
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Melania Gonzalez-Rodriguez
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Sandra Villar-Conde
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Veronica Astillero-Lopez
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | | | | | - Julia Vaamonde-Gamo
- Neurology Service, Ciudad Real General University Hospital, 13005 Ciudad Real, Spain
| | | | - Lucia Gonzalez-Lopez
- Pathology Service, Ciudad Real General University Hospital, 13005 Ciudad Real, Spain
| | | | - Alberto Rabano
- Neuropathology Department and Tissue Bank, CIEN Foundation, Carlos III Health Institute, Madrid, Spain
| | - Alino Martinez-Marcos
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| |
Collapse
|
27
|
Waller R, Mandeya M, Viney E, Simpson JE, Wharton SB. Histological characterization of interneurons in Alzheimer's disease reveals a loss of somatostatin interneurons in the temporal cortex. Neuropathology 2020; 40:336-346. [PMID: 32232904 DOI: 10.1111/neup.12649] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 11/28/2022]
Abstract
Neuronal dysfunction and synaptic loss are major hallmarks of Alzheimer's disease (AD) which correlate with symptom severity. Impairment of the γ-aminobutyric acid (GABA)ergic inhibitory interneurons, which form around 20% of the total neuronal network, may be an early event contributing to neuronal circuit dysfunction in neurodegenerative diseases. This study examined the expression of two of the main classes of inhibitory interneurons, parvalbumin (PV) and somatostatin (SST) interneurons in the temporal cortex and hippocampus of AD and control cases, using immunohistochemistry. We report a significant regional variation in the number of PV and SST interneurons with a higher number identified per mm2 in the temporal cortex compared to the hippocampus. Fewer SST interneurons, but not PV interneurons, were identified per mm2 in the temporal cortex of AD cases compared to control subjects. Our results support regional neuroanatomical effects on selective interneuron classes in AD, and suggest that impairment of the interneuronal circuit may contribute to neuronal dysfunction and cognitive decline in AD.
Collapse
Affiliation(s)
- Rachel Waller
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - Memory Mandeya
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - Edward Viney
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| |
Collapse
|
28
|
Shi A, Petrache AL, Shi J, Ali AB. Preserved Calretinin Interneurons in an App Model of Alzheimer's Disease Disrupt Hippocampal Inhibition via Upregulated P2Y1 Purinoreceptors. Cereb Cortex 2020; 30:1272-1290. [PMID: 31407772 PMCID: PMC7132926 DOI: 10.1093/cercor/bhz165] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/15/2019] [Accepted: 06/24/2019] [Indexed: 12/12/2022] Open
Abstract
To understand the pathogenesis of specific neuronal circuit dysfunction in Alzheimer's disease (AD), we investigated the fate of three subclasses of "modulatory interneurons" in hippocampal CA1 using the AppNL-F/NL-F knock-in mouse model of AD. Cholecystokinin- and somatostatin-expressing interneurons were aberrantly hyperactive preceding the presence of the typical AD hallmarks: neuroinflammation and amyloid-β (Aβ) accumulation. These interneurons showed an age-dependent vulnerability to Aβ penetration and a reduction in density and coexpression of the inhibitory neurotransmitter GABA synthesis enzyme, glutamic acid decarboxylase 67 (GAD67), suggesting a loss in their inhibitory function. However, calretinin (CR) interneurons-specialized to govern only inhibition, showed resilience to Aβ accumulation, preservation of structure, and displayed synaptic hyperinhibition, despite the lack of inhibitory control of CA1 excitatory pyramidal cells from midstages of the disease. This aberrant inhibitory homeostasis observed in CA1 CR cells and pyramidal cells was "normalized" by blocking P2Y1 purinoreceptors, which were "upregulated" and strongly expressed in CR cells and astrocytes in AppNL-F/NL-F mice in the later stages of AD. In summary, AD-associated cell-type selective destruction of inhibitory interneurons and disrupted inhibitory homeostasis rectified by modulation of the upregulated purinoreceptor system may serve as a novel therapeutic strategy to normalize selective dysfunctional synaptic homeostasis during pathogenesis of AD.
Collapse
Affiliation(s)
- Anqi Shi
- UCL School of Pharmacy, London, WC1N 1AX, UK
| | | | - Jiachen Shi
- UCL School of Pharmacy, London, WC1N 1AX, UK
| | - Afia B Ali
- UCL School of Pharmacy, London, WC1N 1AX, UK
| |
Collapse
|
29
|
Kumar D, Sharma A, Sharma L. A Comprehensive Review of Alzheimer's Association with Related Proteins: Pathological Role and Therapeutic Significance. Curr Neuropharmacol 2020; 18:674-695. [PMID: 32172687 PMCID: PMC7536827 DOI: 10.2174/1570159x18666200203101828] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/26/2019] [Accepted: 01/31/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's is an insidious, progressive, chronic neurodegenerative disease which causes the devastation of neurons. Alzheimer's possesses complex pathologies of heterogeneous nature counting proteins as one major factor along with enzymes and mutated genes. Proteins such as amyloid precursor protein (APP), apolipoprotein E (ApoE), presenilin, mortalin, calbindin-D28K, creactive protein, heat shock proteins (HSPs), and prion protein are some of the chief elements in the foremost hypotheses of AD like amyloid-beta (Aβ) cascade hypothesis, tau hypothesis, cholinergic neuron damage, etc. Disturbed expression of these proteins results in synaptic dysfunction, cognitive impairment, memory loss, and neuronal degradation. On the therapeutic ground, attempts of developing anti-amyloid, anti-inflammatory, anti-tau therapies are on peak, having APP and tau as putative targets. Some proteins, e.g., HSPs, which ameliorate oxidative stress, calpains, which help in regulating synaptic plasticity, and calmodulin-like skin protein (CLSP) with its neuroprotective role are few promising future targets for developing anti-AD therapies. On diagnostic grounds of AD C-reactive protein, pentraxins, collapsin response mediator protein-2, and growth-associated protein-43 represent the future of new possible biomarkers for diagnosing AD. The last few decades were concentrated over identifying and studying protein targets of AD. Here, we reviewed the physiological/pathological roles and therapeutic significance of nearly all the proteins associated with AD that addresses putative as well as probable targets for developing effective anti-AD therapies.
Collapse
Affiliation(s)
- Deepak Kumar
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P. India
| | - Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P. India
| | - Lalit Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P. India
| |
Collapse
|
30
|
Sanchez-Mejias E, Nuñez-Diaz C, Sanchez-Varo R, Gomez-Arboledas A, Garcia-Leon JA, Fernandez-Valenzuela JJ, Mejias-Ortega M, Trujillo-Estrada L, Baglietto-Vargas D, Moreno-Gonzalez I, Davila JC, Vitorica J, Gutierrez A. Distinct disease-sensitive GABAergic neurons in the perirhinal cortex of Alzheimer's mice and patients. Brain Pathol 2019; 30:345-363. [PMID: 31491047 PMCID: PMC7064898 DOI: 10.1111/bpa.12785] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 09/02/2019] [Indexed: 12/29/2022] Open
Abstract
Neuronal loss is the best neuropathological substrate that correlates with cortical atrophy and dementia in Alzheimer's disease (AD). Defective GABAergic neuronal functions may lead to cortical network hyperactivity and aberrant neuronal oscillations and in consequence, generate a detrimental alteration in memory processes. In this study, using immunohistochemical and stereological approaches, we report that the two major and non-overlapping groups of inhibitory interneurons (SOM-cells and PV-cells) displayed distinct vulnerability in the perirhinal cortex of APP/PS1 mice and AD patients. SOM-positive neurons were notably sensitive and exhibited a dramatic decrease in the perirhinal cortex of 6-month-old transgenic mice (57% and 61% in areas 36 and 35, respectively) and, most importantly, in AD patients (91% in Braak V-VI cases). In addition, this interneuron degenerative process seems to occur in parallel, and closely related, with the progression of the amyloid pathology. However, the population expressing PV was unaffected in APP/PS1 mice while in AD brains suffered a pronounced and significant loss (69%). As a key component of cortico-hippocampal networks, the perirhinal cortex plays an important role in memory processes, especially in familiarity-based memory recognition. Therefore, disrupted functional connectivity of this cortical region, as a result of the early SOM and PV neurodegeneration, might contribute to the altered brain rhythms and cognitive failures observed in the initial clinical phase of AD patients. Finally, these findings highlight the failure of amyloidogenic AD models to fully recapitulate the selective neuronal degeneration occurring in humans.
Collapse
Affiliation(s)
- Elisabeth Sanchez-Mejias
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Cristina Nuñez-Diaz
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Raquel Sanchez-Varo
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Angela Gomez-Arboledas
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Antonio Garcia-Leon
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Jose Fernandez-Valenzuela
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Marina Mejias-Ortega
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Laura Trujillo-Estrada
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - David Baglietto-Vargas
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ines Moreno-Gonzalez
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jose Carlos Davila
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier Vitorica
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Dpto. Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain.,Instituto de Biomedicina de Sevilla (IBIS)-Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Antonia Gutierrez
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
31
|
Kobro-Flatmoen A, Witter MP. Neuronal chemo-architecture of the entorhinal cortex: A comparative review. Eur J Neurosci 2019; 50:3627-3662. [PMID: 31293027 DOI: 10.1111/ejn.14511] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 06/18/2019] [Accepted: 06/27/2019] [Indexed: 11/29/2022]
Abstract
The identification of neuronal markers, that is, molecules selectively present in subsets of neurons, contributes to our understanding of brain areas and the networks within them. Specifically, recognizing the distribution of different neuronal markers facilitates the identification of borders between functionally distinct brain areas. Detailed knowledge about the localization and physiological significance of neuronal markers may also provide clues to generate new hypotheses concerning aspects of normal and abnormal brain functioning. Here, we provide a comprehensive review on the distribution within the entorhinal cortex of neuronal markers and the morphology of the neurons they reveal. Emphasis is on the comparative distribution of several markers, with a focus on, but not restricted to rodent, monkey and human data, allowing to infer connectional features, across species, associated with these markers, based on what is revealed by mainly rodent data. The overall conclusion from this review is that there is an emerging pattern in the distribution of neuronal markers in the entorhinal cortex when aligning data along a comparable coordinate system in various species.
Collapse
Affiliation(s)
- Asgeir Kobro-Flatmoen
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Menno P Witter
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
32
|
Vico Varela E, Etter G, Williams S. Excitatory-inhibitory imbalance in Alzheimer's disease and therapeutic significance. Neurobiol Dis 2019; 127:605-615. [DOI: 10.1016/j.nbd.2019.04.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 11/29/2022] Open
|
33
|
Unsupervised excitation: GABAergic dysfunctions in Alzheimer’s disease. Brain Res 2019; 1707:216-226. [DOI: 10.1016/j.brainres.2018.11.042] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/31/2018] [Accepted: 11/27/2018] [Indexed: 12/22/2022]
|
34
|
Höfling C, Shehabi E, Kuhn PH, Lichtenthaler SF, Hartlage-Rübsamen M, Roßner S. Cell Type-Specific Human APP Transgene Expression by Hippocampal Interneurons in the Tg2576 Mouse Model of Alzheimer's Disease. Front Neurosci 2019; 13:137. [PMID: 30853883 PMCID: PMC6395433 DOI: 10.3389/fnins.2019.00137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/06/2019] [Indexed: 01/21/2023] Open
Abstract
Amyloid precursor protein (APP) transgenic animal models of Alzheimer’s disease have become versatile tools for basic and translational research. However, there is great heterogeneity of histological, biochemical, and functional data between transgenic mouse lines, which might be due to different transgene expression patterns. Here, the expression of human APP (hAPP) by GABAergic hippocampal interneurons immunoreactive for the calcium binding proteins parvalbumin, calbindin, calretinin, and for the peptide hormone somatostatin was analyzed in Tg2576 mice by double immunofluorescent microscopy. Overall, there was no GABAergic interneuron subpopulation that did not express the transgene. On the other hand, in no case all neurons of such a subpopulation expressed hAPP. In dentate gyrus molecular layer and in stratum lacunosum moleculare less than 10% of hAPP-positive interneurons co-express any of these interneuron markers, whereas in stratum oriens hAPP-expressing neurons frequently co-express these interneuron markers to different proportions. We conclude that these neurons differentially contribute to deficits in young Tg2576 mice before the onset of Abeta plaque pathology. The detailed analysis of distinct brain region and neuron type-specific APP transgene expression patterns is indispensable to understand particular pathological features and mouse line-specific differences in neuronal and systemic functions.
Collapse
Affiliation(s)
- Corinna Höfling
- Paul-Flechsig-Institute for Brain Research, Leipzig University, Leipzig, Germany
| | - Emira Shehabi
- Paul-Flechsig-Institute for Brain Research, Leipzig University, Leipzig, Germany
| | - Peer-Hendrik Kuhn
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | | | - Steffen Roßner
- Paul-Flechsig-Institute for Brain Research, Leipzig University, Leipzig, Germany
| |
Collapse
|
35
|
Early alterations in hippocampal perisomatic GABAergic synapses and network oscillations in a mouse model of Alzheimer's disease amyloidosis. PLoS One 2019; 14:e0209228. [PMID: 30645585 PMCID: PMC6333398 DOI: 10.1371/journal.pone.0209228] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/30/2018] [Indexed: 01/01/2023] Open
Abstract
Several lines of evidence imply changes in inhibitory interneuron connectivity and subsequent alterations in oscillatory network activities in the pathogenesis of Alzheimer’s Disease (AD). Recently, we provided evidence for an increased immunoreactivity of both the postsynaptic scaffold protein gephyrin and the GABAA receptor γ2-subunit in the hippocampus of young (1 and 3 months of age), APPPS1 mice. These mice represent a well-established model of cerebral amyloidosis, which is a hallmark of human AD. In this study, we demonstrate a robust increase of parvalbumin immunoreactivity and accentuated projections of parvalbumin positive (PV+) interneurons, which target perisomatic regions of pyramidal cells within the hippocampal subregions CA1 and CA3 of 3-month-old APPPS1 mice. Colocalisation studies confirmed a significant increase in the density of PV+ projections labeled with antibodies against a presynaptic (vesicular GABA transporter) and a postsynaptic marker (gephyrin) of inhibitory synapses within the pyramidal cell layer of CA1 and CA3. As perisomatic inhibition by PV+-interneurons is crucial for the generation of hippocampal network oscillations involved in spatial processing, learning and memory formation we investigated the impact of the putative enhanced perisomatic inhibition on two types of fast neuronal network oscillations in acute hippocampal slices: 1. spontaneously occurring sharp wave-ripple complexes (SPW-R), and 2. cholinergic γ-oscillations. Interestingly, both network patterns were generally preserved in APPPS1 mice similar to WT mice. However, the comparison of simultaneous CA3 and CA1 recordings revealed that the incidence and amplitude of SPW-Rs were significantly lower in CA1 vs CA3 in APPPS1 slices, whereas the power of γ-oscillations was significantly higher in CA3 vs CA1 in WT-slices indicating an impaired communication between the CA3 and CA1 network activities in APPPS1 mice. Taken together, our data demonstrate an increased GABAergic synaptic output of PV+ interneurons impinging on pyramidal cells of CA1 and CA3, which might limit the coordinated cross-talk between these two hippocampal areas in young APPPS1 mice and mediate long-term changes in synaptic inhibition during progression of amyloidosis.
Collapse
|
36
|
Zallo F, Gardenal E, Verkhratsky A, Rodríguez JJ. Loss of calretinin and parvalbumin positive interneurones in the hippocampal CA1 of aged Alzheimer's disease mice. Neurosci Lett 2018; 681:19-25. [PMID: 29782955 DOI: 10.1016/j.neulet.2018.05.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/30/2018] [Accepted: 05/17/2018] [Indexed: 01/19/2023]
Abstract
Neuronal degeneration associated with Alzheimer's disease (AD), is linked to impaired calcium homeostasis and to changes in calcium-binding proteins (CBPs). The AD-related modification of neuronal CBPs remains controversial. Here we analysed the presence and expression of calretinin (CR) and parvalbumin (PV) in the hippocampal CA1 neurones of 18 months old 3xTg-AD mice compared to non-Tg animals. We found a layer specific decrease in number of interneurones expressing CR and PV (by 33.7% and 52%, respectively). Expression of PV decreased (by 13.8%) in PV-positive neurones, whereas expression of CR did not change in CR positive cells. The loss of specific subpopulations of Ca2+-binding proteins expressing interneurones (CR and PV) together with the decrease of PV in the surviving cells may be linked to their vulnerability to AD pathology. Specific loss of inhibitory interneurones with age could contribute to overall increase in the network excitability associated with AD.
Collapse
Affiliation(s)
- Fatima Zallo
- BioCruces Health Research Institute, 48903, Barakaldo, Spain; Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Spain
| | - Emanuela Gardenal
- Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Spain; Human Histology and Embryology Unit, Medical School, University of Verona, 37134, Verona, Italy
| | - Alexei Verkhratsky
- Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Spain; IKERBASQUE, Basque Foundation for Science, 48013-Bilbao, Medical School, Spain; Achúcarro Basque Center for Neuroscience, 48940, Leioa, Spain; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, United Kingdom; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - José Julio Rodríguez
- BioCruces Health Research Institute, 48903, Barakaldo, Spain; Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Spain; IKERBASQUE, Basque Foundation for Science, 48013-Bilbao, Medical School, Spain.
| |
Collapse
|
37
|
Neurochemical Characterization of PSA-NCAM + Cells in the Human Brain and Phenotypic Quantification in Alzheimer’s Disease Entorhinal Cortex. Neuroscience 2018; 372:289-303. [DOI: 10.1016/j.neuroscience.2017.12.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/22/2017] [Accepted: 12/15/2017] [Indexed: 01/07/2023]
|
38
|
Kitamura T. Driving and regulating temporal association learning coordinated by entorhinal-hippocampal network. Neurosci Res 2017; 121:1-6. [DOI: 10.1016/j.neures.2017.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 03/16/2017] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
|
39
|
Klein AS, Donoso JR, Kempter R, Schmitz D, Beed P. Early Cortical Changes in Gamma Oscillations in Alzheimer's Disease. Front Syst Neurosci 2016; 10:83. [PMID: 27833535 PMCID: PMC5080538 DOI: 10.3389/fnsys.2016.00083] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 10/14/2016] [Indexed: 12/19/2022] Open
Abstract
The entorhinal cortices in the temporal lobe of the brain are key structures relaying memory related information between the neocortex and the hippocampus. The medial entorhinal cortex (MEC) routes spatial information, whereas the lateral entorhinal cortex (LEC) routes predominantly olfactory information to the hippocampus. Gamma oscillations are known to coordinate information transfer between brain regions by precisely timing population activity of neuronal ensembles. Here, we studied the organization of in vitro gamma oscillations in the MEC and LEC of the transgenic (tg) amyloid precursor protein (APP)-presenilin 1 (PS1) mouse model of Alzheimer’s Disease (AD) at 4–5 months of age. In vitro gamma oscillations using the kainate model peaked between 30–50 Hz and therefore we analyzed the oscillatory properties in the 20–60 Hz range. Our results indicate that the LEC shows clear alterations in frequency and power of gamma oscillations at an early stage of AD as compared to the MEC. The gamma-frequency oscillation slows down in the LEC and also the gamma power in dorsal LEC is decreased as early as 4–5 months in the tg APP-PS1 mice. The results of this study suggest that the timing of olfactory inputs from LEC to the hippocampus might be affected at an early stage of AD, resulting in a possible erroneous integration of the information carried by the two input pathways to the hippocampal subfields.
Collapse
Affiliation(s)
| | - José R Donoso
- Department of Biology, Institute for Theoretical Biology, Humboldt UniversityBerlin, Germany; Bernstein Center for Computational NeuroscienceBerlin, Germany
| | - Richard Kempter
- Department of Biology, Institute for Theoretical Biology, Humboldt UniversityBerlin, Germany; Bernstein Center for Computational NeuroscienceBerlin, Germany
| | - Dietmar Schmitz
- Neuroscience Research Center, Charité UniversityBerlin, Germany; Bernstein Center for Computational NeuroscienceBerlin, Germany; Cluster of Excellence "NeuroCure", Charité UniversityBerlin, Germany; DZNE - German Center for Neurodegenerative DiseasesBerlin, Germany; Einstein Foundation BerlinBerlin, Germany
| | - Prateep Beed
- Neuroscience Research Center, Charité UniversityBerlin, Germany; DZNE - German Center for Neurodegenerative DiseasesBerlin, Germany; Berlin Institute of HealthBerlin, Germany
| |
Collapse
|
40
|
Żakowski W. Neurochemistry of the Anterior Thalamic Nuclei. Mol Neurobiol 2016; 54:5248-5263. [DOI: 10.1007/s12035-016-0077-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/23/2016] [Indexed: 01/19/2023]
|
41
|
De la Rosa-Prieto C, Saiz-Sanchez D, Ubeda-Banon I, Flores-Cuadrado A, Martinez-Marcos A. Neurogenesis, Neurodegeneration, Interneuron Vulnerability, and Amyloid-β in the Olfactory Bulb of APP/PS1 Mouse Model of Alzheimer's Disease. Front Neurosci 2016; 10:227. [PMID: 27303258 PMCID: PMC4885141 DOI: 10.3389/fnins.2016.00227] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 05/06/2016] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease, mostly idiopathic and with palliative treatment. Neuropathologically, it is characterized by intracellular neurofibrillary tangles of tau protein and extracellular plaques of amyloid β peptides. The relationship between AD and neurogenesis is unknown, but two facts are particularly relevant. First, early aggregation sites of both proteinopathies include the hippocampal formation and the olfactory bulb (OB), which have been correlated to memory and olfactory deficits, respectively. These areas are well-recognized integration zones of newly-born neurons in the adult brain. Second, molecules, such as amyloid precursor protein (APP) and presenilin-1 are common to both AD etiology and neurogenic development. Adult neurogenesis in AD models has been studied in the hippocampus, but only occasionally addressed in the OB and results are contradictory. To gain insight on the relationship between adult neurogenesis and AD, this work analyzes neurogenesis, neurodegeneration, interneuron vulnerability, and amyloid-β involvement in the OB of an AD model. Control and double-transgenic mice carrying the APP and the presenilin-1 genes, which give rise amyloid β plaques have been used. BrdU-treated animals have been studied at 16, 30, 43, and 56 weeks of age. New-born cell survival (BrdU), neuronal loss (using neuronal markers NeuN and PGP9.5), differential interneuron (calbindin-, parvalbumin-, calretinin- and somatostatin-expressing populations) vulnerability, and involvement by amyloid β have been analyzed. Neurogenesis increases with aging in the granule cell layer of control animals from 16 to 43 weeks. No neuronal loss has been observed after quantifying NeuN or PGP9.5. Regarding interneuron population vulnerability: calbindin-expressing neurons remains unchanged; parvalbumin-expressing neurons trend to increase with aging in transgenic animals; calretinin-expressing neurons increase with aging in transgenic mice and decrease in control animals and neurogenesis is higher in control as compared to transgenic animals at given ages, finally; somatostatin-expressing neurons of transgenic mice decrease with aging and as compared to controls. Amyloid β aggregates with aging in the granule cell layer, which may be related to the particular involvement of somatostatin-expressing cells.
Collapse
Affiliation(s)
- Carlos De la Rosa-Prieto
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical School, Universidad de Castilla-La Mancha Ciudad Real, Spain
| | - Daniel Saiz-Sanchez
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical School, Universidad de Castilla-La Mancha Ciudad Real, Spain
| | - Isabel Ubeda-Banon
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical School, Universidad de Castilla-La Mancha Ciudad Real, Spain
| | - Alicia Flores-Cuadrado
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical School, Universidad de Castilla-La Mancha Ciudad Real, Spain
| | - Alino Martinez-Marcos
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical School, Universidad de Castilla-La Mancha Ciudad Real, Spain
| |
Collapse
|
42
|
Saiz-Sanchez D, Flores-Cuadrado A, Ubeda-Bañon I, de la Rosa-Prieto C, Martinez-Marcos A. Interneurons in the human olfactory system in Alzheimer's disease. Exp Neurol 2015; 276:13-21. [PMID: 26616239 DOI: 10.1016/j.expneurol.2015.11.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/12/2015] [Accepted: 11/21/2015] [Indexed: 01/09/2023]
Abstract
The principal olfactory structures display Alzheimer's disease (AD) related pathology at early stages of the disease. Consequently, olfactory deficits are among the earliest symptoms. Reliable olfactory tests for accurate clinical diagnosis are rarely made. In addition, neuropathological analysis postmortem of olfactory structures is often not made. Therefore, the relationship between the clinical features and the underlying pathology is poorly defined. Traditionally, research into Alzheimer's disease has focused on the degeneration of cortical temporal projection neurons and cholinergic neurons. Recent evidence has demonstrated the neurodegeneration of interneuron populations in AD. This review provides an updated overview of the pathological involvement of interneuron populations in the human olfactory system in Alzheimer's disease.
Collapse
Affiliation(s)
- Daniel Saiz-Sanchez
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Alicia Flores-Cuadrado
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Isabel Ubeda-Bañon
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Carlos de la Rosa-Prieto
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Alino Martinez-Marcos
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, 13005 Ciudad Real, Spain.
| |
Collapse
|
43
|
Vulnerability of calbindin, calretinin and parvalbumin in a transgenic/knock-in APPswe/PS1dE9 mouse model of Alzheimer disease together with disruption of hippocampal neurogenesis. Exp Gerontol 2015; 69:176-88. [DOI: 10.1016/j.exger.2015.06.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 06/16/2015] [Accepted: 06/18/2015] [Indexed: 11/19/2022]
|
44
|
Saiz-Sanchez D, De la Rosa-Prieto C, Ubeda-Banon I, Martinez-Marcos A. Interneurons, tau and amyloid-β in the piriform cortex in Alzheimer's disease. Brain Struct Funct 2014; 220:2011-25. [PMID: 24748561 DOI: 10.1007/s00429-014-0771-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 04/02/2014] [Indexed: 12/14/2022]
Abstract
Impaired olfaction has been described as an early symptom of Alzheimer's disease. Neuroanatomical changes underlying this deficit in the olfactory system are largely unknown. Interestingly, neuropathology begins in the transentorhinal cortex and extends to the neighboring limbic system and basal telencephalic structures that mediate olfactory processing, including the anterior olfactory nucleus and olfactory bulb. The human piriform cortex has been described as a crucial area in odor quality coding; disruption of this region mediates early olfactory deficits in Alzheimer's disease. Most neuropathological investigations have focused on the entorhinal cortex and hippocampus, whereas the piriform cortex has largely been neglected. This work aims to characterize the expression of the neuropathological amyloid-β peptide, tau protein and interneuron population markers (calretinin, parvalbumin and somatostatin) in the piriform cortex of ten Alzheimer-diagnosed (80.4 ± 8.3 years old) and five control (69.6 ± 11.1) cases. Here, we examined the distribution of different interneuronal markers as well as co-localization of interneurons and pathological markers. Results indicated preferential vulnerability of somatostatin- (p = 0.0001 < α = 0.05) and calretinin-positive (p = 0.013 < α = 0.05) cells that colocalized with amyloid-β peptide, while the prevalence of parvalbumin-positive cells was increased (p = 0.045 < α = 0.05) in the Alzheimer's cases. These data may help to reveal the neural basis of olfactory deficits linked to Alzheimer's disease as well as to characterize neuronal populations preferentially vulnerable to neuropathology in regions critically involved in early stages of the disease.
Collapse
Affiliation(s)
- Daniel Saiz-Sanchez
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Avda. de Moledores s/n, 13071, Ciudad Real, Spain
| | | | | | | |
Collapse
|
45
|
Pretreatment with apoaequorin protects hippocampal CA1 neurons from oxygen-glucose deprivation. PLoS One 2013; 8:e79002. [PMID: 24244400 PMCID: PMC3823939 DOI: 10.1371/journal.pone.0079002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 09/26/2013] [Indexed: 12/26/2022] Open
Abstract
Ischemic stroke affects ∼795,000 people each year in the U.S., which results in an estimated annual cost of $73.7 billion. Calcium is pivotal in a variety of neuronal signaling cascades, however, during ischemia, excess calcium influx can trigger excitotoxic cell death. Calcium binding proteins help neurons regulate/buffer intracellular calcium levels during ischemia. Aequorin is a calcium binding protein isolated from the jellyfish Aequorea victoria, and has been used for years as a calcium indicator, but little is known about its neuroprotective properties. The present study used an in vitro rat brain slice preparation to test the hypothesis that an intra-hippocampal infusion of apoaequorin (the calcium binding component of aequorin) protects neurons from ischemic cell death. Bilaterally cannulated rats received an apoaequorin infusion in one hemisphere and vehicle control in the other. Hippocampal slices were then prepared and subjected to 5 minutes of oxygen-glucose deprivation (OGD), and cell death was assayed by trypan blue exclusion. Apoaequorin dose-dependently protected neurons from OGD--doses of 1% and 4% (but not 0.4%) significantly decreased the number of trypan blue-labeled neurons. This effect was also time dependent, lasting up to 48 hours. This time dependent effect was paralleled by changes in cytokine and chemokine expression, indicating that apoaequorin may protect neurons via a neuroimmunomodulatory mechanism. These data support the hypothesis that pretreatment with apoaequorin protects neurons against ischemic cell death, and may be an effective neurotherapeutic.
Collapse
|
46
|
Zakowski W, Bogus-Nowakowska K, Robak A. Embryonic and postnatal development of calcium-binding proteins immunoreactivity in the anterior thalamus of the guinea pig. J Chem Neuroanat 2013; 53:25-32. [PMID: 24060609 DOI: 10.1016/j.jchemneu.2013.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/20/2013] [Accepted: 09/15/2013] [Indexed: 10/26/2022]
Abstract
Our recent studies have shown that the distribution of calretinin (CR) in the anterior thalamic nuclei (ATN) changes significantly during the development of the guinea pig. The present study was designed to reveal the distribution pattern of calcium-binding proteins, i.e. calbindin (CB) and parvalbumin (PV), as well as the colocalization pattern of all three proteins, including CR, in the ATN of guinea pigs ranging from the 40th embryonic day (E40) to the 80th postnatal day (P80). According to these patterns, CB appears exclusively in the perikarya of the anteromedial nucleus (AM) not before P20 and always colocalizes with CR. Moreover, CB and CR colocalize in fibers of thin bundles traversing the anteroventral nucleus (AV) since E50. The ATN also display CB-positive neuropil in all studied stages, especially a strong one in the ventral part of the AV. PV was not observed in the perikarya of the ATN in all the stages, but was abundantly present in the neuropil of the anterodorsal nucleus (AD). No colocalizations exist between PV and the rest of the studied proteins. In conclusion, our study reveals that the distribution of the studied proteins differs greatly. Nevertheless, the postnatal coexistence of CB and CR in the AM perikarya may indicate the cooperation of both of the proteins in some functions of the nucleus. Parvalbumin is limited mostly to the neuropil of the AD, suggesting different functions in comparison to CB and CR.
Collapse
Affiliation(s)
- Witold Zakowski
- Department of Comparative Anatomy, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Pl. Łódzki 3, 10-727 Olsztyn, Poland.
| | | | | |
Collapse
|
47
|
Wilson DIG, Watanabe S, Milner H, Ainge JA. Lateral entorhinal cortex is necessary for associative but not nonassociative recognition memory. Hippocampus 2013; 23:1280-90. [PMID: 23836525 PMCID: PMC4030623 DOI: 10.1002/hipo.22165] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/20/2013] [Accepted: 06/24/2013] [Indexed: 12/28/2022]
Abstract
The lateral entorhinal cortex (LEC) provides one of the two major input pathways to the hippocampus and has been suggested to process the nonspatial contextual details of episodic memory. Combined with spatial information from the medial entorhinal cortex it is hypothesised that this contextual information is used to form an integrated spatially selective, context-specific response in the hippocampus that underlies episodic memory. Recently, we reported that the LEC is required for recognition of objects that have been experienced in a specific context (Wilson et al. (2013) Hippocampus 23:352-366). Here, we sought to extend this work to assess the role of the LEC in recognition of all associative combinations of objects, places and contexts within an episode. Unlike controls, rats with excitotoxic lesions of the LEC showed no evidence of recognizing familiar combinations of object in place, place in context, or object in place and context. However, LEC lesioned rats showed normal recognition of objects and places independently from each other (nonassociative recognition). Together with our previous findings, these data suggest that the LEC is critical for associative recognition memory and may bind together information relating to objects, places, and contexts needed for episodic memory formation.
Collapse
Affiliation(s)
- David I G Wilson
- School of Psychology and Neuroscience, University of St Andrews, St Mary's Quad, St Andrews, Fife, United Kingdom
| | | | | | | |
Collapse
|
48
|
Saiz-Sanchez D, De La Rosa-Prieto C, Ubeda-Bañon I, Martinez-Marcos A. Interneurons and beta-amyloid in the olfactory bulb, anterior olfactory nucleus and olfactory tubercle in APPxPS1 transgenic mice model of Alzheimer's disease. Anat Rec (Hoboken) 2013; 296:1413-23. [PMID: 23904197 DOI: 10.1002/ar.22750] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 06/18/2013] [Indexed: 01/29/2023]
Abstract
Impaired olfaction has been described as an early symptom in Alzheimer's disease (AD). Neuroanatomical changes underlying this deficit in the olfactory system are largely unknown. Given that interneuron populations are crucial in olfactory information processing, we have quantitatively analyzed somatostatin- (SOM), parvalbumin- (PV), and calretinin-expressing (CR) cells in the olfactory bulb, anterior olfactory nucleus, and olfactory tubercle in PS1 x APP double transgenic mice model of AD. The experiments were performed in wild type and double transgenic homozygous animal groups of 2, 4, 6, and 8 months of age to analyze early stages of the pathology. In addition, beta-amyloid (Aβ) expression and its correlation with SOM cells have been quantified under confocal microscopy. The results indicate increasing expressions of Aβ with aging as well as an early fall of SOM and CR expression, whereas PV was decreased later in the disease progression. These observations evidence an early, preferential vulnerability of SOM and CR cells in rostral olfactory structures during AD that may be useful to unravel neural basis of olfactory deficits associated to this neurodegenerative disorder.
Collapse
Affiliation(s)
- Daniel Saiz-Sanchez
- Laboratorio de Neuroplasticidad y Neurodegeneración, Departamento de Ciencias Médicas, Centro Regional de Investigaciones Biomédicas, Facultad de Medicina de Ciudad Real, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
| | | | | | | |
Collapse
|
49
|
Yi SS. Time-dependent changes of calbindin D-28K and parvalbumin immunoreactivity in the hippocampus of rats with streptozotocin-induced type 1 diabetes. J Vet Sci 2013; 14:373-80. [PMID: 23628656 PMCID: PMC3885729 DOI: 10.4142/jvs.2013.14.4.373] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/23/2012] [Indexed: 01/30/2023] Open
Abstract
The hippocampus is affected by various stimuli that include hyperglycemia, depression, and ischemia. Calcium-binding proteins (CaBPs) have protective roles in the response to such stimuli. However, little is known about the expression of CaBPs under diabetic conditions. This study was conducted to examine alterations in the physiological parameters with type 1 diabetes induced with streptozotocin (STZ) as well as time-dependent changes in the expression of two CaBPs changes of were being evaluated. Rats treated with STZ (70 mg/kg) had high blood glucose levels (>21.4 mmol/L) along with increased food intake and water consumption volumes compared to the sham controls. In contrast, body weight of the animals treated with STZ was significantly reduced compared to the sham group. CB-specific immunoreactivity was generally increased in the hippocampal CA1 region and granule cell layer of the dentate gyrus (DG) 2 weeks after STZ treatment, but decreased thereafter in these regions. In contrast, the number of PV-immunoreactive neurons and fibers was unchanged in the hippocampus and DG 2 weeks after STZ treatment. However, this number subsequently decreased over time. These results suggest that CB and PV expression is lowest 3 weeks after STZ administration, and these deficits lead to disturbances in calcium homeostasis.
Collapse
Affiliation(s)
- Sun Shin Yi
- Department of Biomedical Laboratory Science, College of Biomedical Sciences, Soonchunhyang University, Asan 336-745, Korea.
| |
Collapse
|
50
|
Wilson DIG, Langston RF, Schlesiger MI, Wagner M, Watanabe S, Ainge JA. Lateral entorhinal cortex is critical for novel object-context recognition. Hippocampus 2013; 23:352-66. [PMID: 23389958 PMCID: PMC3648979 DOI: 10.1002/hipo.22095] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2013] [Indexed: 11/10/2022]
Abstract
Episodic memory incorporates information about specific events or occasions including spatial locations and the contextual features of the environment in which the event took place. It has been modeled in rats using spontaneous exploration of novel configurations of objects, their locations, and the contexts in which they are presented. While we have a detailed understanding of how spatial location is processed in the brain relatively little is known about where the nonspatial contextual components of episodic memory are processed. Initial experiments measured c-fos expression during an object-context recognition (OCR) task to examine which networks within the brain process contextual features of an event. Increased c-fos expression was found in the lateral entorhinal cortex (LEC; a major hippocampal afferent) during OCR relative to control conditions. In a subsequent experiment it was demonstrated that rats with lesions of LEC were unable to recognize object-context associations yet showed normal object recognition and normal context recognition. These data suggest that contextual features of the environment are integrated with object identity in LEC and demonstrate that recognition of such object-context associations requires the LEC. This is consistent with the suggestion that contextual features of an event are processed in LEC and that this information is combined with spatial information from medial entorhinal cortex to form episodic memory in the hippocampus. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David I G Wilson
- School of Psychology, University of St Andrews, St Mary's Quad, St Andrews, Fife, United Kingdom
| | | | | | | | | | | |
Collapse
|