Skip to main content
Top
Published in: Translational Neurodegeneration 1/2023

Open Access 01-12-2023 | Alzheimer's Disease | Review

Early life adversity as a risk factor for cognitive impairment and Alzheimer’s disease

Authors: Zhihai Huang, J. Dedrick Jordan, Quanguang Zhang

Published in: Translational Neurodegeneration | Issue 1/2023

Login to get access

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.
Literature
1.
go back to reference World Health Organization. Risk reduction of cognitive decline and dementia: WHO guidelines. Geneva; 2019 World Health Organization. Risk reduction of cognitive decline and dementia: WHO guidelines. Geneva; 2019
2.
go back to reference Moon W, Han JW, Bae JB, Suh SW, Kim TH, Kwak KP, et al. Disease burdens of Alzheimer’s disease, vascular dementia, and mild cognitive impairment. J Am Med Dir Assoc. 2021;22:2093-2099.e3.PubMedCrossRef Moon W, Han JW, Bae JB, Suh SW, Kim TH, Kwak KP, et al. Disease burdens of Alzheimer’s disease, vascular dementia, and mild cognitive impairment. J Am Med Dir Assoc. 2021;22:2093-2099.e3.PubMedCrossRef
3.
go back to reference Collaborators GBDD. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: a systematic analysis for the global burden of disease study 2016. Lancet Neurol. 2019;18:88–106.CrossRef Collaborators GBDD. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: a systematic analysis for the global burden of disease study 2016. Lancet Neurol. 2019;18:88–106.CrossRef
4.
5.
go back to reference Kivimaki M, Singh-Manoux A, Pentti J, Sabia S, Nyberg ST, Alfredsson L, et al. Physical inactivity, cardiometabolic disease, and risk of dementia: an individual-participant meta-analysis. BMJ. 2019;365:l1495.PubMedPubMedCentralCrossRef Kivimaki M, Singh-Manoux A, Pentti J, Sabia S, Nyberg ST, Alfredsson L, et al. Physical inactivity, cardiometabolic disease, and risk of dementia: an individual-participant meta-analysis. BMJ. 2019;365:l1495.PubMedPubMedCentralCrossRef
6.
go back to reference Silva MVF, Loures CMG, Alves LCV, de Souza LC, Borges KBG, Carvalho MDG. Alzheimer’s disease: risk factors and potentially protective measures. J Biomed Sci. 2019;26:33.PubMedPubMedCentralCrossRef Silva MVF, Loures CMG, Alves LCV, de Souza LC, Borges KBG, Carvalho MDG. Alzheimer’s disease: risk factors and potentially protective measures. J Biomed Sci. 2019;26:33.PubMedPubMedCentralCrossRef
7.
go back to reference Hoeijmakers L, Lesuis SL, Krugers H, Lucassen PJ, Korosi A. A preclinical perspective on the enhanced vulnerability to Alzheimer’s disease after early-life stress. Neurobiol Stress. 2018;8:172–85.PubMedPubMedCentralCrossRef Hoeijmakers L, Lesuis SL, Krugers H, Lucassen PJ, Korosi A. A preclinical perspective on the enhanced vulnerability to Alzheimer’s disease after early-life stress. Neurobiol Stress. 2018;8:172–85.PubMedPubMedCentralCrossRef
10.
go back to reference Kundakovic M, Champagne FA. Early-life experience, epigenetics, and the developing brain. Neuropsychopharmacology. 2015;40:141–53.PubMedCrossRef Kundakovic M, Champagne FA. Early-life experience, epigenetics, and the developing brain. Neuropsychopharmacology. 2015;40:141–53.PubMedCrossRef
11.
12.
13.
go back to reference Lopez M, Ruiz MO, Rovnaghi CR, Tam GK, Hiscox J, Gotlib IH, et al. The social ecology of childhood and early life adversity. Pediatr Res. 2021;89:353–67.PubMedPubMedCentralCrossRef Lopez M, Ruiz MO, Rovnaghi CR, Tam GK, Hiscox J, Gotlib IH, et al. The social ecology of childhood and early life adversity. Pediatr Res. 2021;89:353–67.PubMedPubMedCentralCrossRef
15.
16.
go back to reference Felitti VJ, Anda RF, Nordenberg D, Williamson DF, Spitz AM, Edwards V, et al. Relationship of childhood abuse and household dysfunction to many of the leading causes of death in adults. The adverse childhood experiences (ACE) study. Am J Prev Med. 1998;14:245–58.PubMedCrossRef Felitti VJ, Anda RF, Nordenberg D, Williamson DF, Spitz AM, Edwards V, et al. Relationship of childhood abuse and household dysfunction to many of the leading causes of death in adults. The adverse childhood experiences (ACE) study. Am J Prev Med. 1998;14:245–58.PubMedCrossRef
18.
go back to reference Loria AS, Ho DH, Pollock JS. A mechanistic look at the effects of adversity early in life on cardiovascular disease risk during adulthood. Acta Physiol (Oxf). 2014;210:277–87.PubMedCrossRef Loria AS, Ho DH, Pollock JS. A mechanistic look at the effects of adversity early in life on cardiovascular disease risk during adulthood. Acta Physiol (Oxf). 2014;210:277–87.PubMedCrossRef
19.
go back to reference Hughes K, Bellis MA, Hardcastle KA, Sethi D, Butchart A, Mikton C, et al. The effect of multiple adverse childhood experiences on health: a systematic review and meta-analysis. Lancet Public Health. 2017;2:e356–e66.PubMedCrossRef Hughes K, Bellis MA, Hardcastle KA, Sethi D, Butchart A, Mikton C, et al. The effect of multiple adverse childhood experiences on health: a systematic review and meta-analysis. Lancet Public Health. 2017;2:e356–e66.PubMedCrossRef
20.
go back to reference Gilbert LK, Breiding MJ, Merrick MT, Thompson WW, Ford DC, Dhingra SS, et al. Childhood adversity and adult chronic disease: an update from ten states and the District of Columbia, 2010. Am J Prev Med. 2015;48:345–9.PubMedCrossRef Gilbert LK, Breiding MJ, Merrick MT, Thompson WW, Ford DC, Dhingra SS, et al. Childhood adversity and adult chronic disease: an update from ten states and the District of Columbia, 2010. Am J Prev Med. 2015;48:345–9.PubMedCrossRef
21.
go back to reference McLaughlin KA, Green JG, Gruber MJ, Sampson NA, Zaslavsky AM, Kessler RC. Childhood adversities and adult psychiatric disorders in the national comorbidity survey replication II: associations with persistence of DSM-IV disorders. Arch Gen Psychiatry. 2010;67:124–32.PubMedPubMedCentralCrossRef McLaughlin KA, Green JG, Gruber MJ, Sampson NA, Zaslavsky AM, Kessler RC. Childhood adversities and adult psychiatric disorders in the national comorbidity survey replication II: associations with persistence of DSM-IV disorders. Arch Gen Psychiatry. 2010;67:124–32.PubMedPubMedCentralCrossRef
22.
go back to reference Yajima H, Haijima A, Khairinisa MA, Shimokawa N, Amano I, Takatsuru Y. Early-life stress induces cognitive disorder in middle-aged mice. Neurobiol Aging. 2018;64:139–46.PubMedCrossRef Yajima H, Haijima A, Khairinisa MA, Shimokawa N, Amano I, Takatsuru Y. Early-life stress induces cognitive disorder in middle-aged mice. Neurobiol Aging. 2018;64:139–46.PubMedCrossRef
23.
go back to reference Holland JF, Khandaker GM, Dauvermann MR, Morris D, Zammit S, Donohoe G. Effects of early life adversity on immune function and cognitive performance: results from the ALSPAC cohort. Soc Psychiatry Psychiatr Epidemiol. 2020;55:723–33.PubMedCrossRef Holland JF, Khandaker GM, Dauvermann MR, Morris D, Zammit S, Donohoe G. Effects of early life adversity on immune function and cognitive performance: results from the ALSPAC cohort. Soc Psychiatry Psychiatr Epidemiol. 2020;55:723–33.PubMedCrossRef
24.
go back to reference Radford K, Delbaere K, Draper B, Mack HA, Daylight G, Cumming R, et al. Childhood stress and adversity is associated with late-life dementia in Aboriginal Australians. Am J Geriatr Psychiatry. 2017;25:1097–106.PubMedCrossRef Radford K, Delbaere K, Draper B, Mack HA, Daylight G, Cumming R, et al. Childhood stress and adversity is associated with late-life dementia in Aboriginal Australians. Am J Geriatr Psychiatry. 2017;25:1097–106.PubMedCrossRef
25.
go back to reference Hedges DW, Woon FL. Early-life stress and cognitive outcome. Psychopharmacology. 2011;214:121–30.PubMedCrossRef Hedges DW, Woon FL. Early-life stress and cognitive outcome. Psychopharmacology. 2011;214:121–30.PubMedCrossRef
26.
go back to reference Muhammad A, Kolb B. Maternal separation altered behavior and neuronal spine density without influencing amphetamine sensitization. Behav Brain Res. 2011;223:7–16.PubMedCrossRef Muhammad A, Kolb B. Maternal separation altered behavior and neuronal spine density without influencing amphetamine sensitization. Behav Brain Res. 2011;223:7–16.PubMedCrossRef
27.
28.
go back to reference Benmhammed H, El Hayek S, Berkik I, Elmostafi H, Bousalham R, Mesfioui A, et al. Animal models of early-life adversity. Methods Mol Biol. 2019;2011:143–61.PubMedCrossRef Benmhammed H, El Hayek S, Berkik I, Elmostafi H, Bousalham R, Mesfioui A, et al. Animal models of early-life adversity. Methods Mol Biol. 2019;2011:143–61.PubMedCrossRef
29.
go back to reference Waters RC, Gould E. Early life adversity and neuropsychiatric disease: differential outcomes and translational relevance of rodent models. Front Syst Neurosci. 2022;16:860847.PubMedPubMedCentralCrossRef Waters RC, Gould E. Early life adversity and neuropsychiatric disease: differential outcomes and translational relevance of rodent models. Front Syst Neurosci. 2022;16:860847.PubMedPubMedCentralCrossRef
30.
31.
go back to reference Lippmann M, Bress A, Nemeroff CB, Plotsky PM, Monteggia LM. Long-term behavioural and molecular alterations associated with maternal separation in rats. Eur J Neurosci. 2007;25:3091–8.PubMedCrossRef Lippmann M, Bress A, Nemeroff CB, Plotsky PM, Monteggia LM. Long-term behavioural and molecular alterations associated with maternal separation in rats. Eur J Neurosci. 2007;25:3091–8.PubMedCrossRef
32.
go back to reference Levis SC, Baram TZ, Mahler SV. Neurodevelopmental origins of substance use disorders: evidence from animal models of early-life adversity and addiction. Eur J Neurosci. 2022;55:2170–95.PubMedCrossRef Levis SC, Baram TZ, Mahler SV. Neurodevelopmental origins of substance use disorders: evidence from animal models of early-life adversity and addiction. Eur J Neurosci. 2022;55:2170–95.PubMedCrossRef
33.
go back to reference Marco EM, Llorente R, Lopez-Gallardo M, Mela V, Llorente-Berzal A, Prada C, et al. The maternal deprivation animal model revisited. Neurosci Biobehav Rev. 2015;51:151–63.PubMedCrossRef Marco EM, Llorente R, Lopez-Gallardo M, Mela V, Llorente-Berzal A, Prada C, et al. The maternal deprivation animal model revisited. Neurosci Biobehav Rev. 2015;51:151–63.PubMedCrossRef
34.
go back to reference Ellenbroek BA, Derks N, Park HJ. Early maternal deprivation retards neurodevelopment in Wistar rats. Stress. 2005;8:247–57.PubMedCrossRef Ellenbroek BA, Derks N, Park HJ. Early maternal deprivation retards neurodevelopment in Wistar rats. Stress. 2005;8:247–57.PubMedCrossRef
35.
go back to reference Rice CJ, Sandman CA, Lenjavi MR, Baram TZ. A novel mouse model for acute and long-lasting consequences of early life stress. Endocrinology. 2008;149:4892–900.PubMedPubMedCentralCrossRef Rice CJ, Sandman CA, Lenjavi MR, Baram TZ. A novel mouse model for acute and long-lasting consequences of early life stress. Endocrinology. 2008;149:4892–900.PubMedPubMedCentralCrossRef
36.
go back to reference Hsiao YM, Tsai TC, Lin YT, Chen CC, Huang CC, Hsu KS. Early life stress dampens stress responsiveness in adolescence: evaluation of neuroendocrine reactivity and coping behavior. Psychoneuroendocrinology. 2016;67:86–99.PubMedCrossRef Hsiao YM, Tsai TC, Lin YT, Chen CC, Huang CC, Hsu KS. Early life stress dampens stress responsiveness in adolescence: evaluation of neuroendocrine reactivity and coping behavior. Psychoneuroendocrinology. 2016;67:86–99.PubMedCrossRef
37.
go back to reference Bolton JL, Schulmann A, Garcia-Curran MM, Regev L, Chen Y, Kamei N, et al. Unexpected transcriptional programs contribute to hippocampal memory deficits and neuronal stunting after early-life adversity. Cell Rep. 2020;33:108511.PubMedPubMedCentralCrossRef Bolton JL, Schulmann A, Garcia-Curran MM, Regev L, Chen Y, Kamei N, et al. Unexpected transcriptional programs contribute to hippocampal memory deficits and neuronal stunting after early-life adversity. Cell Rep. 2020;33:108511.PubMedPubMedCentralCrossRef
38.
go back to reference Bonapersona V, Damsteegt R, Adams ML, van Weert L, Meijer OC, Joels M, et al. Sex-dependent modulation of acute stress reactivity after early life stress in mice: relevance of mineralocorticoid receptor expression. Front Behav Neurosci. 2019;13:181.PubMedPubMedCentralCrossRef Bonapersona V, Damsteegt R, Adams ML, van Weert L, Meijer OC, Joels M, et al. Sex-dependent modulation of acute stress reactivity after early life stress in mice: relevance of mineralocorticoid receptor expression. Front Behav Neurosci. 2019;13:181.PubMedPubMedCentralCrossRef
39.
go back to reference Molet J, Heins K, Zhuo X, Mei YT, Regev L, Baram TZ, et al. Fragmentation and high entropy of neonatal experience predict adolescent emotional outcome. Transl Psychiatry. 2016;6:e702.PubMedPubMedCentralCrossRef Molet J, Heins K, Zhuo X, Mei YT, Regev L, Baram TZ, et al. Fragmentation and high entropy of neonatal experience predict adolescent emotional outcome. Transl Psychiatry. 2016;6:e702.PubMedPubMedCentralCrossRef
40.
go back to reference Gallo M, Shleifer DG, Godoy LD, Ofray D, Olaniyan A, Campbell T, et al. Limited bedding and nesting induces maternal behavior resembling both hypervigilance and abuse. Front Behav Neurosci. 2019;13:167.PubMedPubMedCentralCrossRef Gallo M, Shleifer DG, Godoy LD, Ofray D, Olaniyan A, Campbell T, et al. Limited bedding and nesting induces maternal behavior resembling both hypervigilance and abuse. Front Behav Neurosci. 2019;13:167.PubMedPubMedCentralCrossRef
41.
go back to reference Avital A, Richter-Levin G. Exposure to juvenile stress exacerbates the behavioural consequences of exposure to stress in the adult rat. Int J Neuropsychopharmacol. 2005;8:163–73.PubMedCrossRef Avital A, Richter-Levin G. Exposure to juvenile stress exacerbates the behavioural consequences of exposure to stress in the adult rat. Int J Neuropsychopharmacol. 2005;8:163–73.PubMedCrossRef
42.
go back to reference Huang Z, Zhang Y, Ma X, Feng Y, Zong X, Jordan JD, et al. Photobiomodulation attenuates oligodendrocyte dysfunction and prevents adverse neurological consequences in a rat model of early life adversity. Theranostics. 2023;13:913–30.PubMedPubMedCentralCrossRef Huang Z, Zhang Y, Ma X, Feng Y, Zong X, Jordan JD, et al. Photobiomodulation attenuates oligodendrocyte dysfunction and prevents adverse neurological consequences in a rat model of early life adversity. Theranostics. 2023;13:913–30.PubMedPubMedCentralCrossRef
43.
go back to reference Zardooz H, Sadeghimahalli F, Khodagholi F. Early postnatal stress impairs insulin secretion in response to psychological stress in adult rats. J Endocrinol Invest. 2021;44:277–86.PubMedCrossRef Zardooz H, Sadeghimahalli F, Khodagholi F. Early postnatal stress impairs insulin secretion in response to psychological stress in adult rats. J Endocrinol Invest. 2021;44:277–86.PubMedCrossRef
44.
go back to reference Lu CY, Liu X, Jiang H, Pan F, Ho CS, Ho RC. Effects of traumatic stress induced in the juvenile period on the expression of gamma-aminobutyric acid receptor type a subunits in adult rat brain. Neural Plast. 2017;2017:5715816.PubMedPubMedCentralCrossRef Lu CY, Liu X, Jiang H, Pan F, Ho CS, Ho RC. Effects of traumatic stress induced in the juvenile period on the expression of gamma-aminobutyric acid receptor type a subunits in adult rat brain. Neural Plast. 2017;2017:5715816.PubMedPubMedCentralCrossRef
45.
go back to reference Abbink MR, Naninck EFG, Lucassen PJ, Korosi A. Early-life stress diminishes the increase in neurogenesis after exercise in adult female mice. Hippocampus. 2017;27:839–44.PubMedCrossRef Abbink MR, Naninck EFG, Lucassen PJ, Korosi A. Early-life stress diminishes the increase in neurogenesis after exercise in adult female mice. Hippocampus. 2017;27:839–44.PubMedCrossRef
46.
go back to reference Sadeghimahalli F, Karbaschi R, Zardooz H, Khodagholi F, Rostamkhani F. Effect of early life stress on pancreatic isolated islets’ insulin secretion in young adult male rats subjected to chronic stress. Endocrine. 2015;48:493–503.PubMedCrossRef Sadeghimahalli F, Karbaschi R, Zardooz H, Khodagholi F, Rostamkhani F. Effect of early life stress on pancreatic isolated islets’ insulin secretion in young adult male rats subjected to chronic stress. Endocrine. 2015;48:493–503.PubMedCrossRef
47.
go back to reference Liu D, Diorio J, Day JC, Francis DD, Meaney MJ. Maternal care, hippocampal synaptogenesis and cognitive development in rats. Nat Neurosci. 2000;3:799–806.PubMedCrossRef Liu D, Diorio J, Day JC, Francis DD, Meaney MJ. Maternal care, hippocampal synaptogenesis and cognitive development in rats. Nat Neurosci. 2000;3:799–806.PubMedCrossRef
48.
go back to reference Feldman R, Rosenthal Z, Eidelman AI. Maternal-preterm skin-to-skin contact enhances child physiologic organization and cognitive control across the first 10 years of life. Biol Psychiatry. 2014;75:56–64.PubMedCrossRef Feldman R, Rosenthal Z, Eidelman AI. Maternal-preterm skin-to-skin contact enhances child physiologic organization and cognitive control across the first 10 years of life. Biol Psychiatry. 2014;75:56–64.PubMedCrossRef
49.
go back to reference Marco EM, Valero M, de la Serna O, Aisa B, Borcel E, Ramirez MJ, et al. Maternal deprivation effects on brain plasticity and recognition memory in adolescent male and female rats. Neuropharmacology. 2013;68:223–31.PubMedCrossRef Marco EM, Valero M, de la Serna O, Aisa B, Borcel E, Ramirez MJ, et al. Maternal deprivation effects on brain plasticity and recognition memory in adolescent male and female rats. Neuropharmacology. 2013;68:223–31.PubMedCrossRef
50.
go back to reference Grassi-Oliveira R, Honeycutt JA, Holland FH, Ganguly P, Brenhouse HC. Cognitive impairment effects of early life stress in adolescents can be predicted with early biomarkers: impacts of sex, experience, and cytokines. Psychoneuroendocrinology. 2016;71:19–30.PubMedPubMedCentralCrossRef Grassi-Oliveira R, Honeycutt JA, Holland FH, Ganguly P, Brenhouse HC. Cognitive impairment effects of early life stress in adolescents can be predicted with early biomarkers: impacts of sex, experience, and cytokines. Psychoneuroendocrinology. 2016;71:19–30.PubMedPubMedCentralCrossRef
51.
go back to reference Loi M, Mossink JC, Meerhoff GF, Den Blaauwen JL, Lucassen PJ, Joels M. Effects of early-life stress on cognitive function and hippocampal structure in female rodents. Neuroscience. 2017;342:101–19.PubMedCrossRef Loi M, Mossink JC, Meerhoff GF, Den Blaauwen JL, Lucassen PJ, Joels M. Effects of early-life stress on cognitive function and hippocampal structure in female rodents. Neuroscience. 2017;342:101–19.PubMedCrossRef
52.
go back to reference Feifel AJ, Shair HN, Schmauss C. Lasting effects of early life stress in mice: interaction of maternal environment and infant genes. Genes Brain Behav. 2017;16:768–80.PubMed Feifel AJ, Shair HN, Schmauss C. Lasting effects of early life stress in mice: interaction of maternal environment and infant genes. Genes Brain Behav. 2017;16:768–80.PubMed
53.
go back to reference Janetsian-Fritz SS, Timme NM, Timm MM, McCane AM, Baucum Ii AJ, O’Donnell BF, et al. Maternal deprivation induces alterations in cognitive and cortical function in adulthood. Transl Psychiatry. 2018;8:71.PubMedPubMedCentralCrossRef Janetsian-Fritz SS, Timme NM, Timm MM, McCane AM, Baucum Ii AJ, O’Donnell BF, et al. Maternal deprivation induces alterations in cognitive and cortical function in adulthood. Transl Psychiatry. 2018;8:71.PubMedPubMedCentralCrossRef
54.
go back to reference Reshetnikov VV, Kovner AV, Lepeshko AA, Pavlov KS, Grinkevich LN, Bondar NP. Stress early in life leads to cognitive impairments, reduced numbers of CA3 neurons and altered maternal behavior in adult female mice. Genes Brain Behav. 2020;19:e12541.PubMedCrossRef Reshetnikov VV, Kovner AV, Lepeshko AA, Pavlov KS, Grinkevich LN, Bondar NP. Stress early in life leads to cognitive impairments, reduced numbers of CA3 neurons and altered maternal behavior in adult female mice. Genes Brain Behav. 2020;19:e12541.PubMedCrossRef
55.
go back to reference Xu H, Ye Y, Hao Y, Shi F, Yan Z, Yuan G, et al. Sex differences in associations between maternal deprivation and alterations in hippocampal calcium-binding proteins and cognitive functions in rats. Behav Brain Funct. 2018;14:10.PubMedPubMedCentralCrossRef Xu H, Ye Y, Hao Y, Shi F, Yan Z, Yuan G, et al. Sex differences in associations between maternal deprivation and alterations in hippocampal calcium-binding proteins and cognitive functions in rats. Behav Brain Funct. 2018;14:10.PubMedPubMedCentralCrossRef
56.
go back to reference Wang XD, Rammes G, Kraev I, Wolf M, Liebl C, Scharf SH, et al. Forebrain CRF(1) modulates early-life stress-programmed cognitive deficits. J Neurosci. 2011;31:13625–34.PubMedPubMedCentralCrossRef Wang XD, Rammes G, Kraev I, Wolf M, Liebl C, Scharf SH, et al. Forebrain CRF(1) modulates early-life stress-programmed cognitive deficits. J Neurosci. 2011;31:13625–34.PubMedPubMedCentralCrossRef
57.
go back to reference Brunson KL, Kramar E, Lin B, Chen Y, Colgin LL, Yanagihara TK, et al. Mechanisms of late-onset cognitive decline after early-life stress. J Neurosci. 2005;25:9328–38.PubMedPubMedCentralCrossRef Brunson KL, Kramar E, Lin B, Chen Y, Colgin LL, Yanagihara TK, et al. Mechanisms of late-onset cognitive decline after early-life stress. J Neurosci. 2005;25:9328–38.PubMedPubMedCentralCrossRef
58.
go back to reference Ivy AS, Rex CS, Chen Y, Dube C, Maras PM, Grigoriadis DE, et al. Hippocampal dysfunction and cognitive impairments provoked by chronic early-life stress involve excessive activation of CRH receptors. J Neurosci. 2010;30:13005–15.PubMedPubMedCentralCrossRef Ivy AS, Rex CS, Chen Y, Dube C, Maras PM, Grigoriadis DE, et al. Hippocampal dysfunction and cognitive impairments provoked by chronic early-life stress involve excessive activation of CRH receptors. J Neurosci. 2010;30:13005–15.PubMedPubMedCentralCrossRef
59.
go back to reference Naninck EF, Hoeijmakers L, Kakava-Georgiadou N, Meesters A, Lazic SE, Lucassen PJ, et al. Chronic early life stress alters developmental and adult neurogenesis and impairs cognitive function in mice. Hippocampus. 2015;25:309–28.PubMedCrossRef Naninck EF, Hoeijmakers L, Kakava-Georgiadou N, Meesters A, Lazic SE, Lucassen PJ, et al. Chronic early life stress alters developmental and adult neurogenesis and impairs cognitive function in mice. Hippocampus. 2015;25:309–28.PubMedCrossRef
60.
go back to reference Li C, Liu Y, Yin S, Lu C, Liu D, Jiang H, et al. Long-term effects of early adolescent stress: dysregulation of hypothalamic-pituitary-adrenal axis and central corticotropin releasing factor receptor 1 expression in adult male rats. Behav Brain Res. 2015;288:39–49.PubMedCrossRef Li C, Liu Y, Yin S, Lu C, Liu D, Jiang H, et al. Long-term effects of early adolescent stress: dysregulation of hypothalamic-pituitary-adrenal axis and central corticotropin releasing factor receptor 1 expression in adult male rats. Behav Brain Res. 2015;288:39–49.PubMedCrossRef
61.
go back to reference Liu H, Atrooz F, Salvi A, Salim S. Behavioral and cognitive impact of early life stress: insights from an animal model. Prog Neuropsychopharmacol Biol Psychiatry. 2017;78:88–95.PubMedPubMedCentralCrossRef Liu H, Atrooz F, Salvi A, Salim S. Behavioral and cognitive impact of early life stress: insights from an animal model. Prog Neuropsychopharmacol Biol Psychiatry. 2017;78:88–95.PubMedPubMedCentralCrossRef
62.
63.
go back to reference Schore AN. Effects of a secure attachment relationship on right brain development, affect regulation, and infant mental health. Infant Mental Health J Off Publ World Assoc Infant Mental Health. 2001;22:7–66. Schore AN. Effects of a secure attachment relationship on right brain development, affect regulation, and infant mental health. Infant Mental Health J Off Publ World Assoc Infant Mental Health. 2001;22:7–66.
64.
65.
go back to reference Englund MM, Kuo SI, Puig J, Collins WA. Early roots of adult competence: the significance of close relationships from infancy to early adulthood. Int J Behav Dev. 2011;35:490–6.PubMedPubMedCentralCrossRef Englund MM, Kuo SI, Puig J, Collins WA. Early roots of adult competence: the significance of close relationships from infancy to early adulthood. Int J Behav Dev. 2011;35:490–6.PubMedPubMedCentralCrossRef
66.
go back to reference Gregory A, Rimm-Kaufman S. Positive mother–child interactions in kindergarten: Predictors of school success in high school. Sch Psychol Rev. 2008;37(4):499–515.CrossRef Gregory A, Rimm-Kaufman S. Positive mother–child interactions in kindergarten: Predictors of school success in high school. Sch Psychol Rev. 2008;37(4):499–515.CrossRef
67.
go back to reference Mueller SC, Maheu FS, Dozier M, Peloso E, Mandell D, Leibenluft E, et al. Early-life stress is associated with impairment in cognitive control in adolescence: an fMRI study. Neuropsychologia. 2010;48:3037–44.PubMedPubMedCentralCrossRef Mueller SC, Maheu FS, Dozier M, Peloso E, Mandell D, Leibenluft E, et al. Early-life stress is associated with impairment in cognitive control in adolescence: an fMRI study. Neuropsychologia. 2010;48:3037–44.PubMedPubMedCentralCrossRef
68.
go back to reference Pesonen AK, Eriksson JG, Heinonen K, Kajantie E, Tuovinen S, Alastalo H, et al. Cognitive ability and decline after early life stress exposure. Neurobiol Aging. 2013;34:1674–9.PubMedCrossRef Pesonen AK, Eriksson JG, Heinonen K, Kajantie E, Tuovinen S, Alastalo H, et al. Cognitive ability and decline after early life stress exposure. Neurobiol Aging. 2013;34:1674–9.PubMedCrossRef
69.
go back to reference Hawkins MAW, Layman HM, Ganson KT, Tabler J, Ciciolla L, Tsotsoros CE, et al. Adverse childhood events and cognitive function among young adults: prospective results from the national longitudinal study of adolescent to adult health. Child Abuse Negl. 2021;115:105008.PubMedCrossRef Hawkins MAW, Layman HM, Ganson KT, Tabler J, Ciciolla L, Tsotsoros CE, et al. Adverse childhood events and cognitive function among young adults: prospective results from the national longitudinal study of adolescent to adult health. Child Abuse Negl. 2021;115:105008.PubMedCrossRef
70.
go back to reference Wade M, Zeanah CH, Fox NA, Nelson CA. Global deficits in executive functioning are transdiagnostic mediators between severe childhood neglect and psychopathology in adolescence. Psychol Med. 2020;50:1687–94.PubMedCrossRef Wade M, Zeanah CH, Fox NA, Nelson CA. Global deficits in executive functioning are transdiagnostic mediators between severe childhood neglect and psychopathology in adolescence. Psychol Med. 2020;50:1687–94.PubMedCrossRef
71.
go back to reference Mackes NK, Golm D, Sarkar S, Kumsta R, Rutter M, Fairchild G, et al. Early childhood deprivation is associated with alterations in adult brain structure despite subsequent environmental enrichment. Proc Natl Acad Sci U S A. 2020;117:641–9.PubMedPubMedCentralCrossRef Mackes NK, Golm D, Sarkar S, Kumsta R, Rutter M, Fairchild G, et al. Early childhood deprivation is associated with alterations in adult brain structure despite subsequent environmental enrichment. Proc Natl Acad Sci U S A. 2020;117:641–9.PubMedPubMedCentralCrossRef
72.
go back to reference Ma J, Yang Y, Wan Y, Shen C, Qiu P. The influence of childhood adversities on mid to late cognitive function: from the perspective of life course. PLoS ONE. 2021;16:e0256297.PubMedPubMedCentralCrossRef Ma J, Yang Y, Wan Y, Shen C, Qiu P. The influence of childhood adversities on mid to late cognitive function: from the perspective of life course. PLoS ONE. 2021;16:e0256297.PubMedPubMedCentralCrossRef
73.
go back to reference Zhang K, Zhang W. Adverse childhood experiences and mild cognitive impairment in later life: exploring rural/urban and gender differences using CHARLS. J Appl Gerontol. 2022;41:1454–64.PubMedCrossRef Zhang K, Zhang W. Adverse childhood experiences and mild cognitive impairment in later life: exploring rural/urban and gender differences using CHARLS. J Appl Gerontol. 2022;41:1454–64.PubMedCrossRef
74.
go back to reference Saleh A, Potter GG, McQuoid DR, Boyd B, Turner R, MacFall JR, et al. Effects of early life stress on depression, cognitive performance and brain morphology. Psychol Med. 2017;47:171–81.PubMedCrossRef Saleh A, Potter GG, McQuoid DR, Boyd B, Turner R, MacFall JR, et al. Effects of early life stress on depression, cognitive performance and brain morphology. Psychol Med. 2017;47:171–81.PubMedCrossRef
75.
go back to reference Kalia V, Knauft K, Hayatbini N. Adverse childhood experiences (ACEs) associated with reduced cognitive flexibility in both college and community samples. PLoS ONE. 2021;16:e0260822.PubMedPubMedCentralCrossRef Kalia V, Knauft K, Hayatbini N. Adverse childhood experiences (ACEs) associated with reduced cognitive flexibility in both college and community samples. PLoS ONE. 2021;16:e0260822.PubMedPubMedCentralCrossRef
76.
go back to reference Hoeijmakers L, Ruigrok SR, Amelianchik A, Ivan D, van Dam AM, Lucassen PJ, et al. Early-life stress lastingly alters the neuroinflammatory response to amyloid pathology in an Alzheimer’s disease mouse model. Brain Behav Immun. 2017;63:160–75.PubMedCrossRef Hoeijmakers L, Ruigrok SR, Amelianchik A, Ivan D, van Dam AM, Lucassen PJ, et al. Early-life stress lastingly alters the neuroinflammatory response to amyloid pathology in an Alzheimer’s disease mouse model. Brain Behav Immun. 2017;63:160–75.PubMedCrossRef
77.
go back to reference Abbink MR, Kotah JM, Hoeijmakers L, Mak A, Yvon-Durocher G, van der Gaag B, et al. Characterization of astrocytes throughout life in wildtype and APP/PS1 mice after early-life stress exposure. J Neuroinflammation. 2020;17:91.PubMedPubMedCentralCrossRef Abbink MR, Kotah JM, Hoeijmakers L, Mak A, Yvon-Durocher G, van der Gaag B, et al. Characterization of astrocytes throughout life in wildtype and APP/PS1 mice after early-life stress exposure. J Neuroinflammation. 2020;17:91.PubMedPubMedCentralCrossRef
78.
go back to reference Lesuis SL, Weggen S, Baches S, Lucassen PJ, Krugers HJ. Targeting glucocorticoid receptors prevents the effects of early life stress on amyloid pathology and cognitive performance in APP/PS1 mice. Transl Psychiatry. 2018;8:53.PubMedPubMedCentralCrossRef Lesuis SL, Weggen S, Baches S, Lucassen PJ, Krugers HJ. Targeting glucocorticoid receptors prevents the effects of early life stress on amyloid pathology and cognitive performance in APP/PS1 mice. Transl Psychiatry. 2018;8:53.PubMedPubMedCentralCrossRef
79.
go back to reference Lesuis SL, Kaplick PM, Lucassen PJ, Krugers HJ. Treatment with the glutamate modulator riluzole prevents early life stress-induced cognitive deficits and impairments in synaptic plasticity in APPswe/PS1dE9 mice. Neuropharmacology. 2019;150:175–83.PubMedCrossRef Lesuis SL, Kaplick PM, Lucassen PJ, Krugers HJ. Treatment with the glutamate modulator riluzole prevents early life stress-induced cognitive deficits and impairments in synaptic plasticity in APPswe/PS1dE9 mice. Neuropharmacology. 2019;150:175–83.PubMedCrossRef
80.
go back to reference Hui J, Feng G, Zheng C, Jin H, Jia N. Maternal separation exacerbates Alzheimer’s disease-like behavioral and pathological changes in adult APPswe/PS1dE9 mice. Behav Brain Res. 2017;318:18–23.PubMedCrossRef Hui J, Feng G, Zheng C, Jin H, Jia N. Maternal separation exacerbates Alzheimer’s disease-like behavioral and pathological changes in adult APPswe/PS1dE9 mice. Behav Brain Res. 2017;318:18–23.PubMedCrossRef
81.
go back to reference Tanaka T, Hirai S, Hosokawa M, Saito T, Sakuma H, Saido T, et al. Early-life stress induces the development of Alzheimer’s disease pathology via angiopathy. Exp Neurol. 2021;337:113552.PubMedCrossRef Tanaka T, Hirai S, Hosokawa M, Saito T, Sakuma H, Saido T, et al. Early-life stress induces the development of Alzheimer’s disease pathology via angiopathy. Exp Neurol. 2021;337:113552.PubMedCrossRef
82.
go back to reference Martisova E, Aisa B, Guerenu G, Ramirez MJ. Effects of early maternal separation on biobehavioral and neuropathological markers of Alzheimer’s disease in adult male rats. Curr Alzheimer Res. 2013;10:420–32.PubMedCrossRef Martisova E, Aisa B, Guerenu G, Ramirez MJ. Effects of early maternal separation on biobehavioral and neuropathological markers of Alzheimer’s disease in adult male rats. Curr Alzheimer Res. 2013;10:420–32.PubMedCrossRef
83.
go back to reference Norton MC, Smith KR, Ostbye T, Tschanz JT, Schwartz S, Corcoran C, et al. Early parental death and remarriage of widowed parents as risk factors for Alzheimer disease: the Cache County study. Am J Geriatr Psychiatry. 2011;19:814–24.PubMedPubMedCentralCrossRef Norton MC, Smith KR, Ostbye T, Tschanz JT, Schwartz S, Corcoran C, et al. Early parental death and remarriage of widowed parents as risk factors for Alzheimer disease: the Cache County study. Am J Geriatr Psychiatry. 2011;19:814–24.PubMedPubMedCentralCrossRef
84.
go back to reference Donley GAR, Lonnroos E, Tuomainen TP, Kauhanen J. Association of childhood stress with late-life dementia and Alzheimer’s disease: the KIHD study. Eur J Public Health. 2018;28:1069–73.PubMedCrossRef Donley GAR, Lonnroos E, Tuomainen TP, Kauhanen J. Association of childhood stress with late-life dementia and Alzheimer’s disease: the KIHD study. Eur J Public Health. 2018;28:1069–73.PubMedCrossRef
85.
go back to reference Tani Y, Fujiwara T, Kondo K. Association between adverse childhood experiences and dementia in older japanese adults. JAMA Netw Open. 2020;3:e1920740.PubMedCrossRef Tani Y, Fujiwara T, Kondo K. Association between adverse childhood experiences and dementia in older japanese adults. JAMA Netw Open. 2020;3:e1920740.PubMedCrossRef
86.
go back to reference Tani Y, Fujiwara T, Kondo K. Adverse childhood experiences and dementia: interactions with social capital in the japan gerontological evaluation study cohort. Am J Prev Med. 2021;61:225–34.PubMedCrossRef Tani Y, Fujiwara T, Kondo K. Adverse childhood experiences and dementia: interactions with social capital in the japan gerontological evaluation study cohort. Am J Prev Med. 2021;61:225–34.PubMedCrossRef
87.
go back to reference Schickedanz HB, Jennings LA, Schickedanz A. The association between adverse childhood experiences and positive dementia screen in American older adults. J Gen Intern Med. 2022;37:2398–404.PubMedCrossRef Schickedanz HB, Jennings LA, Schickedanz A. The association between adverse childhood experiences and positive dementia screen in American older adults. J Gen Intern Med. 2022;37:2398–404.PubMedCrossRef
88.
go back to reference Roberts AL, Zafonte R, Chibnik LB, Baggish A, Taylor H, Baker J, et al. Association of adverse childhood experiences with poor neuropsychiatric health and dementia among former professional US football players. JAMA Netw Open. 2022;5:e223299.PubMedPubMedCentralCrossRef Roberts AL, Zafonte R, Chibnik LB, Baggish A, Taylor H, Baker J, et al. Association of adverse childhood experiences with poor neuropsychiatric health and dementia among former professional US football players. JAMA Netw Open. 2022;5:e223299.PubMedPubMedCentralCrossRef
89.
go back to reference Silverman MN, Sternberg EM. Glucocorticoid regulation of inflammation and its functional correlates: from HPA axis to glucocorticoid receptor dysfunction. Ann N Y Acad Sci. 2012;1261:55–63.PubMedPubMedCentralCrossRef Silverman MN, Sternberg EM. Glucocorticoid regulation of inflammation and its functional correlates: from HPA axis to glucocorticoid receptor dysfunction. Ann N Y Acad Sci. 2012;1261:55–63.PubMedPubMedCentralCrossRef
90.
91.
go back to reference Lind K, Edman A, Nordlund A, Olsson T, Wallin A. Increased saliva cortisol awakening response in patients with mild cognitive impairment. Dement Geriatr Cogn Disord. 2007;24:389–95.PubMedCrossRef Lind K, Edman A, Nordlund A, Olsson T, Wallin A. Increased saliva cortisol awakening response in patients with mild cognitive impairment. Dement Geriatr Cogn Disord. 2007;24:389–95.PubMedCrossRef
92.
go back to reference Milligan Armstrong A, Porter T, Quek H, White A, Haynes J, Jackaman C, et al. Chronic stress and Alzheimer’s disease: the interplay between the hypothalamic-pituitary-adrenal axis, genetics and microglia. Biol Rev Camb Philos Soc. 2021;96:2209–28.PubMedCrossRef Milligan Armstrong A, Porter T, Quek H, White A, Haynes J, Jackaman C, et al. Chronic stress and Alzheimer’s disease: the interplay between the hypothalamic-pituitary-adrenal axis, genetics and microglia. Biol Rev Camb Philos Soc. 2021;96:2209–28.PubMedCrossRef
93.
go back to reference Green KN, Billings LM, Roozendaal B, McGaugh JL, LaFerla FM. Glucocorticoids increase amyloid-beta and tau pathology in a mouse model of Alzheimer’s disease. J Neurosci. 2006;26:9047–56.PubMedPubMedCentralCrossRef Green KN, Billings LM, Roozendaal B, McGaugh JL, LaFerla FM. Glucocorticoids increase amyloid-beta and tau pathology in a mouse model of Alzheimer’s disease. J Neurosci. 2006;26:9047–56.PubMedPubMedCentralCrossRef
94.
go back to reference Baglietto-Vargas D, Medeiros R, Martinez-Coria H, LaFerla FM, Green KN. Mifepristone alters amyloid precursor protein processing to preclude amyloid beta and also reduces tau pathology. Biol Psychiatry. 2013;74:357–66.PubMedPubMedCentralCrossRef Baglietto-Vargas D, Medeiros R, Martinez-Coria H, LaFerla FM, Green KN. Mifepristone alters amyloid precursor protein processing to preclude amyloid beta and also reduces tau pathology. Biol Psychiatry. 2013;74:357–66.PubMedPubMedCentralCrossRef
95.
go back to reference Belanoff JK, Jurik J, Schatzberg LD, DeBattista C, Schatzberg AF. Slowing the progression of cognitive decline in Alzheimer’s disease using mifepristone. J Mol Neurosci. 2002;19:201–6.PubMedCrossRef Belanoff JK, Jurik J, Schatzberg LD, DeBattista C, Schatzberg AF. Slowing the progression of cognitive decline in Alzheimer’s disease using mifepristone. J Mol Neurosci. 2002;19:201–6.PubMedCrossRef
96.
go back to reference Banerjee SB, Arterbery AS, Fergus DJ, Adkins-Regan E. Deprivation of maternal care has long-lasting consequences for the hypothalamic-pituitary-adrenal axis of zebra finches. Proc Biol Sci. 2012;279:759–66.PubMed Banerjee SB, Arterbery AS, Fergus DJ, Adkins-Regan E. Deprivation of maternal care has long-lasting consequences for the hypothalamic-pituitary-adrenal axis of zebra finches. Proc Biol Sci. 2012;279:759–66.PubMed
97.
98.
go back to reference Molenaar NM, Tiemeier H, van Rossum EFC, Hillegers MHJ, Bockting CLH, Hoogendijk WJG, et al. Prenatal maternal psychopathology and stress and offspring HPA axis function at 6 years. Psychoneuroendocrinology. 2019;99:120–7.PubMedCrossRef Molenaar NM, Tiemeier H, van Rossum EFC, Hillegers MHJ, Bockting CLH, Hoogendijk WJG, et al. Prenatal maternal psychopathology and stress and offspring HPA axis function at 6 years. Psychoneuroendocrinology. 2019;99:120–7.PubMedCrossRef
99.
go back to reference Vallee M, Mayo W, Dellu F, Le Moal M, Simon H, Maccari S. Prenatal stress induces high anxiety and postnatal handling induces low anxiety in adult offspring: correlation with stress-induced corticosterone secretion. J Neurosci. 1997;17:2626–36.PubMedPubMedCentralCrossRef Vallee M, Mayo W, Dellu F, Le Moal M, Simon H, Maccari S. Prenatal stress induces high anxiety and postnatal handling induces low anxiety in adult offspring: correlation with stress-induced corticosterone secretion. J Neurosci. 1997;17:2626–36.PubMedPubMedCentralCrossRef
100.
go back to reference McIlwrick S, Rechenberg A, Matthes M, Burgstaller J, Schwarzbauer T, Chen A, et al. Genetic predisposition for high stress reactivity amplifies effects of early-life adversity. Psychoneuroendocrinology. 2016;70:85–97.PubMedCrossRef McIlwrick S, Rechenberg A, Matthes M, Burgstaller J, Schwarzbauer T, Chen A, et al. Genetic predisposition for high stress reactivity amplifies effects of early-life adversity. Psychoneuroendocrinology. 2016;70:85–97.PubMedCrossRef
101.
go back to reference Aisa B, Tordera R, Lasheras B, Del Rio J, Ramirez MJ. Cognitive impairment associated to HPA axis hyperactivity after maternal separation in rats. Psychoneuroendocrinology. 2007;32:256–66.PubMedCrossRef Aisa B, Tordera R, Lasheras B, Del Rio J, Ramirez MJ. Cognitive impairment associated to HPA axis hyperactivity after maternal separation in rats. Psychoneuroendocrinology. 2007;32:256–66.PubMedCrossRef
102.
go back to reference Kumsta R, Schlotz W, Golm D, Moser D, Kennedy M, Knights N, et al. HPA axis dysregulation in adult adoptees twenty years after severe institutional deprivation in childhood. Psychoneuroendocrinology. 2017;86:196–202.PubMedCrossRef Kumsta R, Schlotz W, Golm D, Moser D, Kennedy M, Knights N, et al. HPA axis dysregulation in adult adoptees twenty years after severe institutional deprivation in childhood. Psychoneuroendocrinology. 2017;86:196–202.PubMedCrossRef
103.
go back to reference Cryan JF, O’Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. The microbiota-gut-brain axis. Physiol Rev. 2019;99:1877–2013.PubMedCrossRef Cryan JF, O’Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. The microbiota-gut-brain axis. Physiol Rev. 2019;99:1877–2013.PubMedCrossRef
104.
go back to reference Socala K, Doboszewska U, Szopa A, Serefko A, Wlodarczyk M, Zielinska A, et al. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res. 2021;172:105840.PubMedCrossRef Socala K, Doboszewska U, Szopa A, Serefko A, Wlodarczyk M, Zielinska A, et al. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res. 2021;172:105840.PubMedCrossRef
105.
go back to reference Osadchiy V, Martin CR, Mayer EA. The gut-brain axis and the microbiome: mechanisms and clinical implications. Clin Gastroenterol Hepatol. 2019;17:322–32.PubMedCrossRef Osadchiy V, Martin CR, Mayer EA. The gut-brain axis and the microbiome: mechanisms and clinical implications. Clin Gastroenterol Hepatol. 2019;17:322–32.PubMedCrossRef
106.
go back to reference Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney RD, Shanahan F, et al. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry. 2013;18:666–73.PubMedCrossRef Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney RD, Shanahan F, et al. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry. 2013;18:666–73.PubMedCrossRef
107.
go back to reference Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, et al. The first microbial colonizers of the human gut: composition, activities, and health implications of the infant gut microbiota. Microbiol Mol Biol Rev. 2017;81. Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, et al. The first microbial colonizers of the human gut: composition, activities, and health implications of the infant gut microbiota. Microbiol Mol Biol Rev. 2017;81.
108.
go back to reference Zhuang ZQ, Shen LL, Li WW, Fu X, Zeng F, Gui L, et al. Gut microbiota is altered in patients with Alzheimer’s disease. J Alzheimers Dis. 2018;63:1337–46.PubMedCrossRef Zhuang ZQ, Shen LL, Li WW, Fu X, Zeng F, Gui L, et al. Gut microbiota is altered in patients with Alzheimer’s disease. J Alzheimers Dis. 2018;63:1337–46.PubMedCrossRef
109.
go back to reference Guo M, Peng J, Huang X, Xiao L, Huang F, Zuo Z. Gut microbiome features of chinese patients newly diagnosed with Alzheimer’s disease or mild cognitive impairment. J Alzheimers Dis. 2021;80:299–310.PubMedCrossRef Guo M, Peng J, Huang X, Xiao L, Huang F, Zuo Z. Gut microbiome features of chinese patients newly diagnosed with Alzheimer’s disease or mild cognitive impairment. J Alzheimers Dis. 2021;80:299–310.PubMedCrossRef
110.
go back to reference Li B, He Y, Ma J, Huang P, Du J, Cao L, et al. Mild cognitive impairment has similar alterations as Alzheimer’s disease in gut microbiota. Alzheimers Dement. 2019;15:1357–66.PubMedCrossRef Li B, He Y, Ma J, Huang P, Du J, Cao L, et al. Mild cognitive impairment has similar alterations as Alzheimer’s disease in gut microbiota. Alzheimers Dement. 2019;15:1357–66.PubMedCrossRef
112.
go back to reference Chen C, Liao J, Xia Y, Liu X, Jones R, Haran J, et al. Gut microbiota regulate Alzheimer’s disease pathologies and cognitive disorders via PUFA-associated neuroinflammation. Gut. 2022;71:2233–52.PubMedCrossRef Chen C, Liao J, Xia Y, Liu X, Jones R, Haran J, et al. Gut microbiota regulate Alzheimer’s disease pathologies and cognitive disorders via PUFA-associated neuroinflammation. Gut. 2022;71:2233–52.PubMedCrossRef
113.
go back to reference Kim MS, Kim Y, Choi H, Kim W, Park S, Lee D, et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut. 2020;69:283–94.PubMedCrossRef Kim MS, Kim Y, Choi H, Kim W, Park S, Lee D, et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut. 2020;69:283–94.PubMedCrossRef
114.
go back to reference Tran TTT, Corsini S, Kellingray L, Hegarty C, Le Gall G, Narbad A, et al. APOE genotype influences the gut microbiome structure and function in humans and mice: relevance for Alzheimer’s disease pathophysiology. FASEB J. 2019;33:8221–31.PubMedPubMedCentralCrossRef Tran TTT, Corsini S, Kellingray L, Hegarty C, Le Gall G, Narbad A, et al. APOE genotype influences the gut microbiome structure and function in humans and mice: relevance for Alzheimer’s disease pathophysiology. FASEB J. 2019;33:8221–31.PubMedPubMedCentralCrossRef
115.
go back to reference Kim N, Jeon SH, Ju IG, Gee MS, Do J, Oh MS, et al. Transplantation of gut microbiota derived from Alzheimer’s disease mouse model impairs memory function and neurogenesis in C57BL/6 mice. Brain Behav Immun. 2021;98:357–65.PubMedCrossRef Kim N, Jeon SH, Ju IG, Gee MS, Do J, Oh MS, et al. Transplantation of gut microbiota derived from Alzheimer’s disease mouse model impairs memory function and neurogenesis in C57BL/6 mice. Brain Behav Immun. 2021;98:357–65.PubMedCrossRef
116.
go back to reference Yang X, Yu D, Xue L, Li H, Du J. Probiotics modulate the microbiota-gut-brain axis and improve memory deficits in aged SAMP8 mice. Acta Pharm Sin B. 2020;10:475–87.PubMedCrossRef Yang X, Yu D, Xue L, Li H, Du J. Probiotics modulate the microbiota-gut-brain axis and improve memory deficits in aged SAMP8 mice. Acta Pharm Sin B. 2020;10:475–87.PubMedCrossRef
117.
go back to reference Kim CS, Cha L, Sim M, Jung S, Chun WY, Baik HW, et al. Probiotic supplementation improves cognitive function and mood with changes in gut microbiota in community-dwelling older adults: a randomized, double-blind, placebo-controlled, multicenter trial. J Gerontol A Biol Sci Med Sci. 2021;76:32–40.PubMedCrossRef Kim CS, Cha L, Sim M, Jung S, Chun WY, Baik HW, et al. Probiotic supplementation improves cognitive function and mood with changes in gut microbiota in community-dwelling older adults: a randomized, double-blind, placebo-controlled, multicenter trial. J Gerontol A Biol Sci Med Sci. 2021;76:32–40.PubMedCrossRef
118.
go back to reference Liu Q, Xi Y, Wang Q, Liu J, Li P, Meng X, et al. Mannan oligosaccharide attenuates cognitive and behavioral disorders in the 5xFAD Alzheimer’s disease mouse model via regulating the gut microbiota-brain axis. Brain Behav Immun. 2021;95:330–43.PubMedCrossRef Liu Q, Xi Y, Wang Q, Liu J, Li P, Meng X, et al. Mannan oligosaccharide attenuates cognitive and behavioral disorders in the 5xFAD Alzheimer’s disease mouse model via regulating the gut microbiota-brain axis. Brain Behav Immun. 2021;95:330–43.PubMedCrossRef
119.
go back to reference Deng Y, Zhou M, Wang J, Yao J, Yu J, Liu W, et al. Involvement of the microbiota-gut-brain axis in chronic restraint stress: disturbances of the kynurenine metabolic pathway in both the gut and brain. Gut Microbes. 2021;13:1–16.PubMedCrossRef Deng Y, Zhou M, Wang J, Yao J, Yu J, Liu W, et al. Involvement of the microbiota-gut-brain axis in chronic restraint stress: disturbances of the kynurenine metabolic pathway in both the gut and brain. Gut Microbes. 2021;13:1–16.PubMedCrossRef
120.
go back to reference O’Mahony SM, Marchesi JR, Scully P, Codling C, Ceolho AM, Quigley EM, et al. Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnesses. Biol Psychiatry. 2009;65:263–7.PubMedCrossRef O’Mahony SM, Marchesi JR, Scully P, Codling C, Ceolho AM, Quigley EM, et al. Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnesses. Biol Psychiatry. 2009;65:263–7.PubMedCrossRef
121.
go back to reference Enqi W, Jingzhu S, Lingpeng P, Yaqin L. Comparison of the gut microbiota disturbance in rat models of irritable bowel syndrome induced by maternal separation and multiple early-life adversity. Front Cell Infect Microbiol. 2020;10:581974.PubMedCrossRef Enqi W, Jingzhu S, Lingpeng P, Yaqin L. Comparison of the gut microbiota disturbance in rat models of irritable bowel syndrome induced by maternal separation and multiple early-life adversity. Front Cell Infect Microbiol. 2020;10:581974.PubMedCrossRef
122.
go back to reference Rincel M, Aubert P, Chevalier J, Grohard PA, Basso L, Monchaux de Oliveira C, et al. Multi-hit early life adversity affects gut microbiota, brain and behavior in a sex-dependent manner. Brain Behav Immun. 2019;80:179–92.PubMedCrossRef Rincel M, Aubert P, Chevalier J, Grohard PA, Basso L, Monchaux de Oliveira C, et al. Multi-hit early life adversity affects gut microbiota, brain and behavior in a sex-dependent manner. Brain Behav Immun. 2019;80:179–92.PubMedCrossRef
123.
go back to reference Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, et al. Gut microbiome alterations in Alzheimer’s disease. Sci Rep. 2017;7:13537.PubMedPubMedCentralCrossRef Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, et al. Gut microbiome alterations in Alzheimer’s disease. Sci Rep. 2017;7:13537.PubMedPubMedCentralCrossRef
124.
go back to reference Haran JP, Bhattarai SK, Foley SE, Dutta P, Ward DV, Bucci V, et al. Alzheimer’s disease microbiome is associated with dysregulation of the anti-inflammatory P-glycoprotein pathway. mBio. 2019;10. Haran JP, Bhattarai SK, Foley SE, Dutta P, Ward DV, Bucci V, et al. Alzheimer’s disease microbiome is associated with dysregulation of the anti-inflammatory P-glycoprotein pathway. mBio. 2019;10.
125.
go back to reference Zhou Y, Wang Y, Quan M, Zhao H, Jia J. Gut microbiota changes and their correlation with cognitive and neuropsychiatric symptoms in Alzheimer’s disease. J Alzheimers Dis. 2021;81:583–95.PubMedCrossRef Zhou Y, Wang Y, Quan M, Zhao H, Jia J. Gut microbiota changes and their correlation with cognitive and neuropsychiatric symptoms in Alzheimer’s disease. J Alzheimers Dis. 2021;81:583–95.PubMedCrossRef
127.
go back to reference Ma X, Zhang Y, Xu T, Qian M, Yang Z, Zhan X, et al. Early-life intervention using exogenous fecal microbiota alleviates gut injury and reduce inflammation caused by weaning stress in piglets. Front Microbiol. 2021;12:671683.PubMedPubMedCentralCrossRef Ma X, Zhang Y, Xu T, Qian M, Yang Z, Zhan X, et al. Early-life intervention using exogenous fecal microbiota alleviates gut injury and reduce inflammation caused by weaning stress in piglets. Front Microbiol. 2021;12:671683.PubMedPubMedCentralCrossRef
128.
go back to reference Reid BM, Horne R, Donzella B, Szamosi JC, Coe CL, Foster JA, et al. Microbiota-immune alterations in adolescents following early life adversity: a proof of concept study. Dev Psychobiol. 2021;63:851–63.PubMedCrossRef Reid BM, Horne R, Donzella B, Szamosi JC, Coe CL, Foster JA, et al. Microbiota-immune alterations in adolescents following early life adversity: a proof of concept study. Dev Psychobiol. 2021;63:851–63.PubMedCrossRef
129.
go back to reference Coley EJL, Mayer EA, Osadchiy V, Chen Z, Subramanyam V, Zhang Y, et al. Early life adversity predicts brain-gut alterations associated with increased stress and mood. Neurobiol Stress. 2021;15:100348.PubMedPubMedCentralCrossRef Coley EJL, Mayer EA, Osadchiy V, Chen Z, Subramanyam V, Zhang Y, et al. Early life adversity predicts brain-gut alterations associated with increased stress and mood. Neurobiol Stress. 2021;15:100348.PubMedPubMedCentralCrossRef
130.
go back to reference Simons M, Nave KA. Oligodendrocytes: myelination and axonal support. Cold Spring Harb Perspect Biol. 2015;8:a020479.PubMedCrossRef Simons M, Nave KA. Oligodendrocytes: myelination and axonal support. Cold Spring Harb Perspect Biol. 2015;8:a020479.PubMedCrossRef
131.
go back to reference Baumann N, Pham-Dinh D. Biology of oligodendrocyte and myelin in the mammalian central nervous system. Physiol Rev. 2001;81:871–927.PubMedCrossRef Baumann N, Pham-Dinh D. Biology of oligodendrocyte and myelin in the mammalian central nervous system. Physiol Rev. 2001;81:871–927.PubMedCrossRef
132.
133.
go back to reference Wang F, Ren SY, Chen JF, Liu K, Li RX, Li ZF, et al. Myelin degeneration and diminished myelin renewal contribute to age-related deficits in memory. Nat Neurosci. 2020;23:481–6.PubMedPubMedCentralCrossRef Wang F, Ren SY, Chen JF, Liu K, Li RX, Li ZF, et al. Myelin degeneration and diminished myelin renewal contribute to age-related deficits in memory. Nat Neurosci. 2020;23:481–6.PubMedPubMedCentralCrossRef
134.
go back to reference Chen JF, Liu K, Hu B, Li RR, Xin W, Chen H, et al. Enhancing myelin renewal reverses cognitive dysfunction in a murine model of Alzheimer’s disease. Neuron. 2021;109:2292-307.e5.PubMedPubMedCentralCrossRef Chen JF, Liu K, Hu B, Li RR, Xin W, Chen H, et al. Enhancing myelin renewal reverses cognitive dysfunction in a murine model of Alzheimer’s disease. Neuron. 2021;109:2292-307.e5.PubMedPubMedCentralCrossRef
135.
go back to reference Steadman PE, Xia F, Ahmed M, Mocle AJ, Penning ARA, Geraghty AC, et al. Disruption of oligodendrogenesis impairs memory consolidation in adult mice. Neuron. 2020;105:150-64.e6.PubMedCrossRef Steadman PE, Xia F, Ahmed M, Mocle AJ, Penning ARA, Geraghty AC, et al. Disruption of oligodendrogenesis impairs memory consolidation in adult mice. Neuron. 2020;105:150-64.e6.PubMedCrossRef
136.
go back to reference Chen X, Wang F, Gan J, Zhang Z, Liang X, Li T, et al. Myelin deficits caused by Olig2 deficiency lead to cognitive dysfunction and increase vulnerability to social withdrawal in adult mice. Neurosci Bull. 2020;36:419–26.PubMedCrossRef Chen X, Wang F, Gan J, Zhang Z, Liang X, Li T, et al. Myelin deficits caused by Olig2 deficiency lead to cognitive dysfunction and increase vulnerability to social withdrawal in adult mice. Neurosci Bull. 2020;36:419–26.PubMedCrossRef
137.
go back to reference Boda E. Myelin and oligodendrocyte lineage cell dysfunctions: New players in the etiology and treatment of depression and stress-related disorders. Eur J Neurosci. 2021;53:281–97.PubMedCrossRef Boda E. Myelin and oligodendrocyte lineage cell dysfunctions: New players in the etiology and treatment of depression and stress-related disorders. Eur J Neurosci. 2021;53:281–97.PubMedCrossRef
138.
go back to reference Bonnefil V, Dietz K, Amatruda M, Wentling M, Aubry AV, Dupree JL, et al. Region-specific myelin differences define behavioral consequences of chronic social defeat stress in mice. Elife. 2019;8. Bonnefil V, Dietz K, Amatruda M, Wentling M, Aubry AV, Dupree JL, et al. Region-specific myelin differences define behavioral consequences of chronic social defeat stress in mice. Elife. 2019;8.
139.
go back to reference Kokkosis AG, Madeira MM, Mullahy MR, Tsirka SE. Chronic stress disrupts the homeostasis and progeny progression of oligodendroglial lineage cells, associating immune oligodendrocytes with prefrontal cortex hypomyelination. Mol Psychiatry. 2022;27:2833–48.PubMedPubMedCentralCrossRef Kokkosis AG, Madeira MM, Mullahy MR, Tsirka SE. Chronic stress disrupts the homeostasis and progeny progression of oligodendroglial lineage cells, associating immune oligodendrocytes with prefrontal cortex hypomyelination. Mol Psychiatry. 2022;27:2833–48.PubMedPubMedCentralCrossRef
140.
go back to reference Huang CX, Xiao Q, Zhang L, Gao Y, Ma J, Liang X, et al. Stress-induced myelin damage in the hippocampal formation in a rat model of depression. J Psychiatr Res. 2022;155:401–9.PubMedCrossRef Huang CX, Xiao Q, Zhang L, Gao Y, Ma J, Liang X, et al. Stress-induced myelin damage in the hippocampal formation in a rat model of depression. J Psychiatr Res. 2022;155:401–9.PubMedCrossRef
141.
go back to reference Lutz PE, Tanti A, Gasecka A, Barnett-Burns S, Kim JJ, Zhou Y, et al. Association of a history of child abuse with impaired myelination in the anterior cingulate cortex: convergent epigenetic, transcriptional, and morphological evidence. Am J Psychiatry. 2017;174:1185–94.PubMedCrossRef Lutz PE, Tanti A, Gasecka A, Barnett-Burns S, Kim JJ, Zhou Y, et al. Association of a history of child abuse with impaired myelination in the anterior cingulate cortex: convergent epigenetic, transcriptional, and morphological evidence. Am J Psychiatry. 2017;174:1185–94.PubMedCrossRef
142.
go back to reference Ziegler G, Moutoussis M, Hauser TU, Fearon P, Bullmore ET, Goodyer IM, et al. Childhood socio-economic disadvantage predicts reduced myelin growth across adolescence and young adulthood. Hum Brain Mapp. 2020;41:3392–402.PubMedPubMedCentralCrossRef Ziegler G, Moutoussis M, Hauser TU, Fearon P, Bullmore ET, Goodyer IM, et al. Childhood socio-economic disadvantage predicts reduced myelin growth across adolescence and young adulthood. Hum Brain Mapp. 2020;41:3392–402.PubMedPubMedCentralCrossRef
143.
go back to reference Bordner KA, George ED, Carlyle BC, Duque A, Kitchen RR, Lam TT, et al. Functional genomic and proteomic analysis reveals disruption of myelin-related genes and translation in a mouse model of early life neglect. Front Psychiatry. 2011;2:18.PubMedPubMedCentralCrossRef Bordner KA, George ED, Carlyle BC, Duque A, Kitchen RR, Lam TT, et al. Functional genomic and proteomic analysis reveals disruption of myelin-related genes and translation in a mouse model of early life neglect. Front Psychiatry. 2011;2:18.PubMedPubMedCentralCrossRef
144.
go back to reference Yang Y, Cheng Z, Tang H, Jiao H, Sun X, Cui Q, et al. Neonatal maternal separation impairs prefrontal cortical myelination and cognitive functions in rats through activation of wnt signaling. Cereb Cortex. 2017;27:2871–84.PubMed Yang Y, Cheng Z, Tang H, Jiao H, Sun X, Cui Q, et al. Neonatal maternal separation impairs prefrontal cortical myelination and cognitive functions in rats through activation of wnt signaling. Cereb Cortex. 2017;27:2871–84.PubMed
145.
go back to reference Wang Y, Su Y, Yu G, Wang X, Chen X, Yu B, et al. Reduced oligodendrocyte precursor cell impairs astrocytic development in early life stress. Adv Sci (Weinh). 2021;8:e2101181.PubMedCrossRef Wang Y, Su Y, Yu G, Wang X, Chen X, Yu B, et al. Reduced oligodendrocyte precursor cell impairs astrocytic development in early life stress. Adv Sci (Weinh). 2021;8:e2101181.PubMedCrossRef
146.
go back to reference Teissier A, Le Magueresse C, Olusakin J, Andrade da Costa BLS, De Stasi AM, Bacci A, et al. Early-life stress impairs postnatal oligodendrogenesis and adult emotional behaviour through activity-dependent mechanisms. Mol Psychiatry. 2020;25:1159–74.PubMedCrossRef Teissier A, Le Magueresse C, Olusakin J, Andrade da Costa BLS, De Stasi AM, Bacci A, et al. Early-life stress impairs postnatal oligodendrogenesis and adult emotional behaviour through activity-dependent mechanisms. Mol Psychiatry. 2020;25:1159–74.PubMedCrossRef
148.
go back to reference Newcombe EA, Camats-Perna J, Silva ML, Valmas N, Huat TJ, Medeiros R. Inflammation: the link between comorbidities, genetics, and Alzheimer’s disease. J Neuroinflammation. 2018;15:276.PubMedPubMedCentralCrossRef Newcombe EA, Camats-Perna J, Silva ML, Valmas N, Huat TJ, Medeiros R. Inflammation: the link between comorbidities, genetics, and Alzheimer’s disease. J Neuroinflammation. 2018;15:276.PubMedPubMedCentralCrossRef
149.
150.
go back to reference Bradburn S, Murgatroyd C, Ray N. Neuroinflammation in mild cognitive impairment and Alzheimer’s disease: a meta-analysis. Ageing Res Rev. 2019;50:1–8.PubMedCrossRef Bradburn S, Murgatroyd C, Ray N. Neuroinflammation in mild cognitive impairment and Alzheimer’s disease: a meta-analysis. Ageing Res Rev. 2019;50:1–8.PubMedCrossRef
151.
go back to reference Chen X, Hu Y, Cao Z, Liu Q, Cheng Y. Cerebrospinal fluid inflammatory cytokine aberrations in Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis: a systematic review and meta-analysis. Front Immunol. 2018;9:2122.PubMedPubMedCentralCrossRef Chen X, Hu Y, Cao Z, Liu Q, Cheng Y. Cerebrospinal fluid inflammatory cytokine aberrations in Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis: a systematic review and meta-analysis. Front Immunol. 2018;9:2122.PubMedPubMedCentralCrossRef
152.
go back to reference Brosseron F, Traschutz A, Widmann CN, Kummer MP, Tacik P, Santarelli F, et al. Characterization and clinical use of inflammatory cerebrospinal fluid protein markers in Alzheimer’s disease. Alzheimers Res Ther. 2018;10:25.PubMedPubMedCentralCrossRef Brosseron F, Traschutz A, Widmann CN, Kummer MP, Tacik P, Santarelli F, et al. Characterization and clinical use of inflammatory cerebrospinal fluid protein markers in Alzheimer’s disease. Alzheimers Res Ther. 2018;10:25.PubMedPubMedCentralCrossRef
153.
go back to reference Lopez-Rodriguez AB, Hennessy E, Murray CL, Nazmi A, Delaney HJ, Healy D, et al. Acute systemic inflammation exacerbates neuroinflammation in Alzheimer’s disease: IL-1beta drives amplified responses in primed astrocytes and neuronal network dysfunction. Alzheimers Dement. 2021;17:1735–55.PubMedPubMedCentralCrossRef Lopez-Rodriguez AB, Hennessy E, Murray CL, Nazmi A, Delaney HJ, Healy D, et al. Acute systemic inflammation exacerbates neuroinflammation in Alzheimer’s disease: IL-1beta drives amplified responses in primed astrocytes and neuronal network dysfunction. Alzheimers Dement. 2021;17:1735–55.PubMedPubMedCentralCrossRef
154.
go back to reference Ising C, Venegas C, Zhang S, Scheiblich H, Schmidt SV, Vieira-Saecker A, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575:669–73.PubMedPubMedCentralCrossRef Ising C, Venegas C, Zhang S, Scheiblich H, Schmidt SV, Vieira-Saecker A, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575:669–73.PubMedPubMedCentralCrossRef
155.
go back to reference Dong Y, Li S, Lu Y, Li X, Liao Y, Peng Z, et al. Stress-induced NLRP3 inflammasome activation negatively regulates fear memory in mice. J Neuroinflammation. 2020;17:205.PubMedPubMedCentralCrossRef Dong Y, Li S, Lu Y, Li X, Liao Y, Peng Z, et al. Stress-induced NLRP3 inflammasome activation negatively regulates fear memory in mice. J Neuroinflammation. 2020;17:205.PubMedPubMedCentralCrossRef
156.
go back to reference Jolodar SK, Bigdeli M, Moghaddam AH. Hypericin ameliorates maternal separation-induced cognitive deficits and hippocampal inflammation in rats. Mini Rev Med Chem. 2021;21:1144–9.PubMedCrossRef Jolodar SK, Bigdeli M, Moghaddam AH. Hypericin ameliorates maternal separation-induced cognitive deficits and hippocampal inflammation in rats. Mini Rev Med Chem. 2021;21:1144–9.PubMedCrossRef
157.
go back to reference Mi X, Zeng GR, Liu JQ, Luo ZS, Zhang L, Dai XM, et al. Ganoderma lucidum triterpenoids improve maternal separation-induced anxiety- and depression-like behaviors in mice by mitigating inflammation in the periphery and brain. Nutrients. 2022;14:2268.PubMedPubMedCentralCrossRef Mi X, Zeng GR, Liu JQ, Luo ZS, Zhang L, Dai XM, et al. Ganoderma lucidum triterpenoids improve maternal separation-induced anxiety- and depression-like behaviors in mice by mitigating inflammation in the periphery and brain. Nutrients. 2022;14:2268.PubMedPubMedCentralCrossRef
158.
159.
go back to reference Hu X. Microglia/macrophage polarization: Fantasy or evidence of functional diversity? J Cereb Blood Flow Metab. 2020;40:134–S6.CrossRef Hu X. Microglia/macrophage polarization: Fantasy or evidence of functional diversity? J Cereb Blood Flow Metab. 2020;40:134–S6.CrossRef
160.
go back to reference Wang YL, Han QQ, Gong WQ, Pan DH, Wang LZ, Hu W, et al. Microglial activation mediates chronic mild stress-induced depressive- and anxiety-like behavior in adult rats. J Neuroinflammation. 2018;15:21.PubMedPubMedCentralCrossRef Wang YL, Han QQ, Gong WQ, Pan DH, Wang LZ, Hu W, et al. Microglial activation mediates chronic mild stress-induced depressive- and anxiety-like behavior in adult rats. J Neuroinflammation. 2018;15:21.PubMedPubMedCentralCrossRef
161.
go back to reference Sugama S, Takenouchi T, Hashimoto M, Ohata H, Takenaka Y, Kakinuma Y. Stress-induced microglial activation occurs through beta-adrenergic receptor: noradrenaline as a key neurotransmitter in microglial activation. J Neuroinflammation. 2019;16:266.PubMedPubMedCentralCrossRef Sugama S, Takenouchi T, Hashimoto M, Ohata H, Takenaka Y, Kakinuma Y. Stress-induced microglial activation occurs through beta-adrenergic receptor: noradrenaline as a key neurotransmitter in microglial activation. J Neuroinflammation. 2019;16:266.PubMedPubMedCentralCrossRef
162.
go back to reference Roque A, Ochoa-Zarzosa A, Torner L. Maternal separation activates microglial cells and induces an inflammatory response in the hippocampus of male rat pups, independently of hypothalamic and peripheral cytokine levels. Brain Behav Immun. 2016;55:39–48.PubMedCrossRef Roque A, Ochoa-Zarzosa A, Torner L. Maternal separation activates microglial cells and induces an inflammatory response in the hippocampus of male rat pups, independently of hypothalamic and peripheral cytokine levels. Brain Behav Immun. 2016;55:39–48.PubMedCrossRef
163.
go back to reference Gomez-Gonzalez B, Escobar A. Altered functional development of the blood-brain barrier after early life stress in the rat. Brain Res Bull. 2009;79:376–87.PubMedCrossRef Gomez-Gonzalez B, Escobar A. Altered functional development of the blood-brain barrier after early life stress in the rat. Brain Res Bull. 2009;79:376–87.PubMedCrossRef
164.
go back to reference Welcome MO, Mastorakis NE. Stress-induced blood brain barrier disruption: molecular mechanisms and signaling pathways. Pharmacol Res. 2020;157:104769.PubMedCrossRef Welcome MO, Mastorakis NE. Stress-induced blood brain barrier disruption: molecular mechanisms and signaling pathways. Pharmacol Res. 2020;157:104769.PubMedCrossRef
165.
go back to reference Bolton JL, Short AK, Othy S, Kooiker CL, Shao M, Gunn BG, et al. Early stress-induced impaired microglial pruning of excitatory synapses on immature CRH-expressing neurons provokes aberrant adult stress responses. Cell Rep. 2022;38:110600.PubMedPubMedCentralCrossRef Bolton JL, Short AK, Othy S, Kooiker CL, Shao M, Gunn BG, et al. Early stress-induced impaired microglial pruning of excitatory synapses on immature CRH-expressing neurons provokes aberrant adult stress responses. Cell Rep. 2022;38:110600.PubMedPubMedCentralCrossRef
166.
167.
go back to reference Moreno-Jimenez EP, Terreros-Roncal J, Flor-Garcia M, Rabano A, Llorens-Martin M. Evidences for adult hippocampal neurogenesis in humans. J Neurosci. 2021;41:2541–53.PubMedPubMedCentralCrossRef Moreno-Jimenez EP, Terreros-Roncal J, Flor-Garcia M, Rabano A, Llorens-Martin M. Evidences for adult hippocampal neurogenesis in humans. J Neurosci. 2021;41:2541–53.PubMedPubMedCentralCrossRef
169.
go back to reference Moreno-Jimenez EP, Flor-Garcia M, Terreros-Roncal J, Rabano A, Cafini F, Pallas-Bazarra N, et al. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat Med. 2019;25:554–60.PubMedCrossRef Moreno-Jimenez EP, Flor-Garcia M, Terreros-Roncal J, Rabano A, Cafini F, Pallas-Bazarra N, et al. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat Med. 2019;25:554–60.PubMedCrossRef
170.
go back to reference Hollands C, Tobin MK, Hsu M, Musaraca K, Yu TS, Mishra R, et al. Depletion of adult neurogenesis exacerbates cognitive deficits in Alzheimer’s disease by compromising hippocampal inhibition. Mol Neurodegener. 2017;12:64.PubMedPubMedCentralCrossRef Hollands C, Tobin MK, Hsu M, Musaraca K, Yu TS, Mishra R, et al. Depletion of adult neurogenesis exacerbates cognitive deficits in Alzheimer’s disease by compromising hippocampal inhibition. Mol Neurodegener. 2017;12:64.PubMedPubMedCentralCrossRef
171.
go back to reference Xu L, Guo Y, Wang G, Sun G, Sun W, Li J, et al. Inhibition of adult hippocampal neurogenesis plays a role in sevoflurane-induced cognitive impairment in aged mice through brain-derived neurotrophic factor/tyrosine receptor kinase B and neurotrophin-3/tropomyosin receptor kinase C pathways. Front Aging Neurosci. 2022;14:782932.PubMedPubMedCentralCrossRef Xu L, Guo Y, Wang G, Sun G, Sun W, Li J, et al. Inhibition of adult hippocampal neurogenesis plays a role in sevoflurane-induced cognitive impairment in aged mice through brain-derived neurotrophic factor/tyrosine receptor kinase B and neurotrophin-3/tropomyosin receptor kinase C pathways. Front Aging Neurosci. 2022;14:782932.PubMedPubMedCentralCrossRef
172.
go back to reference Choi SH, Bylykbashi E, Chatila ZK, Lee SW, Pulli B, Clemenson GD, et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer’s mouse model. Science. 2018;361:eaan8821.PubMedPubMedCentralCrossRef Choi SH, Bylykbashi E, Chatila ZK, Lee SW, Pulli B, Clemenson GD, et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer’s mouse model. Science. 2018;361:eaan8821.PubMedPubMedCentralCrossRef
173.
go back to reference Mirescu C, Peters JD, Gould E. Early life experience alters response of adult neurogenesis to stress. Nat Neurosci. 2004;7:841–6.PubMedCrossRef Mirescu C, Peters JD, Gould E. Early life experience alters response of adult neurogenesis to stress. Nat Neurosci. 2004;7:841–6.PubMedCrossRef
174.
go back to reference Ruiz R, Roque A, Pineda E, Licona-Limon P, Jose Valdez-Alarcon J, Lajud N. Early life stress accelerates age-induced effects on neurogenesis, depression, and metabolic risk. Psychoneuroendocrinology. 2018;96:203–11.PubMedCrossRef Ruiz R, Roque A, Pineda E, Licona-Limon P, Jose Valdez-Alarcon J, Lajud N. Early life stress accelerates age-induced effects on neurogenesis, depression, and metabolic risk. Psychoneuroendocrinology. 2018;96:203–11.PubMedCrossRef
175.
go back to reference Suri D, Veenit V, Sarkar A, Thiagarajan D, Kumar A, Nestler EJ, et al. Early stress evokes age-dependent biphasic changes in hippocampal neurogenesis, BDNF expression, and cognition. Biol Psychiatry. 2013;73:658–66.PubMedCrossRef Suri D, Veenit V, Sarkar A, Thiagarajan D, Kumar A, Nestler EJ, et al. Early stress evokes age-dependent biphasic changes in hippocampal neurogenesis, BDNF expression, and cognition. Biol Psychiatry. 2013;73:658–66.PubMedCrossRef
176.
go back to reference Youssef M, Atsak P, Cardenas J, Kosmidis S, Leonardo ED, Dranovsky A. Early life stress delays hippocampal development and diminishes the adult stem cell pool in mice. Sci Rep. 2019;9:4120.PubMedPubMedCentralCrossRef Youssef M, Atsak P, Cardenas J, Kosmidis S, Leonardo ED, Dranovsky A. Early life stress delays hippocampal development and diminishes the adult stem cell pool in mice. Sci Rep. 2019;9:4120.PubMedPubMedCentralCrossRef
177.
go back to reference Frank MG, Miguel ZD, Watkins LR, Maier SF. Prior exposure to glucocorticoids sensitizes the neuroinflammatory and peripheral inflammatory responses to E. coli lipopolysaccharide. Brain Behav Immun. 2010;24:19–30.PubMedCrossRef Frank MG, Miguel ZD, Watkins LR, Maier SF. Prior exposure to glucocorticoids sensitizes the neuroinflammatory and peripheral inflammatory responses to E. coli lipopolysaccharide. Brain Behav Immun. 2010;24:19–30.PubMedCrossRef
178.
go back to reference Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O. Inflammation is detrimental for neurogenesis in adult brain. Proc Natl Acad Sci U S A. 2003;100:13632–7.PubMedPubMedCentralCrossRef Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O. Inflammation is detrimental for neurogenesis in adult brain. Proc Natl Acad Sci U S A. 2003;100:13632–7.PubMedPubMedCentralCrossRef
179.
180.
go back to reference Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18:965–77.PubMedPubMedCentralCrossRef Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18:965–77.PubMedPubMedCentralCrossRef
181.
go back to reference Hoban AE, Stilling RM, Ryan FJ, Shanahan F, Dinan TG, Claesson MJ, et al. Regulation of prefrontal cortex myelination by the microbiota. Transl Psychiatry. 2016;6:e774.PubMedPubMedCentralCrossRef Hoban AE, Stilling RM, Ryan FJ, Shanahan F, Dinan TG, Claesson MJ, et al. Regulation of prefrontal cortex myelination by the microbiota. Transl Psychiatry. 2016;6:e774.PubMedPubMedCentralCrossRef
182.
go back to reference Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell. 2016;167:1469-80.e12.PubMedPubMedCentralCrossRef Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell. 2016;167:1469-80.e12.PubMedPubMedCentralCrossRef
183.
go back to reference Sege RD, Hatmaker-Flanigan E, De Vos E, Levin-Goodman R, Spivak H. Anticipatory guidance and violence prevention: results from family and pediatrician focus groups. Pediatrics. 2006;117:455–63.PubMedCrossRef Sege RD, Hatmaker-Flanigan E, De Vos E, Levin-Goodman R, Spivak H. Anticipatory guidance and violence prevention: results from family and pediatrician focus groups. Pediatrics. 2006;117:455–63.PubMedCrossRef
184.
go back to reference Shaefer HL, Collyer S, Duncan G, Edin K, Garfinkel I, Harris D, et al. A universal child allowance: a plan to reduce poverty and income instability among children in the United States. RSF. 2018;4:22–42.PubMedPubMedCentralCrossRef Shaefer HL, Collyer S, Duncan G, Edin K, Garfinkel I, Harris D, et al. A universal child allowance: a plan to reduce poverty and income instability among children in the United States. RSF. 2018;4:22–42.PubMedPubMedCentralCrossRef
185.
go back to reference Kempermann G. Environmental enrichment, new neurons and the neurobiology of individuality. Nat Rev Neurosci. 2019;20:235–45.PubMedCrossRef Kempermann G. Environmental enrichment, new neurons and the neurobiology of individuality. Nat Rev Neurosci. 2019;20:235–45.PubMedCrossRef
186.
go back to reference Ohline SM, Abraham WC. Environmental enrichment effects on synaptic and cellular physiology of hippocampal neurons. Neuropharmacology. 2019;145:3–12.PubMedCrossRef Ohline SM, Abraham WC. Environmental enrichment effects on synaptic and cellular physiology of hippocampal neurons. Neuropharmacology. 2019;145:3–12.PubMedCrossRef
187.
go back to reference do Prado CH, Narahari T, Holland FH, Lee HN, Murthy SK, Brenhouse HC. Effects of early adolescent environmental enrichment on cognitive dysfunction, prefrontal cortex development, and inflammatory cytokines after early life stress. Dev Psychobiol. 2016;58:482–91.PubMedCrossRef do Prado CH, Narahari T, Holland FH, Lee HN, Murthy SK, Brenhouse HC. Effects of early adolescent environmental enrichment on cognitive dysfunction, prefrontal cortex development, and inflammatory cytokines after early life stress. Dev Psychobiol. 2016;58:482–91.PubMedCrossRef
188.
go back to reference Rule L, Yang J, Watkin H, Hall J, Brydges NM. Environmental enrichment rescues survival and function of adult-born neurons following early life stress. Mol Psychiatry. 2021;26:1898–908.PubMedCrossRef Rule L, Yang J, Watkin H, Hall J, Brydges NM. Environmental enrichment rescues survival and function of adult-born neurons following early life stress. Mol Psychiatry. 2021;26:1898–908.PubMedCrossRef
189.
go back to reference Menezes J, Souto das Neves BH, Goncalves R, Benetti F, Mello-Carpes PB. Maternal deprivation impairs memory and cognitive flexibility, effect that is avoided by environmental enrichment. Behav Brain Res. 2020;381:112468.PubMedCrossRef Menezes J, Souto das Neves BH, Goncalves R, Benetti F, Mello-Carpes PB. Maternal deprivation impairs memory and cognitive flexibility, effect that is avoided by environmental enrichment. Behav Brain Res. 2020;381:112468.PubMedCrossRef
190.
go back to reference Joushi S, Esmaeilpour K, Masoumi-Ardakani Y, Esmaeili-Mahani S, Sheibani V. Effects of short environmental enrichment on early-life adversity induced cognitive alternations in adolescent rats. J Neurosci Res. 2021;99:3373–91.PubMedCrossRef Joushi S, Esmaeilpour K, Masoumi-Ardakani Y, Esmaeili-Mahani S, Sheibani V. Effects of short environmental enrichment on early-life adversity induced cognitive alternations in adolescent rats. J Neurosci Res. 2021;99:3373–91.PubMedCrossRef
191.
go back to reference Shahidi F, Ambigaipalan P. Omega-3 polyunsaturated fatty acids and their health benefits. Annu Rev Food Sci Technol. 2018;9:345–81.PubMedCrossRef Shahidi F, Ambigaipalan P. Omega-3 polyunsaturated fatty acids and their health benefits. Annu Rev Food Sci Technol. 2018;9:345–81.PubMedCrossRef
193.
194.
go back to reference Yam KY, Schipper L, Reemst K, Ruigrok SR, Abbink MR, Hoeijmakers L, et al. Increasing availability of omega-3 fatty acid in the early-life diet prevents the early-life stress-induced cognitive impairments without affecting metabolic alterations. FASEB J. 2019;33:5729–40.PubMedCrossRef Yam KY, Schipper L, Reemst K, Ruigrok SR, Abbink MR, Hoeijmakers L, et al. Increasing availability of omega-3 fatty acid in the early-life diet prevents the early-life stress-induced cognitive impairments without affecting metabolic alterations. FASEB J. 2019;33:5729–40.PubMedCrossRef
195.
go back to reference Pusceddu MM, Kelly P, Ariffin N, Cryan JF, Clarke G, Dinan TG. n-3 PUFAs have beneficial effects on anxiety and cognition in female rats: effects of early life stress. Psychoneuroendocrinology. 2015;58:79–90.PubMedCrossRef Pusceddu MM, Kelly P, Ariffin N, Cryan JF, Clarke G, Dinan TG. n-3 PUFAs have beneficial effects on anxiety and cognition in female rats: effects of early life stress. Psychoneuroendocrinology. 2015;58:79–90.PubMedCrossRef
196.
go back to reference Pusceddu MM, El Aidy S, Crispie F, O’Sullivan O, Cotter P, Stanton C, et al. N-3 polyunsaturated fatty acids (PUFAs) reverse the impact of early-life stress on the gut microbiota. PLoS ONE. 2015;10:e0139721.PubMedPubMedCentralCrossRef Pusceddu MM, El Aidy S, Crispie F, O’Sullivan O, Cotter P, Stanton C, et al. N-3 polyunsaturated fatty acids (PUFAs) reverse the impact of early-life stress on the gut microbiota. PLoS ONE. 2015;10:e0139721.PubMedPubMedCentralCrossRef
197.
go back to reference Liao JF, Hsu CC, Chou GT, Hsu JS, Liong MT, Tsai YC. Lactobacillus paracasei PS23 reduced early-life stress abnormalities in maternal separation mouse model. Benef Microbes. 2019;10:425–36.PubMedCrossRef Liao JF, Hsu CC, Chou GT, Hsu JS, Liong MT, Tsai YC. Lactobacillus paracasei PS23 reduced early-life stress abnormalities in maternal separation mouse model. Benef Microbes. 2019;10:425–36.PubMedCrossRef
198.
go back to reference Cowan CSM, Stylianakis AA, Richardson R. Early-life stress, microbiota, and brain development: probiotics reverse the effects of maternal separation on neural circuits underpinning fear expression and extinction in infant rats. Dev Cogn Neurosci. 2019;37:100627.PubMedPubMedCentralCrossRef Cowan CSM, Stylianakis AA, Richardson R. Early-life stress, microbiota, and brain development: probiotics reverse the effects of maternal separation on neural circuits underpinning fear expression and extinction in infant rats. Dev Cogn Neurosci. 2019;37:100627.PubMedPubMedCentralCrossRef
199.
go back to reference Yang L, Wu C, Li Y, Dong Y, Wu CY, Lee RH, et al. Long-term exercise pre-training attenuates Alzheimer’s disease-related pathology in a transgenic rat model of Alzheimer’s disease. Geroscience. 2022;44:1457–77.PubMedPubMedCentralCrossRef Yang L, Wu C, Li Y, Dong Y, Wu CY, Lee RH, et al. Long-term exercise pre-training attenuates Alzheimer’s disease-related pathology in a transgenic rat model of Alzheimer’s disease. Geroscience. 2022;44:1457–77.PubMedPubMedCentralCrossRef
200.
go back to reference Huang X, Zhao X, Li B, Cai Y, Zhang S, Wan Q, et al. Comparative efficacy of various exercise interventions on cognitive function in patients with mild cognitive impairment or dementia: a systematic review and network meta-analysis. J Sport Health Sci. 2022;11:212–23.PubMedCrossRef Huang X, Zhao X, Li B, Cai Y, Zhang S, Wan Q, et al. Comparative efficacy of various exercise interventions on cognitive function in patients with mild cognitive impairment or dementia: a systematic review and network meta-analysis. J Sport Health Sci. 2022;11:212–23.PubMedCrossRef
201.
go back to reference Sadeghi M, Peeri M, Hosseini MJ. Adolescent voluntary exercise attenuated hippocampal innate immunity responses and depressive-like behaviors following maternal separation stress in male rats. Physiol Behav. 2016;163:177–83.PubMedCrossRef Sadeghi M, Peeri M, Hosseini MJ. Adolescent voluntary exercise attenuated hippocampal innate immunity responses and depressive-like behaviors following maternal separation stress in male rats. Physiol Behav. 2016;163:177–83.PubMedCrossRef
202.
go back to reference Zolfaghari FS, Pirri F, Gauvin E, Peeri M, Amiri S. Exercise and fluoxetine treatment during adolescence protect against early life stress-induced behavioral abnormalities in adult rats. Pharmacol Biochem Behav. 2021;205:173190.PubMedCrossRef Zolfaghari FS, Pirri F, Gauvin E, Peeri M, Amiri S. Exercise and fluoxetine treatment during adolescence protect against early life stress-induced behavioral abnormalities in adult rats. Pharmacol Biochem Behav. 2021;205:173190.PubMedCrossRef
203.
go back to reference Park SS, Kim TW, Park HS, Seo TB, Kim YP. Effects of treadmill exercise on activity, short-term memory, vascular dysfunction in maternal separation rats. J Exerc Rehabil. 2020;16:118–23.PubMedPubMedCentralCrossRef Park SS, Kim TW, Park HS, Seo TB, Kim YP. Effects of treadmill exercise on activity, short-term memory, vascular dysfunction in maternal separation rats. J Exerc Rehabil. 2020;16:118–23.PubMedPubMedCentralCrossRef
Metadata
Title
Early life adversity as a risk factor for cognitive impairment and Alzheimer’s disease
Authors
Zhihai Huang
J. Dedrick Jordan
Quanguang Zhang
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Translational Neurodegeneration / Issue 1/2023
Electronic ISSN: 2047-9158
DOI
https://doi.org/10.1186/s40035-023-00355-z

Other articles of this Issue 1/2023

Translational Neurodegeneration 1/2023 Go to the issue