1
|
Lucassen PJ, Korosi A, de Rooij SR, Smit AB, Van Dam AM, Daskalakis NP, Van Kesteren RE, Verheijen MHG, Lesuis SL, Kessels HW, Krugers HJ. How Can Early Stress Influence Later Alzheimer's Disease Risk? Possible Mediators and Underlying Mechanisms. Biol Psychiatry 2024:S0006-3223(24)01757-8. [PMID: 39577793 DOI: 10.1016/j.biopsych.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Alzheimer's disease (AD) is a progressive, age-related neurodegenerative disorder to which genetic mutations and risk factors contribute. Evidence is increasing that environmental and lifestyle-related factors, such as exercise, nutrition, education, and exposure to (early-life) stress modify the onset, incidence, and progression of AD. Here, we discuss recent preclinical findings on putative substrates that can explain or contribute to the effects of stress early in life on the risk of developing AD. We focus in particular on stress hormones, neural networks, synapses, mitochondria, nutrient and lipid metabolism, adult neurogenesis, engram cell ensembles, and neuroinflammation. We discuss the idea that stress exposure early in life can alter these processes, either combined or in isolation, thereby reducing the capacity of the brain to resist deleterious consequences of, for example, amyloid-β accumulation, thereby accelerating cognitive decline and progression of Alzheimer-related changes in model systems of the disease. A better understanding of whether experiences early in life also modify trajectories of cognitive decline and pathology in AD and how the substrates discussed translate to humans may help develop novel preventive and/or therapeutic strategies to mitigate the consequences of stressors early in life and increase resilience to developing dementia.
Collapse
Affiliation(s)
- Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands.
| | - Aniko Korosi
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Susanne R de Rooij
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Centre for Neurogenomics and Cognitive Research, Department of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Anne-Marie Van Dam
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Nikolaos P Daskalakis
- Neurogenomics and Translational Bioinformatics Laboratory, McLean Hospital, Harvard University, Boston, Massachusetts
| | - Ronald E Van Kesteren
- Department of Molecular and Cellular Neurobiology, Centre for Neurogenomics and Cognitive Research, Department of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Mark H G Verheijen
- Department of Molecular and Cellular Neurobiology, Centre for Neurogenomics and Cognitive Research, Department of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sylvie L Lesuis
- Department of Cellular & Computational Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Helmut W Kessels
- Department of Cellular & Computational Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Reid BM. Early life stress and iron metabolism in developmental psychoneuroimmunology. Brain Behav Immun Health 2024; 40:100824. [PMID: 39161875 PMCID: PMC11331713 DOI: 10.1016/j.bbih.2024.100824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 06/03/2024] [Accepted: 07/15/2024] [Indexed: 08/21/2024] Open
Abstract
An estimated 250 million children face adverse health outcomes from early life exposure to severe or chronic social, economic, and nutritional adversity, highlighting/emphasizing the pressing concern about the link between ELS and long-term implications on mental and physical health. There is significant overlap between populations experiencing high levels of chronic stress and those experiencing iron deficiency, spotlighting the potential role of iron as a key mediator in this association. Iron, an essential micronutrient for brain development and immune function, is often depleted in stress conditions. Iron deficiency among the most common nutrient deficiencies in the world. Fetal and infant iron status may thus serve as a crucial intermediary between early chronic psychological stress and subsequent immune system changes to impact neurodevelopment. The review presents a hypothesized pathway between early life stress (ELS), iron deficiency, and neurodevelopment through the hypothalamic-pituitary-adrenocortical (HPA) axis and the IL-6-hepcidin axis. This hypothesis is derived from (1) evidence that stress impacts iron status (2) long-term neurodevelopmental outcomes that are shared by ELS and iron deficiency exposure, and (3) possible mechanisms for how iron may mediate the relation between ELS and iron deficiency through alterations in the developing immune system. The article concludes by proposing future research directions, emphasizing the need for rigorous studies to elucidate how stress and iron metabolism interact to modify the developing immune system. Understanding these mechanisms could open new avenues for improving human health and neurodevelopment for women and children globally, making it a timely and vital area of study in psychoneuroimmunology research.
Collapse
Affiliation(s)
- Brie M. Reid
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, USA
- Center for Behavioral and Preventive Medicine, The Miriam Hospital, USA
- Department of Psychology, Department of Health Sciences, Northeastern University, USA
| |
Collapse
|
3
|
Snowden SG, Koulman A, Gaser C, la Fleur SE, Roseboom TJ, Korosi A, de Rooij SR. Prenatal exposure to undernutrition is associated with a specific lipid profile predicting future brain aging. NPJ AGING 2024; 10:42. [PMID: 39349457 PMCID: PMC11442854 DOI: 10.1038/s41514-024-00169-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/05/2024] [Indexed: 10/02/2024]
Abstract
Prenatal adversity affects cognitive and brain aging. Both lipid and leptin concentrations may be involved. We investigated if prenatal undernutrition is associated with a specific blood lipid profile and/or leptin concentrations, and if these relate to cognitive function and brain aging. 801 plasma samples of members of the Dutch famine birth cohort were assessed for lipidomics and leptin at age 58. Cognitive performance was measured with a Stroop task at 58, and MRI-based BrainAGE was derived in a subsample at 68. Out of 259 lipid signals, a signature of five identified individuals who were undernourished prenatally. These five lipids were not associated with cognitive performance, but three were predictive of BrainAGE. Leptin was not associated with prenatal famine exposure, Stroop performance, or BrainAGE. In conclusion, prenatal undernutrition was associated with an altered lipid profile predictive of BrainAGE 10 years later, demonstrating the potential of lipid profiles as early biomarkers for accelerated brain aging.
Collapse
Affiliation(s)
- Stuart G Snowden
- Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Level 4 Pathology, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- Department of Biological Sciences, Royal Holloway University of London, Egham, Surrey, TW20 0EX, UK
| | - Albert Koulman
- Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Level 4 Pathology, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Christian Gaser
- Structural Brain Mapping Group, Department of Neurology, Jena University Hospital, Jena, Germany
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Susanne E la Fleur
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Cellular and Molecular Mechanisms, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology & Metabolism, Amsterdam, The Netherlands
| | - Tessa J Roseboom
- Department of Epidemiology and Data Science, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Aniko Korosi
- Centre for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne R de Rooij
- Department of Epidemiology and Data Science, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands.
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands.
- Amsterdam Public Health research institute, Aging & Later life, Health Behaviors & Chronic Diseases, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Singhaarachchi PH, Antal P, Calon F, Culmsee C, Delpech JC, Feldotto M, Geertsema J, Hoeksema EE, Korosi A, Layé S, McQualter J, de Rooij SR, Rummel C, Slayo M, Sominsky L, Spencer SJ. Aging, sex, metabolic and life experience factors: Contributions to neuro-inflammaging in Alzheimer's disease research. Neurosci Biobehav Rev 2024; 162:105724. [PMID: 38762130 DOI: 10.1016/j.neubiorev.2024.105724] [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: 02/28/2024] [Revised: 04/24/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Alzheimer's disease (AD) is prevalent around the world, yet our understanding of the disease is still very limited. Recent work suggests that the cornerstone of AD may include the inflammation that accompanies it. Failure of a normal pro-inflammatory immune response to resolve may lead to persistent central inflammation that contributes to unsuccessful clearance of amyloid-beta plaques as they form, neuronal death, and ultimately cognitive decline. Individual metabolic, and dietary (lipid) profiles can differentially regulate this inflammatory process with aging, obesity, poor diet, early life stress and other inflammatory factors contributing to a greater risk of developing AD. Here, we integrate evidence for the interface between these factors, and how they contribute to a pro-inflammatory brain milieu. In particular, we discuss the importance of appropriate polyunsaturated fatty acids (PUFA) in the diet for the metabolism of specialised pro-resolving mediators (SPMs); raising the possibility for dietary strategies to improve AD outlook.
Collapse
Affiliation(s)
| | - Peter Antal
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, 1111, Hungary
| | - Frédéric Calon
- Faculty of Pharmacy, Centre de Recherche du CHU de Québec-Laval University, Quebec G1V0A6, Canada; International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, Philipps University of Marburg, Marburg D-35032, Germany; Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany
| | - Jean-Christophe Delpech
- International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Martin Feldotto
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Jorine Geertsema
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Emmy E Hoeksema
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Sophie Layé
- International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Jonathan McQualter
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Susanne R de Rooij
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, University of Amsterdam, 1018, the Netherlands
| | - Christoph Rummel
- Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany; Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Mary Slayo
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia; Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany; Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Luba Sominsky
- Barwon Health, Geelong, Victoria 3220, Australia; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria 3217, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia.
| |
Collapse
|
5
|
Kelly CE, Thompson DK, Adamson CL, Ball G, Dhollander T, Beare R, Matthews LG, Alexander B, Cheong JLY, Doyle LW, Anderson PJ, Inder TE. Cortical growth from infancy to adolescence in preterm and term-born children. Brain 2024; 147:1526-1538. [PMID: 37816305 PMCID: PMC10994536 DOI: 10.1093/brain/awad348] [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] [Received: 01/20/2023] [Revised: 08/10/2023] [Accepted: 09/30/2023] [Indexed: 10/12/2023] Open
Abstract
Early life experiences can exert a significant influence on cortical and cognitive development. Very preterm birth exposes infants to several adverse environmental factors during hospital admission, which affect cortical architecture. However, the subsequent consequence of very preterm birth on cortical growth from infancy to adolescence has never been defined; despite knowledge of critical periods during childhood for establishment of cortical networks. Our aims were to: chart typical longitudinal cortical development and sex differences in cortical development from birth to adolescence in healthy term-born children; estimate differences in cortical development between children born at term and very preterm; and estimate differences in cortical development between children with normal and impaired cognition in adolescence. This longitudinal cohort study included children born at term (≥37 weeks' gestation) and very preterm (<30 weeks' gestation) with MRI scans at ages 0, 7 and 13 years (n = 66 term-born participants comprising 34 with one scan, 18 with two scans and 14 with three scans; n = 201 very preterm participants comprising 56 with one scan, 88 with two scans and 57 with three scans). Cognitive assessments were performed at age 13 years. Cortical surface reconstruction and parcellation were performed with state-of-the-art, equivalent MRI analysis pipelines for all time points, resulting in longitudinal cortical volume, surface area and thickness measurements for 62 cortical regions. Developmental trajectories for each region were modelled in term-born children, contrasted between children born at term and very preterm, and contrasted between all children with normal and impaired cognition. In typically developing term-born children, we documented anticipated patterns of rapidly increasing cortical volume, area and thickness in early childhood, followed by more subtle changes in later childhood, with smaller cortical size in females than males. In contrast, children born very preterm exhibited increasingly reduced cortical volumes, relative to term-born children, particularly during ages 0-7 years in temporal cortical regions. This reduction in cortical volume in children born very preterm was largely driven by increasingly reduced cortical thickness rather than area. This resulted in amplified cortical volume and thickness reductions by age 13 years in individuals born very preterm. Alterations in cortical thickness development were found in children with impaired language and memory. This study shows that the neurobiological impact of very preterm birth on cortical growth is amplified from infancy to adolescence. These data further inform the long-lasting impact on cortical development from very preterm birth, providing broader insights into neurodevelopmental consequences of early life experiences.
Collapse
Affiliation(s)
- Claire E Kelly
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC 3800, Australia
- Victorian Infant Brain Studies (VIBeS), Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Developmental Imaging, Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
| | - Deanne K Thompson
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC 3800, Australia
- Victorian Infant Brain Studies (VIBeS), Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Developmental Imaging, Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Chris L Adamson
- Developmental Imaging, Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
| | - Gareth Ball
- Developmental Imaging, Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Thijs Dhollander
- Developmental Imaging, Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
| | - Richard Beare
- Developmental Imaging, Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- National Centre for Healthy Ageing and Peninsula Clinical School, Faculty of Medicine, Monash University, Melbourne, VIC 3199, Australia
| | - Lillian G Matthews
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC 3800, Australia
- Victorian Infant Brain Studies (VIBeS), Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bonnie Alexander
- Developmental Imaging, Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Neurosurgery, The Royal Children’s Hospital, Melbourne, VIC 3052, Australia
| | - Jeanie L Y Cheong
- Victorian Infant Brain Studies (VIBeS), Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC 3052, Australia
- Newborn Research, The Royal Women’s Hospital, Melbourne, VIC 3052, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lex W Doyle
- Victorian Infant Brain Studies (VIBeS), Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
- Newborn Research, The Royal Women’s Hospital, Melbourne, VIC 3052, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Peter J Anderson
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC 3800, Australia
- Victorian Infant Brain Studies (VIBeS), Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
| | - Terrie E Inder
- Center for Neonatal Research, Children's Hospital of Orange County, Orange, CA 92868, USA
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
6
|
Hartmann J, Bajaj T, Otten J, Klengel C, Ebert T, Gellner AK, Junglas E, Hafner K, Anderzhanova EA, Tang F, Missig G, Rexrode L, Trussell DT, Li KX, Pöhlmann ML, Mackert S, Geiger TM, Heinz DE, Lardenoije R, Dedic N, McCullough KM, Próchnicki T, Rhomberg T, Martinelli S, Payton A, Robinson AC, Stein V, Latz E, Carlezon WA, Hausch F, Schmidt MV, Murgatroyd C, Berretta S, Klengel T, Pantazopoulos H, Ressler KJ, Gassen NC. SKA2 regulated hyperactive secretory autophagy drives neuroinflammation-induced neurodegeneration. Nat Commun 2024; 15:2635. [PMID: 38528004 PMCID: PMC10963788 DOI: 10.1038/s41467-024-46953-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
High levels of proinflammatory cytokines induce neurotoxicity and catalyze inflammation-driven neurodegeneration, but the specific release mechanisms from microglia remain elusive. Here we show that secretory autophagy (SA), a non-lytic modality of autophagy for secretion of vesicular cargo, regulates neuroinflammation-mediated neurodegeneration via SKA2 and FKBP5 signaling. SKA2 inhibits SA-dependent IL-1β release by counteracting FKBP5 function. Hippocampal Ska2 knockdown in male mice hyperactivates SA resulting in neuroinflammation, subsequent neurodegeneration and complete hippocampal atrophy within six weeks. The hyperactivation of SA increases IL-1β release, contributing to an inflammatory feed-forward vicious cycle including NLRP3-inflammasome activation and Gasdermin D-mediated neurotoxicity, which ultimately drives neurodegeneration. Results from protein expression and co-immunoprecipitation analyses of male and female postmortem human brains demonstrate that SA is hyperactivated in Alzheimer's disease. Overall, our findings suggest that SKA2-regulated, hyperactive SA facilitates neuroinflammation and is linked to Alzheimer's disease, providing mechanistic insight into the biology of neuroinflammation.
Collapse
Affiliation(s)
- Jakob Hartmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA.
| | - Thomas Bajaj
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Joy Otten
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Claudia Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Tim Ebert
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Anne-Kathrin Gellner
- Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Ellen Junglas
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Elmira A Anderzhanova
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Fiona Tang
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Galen Missig
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Lindsay Rexrode
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Daniel T Trussell
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Katelyn X Li
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Max L Pöhlmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Sarah Mackert
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Thomas M Geiger
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, 64287, Darmstadt, Germany
| | - Daniel E Heinz
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Roy Lardenoije
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Nina Dedic
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Kenneth M McCullough
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Tomasz Próchnicki
- Institute of Innate Immunity, University Hospital Bonn, 53127, Bonn, Germany
| | - Thomas Rhomberg
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Silvia Martinelli
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Antony Payton
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, M13 9PL, UK
| | - Andrew C Robinson
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Salford Royal Hospital, Salford, M6 8HD, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre (MAHSC), Manchester, UK
| | - Valentin Stein
- Institute of Physiology II, University of Bonn, 53127, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, 53127, Bonn, Germany
- Deutsches Rheuma Forschungszentrum Berlin (DRFZ), 10117, Berlin, Germany
| | - William A Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Felix Hausch
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, 64287, Darmstadt, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Chris Murgatroyd
- Department of Life Sciences, Manchester Metropolitan University, Manchester, M15 6BH, UK
| | - Sabina Berretta
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA.
| | - Nils C Gassen
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany.
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804, Munich, Germany.
| |
Collapse
|
7
|
Kotah JM, Kater MSJ, Brosens N, Lesuis SL, Tandari R, Blok TM, Marchetto L, Yusaf E, Koopmans FTW, Smit AB, Lucassen PJ, Krugers HJ, Verheijen MHG, Korosi A. Early-life stress and amyloidosis in mice share pathogenic pathways involving synaptic mitochondria and lipid metabolism. Alzheimers Dement 2024; 20:1637-1655. [PMID: 38055782 PMCID: PMC10984508 DOI: 10.1002/alz.13569] [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] [Received: 05/30/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 12/08/2023]
Abstract
INTRODUCTION Early-life stress (ES) increases the risk for Alzheimer's disease (AD). We and others have shown that ES aggravates amyloid-beta (Aβ) pathology and promotes cognitive dysfunction in APP/PS1 mice, but underlying mechanisms remain unclear. METHODS We studied how ES affects the hippocampal synaptic proteome in wild-type (WT) and APP/PS1 mice at early and late pathological stages, and validated hits using electron microscopy and immunofluorescence. RESULTS The hippocampal synaptosomes of both ES-exposed WT and early-stage APP/PS1 mice showed a relative decrease in actin dynamics-related proteins and a relative increase in mitochondrial proteins. ES had minimal effects on older WT mice, while strongly affecting the synaptic proteome of advanced stage APP/PS1 mice, particularly the expression of astrocytic and mitochondrial proteins. DISCUSSION Our data show that ES and amyloidosis share pathogenic pathways involving synaptic mitochondrial dysfunction and lipid metabolism, which may underlie the observed impact of ES on the trajectory of AD.
Collapse
Affiliation(s)
- Janssen M. Kotah
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Mandy S. J. Kater
- Department of Molecular and Cellular NeurobiologyCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Niek Brosens
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Sylvie L. Lesuis
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Roberta Tandari
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Thomas M. Blok
- Department of Molecular and Cellular NeurobiologyCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Luca Marchetto
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Ella Yusaf
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Frank T. W. Koopmans
- Department of Molecular and Cellular NeurobiologyCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - August B. Smit
- Department of Molecular and Cellular NeurobiologyCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Paul J. Lucassen
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Harm J. Krugers
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Mark H. G. Verheijen
- Department of Molecular and Cellular NeurobiologyCenter for Neurogenomics and Cognitive ResearchAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Aniko Korosi
- Brain Plasticity GroupSwammerdam Institute for Life Sciences – Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
8
|
Reemst K, Lopizzo N, Abbink MR, Engelenburg HJ, Cattaneo A, Korosi A. Molecular underpinnings of programming by early-life stress and the protective effects of early dietary ω6/ω3 ratio, basally and in response to LPS: Integrated mRNA-miRNAs approach. Brain Behav Immun 2024; 117:283-297. [PMID: 38242369 DOI: 10.1016/j.bbi.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 12/22/2023] [Accepted: 01/14/2024] [Indexed: 01/21/2024] Open
Abstract
Early-life stress (ELS) exposure increases the risk for mental disorders, including cognitive impairments later in life. We have previously demonstrated that an early diet with low ω6/ω3 polyunsaturated fatty acid (PUFA) ratio protects against ELS-induced cognitive impairments. Several studies have implicated the neuroimmune system in the ELS and diet mediated effects, but currently the molecular pathways via which ELS and early diet exert their long-term impact are not yet fully understood. Here we study the effects of ELS and dietary PUFA ratio on hippocampal mRNA and miRNA expression in adulthood, both under basal as well as inflammatory conditions. Male mice were exposed to chronic ELS by the limiting bedding and nesting material paradigm from postnatal day(P)2 to P9, and provided with a diet containing a standard (high (15:1.1)) or protective (low (1.1:1)) ω6 linoleic acid to ω3 alpha-linolenic acid ratio from P2 to P42. At P120, memory was assessed using the object location task. Subsequently, a single lipopolysaccharide (LPS) injection was given and 24 h later hippocampal genome-wide mRNA and microRNA (miRNA) expression was measured using microarray. Spatial learning deficits induced by ELS in mice fed the standard (high ω6/ω3) diet were reversed by the early-life protective (low ω6/ω3) diet. An integrated miRNA - mRNA analysis revealed that ELS and early diet induced miRNA driven mRNA expression changes into adulthood. Under basal conditions both ELS and the diet affected molecular pathways related to hippocampal plasticity, with the protective (low ω6/ω3 ratio) diet leading to activation of molecular pathways associated with improved hippocampal plasticity and learning and memory in mice previously exposed to ELS (e.g., CREB signaling and endocannabinoid neuronal synapse pathway). LPS induced miRNA and mRNA expression was strongly dependent on both ELS and early diet. In mice fed the standard (high ω6/ω3) diet, LPS increased miRNA expression leading to activation of inflammatory pathways. In contrast, in mice fed the protective diet, LPS reduced miRNA expression and altered target mRNA expression inhibiting inflammatory signaling pathways and pathways associated with hippocampal plasticity, which was especially apparent in mice previously exposed to ELS. This data provides molecular insights into how the protective (low ω6/ω3) diet during development could exert its long-lasting beneficial effects on hippocampal plasticity and learning and memory especially in a vulnerable population exposed to stress early in life, providing the basis for the development of intervention strategies.
Collapse
Affiliation(s)
- Kitty Reemst
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands
| | - Nicola Lopizzo
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Maralinde R Abbink
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands
| | - Hendrik J Engelenburg
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands.
| |
Collapse
|
9
|
Song J, Li Z, Xue X, Meng J, Zhu W, Hu S, Xu G, Wang L. Neonatal stress disrupts the glymphatic system development and increases the susceptibility to Parkinson's disease in later life. CNS Neurosci Ther 2024; 30:e14587. [PMID: 38421142 PMCID: PMC10851323 DOI: 10.1111/cns.14587] [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] [Received: 07/07/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION Neonatal stress disrupts brain development and increases the risk of neurological disorders later in life. However, the impact of neonatal stress on the development of the glymphatic system and susceptibility to Parkinson's disease (PD) remains largely unknown. METHODS Neonatal maternal deprivation (NMD) was performed on mice for 14 consecutive days to model chronic neonatal stress. Adeno-associated virus expressing A53T-α-synuclein (α-syn) was injected into the substantia nigra to establish PD model mice. Glymphatic activity was determined using in vivo magnetic resonance imaging, ex vivo fluorescence imaging and microplate assay. The transcription and expression of aquaporin-4 (AQP4) and other molecules were evaluated by qPCR, western blotting, and immunofluorescence. Animal's responses to NMD and α-syn overexpression were observed using behavioral tests. RESULTS Glymphatic activity was impaired in adult NMD mice. AQP4 polarization and platelet-derived growth factor B (PDGF-B) signaling were reduced in the frontal cortex and hippocampus of both young and adult NMD mice. Furthermore, exogenous α-syn accumulation was increased and PD-like symptoms were aggravated in adult NMD mice. CONCLUSION The results demonstrated that NMD could disrupt the development of the glymphatic system through PDGF-B signaling and increase the risk of PD later in life, indicating that alleviating neonatal stress could be beneficial in protecting the glymphatic system and reducing susceptibility to neurodegeneration.
Collapse
Affiliation(s)
- Jian Song
- Department of Physiology and NeurobiologySuzhou Medical College of Soochow UniversitySuzhouChina
| | - Zhen‐Hua Li
- Department of Physiology and NeurobiologySuzhou Medical College of Soochow UniversitySuzhouChina
| | - Xin‐Yu Xue
- Department of Physiology and NeurobiologySuzhou Medical College of Soochow UniversitySuzhouChina
| | - Jing‐Cai Meng
- Department of Physiology and NeurobiologySuzhou Medical College of Soochow UniversitySuzhouChina
| | - Wen‐Xin Zhu
- Department of Physiology and NeurobiologySuzhou Medical College of Soochow UniversitySuzhouChina
| | - Shufen Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of NeuroscienceSoochow UniversitySuzhouChina
| | - Guang‐Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of NeuroscienceSoochow UniversitySuzhouChina
| | - Lin‐Hui Wang
- Department of Physiology and NeurobiologySuzhou Medical College of Soochow UniversitySuzhouChina
| |
Collapse
|
10
|
Bozzay ML, Joyce HE, Jiang L, De Vito AN, Emrani S, Browne J, Bayer TA, Quinn MJ, Primack JM, Kelso CM, Wu WC, Rudolph JL, McGeary JE, Kunicki ZJ. Time to Dementia Diagnosis Among Veterans with Comorbid Insomnia and Depressive Episodes. J Alzheimers Dis 2024; 100:899-909. [PMID: 38995783 DOI: 10.3233/jad-240080] [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: 07/14/2024]
Abstract
Background Older adults with heart failure are at elevated risk of Alzheimer's disease and related dementias (AD/ADRD). Research suggests that insomnia and depressive episodes contribute somewhat dissociable impacts on risk for AD/ADRD in this patient population, although the temporal ordering of effects is unknown. Objective This study examined time to dementia diagnosis among patients with comorbid insomnia and/or depressive episodes in an epidemiological sample. Methods Secondary data analyses were conducted using a cohort study of 203,819 Veterans with a primary admission diagnosis of heart failure in 129 VA Medical Centers. Results Patients with diagnoses of both insomnia and depressive episodes had the shortest time to a dementia diagnosis at both 1-year (Hazard ratio = 1.43, 95% CI [1.36, 1.51]) and 3-year follow-up time points (Hazard ratio = 1.40, 95% CI [1.34, 1.47]) versus patients with one or neither comorbidity. Conclusions Individuals with both comorbidities had the shortest time to dementia onset. Screening for these comorbidities may help to identify patients at elevated risk of dementia who could benefit from enhanced monitoring or early intervention strategies for more rapid detection and management of dementia symptoms.
Collapse
Affiliation(s)
- Melanie L Bozzay
- Department of Psychiatry and Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Lan Jiang
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
| | - Alyssa N De Vito
- Butler Hospital, Providence, RI, USA
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Sheina Emrani
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Julia Browne
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Thomas A Bayer
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - McKenzie J Quinn
- VA RR& D Center for Neurorestoration and Neurotechnology, Providence VA Medical Center, Providence, RI, USA
| | - Jennifer M Primack
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Catherine M Kelso
- VA RR& D Center for Neurorestoration and Neurotechnology, Providence VA Medical Center, Providence, RI, USA
- Veterans Health Administration, Office of Patient Care Services, Geriatrics and Extended Care, Washington, DC, USA
| | - Wen-Chih Wu
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
| | - James L Rudolph
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - John E McGeary
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Zachary J Kunicki
- VA Center of Innovation in Long Term Services, Providence VA Medical Center, Providence, RI, USA
- Department of Psychiatry & Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
11
|
Diaconu B, Kohls G, Rogers JC, Pauli R, Cornwell H, Bernhard A, Martinelli A, Ackermann K, Fann N, Fernandez-Rivas A, Gonzalez-Torres MA, Gonzalez de Artaza-Lavesa M, Hervas A, Stadler C, Konrad K, Freitag CM, Fairchild G, Rotshtein P, De Brito SA. Emotion processing in maltreated boys and girls: Evidence for latent vulnerability. Eur Child Adolesc Psychiatry 2023; 32:2523-2536. [PMID: 36738328 PMCID: PMC10682268 DOI: 10.1007/s00787-022-02132-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 12/14/2022] [Indexed: 02/05/2023]
Abstract
Evidence of alterations in emotion processing in maltreated youth has been hypothesized to reflect latent vulnerability for psychopathology. However, previous studies have not systematically examined the influence of psychopathology on the results. Here, we examined emotion recognition and learning in youth who differed in terms of presence vs. absence of maltreatment and psychopathology and tested for potential sex effects. Maltreatment and psychopathology were assessed in 828 youth (514 females) aged 9-18 years using diagnostic interviews and self- and parent-report questionnaires. Emotion recognition was assessed via identification of morphed facial expressions of six universal emotions. For emotion learning, reward and punishment values were assigned to novel stimuli and participants had to learn to correctly respond/withhold response to stimuli to maximize points. A three-way interaction of maltreatment by psychopathology by emotion indicated that when psychopathology was low, maltreated youth were less accurate than non-maltreated youth for happy, fear and disgust. A three-way interaction of sex, maltreatment and emotion indicated that maltreated girls and boys were impaired for fear, but girls showed an impairment for happy, while boys for disgust. There were no effects of maltreatment, psychopathology, or sex on reward learning. However, a two-way interaction between sex and maltreatment showed that maltreated girls were worse at learning from punishment relative to non-maltreated girls, while maltreated boys were better than non-maltreated boys. The study provides the first clear evidence of latent-vulnerability in emotion recognition in maltreated youth and suggests that girls and boys might be characterized by distinct profiles of emotion recognition and learning following maltreatment.
Collapse
Affiliation(s)
- Bianca Diaconu
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Gregor Kohls
- Child Neuropsychology Section, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, RWTH Aachen, Aachen, Germany
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Jack C Rogers
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Birmingham, B15 2TT, UK
- Institute for Mental Health, School of Psychology, University of Birmingham, Birmingham, UK
| | - Ruth Pauli
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Birmingham, B15 2TT, UK
| | | | - Anka Bernhard
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Anne Martinelli
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
- School of Psychology, Fresenius University of Applied Sciences, Frankfurt am Main, Germany
| | - Katharina Ackermann
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Nikola Fann
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | | | | | | | - Amaia Hervas
- University Hospital Mutua Terrassa, Barcelona, Spain
- Global Institute of Neurodevelopment Integrated Care (IGAIN), Barcelona, Spain
| | - Christina Stadler
- Department of Child and Adolescent Psychiatry, Psychiatric University Hospital, University of Basel, Basel, Switzerland
| | - Kerstin Konrad
- Child Neuropsychology Section, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, RWTH Aachen, Aachen, Germany
- JARA-Brain Institute II, Molecular Neuroscience and Neuroimaging, RWTH Aachen & Research Centre Juelich, Juelich, Germany
| | - Christine M Freitag
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | | | - Pia Rotshtein
- Neuroimaging Research Unit, University of Haifa, Haifa, Israel
| | - Stephane A De Brito
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
12
|
Kotlinska JH, Grochecki P, Michalak A, Pankowska A, Kochalska K, Suder P, Ner-Kluza J, Matosiuk D, Marszalek-Grabska M. Neonatal Maternal Separation Induces Sexual Dimorphism in Brain Development: The Influence on Amino Acid Levels and Cognitive Disorders. Biomolecules 2023; 13:1449. [PMID: 37892131 PMCID: PMC10605115 DOI: 10.3390/biom13101449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/09/2023] [Accepted: 09/23/2023] [Indexed: 10/29/2023] Open
Abstract
Repeated maternal separation (MS) is a useful experimental model in rodents for studying the long-term influence of early-life stress on brain neurophysiology. In our work, we assessed the effect of repeated MS (postnatal day (PND)1-21, 180 min/day) on the postnatal development of rat brain regions involved in memory using proton magnetic resonance spectroscopy (1HMRS) for tissue volume and the level of amino acids such as glutamate, aspartate, glutamine, glycine and gamma-aminobutyric acid (GABA) in the hippocampus. We assessed whether these effects are sex dependent. We also use novel object recognition (NOR) task to examine the effect of MS on memory and the effect of ethanol on it. Finally, we attempted to ameliorate postnatal stress-induced memory deficits by using VU-29, a positive allosteric modulator (PAM) of the metabotropic glutamate type 5 (mGlu5) receptor. In males, we noted deficits in the levels of glutamate, glycine and glutamine and increases in GABA in the hippocampus. In addition, the values of perirhinal cortex, prefrontal cortex and insular cortex and CA3 were decreased in these animals. MS females, in contrast, demonstrated significant increase in glutamate levels and decrease in GABA levels in the hippocampus. Here, the CA1 values alone were increased. VU-29 administration ameliorated these cognitive deficits. Thus, MS stress disturbs amino acids levels mainly in the hippocampus of adult male rats, and enhancement of glutamate neurotransmission reversed recognition memory deficits in these animals.
Collapse
Affiliation(s)
- Jolanta H. Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University, Chodzki 4A, 20-093 Lublin, Poland;
| | - Pawel Grochecki
- Department of Pharmacology and Pharmacodynamics, Medical University, Chodzki 4A, 20-093 Lublin, Poland;
| | - Agnieszka Michalak
- Independent Laboratory of Behavioral Studies, Medical University, Chodzki 4A, 20-093 Lublin, Poland;
| | - Anna Pankowska
- Department of Radiography, Medical University, Staszica 16, 20-081 Lublin, Poland; (A.P.); (K.K.)
| | - Katarzyna Kochalska
- Department of Radiography, Medical University, Staszica 16, 20-081 Lublin, Poland; (A.P.); (K.K.)
| | - Piotr Suder
- Department of Analytical Chemistry and Biochemistry, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, A. Mickiewicza 30, 30-059 Krakow, Poland; (P.S.); (J.N.-K.)
| | - Joanna Ner-Kluza
- Department of Analytical Chemistry and Biochemistry, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, A. Mickiewicza 30, 30-059 Krakow, Poland; (P.S.); (J.N.-K.)
| | - Dariusz Matosiuk
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modelling Lab, Medical University, Chodzki 4A, 20-093 Lublin, Poland;
| | - Marta Marszalek-Grabska
- Department of Experimental and Clinical Pharmacology, Medical University, Jaczewskiego 8B, 20-090 Lublin, Poland;
| |
Collapse
|
13
|
Jeanneteau F. Stress and the risk of Alzheimer dementia: Can deconstructed engrams be rebuilt? J Neuroendocrinol 2023; 35:e13235. [PMID: 36775895 DOI: 10.1111/jne.13235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
The exact neuropathological mechanism by which the dementia process unfolds is under intense scrutiny. The disease affects about 38 million people worldwide, 70% of which are clinically diagnosed with Alzheimer's disease (AD). If the destruction of synapses essential for learning, planning and decision-making is part of the problem, must the restoration of previously lost synapses be part of the solution? It is plausible that neuronal capacity to restitute information corresponds with the adaptive capacity of its connectivity reserve. A challenge will be to promote the functional connectivity that can compensate for the lost one. This will require better clarification of the remodeling of functional connectivity during the progression of AD dementia and its reversal upon experimental treatment. A major difficulty is to promote the neural pathways that are atrophied in AD dementia while suppressing others that are bolstered. Therapeutic strategies should aim at scaling functional connectivity to a just balance between the atrophic and hypertrophic systems. However, the exact factors that can help reach this objective are still unclear. Similarities between the effects of chronic stress and some neuropathological mechanisms underlying AD dementia support the idea that common components deserve prime attention as therapeutic targets.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
14
|
Ren Z, Su B, Du Y, Zhou T, Zheng X, Liu J. Effect modifications of BMI transition and trajectory in the associations of adverse childhood experiences with new-onset dementia and its subtypes in older US adults. Gen Psychiatr 2023; 36:e101092. [PMID: 37622031 PMCID: PMC10445386 DOI: 10.1136/gpsych-2023-101092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Background Adverse childhood experiences (ACEs) and dementia are associated and comorbid with obesity. However, according to emerging research, the role of obesity in the association between ACEs and dementia seems controversial. Aims This analysis aimed to explore the associations between ACEs and different dementia subtypes and the effect modification of long-term body mass index (BMI). Methods Data were obtained from the US Health and Retirement Study. Six ACEs were categorised as 0, 1 and 2 or more. All-cause dementia, Alzheimer's disease (AD) and other dementias were defined by self-reported or proxy-reported physician diagnosis. Cox proportional hazards regression was used to explore the associations of ACEs with new-onset all-cause dementia, AD and other dementias from 2010 to 2020. Effect modification of BMI in 2010 and BMI transition and trajectory (fitted by group-based trajectory modelling) from 2004 to 2010 were assessed. Results 15 282 participants with a mean age of 67.0 years (58.0-75.0) were included in the 2010 data analysis. Significant interactions of ACEs with baseline BMI, BMI transition and BMI trajectory in their associations with new-onset all-cause dementia and AD were observed (all p<0.05). For instance, positive associations of two or more ACEs (vs none) with all-cause dementia and AD were found in those with a BMI trajectory of maintaining ≥30 kg/m2 (maintain obesity) rather than a decline to or maintaining <25 kg/m2 (decline to or maintain normal weight), with hazard ratios (HRs) of 1.87 (95% confidence interval (CI): 1.45 to 2.42) and 1.85 (95% CI: 1.22 to 2.80), respectively. Conclusions ACEs were associated with dementia and AD in US adults with long-term abnormally elevated BMI but not with long-term normal or decreasing BMI. Integrated weight management throughout life could prevent dementia among those with childhood adversity.
Collapse
Affiliation(s)
- Ziyang Ren
- Institute of Reproductive and Child Health/National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Binbin Su
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yushan Du
- Institute of Reproductive and Child Health/National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Tianjing Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xiaoying Zheng
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jufen Liu
- Institute of Reproductive and Child Health/National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- APEC Health Science Academy (HeSAY), Peking University, Beijing, China
| |
Collapse
|
15
|
Huang Z, Jordan JD, Zhang Q. Early life adversity as a risk factor for cognitive impairment and Alzheimer's disease. Transl Neurodegener 2023; 12:25. [PMID: 37173751 PMCID: PMC10182702 DOI: 10.1186/s40035-023-00355-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
Neurological conditions, including cognitive impairment and Alzheimer's disease (AD), impose a huge burden on society, affecting millions of people globally. In addition to genetic factors, recent studies indicate that environmental and experiential factors may contribute to the pathogenesis of these diseases. Early life adversity (ELA) has a profound impact on brain function and health later in life. In rodent models, exposure to ELA results in specific cognitive deficits and aggravated AD pathology. Extensive concerns have been raised regarding the higher risk of developing cognitive impairments in people with a history of ELA. In this review, we scrutinize findings from human and animal studies focusing on the connection of ELA with cognitive impairment and AD. These discoveries suggest that ELA, especially at early postnatal stages, increases susceptibility to cognitive impairment and AD later in life. In terms of mechanisms, ELA could lead to dysregulation of the hypothalamus-pituitary-adrenal axis, altered gut microbiome, persistent inflammation, oligodendrocyte dysfunction, hypomyelination, and aberrant adult hippocampal neurogenesis. Crosstalks among these events may synergistically contribute to cognitive impairment later in life. Additionally, we discuss several interventions that may alleviate adverse consequences of ELA. Further investigation into this crucial area will help improve ELA management and reduce the burden of related neurological conditions.
Collapse
Affiliation(s)
- Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - J Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| |
Collapse
|
16
|
Wiegersma AM, Boots A, Langendam MW, Limpens J, Shenkin SD, Korosi A, Roseboom TJ, de Rooij SR. Do prenatal factors shape the risk for dementia?: A systematic review of the epidemiological evidence for the prenatal origins of dementia. Soc Psychiatry Psychiatr Epidemiol 2023:10.1007/s00127-023-02471-7. [PMID: 37029828 DOI: 10.1007/s00127-023-02471-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 03/30/2023] [Indexed: 04/09/2023]
Abstract
PURPOSE Prenatal factors such as maternal stress, infection and nutrition affect fetal brain development and may also influence later risk for dementia. The purpose of this systematic review was to provide an overview of all studies which investigated the association between prenatal factors and later risk for dementia. METHODS We systematically searched MEDLINE and Embase for original human studies reporting on associations between prenatal factors and dementia from inception to 23 November 2022. Prenatal factors could be any factor assessed during pregnancy, at birth or postnatally, provided they were indicative of a prenatal exposure. Risk of bias was assessed using the Newcastle Ottawa Scale. We followed PRISMA guidelines for reporting. RESULTS A total of 68 studies met eligibility criteria (including millions of individuals), assessing maternal age (N = 30), paternal age (N = 22), birth order (N = 15), season of birth (N = 16), place of birth (N = 13), prenatal influenza pandemic (N = 1) or Chinese famine exposure (N = 1), birth characteristics (N = 3) and prenatal hormone exposure (N = 4). We observed consistent results for birth in a generally less optimal environment (e.g. high infant mortality area) being associated with higher dementia risk. Lower and higher birth weight and prenatal famine exposure were associated with higher dementia risk. The studies on season of birth, digit ratio, prenatal influenza pandemic exposure, parental age and birth order showed inconsistent results and were hampered by relatively high risk of bias. CONCLUSION Our findings suggest that some prenatal factors, especially those related to a suboptimal prenatal environment, are associated with an increased dementia risk. As these associations may be confounded by factors such as parental socioeconomic status, more research is needed to examine the potential causal role of the prenatal environment in dementia.
Collapse
Affiliation(s)
- Aline Marileen Wiegersma
- Epidemiology and Data Science, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Public Health Research Institute, Aging & Later Life, Health Behaviors & Chronic Diseases, Amsterdam, The Netherlands.
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands.
| | - Amber Boots
- Epidemiology and Data Science, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Aging & Later Life, Health Behaviors & Chronic Diseases, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Miranda W Langendam
- Epidemiology and Data Science, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Methodology, Amsterdam, The Netherlands
| | - Jacqueline Limpens
- Medical Library, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Susan D Shenkin
- Geriatric Medicine, Usher Institute, University of Edinburgh, Edinburgh, Scotland, UK
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Tessa J Roseboom
- Epidemiology and Data Science, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Obstetrics and Gynaecology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Aging & Later Life, Health Behaviors & Chronic Diseases, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Susanne R de Rooij
- Epidemiology and Data Science, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Aging & Later Life, Health Behaviors & Chronic Diseases, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Hartmann J, Bajaj T, Otten J, Klengel C, Gellner AK, Junglas E, Hafner K, Anderzhanova EA, Tang F, Missig G, Rexrode L, Li K, Pöhlmann ML, Heinz DE, Lardenoije R, Dedic N, McCullough KM, Próchnicki T, Rhomberg T, Martinelli S, Payton A, Robinson AC, Stein V, Latz E, Carlezon WA, Schmidt MV, Murgatroyd C, Berretta S, Klengel T, Pantazopoulos H, Ressler KJ, Gassen NC. SKA2 regulated hyperactive secretory autophagy drives neuroinflammation-induced neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.534570. [PMID: 37066393 PMCID: PMC10103985 DOI: 10.1101/2023.04.03.534570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
High levels of proinflammatory cytokines induce neurotoxicity and catalyze inflammation-driven neurodegeneration, but the specific release mechanisms from microglia remain elusive. We demonstrate that secretory autophagy (SA), a non-lytic modality of autophagy for secretion of vesicular cargo, regulates neuroinflammation-mediated neurodegeneration via SKA2 and FKBP5 signaling. SKA2 inhibits SA-dependent IL-1β release by counteracting FKBP5 function. Hippocampal Ska2 knockdown in mice hyperactivates SA resulting in neuroinflammation, subsequent neurodegeneration and complete hippocampal atrophy within six weeks. The hyperactivation of SA increases IL-1β release, initiating an inflammatory feed-forward vicious cycle including NLRP3-inflammasome activation and Gasdermin D (GSDMD)-mediated neurotoxicity, which ultimately drives neurodegeneration. Results from protein expression and co-immunoprecipitation analyses of postmortem brains demonstrate that SA is hyperactivated in Alzheimer's disease. Overall, our findings suggest that SKA2-regulated, hyperactive SA facilitates neuroinflammation and is linked to Alzheimer's disease, providing new mechanistic insight into the biology of neuroinflammation.
Collapse
|
18
|
Kelly CE, Shaul M, Thompson DK, Mainzer RM, Yang JY, Dhollander T, Cheong JL, Inder TE, Doyle LW, Anderson PJ. Long-lasting effects of very preterm birth on brain structure in adulthood: A systematic review and meta-analysis. Neurosci Biobehav Rev 2023; 147:105082. [PMID: 36775083 DOI: 10.1016/j.neubiorev.2023.105082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/01/2023] [Accepted: 02/05/2023] [Indexed: 02/12/2023]
Abstract
Early life experiences, such as very preterm (VP) birth, can affect brain and cognitive development. Several prior studies investigated brain structure in adults born VP; synthesising these studies may help to provide a clearer understanding of long-term effects of VP birth on the brain. We systematically searched Medline and Embase for articles that investigated brain structure using MRI in adulthood in individuals born VP (<32 weeks' gestation) or with very low birth weight (VLBW; <1500 g), and controls born at term or with normal birth weight. In total, 77 studies met the review inclusion criteria, of which 28 studies were eligible for meta-analyses, including data from up to 797 VP/VLBW participants and 518 controls, aged 18-33 years. VP/VLBW adults exhibited volumetric, morphologic and microstructural alterations in subcortical and temporal cortical regions compared with controls, with pooled standardised mean differences up to - 1.0 (95% confidence interval: -1.2, -0.8). This study suggests there is a persisting neurological impact of VP birth, which may provide developmental neurobiological insights for adult cognition in high-risk populations.
Collapse
Affiliation(s)
- Claire E Kelly
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, Australia; Victorian Infant Brain Studies (VIBeS), Murdoch Children's Research Institute, Melbourne, Australia; Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Australia.
| | - Michelle Shaul
- Victorian Infant Brain Studies (VIBeS), Murdoch Children's Research Institute, Melbourne, Australia; Deakin University, Melbourne, Australia
| | - Deanne K Thompson
- Victorian Infant Brain Studies (VIBeS), Murdoch Children's Research Institute, Melbourne, Australia; Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Rheanna M Mainzer
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia; Clinical Epidemiology and Biostatistics Unit, Population Health, Murdoch Children's Research Institute, Melbourne, Australia
| | - Joseph Ym Yang
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Australia; Neuroscience Advanced Clinical Imaging Service (NACIS), Department of Neurosurgery, The Royal Children's Hospital, Melbourne, Australia; Neuroscience Research, Murdoch Children's Research Institute, Melbourne, Australia
| | - Thijs Dhollander
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Australia
| | - Jeanie Ly Cheong
- Victorian Infant Brain Studies (VIBeS), Murdoch Children's Research Institute, Melbourne, Australia; The Royal Women's Hospital, Melbourne, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Australia
| | - Terrie E Inder
- Department of Pediatrics, Children's Hospital of Orange County, University of California Irvine, CA, USA
| | - Lex W Doyle
- Victorian Infant Brain Studies (VIBeS), Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Australia; The Royal Women's Hospital, Melbourne, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Australia
| | - Peter J Anderson
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, Australia; Victorian Infant Brain Studies (VIBeS), Murdoch Children's Research Institute, Melbourne, Australia
| |
Collapse
|
19
|
Barrett-Young A, Ambler A, Cheyne K, Guiney H, Kokaua J, Tham YC, Williams MJA, Wilson GA, Wong TY, Poulton R. Childhood Social Isolation as a Predictor of Retinal Neuronal Thickness in Middle Age: A Lifecourse Birth Cohort Study. Psychosom Med 2023; 85:238-249. [PMID: 36800261 PMCID: PMC10073287 DOI: 10.1097/psy.0000000000001177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
OBJECTIVE We investigated whether childhood social isolation was associated with retinal neural layer changes in adulthood, and whether this association was independent of other childhood or adulthood risk factors, including adult social isolation. METHODS Participants were members of the Dunedin Multidisciplinary Health and Development Study, a longitudinal population-based birth cohort from Aotearoa New Zealand ( n = 1037), born 1972 to 1973 and followed until age 45 years, with 94% of the living cohort still participating. Social isolation was recorded prospectively at ages 5, 7, 9, and 11 years, from teacher and parent report. Retinal nerve fiber layer (RNFL) and ganglion cell-inner plexiform layer thicknesses were measured via optical coherence tomography at age 45 years. RESULTS Childhood social isolation was associated with thinner average RNFL ( B = -0.739, p = .02), nasal RNFL ( B = -1.118, p = .005), and inferior RNFL ( B = -1.524, p = .007), although only nasal RNFL remained significant after adjustment. These associations were not fully explained by other psychosocial or physical health risk factors in childhood or adulthood, nor were they mediated by adult loneliness or social support. CONCLUSIONS Childhood social isolation was an independent predictor of RNFL thickness in middle age. Highlighting prospective links between childhood psychosocial adversity and retinal neuronal measures will help to inform future research into the utility of retinal neuronal thickness as a biomarker for neurodegeneration.
Collapse
Affiliation(s)
- Ashleigh Barrett-Young
- Dunedin Multidisciplinary Health and Development Research Unit, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Antony Ambler
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Kirsten Cheyne
- Dunedin Multidisciplinary Health and Development Research Unit, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Hayley Guiney
- Dunedin Multidisciplinary Health and Development Research Unit, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Jesse Kokaua
- Dunedin Multidisciplinary Health and Development Research Unit, University of Otago, Dunedin, New Zealand
- Va’a O Tautai—Centre for Pacific Health, University of Otago, Dunedin, New Zealand
| | - Yih Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Duke-NUS Medical School, Singapore
| | | | - Graham A. Wilson
- Department of Medicine, Otago Medical School, University of Otago, Dunedin, New Zealand
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Duke-NUS Medical School, Singapore
| | - Richie Poulton
- Dunedin Multidisciplinary Health and Development Research Unit, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
20
|
Minné D, Marnewick JL, Engel-Hills P. Early Chronic Stress Induced Changes within the Locus Coeruleus in Sporadic Alzheimer's Disease. Curr Alzheimer Res 2023; 20:301-317. [PMID: 37872793 DOI: 10.2174/1567205020666230811092956] [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] [Received: 02/27/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 10/25/2023]
Abstract
Chronic exposure to stress throughout the lifespan has been the focus of many studies on Alzheimer's disease (AD) because of the similarities between the biological mechanisms involved in chronic stress and the pathophysiology of AD. In fact, the earliest abnormality associated with the disease is the presence of phosphorylated tau protein in locus coeruleus neurons, a brain structure highly responsive to stress and perceived threat. Here, we introduce allostatic load as a useful concept for understanding many of the complex, interacting neuropathological changes involved in the AD degenerative process. In response to chronic stress, aberrant tau proteins that begin to accumulate within the locus coeruleus decades prior to symptom onset appear to represent a primary pathological event in the AD cascade, triggering a wide range of interacting brain changes involving neuronal excitotoxicity, endocrine alterations, inflammation, oxidative stress, and amyloid plaque exacerbation. While it is acknowledged that stress will not necessarily be the major precipitating factor in all cases, early tau-induced changes within the locus coeruleus-norepinephrine pathway suggests that a therapeutic window might exist for preventative measures aimed at managing stress and restoring balance within the HPA axis.
Collapse
Affiliation(s)
- Donné Minné
- Applied Microbial & Health Biotechnology Institute, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
- Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| | - Jeanine L Marnewick
- Applied Microbial & Health Biotechnology Institute, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| | - Penelope Engel-Hills
- Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| |
Collapse
|
21
|
Brosens N, Samouil D, Stolker S, Katsika EV, Weggen S, Lucassen PJ, Krugers HJ. Early Life Stress Enhances Cognitive Decline and Alters Synapse Function and Interneuron Numbers in Young Male APP/PS1 Mice. J Alzheimers Dis 2023; 96:1097-1113. [PMID: 37980670 PMCID: PMC10741326 DOI: 10.3233/jad-230727] [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] [Accepted: 09/12/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Exposure to stress early in life increases the susceptibility to Alzheimer's disease (AD) pathology in aged AD mouse models. So far, the underlying mechanisms have remained elusive. OBJECTIVE To investigate 1) effects of early life stress (ELS) on early functional signs that precede the advanced neuropathological changes, and 2) correlate synaptosomal protein content with cognition to identify neural correlates of AD. METHODS APPswe/PS1dE9 mice and littermates were subjected to ELS by housing dams and pups with limited bedding and nesting material from postnatal days 2-9. At 3 months of age, an age where no cognitive loss or amyloid-β (Aβ) pathology is typically reported in this model, we assessed hippocampal Aβ pathology, synaptic strength and synapse composition and interneuron populations. Moreover, cognitive flexibility was assessed and correlated with synaptosomal protein content. RESULTS While ELS did not affect Aβ pathology, it increased synaptic strength and decreased the number of calretinin+ interneurons in the hippocampal dentate gyrus. Both genotype and condition further affected the level of postsynaptic glutamatergic protein content. Finally, APP/PS1 mice were significantly impaired in cognitive flexibility at 3 months of age, and ELS exacerbated this impairment, but only at relatively high learning criteria. CONCLUSIONS ELS reduced cognitive flexibility in young APP/PS1 mice and altered markers for synapse and network function. These findings at an early disease stage provide novel insights in AD etiology and in how ELS could increase AD susceptibility.
Collapse
Affiliation(s)
- Niek Brosens
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Dimitris Samouil
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Sabine Stolker
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Sascha Weggen
- Department of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Paul J. Lucassen
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Reemst K, Kracht L, Kotah JM, Rahimian R, van Irsen AAS, Congrains Sotomayor G, Verboon LN, Brouwer N, Simard S, Turecki G, Mechawar N, Kooistra SM, Eggen BJL, Korosi A. Early-life stress lastingly impacts microglial transcriptome and function under basal and immune-challenged conditions. Transl Psychiatry 2022; 12:507. [PMID: 36481769 PMCID: PMC9731997 DOI: 10.1038/s41398-022-02265-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Early-life stress (ELS) leads to increased vulnerability to psychiatric disorders including depression later in life. Neuroinflammatory processes have been implicated in ELS-induced negative health outcomes, but how ELS impacts microglia, the main tissue-resident macrophages of the central nervous system, is unknown. Here, we determined the effects of ELS-induced by limited bedding and nesting material during the first week of life (postnatal days [P]2-9) on microglial (i) morphology; (ii) hippocampal gene expression; and (iii) synaptosome phagocytic capacity in male pups (P9) and adult (P200) mice. The hippocampus of ELS-exposed adult mice displayed altered proportions of morphological subtypes of microglia, as well as microglial transcriptomic changes related to the tumor necrosis factor response and protein ubiquitination. ELS exposure leads to distinct gene expression profiles during microglial development from P9 to P200 and in response to an LPS challenge at P200. Functionally, synaptosomes from ELS-exposed mice were phagocytosed less by age-matched microglia. At P200, but not P9, ELS microglia showed reduced synaptosome phagocytic capacity when compared to control microglia. Lastly, we confirmed the ELS-induced increased expression of the phagocytosis-related gene GAS6 that we observed in mice, in the dentate gyrus of individuals with a history of child abuse using in situ hybridization. These findings reveal persistent effects of ELS on microglial function and suggest that altered microglial phagocytic capacity is a key contributor to ELS-induced phenotypes.
Collapse
Affiliation(s)
- Kitty Reemst
- grid.7177.60000000084992262Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH The Netherlands
| | - Laura Kracht
- grid.4494.d0000 0000 9558 4598Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Janssen M. Kotah
- grid.7177.60000000084992262Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH The Netherlands
| | - Reza Rahimian
- grid.412078.80000 0001 2353 5268McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal, QC H4H 1R3 Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC H3A 1A1 Canada
| | - Astrid A. S. van Irsen
- grid.7177.60000000084992262Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH The Netherlands
| | - Gonzalo Congrains Sotomayor
- grid.7177.60000000084992262Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH The Netherlands
| | - Laura N. Verboon
- grid.7177.60000000084992262Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH The Netherlands
| | - Nieske Brouwer
- grid.4494.d0000 0000 9558 4598Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sophie Simard
- grid.412078.80000 0001 2353 5268McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal, QC H4H 1R3 Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC H3A 1A1 Canada
| | - Gustavo Turecki
- grid.412078.80000 0001 2353 5268McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal, QC H4H 1R3 Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC H3A 1A1 Canada
| | - Naguib Mechawar
- grid.412078.80000 0001 2353 5268McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal, QC H4H 1R3 Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC H3A 1A1 Canada
| | - Susanne M. Kooistra
- grid.4494.d0000 0000 9558 4598Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Bart J. L. Eggen
- grid.4494.d0000 0000 9558 4598Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity Group, University of Amsterdam, Amsterdam, Science Park 904, 1098 XH, The Netherlands.
| |
Collapse
|
23
|
Reemst K, Broos JY, Abbink MR, Cimetti C, Giera M, Kooij G, Korosi A. Early-life stress and dietary fatty acids impact the brain lipid/oxylipin profile into adulthood, basally and in response to LPS. Front Immunol 2022; 13:967437. [PMID: 36131915 PMCID: PMC9484596 DOI: 10.3389/fimmu.2022.967437] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/04/2022] [Indexed: 01/06/2023] Open
Abstract
Brain lipid dysregulation is a hallmark of depression and Alzheimer's disease, also marked by chronic inflammation. Early-life stress (ELS) and dietary intake of polyunsaturated fatty acids (PUFAs) are risk factors for these pathologies and are known to impact inflammatory processes. However, if these early-life factors alter brain lipid homeostasis on the long-term and thereby contribute to this risk remains to be elucidated. We have recently shown that an early diet enriched in omega(ω)-3 PUFAs protected against the long-term negative effects of ELS on cognition and neuroinflammation. Here, we aim to understand if modulation of brain lipid and oxylipin profiles contributes to the detrimental effects of ELS and the protective ones of the diet. We therefore studied if and how ELS and early dietary PUFAs modulate the brain lipid and oxylipin profile, basally as well as in response to an inflammatory challenge, to unmask possible latent effects. Male mice were exposed to ELS via the limited bedding and nesting paradigm, received an early diet with high or low ω6/ω3 ratio (HRD and LRD) and were injected with saline or lipopolysaccharide (LPS) in adulthood. Twenty-four hours later plasma cytokines (Multiplex) and hypothalamic lipids and oxylipins (liquid chromatography tandem mass spectrometry) were measured. ELS exacerbated the LPS-induced increase in IL-6, CXCL1 and CCL2. Both ELS and diet affected the lipid/oxylipin profile long-term. For example, ELS increased diacylglycerol and LRD reduced triacylglycerol, free fatty acids and ceramides. Importantly, the ELS-induced alterations were strongly influenced by the early diet. For example, the ELS-induced decrease in eicosapentaenoic acid was reversed when fed LRD. Similarly, the majority of the LPS-induced alterations were distinct for control and ELS exposed mice and unique for mice fed with LRD or HRD. LPS decreased ceramides and lysophosphotidylcholine, increased hexosylceramides and prostaglandin E2, reduced triacylglycerol species and ω6-derived oxylipins only in mice fed LRD and ELS reduced the LPS-induced increase in phosphatidylcholine. These data give further insights into the alterations in brain lipids and oxylipins that might contribute to the detrimental effects of ELS, to the protective ones of LRD and the possible early-origin of brain lipid dyshomeostasis characterizing ELS-related psychopathologies.
Collapse
Affiliation(s)
- Kitty Reemst
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park, Amsterdam, Netherlands
| | - Jelle Y. Broos
- Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Multiple Sclerosis (MS) Center Amsterdam, Amsterdam, Netherlands,Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Maralinde R. Abbink
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park, Amsterdam, Netherlands
| | - Chiara Cimetti
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park, Amsterdam, Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Gijs Kooij
- Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Multiple Sclerosis (MS) Center Amsterdam, Amsterdam, Netherlands
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park, Amsterdam, Netherlands,*Correspondence: Aniko Korosi,
| |
Collapse
|
24
|
Bachiller S, Hidalgo I, Garcia MG, Boza-Serrano A, Paulus A, Denis Q, Haikal C, Manouchehrian O, Klementieva O, Li JY, Pronk CJ, Gouras GK, Deierborg T. Early-life stress elicits peripheral and brain immune activation differently in wild type and 5xFAD mice in a sex-specific manner. J Neuroinflammation 2022; 19:151. [PMID: 35705972 PMCID: PMC9199174 DOI: 10.1186/s12974-022-02515-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 06/02/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The risk of developing Alzheimer's disease (AD) is modulated by genetic and environmental factors. Early-life stress (ELS) exposure during critical periods of brain development can impact later brain function and health, including increasing the risk of developing AD. Microglial dysfunction and neuroinflammation have been implicated as playing a role in AD pathology and may be modulated by ELS. To complicate matters further, sex-specific effects have been noted in response to ELS and in the incidence and progression of AD. METHODS Here, we subjected male and female mice with either a wild type or 5xFAD familial AD-model background to maternal separation (MS) from postnatal day 2 to 14 to induce ELS. RESULTS We detected hippocampal neuroinflammatory alterations already at postnatal day 15. By 4 months of age, MS mice presented increased immobility time in the forced swim test and a lower discrimination index in the novel object recognition memory test compared to controls. We found altered Bdnf and Arc expression in the hippocampus and increased microglial activation in the prefrontal cortex due to MS in a sex-dependent manner. In 5xFAD mice specifically, MS exacerbated amyloid-beta deposition, particularly in females. In the periphery, the immune cell population was altered by MS exposure. CONCLUSION Overall, our results demonstrate that MS has both short- and long-term effects on brain regions related to memory and on the inflammatory system, both in the brain and periphery. These ELS-related effects that are detectable even in adulthood may exacerbate pathology and increase the risk of developing AD via sex-specific mechanisms.
Collapse
Affiliation(s)
- S. Bachiller
- grid.4514.40000 0001 0930 2361Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden ,grid.411109.c0000 0000 9542 1158Present Address: Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocío University Hospital, CSIC, University of Sevilla, Seville, Spain
| | - I. Hidalgo
- grid.4514.40000 0001 0930 2361Division of Molecular Hematology, Institution of Laboratory Medicine, Lund University, Lund, Sweden
| | - M. G. Garcia
- grid.4514.40000 0001 0930 2361Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden ,grid.4514.40000 0001 0930 2361Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - A. Boza-Serrano
- grid.4514.40000 0001 0930 2361Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden ,grid.9224.d0000 0001 2168 1229Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - A. Paulus
- grid.4514.40000 0001 0930 2361Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden ,grid.4514.40000 0001 0930 2361Medical Microspectroscopy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Q. Denis
- grid.4514.40000 0001 0930 2361Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - C. Haikal
- grid.4514.40000 0001 0930 2361Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - O. Manouchehrian
- grid.4514.40000 0001 0930 2361Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - O. Klementieva
- grid.4514.40000 0001 0930 2361Medical Microspectroscopy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - J. Y. Li
- grid.4514.40000 0001 0930 2361Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - C. J. Pronk
- grid.4514.40000 0001 0930 2361Division of Molecular Hematology, Institution of Laboratory Medicine, Lund University, Lund, Sweden
| | - G. K. Gouras
- grid.4514.40000 0001 0930 2361Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - T. Deierborg
- grid.4514.40000 0001 0930 2361Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
25
|
Niedens M, Yeager A, Vidoni ED, Barton K, Perales-Puchalt J, Dealey RP, Quinn D, Gage LA. A Collaborative Approach to Dementia Inclusion in Social Work Education: The Dementia Intensive. JOURNAL OF SOCIAL WORK EDUCATION 2022; 59:493-505. [PMID: 37397072 PMCID: PMC10309142 DOI: 10.1080/10437797.2022.2039820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 07/04/2023]
Abstract
There are 5.8 million Americans with Alzheimer's disease and this number is rising. Social Work can play a key role. Yet, like other disciplines, the field is ill prepared for the growing number of individuals and family members who are impacted physically, emotionally and financially. Compounding the challenge, the number of social work students identifying interest in the field is low. This mixed methods concurrent study assessed the preliminary efficacy of a day-long education event among social work students from eight social work programs. Pre- post-training survey included: 1) dementia knowledge, assessed with the Dementia Knowledge Assessment Scale, and 2) negative attitudes towards dementia, assessed by asking students to identify three words that reflected their thoughts on dementia, which were later rated as positive, negative or neutral by three external raters. Bivariate analyses showed that dementia knowledge (mean difference= 9.9) and attitudes (10% lower) improved from pre- to post-training (p<0.05). Collaboration between social work programs can increase student access to strength-based dementia education. Such programs hold the potential of improving dementia capability within the field of Social Work.
Collapse
Affiliation(s)
| | - Amy Yeager
- KU Alzheimer's Disease Research Center, Department of Neurology
| | - Eric D Vidoni
- KU Alzheimer's Disease Research Center, Department of Neurology
| | | | | | | | - Dory Quinn
- Pittsburg State University, Department of History, Philosophy & Social Sciences
| | - L Ashley Gage
- University of Central Missouri, Department of Social Work
| |
Collapse
|
26
|
Graham KL, Paun O, Stillerman A. The Impact of Adverse Childhood Experiences on Cognition in African American Older Adults: An Integrated Literature Review. Res Gerontol Nurs 2021; 14:265-272. [PMID: 34542345 DOI: 10.3928/19404921-20210825-04] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The current integrative literature review examined the existing evidence on the connection between adverse childhood experiences (ACEs) and cognitive changes in African American older adults. Using the Covidence platform, several databases were searched, resulting in 266 publications dated 2008-2020. Ten articles met inclusion criteria and were reviewed. Findings indicate that four ACEs (physical, sexual, and verbal abuse, and low socioeconomic status) are associated with impaired cognition in African American older adults. Four gaps were identified: lack of (a) older adult participants in research investigating original and expanded ACEs; (b) exclusively African American samples of participants in studies examining the relationship between ACEs and cognition; (c) consensus about what specific ACEs contribute to changes in cognition in older adults; and (d) information about successful interventions created to prevent and mitigate the effects of ACEs in older adults. This review provides a synthesis of the limited evidence on the effects of ACEs on cognition among other outcomes. Findings on the effects of ACEs on African American older adults' cognition are limited, thus making a compelling case for further investigating the role of childhood adversity in the disparity of cognitive changes in African American communities. [Research in Gerontological Nursing, 14(5), 265-272.].
Collapse
|
27
|
Sierra-Fonseca JA, Hamdan JN, Cohen AA, Cardenas SM, Saucedo S, Lodoza GA, Gosselink KL. Neonatal Maternal Separation Modifies Proteostasis Marker Expression in the Adult Hippocampus. Front Mol Neurosci 2021; 14:661993. [PMID: 34447296 PMCID: PMC8383781 DOI: 10.3389/fnmol.2021.661993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 07/05/2021] [Indexed: 01/01/2023] Open
Abstract
Exposure to early-life stress (ELS) can persistently modify neuronal circuits and functions, and contribute to the expression of misfolded and aggregated proteins that are hallmarks of several neurodegenerative diseases. The healthy brain is able to clear dysfunctional proteins through the ubiquitin-proteasome system (UPS) and the autophagy-lysosomal pathway (ALP). Accumulating evidence indicates that impairment of these pathways contributes to enhanced protein aggregation and neurodegeneration. While stress is a known precipitant of neurological decline, few specific mechanistic links underlying this relationship have been identified. We hypothesized that neonatal maternal separation (MatSep), a well-established model of ELS, has the ability to alter the levels of UPS and ALP components in the brain, and thus has the potential to disrupt proteostasis. The expression of proteostasis-associated protein markers was evaluated by immunoblotting in the hippocampus and cortex of adult Wistar rats that were previously subjected to MatSep. We observed multiple sex- and MatSep-specific changes in the expression of proteins in the ALP, mitophagy, and UPS pathways, particularly in the hippocampus of adult animals. In contrast, MatSep had limited influence on proteostasis marker expression in the cortex of adult animals. Our results indicate that MatSep can selectively modify the intracellular protein degradation machinery in ways that may impact the development and progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Jorge A Sierra-Fonseca
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States
| | - Jameel N Hamdan
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States
| | - Alexis A Cohen
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States.,Neuroscience Program, Smith College, Northampton, MA, United States
| | - Sonia M Cardenas
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States
| | - Sigifredo Saucedo
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States
| | - Gabriel A Lodoza
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States
| | - Kristin L Gosselink
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States.,Department of Physiology and Pathology, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| |
Collapse
|
28
|
Early Life Stress and Metabolic Plasticity of Brain Cells: Impact on Neurogenesis and Angiogenesis. Biomedicines 2021; 9:biomedicines9091092. [PMID: 34572278 PMCID: PMC8470044 DOI: 10.3390/biomedicines9091092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/15/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
Early life stress (ELS) causes long-lasting changes in brain plasticity induced by the exposure to stress factors acting prenatally or in the early postnatal ontogenesis due to hyperactivation of hypothalamic-pituitary-adrenal axis and sympathetic nervous system, development of neuroinflammation, aberrant neurogenesis and angiogenesis, and significant alterations in brain metabolism that lead to neurological deficits and higher susceptibility to development of brain disorders later in the life. As a key component of complex pathogenesis, ELS-mediated changes in brain metabolism associate with development of mitochondrial dysfunction, loss of appropriate mitochondria quality control and mitochondrial dynamics, deregulation of metabolic reprogramming. These mechanisms are particularly critical for maintaining the pool and development of brain cells within neurogenic and angiogenic niches. In this review, we focus on brain mitochondria and energy metabolism related to tightly coupled neurogenic and angiogenic events in healthy and ELS-affected brain, and new opportunities to develop efficient therapeutic strategies aimed to restore brain metabolism and reduce ELS-induced impairments of brain plasticity.
Collapse
|
29
|
Milligan Armstrong A, Porter T, Quek H, White A, Haynes J, Jackaman C, Villemagne V, Munyard K, Laws SM, Verdile G, Groth D. Chronic stress and Alzheimer's disease: the interplay between the hypothalamic-pituitary-adrenal axis, genetics and microglia. Biol Rev Camb Philos Soc 2021; 96:2209-2228. [PMID: 34159699 DOI: 10.1111/brv.12750] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/21/2022]
Abstract
Chronic psychosocial stress is increasingly being recognised as a risk factor for sporadic Alzheimer's disease (AD). The hypothalamic-pituitary-adrenal axis (HPA axis) is the major stress response pathway in the body and tightly regulates the production of cortisol, a glucocorticoid hormone. Dysregulation of the HPA axis and increased levels of cortisol are commonly found in AD patients and make a major contribution to the disease process. The underlying mechanisms remain poorly understood. In addition, within the general population there are interindividual differences in sensitivities to glucocorticoid and stress responses, which are thought to be due to a combination of genetic and environmental factors. These differences could ultimately impact an individuals' risk of AD. The purpose of this review is first to summarise the literature describing environmental and genetic factors that can impact an individual's HPA axis reactivity and function and ultimately AD risk. Secondly, we propose a mechanism by which genetic factors that influence HPA axis reactivity may also impact inflammation, a key driver of neurodegeneration. We hypothesize that these factors can mediate glucocorticoid priming of the immune cells of the brain, microglia, to become pro-inflammatory and promote a neurotoxic environment resulting in neurodegeneration. Understanding the underlying molecular mechanisms and identifying these genetic factors has implications for evaluating stress-related risk/progression to neurodegeneration, informing the success of interventions based on stress management and potential risks associated with the common use of glucocorticoids.
Collapse
Affiliation(s)
- Ayeisha Milligan Armstrong
- Curtin Health Innovation Research Institute, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia
| | - Tenielle Porter
- Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
| | - Hazel Quek
- QIMR Berghofer Medical Institute, 300 Herston Rd, Herston, QLD, Australia
| | - Anthony White
- QIMR Berghofer Medical Institute, 300 Herston Rd, Herston, QLD, Australia
| | - John Haynes
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Connie Jackaman
- Curtin Health Innovation Research Institute, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia
| | - Victor Villemagne
- School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Kylie Munyard
- Curtin Health Innovation Research Institute, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia
| | - Simon M Laws
- Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
| | - Giuseppe Verdile
- Curtin Health Innovation Research Institute, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia.,School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
| | - David Groth
- Curtin Health Innovation Research Institute, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia
| |
Collapse
|
30
|
Solarz A, Majcher-Maślanka I, Kryst J, Chocyk A. A Search for Biomarkers of Early-life Stress-related Psychopathology: Focus on 70-kDa Heat Shock Proteins. Neuroscience 2021; 463:238-253. [PMID: 33662529 DOI: 10.1016/j.neuroscience.2021.02.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/12/2021] [Accepted: 02/21/2021] [Indexed: 12/31/2022]
Abstract
Clinical studies clearly indicate that early-life stress (ELS) may cause physical and mental health problems later in life. Therefore, the identification of universal biomarkers of ELS-related diseases is very important. The 70-kDa heat shock proteins (HSP70s), specifically HSPA5 and HSPA1B, have been recently shown to be potentially associated with occurrence of anxiety, mood disorders, and schizophrenia; thus, we hypothesized that HSP70s are potential candidate biomarkers of ELS-induced psychopathologies. A maternal separation (MS) procedure in rats was used to model ELS, and the expression of HSPA5 and HSPA1B was investigated in the blood, medial prefrontal cortex (mPFC), and hippocampus of juvenile, preadolescent, and adult animals. We also studied the effects of MS on the long-term potentiation (LTP) and behavioral phenotypes of adult rats. We found that MS enhanced the expression of HSPA1B mRNA in the blood and mPFC of juvenile and preadolescent rats. This increase was accompanied by an increase in the HSPA1A/1B protein levels in the mPFC and hippocampus of juvenile rats that persisted in the mPFC until adulthood. MS juvenile and adult rats showed enhanced HSPA5 mRNA expression in the blood and increased HSPA5 protein expression in the mPFC (juveniles) and hippocampus (adults). Concurrently, MS adult rats exhibited aberrations in LTP in the mPFC and hippocampus and a less anxious behavioral phenotype. These results indicate that MS may produce enduring overexpression of HSPA1B and HSPA5 in the brain and blood. Therefore, both HSP70 family members may be potential candidate peripheral and brain biomarkers of ELS-induced changes in brain functioning.
Collapse
Affiliation(s)
- Anna Solarz
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland
| | - Iwona Majcher-Maślanka
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland
| | - Joanna Kryst
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland
| | - Agnieszka Chocyk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland.
| |
Collapse
|
31
|
Chi H, Zhai Q, Zhang M, Su D, Cao W, Li W, She X, Yang H, Wang K, Gao X, Ma K, Cui B, Qiu Y. APP/PS1 Gene-Environment Noise Interaction Aggravates AD-like Neuropathology in Hippocampus Via Activation of the VDAC1 Positive Feedback Loop. Curr Alzheimer Res 2021; 18:14-24. [PMID: 33761858 DOI: 10.2174/1567205018666210324114153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 09/03/2020] [Accepted: 02/16/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Environmental risk factors, including environmental noise stress, and genetic factors, have been associated with the occurrence and development of Alzheimer's disease (AD). However, the exact role and mechanism of AD-like pathology induced by environment-gene interactions between environmental noise and APP/PS1 gene remain elusive. METHODS Herein, we investigated the impact of chronic noise exposure on AD-like neuropathology in APP/PS1 transgenic mice. The Morris water maze (MWM) task was conducted to evaluate AD-like changes. The hippocampal phosphorylated Tau, amyloid-β (Aβ), and neuroinflammation were assessed. We also assessed changes in positive feedback loop signaling of the voltage-dependent anion channel 1 (VDAC1) to explore the potential underlying mechanism linking AD-like neuropathology to noise-APP/PS1 interactions. RESULTS Long-term noise exposure significantly increased the escape latency and the number of platform crossings in the MWM task. The Aβ overproduction was induced in the hippocampus of APP/PS1 mice, along with the increase of Tau phosphorylation at Ser396 and Thr231 and the increase of the microglia and astrocytes markers expression. Moreover, the VDAC1-AKT (protein kinase B)-GSK3β (glycogen synthase kinase 3 beta)-VDAC1 signaling pathway was abnormally activated in the hippocampus of APP/PS1 mice after noise exposure. CONCLUSION Chronic noise exposure and APP/PS1 overexpression may synergistically exacerbate cognitive impairment and neuropathological changes that occur in AD. This interaction may be mediated by the positive feedback loop of the VDAC1-AKT-GSK3β-VDAC1 signaling pathway.
Collapse
Affiliation(s)
- Huimin Chi
- Weifang Medical University, Weifang,China
| | | | - Ming Zhang
- Tianjin Centers for Disease Control and Prevention, Tianjin,China
| | - Donghong Su
- Tianjin Institute of Environmental and Operational Medicine, Tianjin,China
| | - Wa Cao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin,China
| | - Wenlong Li
- Weifang Medical University, Weifang,China
| | - Xiaojun She
- Tianjin Institute of Environmental and Operational Medicine, Tianjin,China
| | - Honglian Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin,China
| | - Kun Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin,China
| | - Xiujie Gao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin,China
| | - Kefeng Ma
- Tianjin Institute of Environmental and Operational Medicine, Tianjin,China
| | - Bo Cui
- Tianjin Institute of Environmental and Operational Medicine, Tianjin,China
| | - Yugang Qiu
- Weifang Medical University, Weifang,China
| |
Collapse
|
32
|
Gomes PRL, Motta-Teixeira LC, Gallo CC, Carmo Buonfiglio DD, Camargo LSD, Quintela T, Reiter RJ, Amaral FGD, Cipolla-Neto J. Maternal pineal melatonin in gestation and lactation physiology, and in fetal development and programming. Gen Comp Endocrinol 2021; 300:113633. [PMID: 33031801 DOI: 10.1016/j.ygcen.2020.113633] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/20/2020] [Indexed: 12/21/2022]
Abstract
Pregnancy and lactation are reproductive processes that rely on physiological adaptations that should be timely and adequately triggered to guarantee both maternal and fetal health. Pineal melatonin is a hormone that presents daily and seasonal variations that synchronizes the organism's physiology to the different demands across time through its specific mechanisms and ways of action. The reproductive system is a notable target for melatonin as it actively participates on reproductive physiology and regulates the hypothalamus-pituitary-gonads axis, influencing gonadotropins and sexual hormones synthesis and release. For its antioxidant properties, melatonin is also vital for the oocytes and spermatozoa quality and viability, and for blastocyst development. Maternal pineal melatonin blood levels increase during pregnancy and triggers the maternal physiological alterations in energy metabolism both during pregnancy and lactation to cope with the energy demands of both periods and to promote adequate mammary gland development. Moreover, maternal melatonin freely crosses the placenta and is the only source of this hormone to the fetus. It importantly times the conceptus physiology and influences its development and programing of several functions that depend on neural and brain development, ultimately priming adult behavior and energy and glucose metabolism. The present review aims to explain the above listed melatonin functions, including the potential alterations observed in the progeny gestated under maternal chronodisruption and/or hypomelatoninemia.
Collapse
Affiliation(s)
- Patrícia Rodrigues Lourenço Gomes
- Neurobiology Lab, Department of Physiology and Biophysics, 1524 Prof. Lineu Prestes Ave., Institute of Biomedical Sciences, Bldg 1, Lab 118, University of São Paulo, São Paulo 05508-000, Brazil
| | - Lívia Clemente Motta-Teixeira
- Neurobiology Lab, Department of Physiology and Biophysics, 1524 Prof. Lineu Prestes Ave., Institute of Biomedical Sciences, Bldg 1, Lab 118, University of São Paulo, São Paulo 05508-000, Brazil
| | - Camila Congentino Gallo
- Pineal Neurobiology Lab, Department of Physiology, 862 Botucatu St., 5th floor, Federal University of São Paulo, São Paulo 04023-901, Brazil.
| | - Daniella do Carmo Buonfiglio
- Neurobiology Lab, Department of Physiology and Biophysics, 1524 Prof. Lineu Prestes Ave., Institute of Biomedical Sciences, Bldg 1, Lab 118, University of São Paulo, São Paulo 05508-000, Brazil
| | - Ludmilla Scodeler de Camargo
- Pineal Neurobiology Lab, Department of Physiology, 862 Botucatu St., 5th floor, Federal University of São Paulo, São Paulo 04023-901, Brazil.
| | - Telma Quintela
- CICS-UBI - Health Sciences Research Center, Infante D. Henrique Ave, University of Beira Interior, Covilhã 6200-506, Portugal.
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, 7703 Floyd Curl Drive, UT Health San Antonio, San Antonio, TX 78229, USA.
| | - Fernanda Gaspar do Amaral
- Pineal Neurobiology Lab, Department of Physiology, 862 Botucatu St., 5th floor, Federal University of São Paulo, São Paulo 04023-901, Brazil.
| | - José Cipolla-Neto
- Neurobiology Lab, Department of Physiology and Biophysics, 1524 Prof. Lineu Prestes Ave., Institute of Biomedical Sciences, Bldg 1, Lab 118, University of São Paulo, São Paulo 05508-000, Brazil.
| |
Collapse
|
33
|
Richardson R, Bowers J, Callaghan BL, Baker KD. Does maternal separation accelerate maturation of perineuronal nets and parvalbumin-containing inhibitory interneurons in male and female rats? Dev Cogn Neurosci 2020; 47:100905. [PMID: 33385787 PMCID: PMC7786030 DOI: 10.1016/j.dcn.2020.100905] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 12/14/2020] [Accepted: 12/22/2020] [Indexed: 11/22/2022] Open
Abstract
Maternal separation did not accelerate maturation of PNNs in amygdala or PFC. Maternal separation did not affect PV density in infant and juveniles. No sex differences were observed in effects of maternal separation on PNNs or PV. Impact of early adversity may be more easily seen with functional neural measures.
Early life adversity impacts on a range of emotional, cognitive, and psychological processes. A recent theoretical model suggests that at least some of these effects are due to accelerated maturation of specific physiological systems and/or neural circuits. For example, maternal separation (MS), a model of early life adversity in rodents, accelerates maturation of memory systems, and here we examined its impact on maturation of perineuronal nets (PNNs) and parvalbumin (PV)-containing inhibitory interneurons. PNNs are specialized extracellular matrix structures suggested to be involved in stabilizing long-term memories and in the closure of a sensitive period in memory development. PV-containing inhibitory interneurons are the type of cell that PNNs preferentially surround, and are also thought to be involved in memory. In Experiment 1, with male rats, there was an increase in PNNs in both the amygdala and prefrontal cortex with age from infancy to juvenility. Contrary to prediction, MS had no impact on either PNN or PV expression. The same pattern was observed in female rats in Experiment 2. Taken together, these data show that the early maturation of memory in MS infants is not due to an accelerated maturation of PNNs or PV-containing cells in either the amygdala or prefrontal cortex.
Collapse
Affiliation(s)
| | - Jeremy Bowers
- School of Psychology, UNSW Sydney, NSW, 2052, Australia
| | - Bridget L Callaghan
- Department of Psychology, University of California - Los Angeles, Los Angeles, CA, United States
| | | |
Collapse
|
34
|
Karem H, Mehla J, Kolb BE, Mohajerani MH. Traffic noise exposure, cognitive decline, and amyloid-beta pathology in an AD mouse model. Synapse 2020; 75:e22192. [PMID: 33096582 DOI: 10.1002/syn.22192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 11/11/2022]
Abstract
Concerns are growing that exposure to environmental pollutants, such as traffic noise, might cause cognitive impairments and predispose individuals toward the development of Alzheimer's disease (AD) dementia. In this study in a knock-in mouse model of AD, we investigated how chronic traffic noise exposure (CTNE) impacts cognitive performance and amyloid-beta (Aβ) pathology. A group of APPNL-G-F/NL-G-F mice was exposed to CTNE (70 dBA , 8 hr/day for 1 month) and compared with nonexposed counterparts. Following CTNE, an increase in hypothalamic-pituitary-adrenal (HPA) axis responsivity was observed by corticosterone assay of the blood. One month after CTNE, the CTNE group demonstrated impairments in cognitive and motor functions, and indications of anxiety-like behavior, relative to the control animals. The noise-exposed group also showed elevated Aβ aggregation, as inferred by a greater number of plaques and larger average plaque size in various regions of the brain, including regions involved in stress regulation. The results support that noise-associated dysregulation of the neuroendocrine system as a potential risk factor for developing cognitive impairment and Aβ pathology, which should be further investigated in human studies.
Collapse
Affiliation(s)
- Hadil Karem
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Jogender Mehla
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Bryan E Kolb
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
35
|
Jafari Z, Mehla J, Kolb BE, Mohajerani MH. Gestational Stress Augments Postpartum β-Amyloid Pathology and Cognitive Decline in a Mouse Model of Alzheimer's Disease. Cereb Cortex 2020; 29:3712-3724. [PMID: 30561536 DOI: 10.1093/cercor/bhy251] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/02/2018] [Indexed: 12/14/2022] Open
Abstract
Besides well-known risk factors for Alzheimer's disease (AD), stress, and in particular noise stress (NS), is a lifestyle risk factor common today. It is known that females are at a significantly greater risk of developing AD than males, and given that stress is a common adversity in females during pregnancy, we hypothesized that gestational noise exposure could exacerbate the postpartum development of the AD-like neuropathological changes during the life span. Pregnant APPNL-G-F/NL-G-F mice were randomly assigned to either the stress condition or control group. The stress group was exposed to the NS on gestational days 12-16, which resulted in a markedly higher hypothalamic-pituitary-adrenal (HPA) axis responsivity during the postpartum stage. Higher amyloid-β (Aβ) deposition and larger Aβ plaque size in the olfactory area were the early onset impacts of the gestational stress (GS) seen at the age of 4 months. This pattern of increased Aβ aggregation and larger plaque size were observed in various brain areas involved in both AD and stress regulation, especially in limbic structures, at the age of 6 months. The GS also produced anxiety-like behavior, deficits in learning and memory, and impaired motor coordination. The findings suggest that environmental stresses during pregnancy pose a potential risk factor in accelerating postpartum cognitive decline and AD-like neuropathological changes in the dams (mothers) later in life.
Collapse
Affiliation(s)
- Zahra Jafari
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience (CCBN), University of Lethbridge, Lethbridge, Canada.,Department of Basic Sciences in Rehabilitation, School of Rehabilitation Sciences, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Jogender Mehla
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience (CCBN), University of Lethbridge, Lethbridge, Canada
| | - Bryan E Kolb
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience (CCBN), University of Lethbridge, Lethbridge, Canada
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience (CCBN), University of Lethbridge, Lethbridge, Canada
| |
Collapse
|
36
|
Jafari Z, Kolb BE, Mohajerani MH. Noise exposure accelerates the risk of cognitive impairment and Alzheimer’s disease: Adulthood, gestational, and prenatal mechanistic evidence from animal studies. Neurosci Biobehav Rev 2020; 117:110-128. [DOI: 10.1016/j.neubiorev.2019.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/18/2019] [Accepted: 04/02/2019] [Indexed: 12/25/2022]
|
37
|
Conde-Sala JL, Garre-Olmo J. Early parental death and psychosocial risk factors for dementia: A case-control study in Europe. Int J Geriatr Psychiatry 2020; 35:1051-1059. [PMID: 32392630 DOI: 10.1002/gps.5328] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/29/2020] [Accepted: 05/03/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To assess the association between early parental death and the risk of dementia in adult life and to examine the risk factors associated with early parental death in people with and without dementia. METHODS/DESIGN A population-based case-control study of a sample of 65 997 participants from the Survey of Health, Ageing and Retirement in Europe study. Early parental death was operationalized as parental death at the age of ≤16 years. Main analyses were conducted using bivariate and multivariate logistic regression analyses. RESULTS The odds ratio (OR) for dementia in individuals who experienced early parental death (father or mother) at the age of ≤16 years was 1.83 (95%CI 1.61-2.09) and 1.54 (95%CI 1.35-1.76) adjusted for age, gender and education. In the multivariate logistic regression analysis carried out with the whole sample, early parental death increased the risk of dementia (OR = 1.50, 95%CI 1.31-1.72), along with older age (OR = 5.92, 95%CI 4.86-7.17), neuroticism (OR = 2.94, 95%CI 2.61-3.31), low education level (OR = 1.84, 95%CI 1.64-2.05) and low income (OR = 1.49, 95%CI 1.34-1.67). DISCUSSION Early parental death (≤16 years) was associated with an increased risk of dementia. We discuss the neurobiological markers associated with adverse childhood experiences (ACEs) and dementia as well as interventions to counteract the negative health effects on adults. J Am Geriatr Soc 68:-, 2020.
Collapse
Affiliation(s)
- Josep L Conde-Sala
- Institute of Neurosciences, University of Barcelona, Catalonia, Spain.,Aging, Disability and Health Research Group, Girona Biomedical Research Institute (IdIBGi), Catalonia, Spain
| | - Josep Garre-Olmo
- Aging, Disability and Health Research Group, Girona Biomedical Research Institute (IdIBGi), Catalonia, Spain.,Department of Medical Sciences, University of Girona, Spain
| |
Collapse
|
38
|
Abbink MR, Kotah JM, Hoeijmakers L, Mak A, Yvon-Durocher G, van der Gaag B, Lucassen PJ, Korosi A. Characterization of astrocytes throughout life in wildtype and APP/PS1 mice after early-life stress exposure. J Neuroinflammation 2020; 17:91. [PMID: 32197653 PMCID: PMC7083036 DOI: 10.1186/s12974-020-01762-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background Early-life stress (ES) is an emerging risk factor for later life development of Alzheimer’s disease (AD). We have previously shown that ES modulates amyloid-beta pathology and the microglial response to it in the APPswe/PS1dE9 mouse model. Because astrocytes are key players in the pathogenesis of AD, we studied here if and how ES affects astrocytes in wildtype (WT) and APP/PS1 mice and how these relate to the previously reported amyloid pathology and microglial profile. Methods We induced ES by limiting nesting and bedding material from postnatal days (P) 2–9. We studied in WT mice (at P9, P30, and 6 months) and in APP/PS1 mice (at 4 and 10 months) (i) GFAP coverage, cell density, and complexity in hippocampus (HPC) and entorhinal cortex (EC); (ii) hippocampal gene expression of astrocyte markers; and (iii) the relationship between astrocyte, microglia, and amyloid markers. Results In WT mice, ES increased GFAP coverage in HPC subregions at P9 and decreased it at 10 months. APP/PS1 mice at 10 months exhibited both individual cell as well as clustered GFAP signals. APP/PS1 mice when compared to WT exhibited reduced total GFAP coverage in HPC, which is increased in the EC, while coverage of the clustered GFAP signal in the HPC was increased and accompanied by increased expression of several astrocytic genes. While measured astrocytic parameters in APP/PS1 mice appear not be further modulated by ES, analyzing these in the context of ES-induced alterations to amyloid pathology and microglial shows alterations at both 4 and 10 months of age. Conclusions Our data suggest that ES leads to alterations to the astrocytic response to amyloid-β pathology.
Collapse
Affiliation(s)
- Maralinde R Abbink
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Janssen M Kotah
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Lianne Hoeijmakers
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Aline Mak
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Genevieve Yvon-Durocher
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Bram van der Gaag
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Aniko Korosi
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands.
| |
Collapse
|
39
|
Impact of Maternal Separation on Dopamine System and its Association with Parkinson's Disease. Neuromolecular Med 2020; 22:335-340. [PMID: 31933131 DOI: 10.1007/s12017-019-08587-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/25/2019] [Indexed: 01/09/2023]
Abstract
As a type of stress, maternal separation (MS) has been one of the most widely used models in neuropsychiatric research. An increasing number of studies has found that MS not only affects the function of the hypothalamic-pituitary-adrenal axis and hippocampal 5-hydroxytryptamine system, but also causes dysfunction of the central dopamine (DA) system and increases the susceptibility of dopaminergic neurons to pathogenic factors of Parkinson's disease (PD), for instance, 6-hydroxydopamine, thus impairing motor function. We reviewed the impact of MS on the DA system and its correlation with PD and found the following: (1) discrepant effects of MS on the DA system have been reported; (2) MS is a good model to study the impact of stress on the occurrence and development of PD, however, unified modeling criteria of MS are required; (3) correlation between MS and PD may involve the impact of MS on the DA system, which however is not the only connection; (4) intervening measures can block pathways between MS and PD, which provides reference for the prevention of PD in specific populations such as left-behind children.
Collapse
|
40
|
Microglia, Lifestyle Stress, and Neurodegeneration. Immunity 2020; 52:222-240. [PMID: 31924476 DOI: 10.1016/j.immuni.2019.12.003] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/26/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023]
Abstract
Recent years have witnessed a revolution in our understanding of microglia biology, including their major role in the etiology and pathogenesis of neurodegenerative diseases. Technological advances have enabled the identification of microglial signatures in health and disease, including the development of new models to investigate and manipulate human microglia in vivo in the context of disease. In parallel, genetic association studies have identified several gene risk factors associated with Alzheimer's disease that are specifically or highly expressed by microglia in the central nervous system (CNS). Here, we discuss evidence for the effect of stress, diet, sleep patterns, physical activity, and microbiota composition on microglia biology and consider how lifestyle might influence an individual's predisposition to neurodegenerative diseases. We discuss how different lifestyles and environmental factors might regulate microglia, potentially leading to increased susceptibility to neurodegenerative disease, and we highlight the need to investigate the contribution of modern environmental factors on microglia modulation in neurodegeneration.
Collapse
|
41
|
Cortese A, Delgado-Morales R, Almeida OFX, Romberg C. The Arctic/Swedish APP mutation alters the impact of chronic stress on cognition in mice. Eur J Neurosci 2019; 50:2773-2785. [PMID: 31231836 PMCID: PMC6852344 DOI: 10.1111/ejn.14500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 12/14/2022]
Abstract
Chronic stress is a major risk factor for developing Alzheimer's disease (AD) and promotes the processing of amyloid precursor protein (APP) to β-amyloid (Aβ). However, the precise relationship of stress and disease-typical cognitive decline is presently not well understood. The aim of this study was to investigate how early life stress may affect cognition in adult mice with and without soluble Aβ pathology typical for the early stages of the disease. We focussed on sustained attention and response control, aspects of cognition mediated by the prefrontal cortex that are consistently impaired both in early AD and after chronic stress exposure. Young wild-type mice as well as transgenic arcAβ mice overexpressing the hAPParc/swe transgene were exposed to a chronic unpredictable stress paradigm (age 3-8 weeks). At 15 weeks, these mice were tested on the 5-choice serial reaction time task, a test of sustained attention and executive control. We found that, expectedly, chronic stress increased impulsive choices and impaired sustained attention in wild-type mice. However, the same treatment reduced impulsivity and did not interfere with sustained attention in arcAβ mice. These findings suggest an unexpected interaction between chronic stress and Aβ whereby Aβ-pathology caused by the hAPParc/swe mutation prevented and/or reversed stress-induced cognitive changes through mechanisms that deserve further investigation. They also indicate that Aβ, in modest amounts, may have a beneficial role for cognitive stability, for example by protecting neural networks from the impact of further physiological or behavioural stress.
Collapse
Affiliation(s)
- Aurelio Cortese
- Max-Planck-Institute for Psychiatry, Munich, Germany.,Computational Neuroscience Laboratories, ATR Institute International, Kyoto, Japan
| | | | | | | |
Collapse
|
42
|
Lesuis SL, Lucassen PJ, Krugers HJ. Early life stress amplifies fear responses and hippocampal synaptic potentiation in the APPswe/PS1dE9 Alzheimer mouse model. Neuroscience 2019; 454:151-161. [PMID: 31302265 DOI: 10.1016/j.neuroscience.2019.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/25/2019] [Accepted: 07/02/2019] [Indexed: 01/04/2023]
Abstract
Cognitive deficits and alterations in emotional behaviour are typical features of Alzheimer's disease (AD). Moreover, exposure to stress or adversity during the early life period has been associated with an acceleration of cognitive deficits and increased AD pathology in transgenic AD mouse models. Whether and how early life adversity affects fear memory in AD mice remains elusive. We therefore investigated whether exposure to early life stress (ELS) alters fear learning in APPswe/PS1dE9 mice, a classic mouse model for AD, and whether this is accompanied by alterations in hippocampal synaptic potentiation, an important cellular substrate for learning and memory. Transgenic APPswe/PS1dE9 mice were subjected to ELS by housing the dams and her pups with limited nesting and bedding material from postnatal days 2-9. Following a fear conditioning paradigm, 12-month-old ELS-exposed APPswe/PS1dE9 mice displayed enhanced contextual freezing behaviour, both in the conditioning context and in a novel context. ELS-exposed APPswe/PS1dE9 mice also displayed enhanced hippocampal synaptic potentiation, even in the presence of the GluN2B antagonist Ro25-6981 (which prevented synaptic potentiation in control mice). No differences in the level of PSD-95 or synaptophysin were observed between the groups. We conclude that in APPswe/PS1dE9 mice, ELS increases fear memory in the conditioning context as well as a novel context, which is accompanied by aberrant hippocampal synaptic potentiation. These results may help to understand how individual differences in the vulnerability to develop AD arise and emphasise the importance of the early postnatal time window in these differences. This article is part of Special Issue entitled: Lifestyle and Brain Metaplasticity.
Collapse
Affiliation(s)
- Sylvie L Lesuis
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, the Netherlands.
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, the Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, the Netherlands
| |
Collapse
|
43
|
Walsh E, Blake Y, Donati A, Stoop R, von Gunten A. Early Secure Attachment as a Protective Factor Against Later Cognitive Decline and Dementia. Front Aging Neurosci 2019; 11:161. [PMID: 31333443 PMCID: PMC6622219 DOI: 10.3389/fnagi.2019.00161] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 06/12/2019] [Indexed: 01/07/2023] Open
Abstract
The etiology of neurodegenerative disorders such as dementia is complex and incompletely understood. Interest in a developmental perspective to these pathologies is gaining momentum. An early supportive social environment seems to have important implications for social, affective and cognitive abilities across the lifespan. Attachment theory may help to explain the link between these early experiences and later outcomes. This theory considers early interactions between an infant and its caregiver to be crucial to shaping social behavior and emotion regulation strategies throughout adult life. Furthermore, research has demonstrated that such early attachment experiences can, potentially through epigenetic mechanisms, have profound neurobiological and cognitive consequences. Here we discuss how early attachment might influence the development of affective, cognitive, and neurobiological resources that could protect against cognitive decline and dementia. We argue that social relations, both early and late in life, are vital to ensuring cognitive and neurobiological health. The concepts of brain and cognitive reserve are crucial to understanding how environmental factors may impact cognitive decline. We examine the role that attachment might play in fostering brain and cognitive reserve in old age. Finally, we put forward the concept of affective reserve, to more directly frame the socio-affective consequences of early attachment as protectors against cognitive decline. We thereby aim to highlight that, in the study of aging, cognitive decline and dementia, it is crucial to consider the role of affective and social factors such as attachment.
Collapse
Affiliation(s)
- Emilie Walsh
- Service of Old Age Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Yvonne Blake
- Center for Psychiatric Neurosciences, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Alessia Donati
- Service of Old Age Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Ron Stoop
- Center for Psychiatric Neurosciences, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Armin von Gunten
- Service of Old Age Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
44
|
Jafari Z, Okuma M, Karem H, Mehla J, Kolb BE, Mohajerani MH. Prenatal noise stress aggravates cognitive decline and the onset and progression of beta amyloid pathology in a mouse model of Alzheimer's disease. Neurobiol Aging 2019; 77:66-86. [DOI: 10.1016/j.neurobiolaging.2019.01.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 01/08/2019] [Accepted: 01/21/2019] [Indexed: 12/18/2022]
|
45
|
Radford K, Lavrencic LM, Delbaere K, Draper B, Cumming R, Daylight G, Mack HA, Chalkley S, Bennett H, Garvey G, Hill TY, Lasschuit D, Broe GA. Factors Associated with the High Prevalence of Dementia in Older Aboriginal Australians. J Alzheimers Dis 2019; 70:S75-S85. [PMID: 30507573 PMCID: PMC6700619 DOI: 10.3233/jad-180573] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2018] [Indexed: 01/12/2023]
Abstract
Dementia prevalence in Aboriginal and Torres Strait Islander Australians is three to five times higher than the general Australian population. A better understanding of the underlying biomedical and social risk factors is needed to guide dementia prevention in Aboriginal Australians. The current study is the first to examine potential risk factors for dementia in the majority urban and regional population, with a representative sample of 336 Aboriginal Australians aged 60 years and older. Participants included 45 people with a dementia diagnosis (n = 27 probable/possible Alzheimer's disease); and 286 people without dementia. Univariate logistic regression analyses (controlling for age) identified childhood trauma, mid-life factors (history of unskilled work, past high-risk alcohol use), and medical factors (history of stroke, head injury with loss of consciousness, epilepsy) as risk factors for dementia. Multivariable analysis revealed age, childhood trauma, unskilled work, stroke, and head injury as independent predictors of all-cause dementia. A range of comorbid factors related to dementia was also identified (i.e., functional impairment, incontinence, recent hospital admission, low body mass index, living in residential care, depression, current high-risk alcohol use, social isolation, low physical activity levels). These findings extend previous outcomes in a remote Aboriginal population by highlighting that life-course social determinants of health, in addition to neurological disorders, likely play an important role in elevating dementia risk. Certain psychosocial and medical exposures are highly prevalent in Aboriginal Australians, similar to other indigenous populations, and should be considered when designing targeted and culturally appropriate prevention initiatives to reduce the burden of dementia.
Collapse
Affiliation(s)
- Kylie Radford
- Neuroscience Research Australia, Randwick, NSW, Australia
- University of New South Wales, Sydney, NSW, Australia
| | | | - Kim Delbaere
- Neuroscience Research Australia, Randwick, NSW, Australia
- University of New South Wales, Sydney, NSW, Australia
| | - Brian Draper
- University of New South Wales, Sydney, NSW, Australia
- Prince of Wales Hospital, Randwick, NSW, Australia
| | | | - Gail Daylight
- Neuroscience Research Australia, Randwick, NSW, Australia
| | - Holly A. Mack
- University of Technology Sydney, Ultimo, NSW, Australia
| | - Simon Chalkley
- Neuroscience Research Australia, Randwick, NSW, Australia
| | - Hayley Bennett
- Neuroscience Research Australia, Randwick, NSW, Australia
| | - Gail Garvey
- Menzies School of Health Research, Brisbane, QLD, Australia
| | - Thi Yen Hill
- Neuroscience Research Australia, Randwick, NSW, Australia
- Prince of Wales Hospital, Randwick, NSW, Australia
| | - Danielle Lasschuit
- Neuroscience Research Australia, Randwick, NSW, Australia
- Prince of Wales Hospital, Randwick, NSW, Australia
| | - Gerald A. Broe
- Neuroscience Research Australia, Randwick, NSW, Australia
- University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
46
|
Henriques-Calado J, Duarte-Silva ME, Sousa Ferreira A. Depressive vulnerability in women with Alzheimer's disease: Relationship with personality traits and abnormal personality dimensions. J Affect Disord 2018; 241:182-191. [PMID: 30125822 DOI: 10.1016/j.jad.2018.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/18/2018] [Accepted: 08/04/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND This study sought to determine the evaluation of current and pre-morbid depressive vulnerability dimensions in Alzheimer's disease. Sidney Blatt´s personality developmental perspective, the Five-Factor model and Axis II personality disorders were taken as references. METHODS The study was conducted with two groups which were assessed using the Depressive Experiences Questionnaire, the NEO-FFI and the Personality Diagnostic Questionnaire-4+, in the form of individual interview sessions. Current personality measure: Alzheimer's disease Group, consisting of 44 female participants (MAge = 81.36 years); Pre-morbid personality measure: Alzheimer's disease Group Informants (n = 40). RESULTS Self-Criticism personality vulnerability is a general indicator of psychopathology. In pre-morbidity, Neuroticism (β = 0.41), Agreeableness (β = -0.63) and Conscientiousness (β = -0.08) predicted Self-Criticism, explaining 64% of the variance; additionally, Self-Criticism (β = 0.72) and Neediness (β = 2.05) predicted the PDQ-4+ total, explaining 58% of the variance. In terms of current personality, the PDQ-4+ total was predicted by Self-Criticism (β = 0.55), explaining 30% of the variance. LIMITATIONS The small size of the samples, especially since it is difficult to access individuals diagnosed with AD at the onset or in its early stages; measuring personality changes by means of retrospective assessment by proxies may have introduced some memory bias. CONCLUSIONS These findings are relevant to research relating depressive vulnerability to personality traits and psychopathology in Alzheimer's disease.
Collapse
Affiliation(s)
- Joana Henriques-Calado
- Faculdade de Psicologia, Universidade de Lisboa, Alameda da Universidade, 1649-013 Lisboa, Portugal; CICPSI, Faculdade de Psicologia, Universidade de Lisboa, Alameda da Universidade, 1649-013 Lisboa, Portugal.
| | | | - Ana Sousa Ferreira
- Faculdade de Psicologia, Universidade de Lisboa, Alameda da Universidade, 1649-013 Lisboa, Portugal; Instituto Universitário de Lisboa - Business Research Unit (BRU-IUL), Portugal.
| |
Collapse
|