1
|
Tian X, Russo SJ, Li L. Behavioral Animal Models and Neural-Circuit Framework of Depressive Disorder. Neurosci Bull 2024:10.1007/s12264-024-01270-7. [PMID: 39120643 DOI: 10.1007/s12264-024-01270-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/26/2024] [Indexed: 08/10/2024] Open
Abstract
Depressive disorder is a chronic, recurring, and potentially life-endangering neuropsychiatric disease. According to a report by the World Health Organization, the global population suffering from depression is experiencing a significant annual increase. Despite its prevalence and considerable impact on people, little is known about its pathogenesis. One major reason is the scarcity of reliable animal models due to the absence of consensus on the pathology and etiology of depression. Furthermore, the neural circuit mechanism of depression induced by various factors is particularly complex. Considering the variability in depressive behavior patterns and neurobiological mechanisms among different animal models of depression, a comparison between the neural circuits of depression induced by various factors is essential for its treatment. In this review, we mainly summarize the most widely used behavioral animal models and neural circuits under different triggers of depression, aiming to provide a theoretical basis for depression prevention.
Collapse
Affiliation(s)
- Xiangyun Tian
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Long Li
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
2
|
Reguilón MD, Manzanedo C, Miñarro J, Rodríguez-Arias M. Stress inoculation during adolescence attenuates social stress-induced increase in ethanol intake in adult male mice. Neuropharmacology 2024; 246:109838. [PMID: 38199295 DOI: 10.1016/j.neuropharm.2024.109838] [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: 10/10/2023] [Revised: 12/05/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024]
Abstract
Social stress exposure heightens the risk of substance abuse disorder development, especially when endured during adolescence, influencing long-term mental health. This study investigates early-life stress's potential to confer resilience against later-life stressors. To investigate this hypothesis, we examined the impact of a single social defeat (SD) incident during adolescent mice's lives on subsequent voluntary ethanol consumption following repeated adult social stress exposure. Half of the adolescent mice experienced SD at postnatal day 28. Three weeks later (postnatal day 49), defeated groups encountered four confrontations with aggressive residents every 72 h, while control groups were exposed to non-resident exploration. A day after the last SD, defeated mice were classified as resilient or susceptible based on their response to a social interaction test (SIT), a model for depressive behavior. To assess ethanol consumption during young adulthood, researchers used the 'drinking in the dark' and oral ethanol self-administration paradigms. Stress inoculation (IS) slightly increased resilient animals in the SIT. In mice without IS exposure during adolescence, susceptible defeated mice displayed higher ethanol consumption and motivation than control and resilient mice. IS in adolescence effectively counteracted this effect, as IS-SD groups, whether resilient or susceptible, showed no increase in ethanol intake. These groups also exhibited similar motivation to control, measured by the progressive ratio. Notably, elevated IL-6 levels seen in SD-S mice were absent in IS-exposed mice. Additionally, IS-exposed groups had lower prefrontal cortex IL-6 and CX3CL1 levels. These findings support the hypothesis that IS, induced by moderate-intensity stress during adolescence, can enhance resilience to more severe stressors in adulthood.
Collapse
Affiliation(s)
- Marina D Reguilón
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain
| | - Carmen Manzanedo
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain
| | - José Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain
| | - Marta Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de València, Valencia, Spain.
| |
Collapse
|
3
|
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
|
4
|
Cifre M, Palou A, Oliver P. The Metabolically Obese, Normal-Weight Phenotype in Young Rats Is Associated with Cognitive Impairment and Partially Preventable with Leptin Intake during Lactation. Int J Mol Sci 2023; 25:228. [PMID: 38203399 PMCID: PMC10778589 DOI: 10.3390/ijms25010228] [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: 11/14/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024] Open
Abstract
The intake of high-fat diets (HFDs) and obesity are linked to cognitive impairment. Here, we aimed to investigate whether an early metabolically obese, normal-weight (MONW) phenotype, induced with an HFD in young rats, also leads to cognitive dysfunction and to evaluate the potential cognitive benefits of neonatal intake of leptin. To achieve this, Wistar rats orally received physiological doses of leptin or its vehicle during lactation, followed by 11 weeks of pair-feeding with an HFD or control diet post-weaning. Working memory was assessed using a T-maze, and gene expression in the hippocampus and peripheral blood mononuclear cells (PBMCs) was assessed with real-time RT-qPCR to identify cognition biomarkers. Young MONW-like rats showed hippocampal gene expression changes and decreased working memory. Animals receiving leptin during lactation presented similar gene expression changes but preserved working memory despite HFD intake, partly due to improved insulin sensitivity. Notably, PBMC Syn1 expression appears as an accessible biomarker of cognitive health, reflecting both the detrimental effect of HFD intake at early ages despite the absence of obesity and the positive effects of neonatal leptin treatment on cognition. Thus, the MONW phenotype developed at a young age is linked to cognitive dysfunction, which is reflected at the transcriptomic level in PBMCs. Neonatal leptin intake can partly counteract this impaired cognition resulting from early HFD consumption.
Collapse
Affiliation(s)
- Margalida Cifre
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands (UIB), 07122 Palma, Spain (A.P.)
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Andreu Palou
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands (UIB), 07122 Palma, Spain (A.P.)
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
| | - Paula Oliver
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands (UIB), 07122 Palma, Spain (A.P.)
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
| |
Collapse
|
5
|
Hong JY, Medzhitov R. On developmental programming of the immune system. Trends Immunol 2023; 44:877-889. [PMID: 37852863 DOI: 10.1016/j.it.2023.09.004] [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: 09/05/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 10/20/2023]
Abstract
Early-life environmental exposures play a significant role in shaping long-lasting immune phenotypes and disease susceptibility. Nevertheless, comprehensive understanding of the developmental programming of immunity is limited. We propose that the vertebrate immune system contains durable programmable components established through early environmental interactions and maintained in a stable and homeostatic manner. Some immune components, such as immunological memory, are intrinsically programmable. Others are influenced by conditions during critical developmental windows in early life, including microbiota, hormones, metabolites, and environmental stress, which impact programming. Developmental immune programming can promote adaptation to an anticipated future environment. However, mismatches between predicted and actual environments can result in disease. This is relevant because understanding programming mechanisms can offer insights into the origin of inflammatory diseases, ideally enabling effective prevention and treatment strategies.
Collapse
Affiliation(s)
- Jun Young Hong
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
6
|
Bekdash RA. Methyl Donors, Epigenetic Alterations, and Brain Health: Understanding the Connection. Int J Mol Sci 2023; 24:ijms24032346. [PMID: 36768667 PMCID: PMC9917111 DOI: 10.3390/ijms24032346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Methyl donors such as choline, betaine, folic acid, methionine, and vitamins B6 and B12 are critical players in the one-carbon metabolism and have neuroprotective functions. The one-carbon metabolism comprises a series of interconnected chemical pathways that are important for normal cellular functions. Among these pathways are those of the methionine and folate cycles, which contribute to the formation of S-adenosylmethionine (SAM). SAM is the universal methyl donor of methylation reactions such as histone and DNA methylation, two epigenetic mechanisms that regulate gene expression and play roles in human health and disease. Epigenetic mechanisms have been considered a bridge between the effects of environmental factors, such as nutrition, and phenotype. Studies in human and animal models have indicated the importance of the optimal levels of methyl donors on brain health and behavior across the lifespan. Imbalances in the levels of these micronutrients during critical periods of brain development have been linked to epigenetic alterations in the expression of genes that regulate normal brain function. We present studies that support the link between imbalances in the levels of methyl donors, epigenetic alterations, and stress-related disorders. Appropriate levels of these micronutrients should then be monitored at all stages of development for a healthier brain.
Collapse
Affiliation(s)
- Rola A Bekdash
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| |
Collapse
|
7
|
Natale F, Spinelli M, Rinaudo M, Cocco S, Nifo Sarrapochiello I, Fusco S, Grassi C. Maternal High Fat Diet Anticipates the AD-like Phenotype in 3xTg-AD Mice by Epigenetic Dysregulation of Aβ Metabolism. Cells 2023; 12:cells12020220. [PMID: 36672155 PMCID: PMC9856666 DOI: 10.3390/cells12020220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/22/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Maternal overnutrition has been reported to affect brain plasticity of the offspring by altering gene expression, regulating both synaptic plasticity and adult neurogenesis. However, whether perinatal metabolic stress may influence the accumulation of misfolded proteins and the development of neurodegeneration remains to be clarified. We investigated the impact of maternal high fat diet (HFD) in an experimental model of Alzheimer's disease (AD). The 3xTg-AD mice born to overfed mothers showed an impairment of synaptic plasticity and cognitive deficits earlier than controls. Maternal HFD also altered the expression of genes regulating amyloid-β-protein (Aβ) metabolism (i.e., Bace1, Ern1, Ide and Nicastrin) and enhanced Aβ deposition in the hippocampus. Finally, we found an epigenetic derangement and an aberrant recruitment of transcription factors NF-kB and STAT3 and chromatin remodeler HDAC2 on the regulatory sequences of the same genes. Collectively, our data indicate that early life metabolic stress worsens the AD phenotype via epigenetic alteration of genes regulating Aβ synthesis and clearance.
Collapse
Affiliation(s)
- Francesca Natale
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Matteo Spinelli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Marco Rinaudo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Sara Cocco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Correspondence:
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
8
|
Melgar-Locatelli S, de Ceglia M, Mañas-Padilla MC, Rodriguez-Pérez C, Castilla-Ortega E, Castro-Zavala A, Rivera P. Nutrition and adult neurogenesis in the hippocampus: Does what you eat help you remember? Front Neurosci 2023; 17:1147269. [PMID: 36908779 PMCID: PMC9995971 DOI: 10.3389/fnins.2023.1147269] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Neurogenesis is a complex process by which neural progenitor cells (NPCs)/neural stem cells (NSCs) proliferate and differentiate into new neurons and other brain cells. In adulthood, the hippocampus is one of the areas with more neurogenesis activity, which is involved in the modulation of both emotional and cognitive hippocampal functions. This complex process is affected by many intrinsic and extrinsic factors, including nutrition. In this regard, preclinical studies performed in rats and mice demonstrate that high fats and/or sugars diets have a negative effect on adult hippocampal neurogenesis (AHN). In contrast, diets enriched with bioactive compounds, such as polyunsaturated fatty acids and polyphenols, as well as intermittent fasting or caloric restriction, can induce AHN. Interestingly, there is also growing evidence demonstrating that offspring AHN can be affected by maternal nutrition in the perinatal period. Therefore, nutritional interventions from early stages and throughout life are a promising perspective to alleviate neurodegenerative diseases by stimulating neurogenesis. The underlying mechanisms by which nutrients and dietary factors affect AHN are still being studied. Interestingly, recent evidence suggests that additional peripheral mediators may be involved. In this sense, the microbiota-gut-brain axis mediates bidirectional communication between the gut and the brain and could act as a link between nutritional factors and AHN. The aim of this mini-review is to summarize, the most recent findings related to the influence of nutrition and diet in the modulation of AHN. The importance of maternal nutrition in the AHN of the offspring and the role of the microbiota-gut-brain axis in the nutrition-neurogenesis relationship have also been included.
Collapse
Affiliation(s)
- Sonia Melgar-Locatelli
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain.,Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| | - Marialuisa de Ceglia
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain.,UGC Salud Mental, Hospital Universitario Regional de Málaga, Málaga, Spain
| | - M Carmen Mañas-Padilla
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain.,Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| | - Celia Rodriguez-Pérez
- Departamento de Nutrición y Bromatología, Facultad de Ciencias de la Salud, Universidad de Granada, Granada, Spain.,Instituto de Nutrición y Tecnología de los Alimentos 'José Mataix', Universidad de Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Estela Castilla-Ortega
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain.,Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| | - Adriana Castro-Zavala
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain.,Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| | - Patricia Rivera
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain.,UGC Salud Mental, Hospital Universitario Regional de Málaga, Málaga, Spain
| |
Collapse
|
9
|
Juncker HG, Naninck EFG, Schipper L, Lucassen PJ, van Goudoever JB, de Rooij SR, Korosi A. Maternal stress in the postpartum period is associated with altered human milk fatty acid composition. Clin Nutr 2022; 41:2517-2528. [PMID: 36223713 DOI: 10.1016/j.clnu.2022.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS Maternal stress in the postpartum period affects not only the mother, but also her newborn child who is at increased risk for a wide range of disorders later in life. The mechanisms underlying transmission of maternal stress to the child remain elusive. Human milk (HM) is a potential candidate and is an important source of fatty acid (FA), which are crucial for child (neuro)development. This study aims to investigate whether maternal psychological and biological stress influences HM FA composition over the first month postpartum. METHODS The Amsterdam Mother's Milk study is a prospective cohort study. We included lactating women who delivered at term with a large range of stress levels: a high stress (HS) group, women whose child was hospitalized for a minimum of 2 days (n=23) and a control (CTL) group, women who gave birth to a healthy child (n=73). HM was collected three times a day at postpartum days 10, 17 and 24. Perceived psychological stress was measured using multiple validated questionnaires, while biological stress measures were based on cortisol in hair, saliva and HM. HM FAs were analyzed by gas-chromatography and compared between groups. RESULTS Maternal perceived stress scores were significantly higher in the HS group (p < 0.01), whereas cortisol measurements did not differ between groups. The absolute concentrations of total FA in HM (p=0.023), including the total amount of poly unsaturated fatty acids (PUFAs) (p=0.022) and omega-6 PUFAs (p=0.018), were lower in the HS group compared to the CTL group. Relative values of FAs did not differ between groups. CONCLUSION Maternal stress in the first month postpartum was associated with overall lower levels of FA in HM. This possibly indicates a route of transmission of maternal stress signals to the infant. Future research should investigate if these stress-induced changes in HM FAs have consequences for child development.
Collapse
Affiliation(s)
- H G Juncker
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Amsterdam Reproduction and Development Institute, Amsterdam, the Netherlands
| | - E F G Naninck
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Amsterdam Reproduction and Development Institute, Amsterdam, the Netherlands
| | - L Schipper
- Danone Nutricia Research, Utrecht, the Netherlands
| | - P J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - J B van Goudoever
- Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Amsterdam Reproduction and Development Institute, Amsterdam, the Netherlands
| | - S R de Rooij
- Amsterdam UMC, University of Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - A Korosi
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
10
|
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: 1.5] [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
|
11
|
High Fat Diet Multigenerationally Affects Hippocampal Neural Stem Cell Proliferation via Epigenetic Mechanisms. Cells 2022; 11:cells11172661. [PMID: 36078069 PMCID: PMC9454549 DOI: 10.3390/cells11172661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Early-life metabolic stress has been demonstrated to affect brain development, persistently influence brain plasticity and to exert multigenerational effects on cognitive functions. However, the impact of an ancestor’s diet on the adult neurogenesis of their descendants has not yet been investigated. Here, we studied the effects of maternal high fat diet (HFD) on hippocampal adult neurogenesis and the proliferation of neural stem and progenitor cells (NSPCs) derived from the hippocampus of both the second and the third generations of progeny (F2HFD and F3HFD). Maternal HFD caused a multigenerational depletion of neurogenic niche in F2HFD and F3HFD mice. Moreover, NSPCs derived from HFD descendants showed altered expression of genes regulating stem cell proliferation and neurodifferentiation (i.e., Hes1, NeuroD1, Bdnf). Finally, ancestor HFD-related hyper-activation of both STAT3 and STAT5 induced enhancement of their binding on the regulatory sequences of Gfap gene and an epigenetic switch from permissive to repressive chromatin on the promoter of the NeuroD1 gene. Collectively, our data indicate that maternal HFD multigenerationally affects hippocampal adult neurogenesis via an epigenetic derangement of pro-neurogenic gene expression in NSPCs.
Collapse
|
12
|
Early life adversity shapes neural circuit function during sensitive postnatal developmental periods. Transl Psychiatry 2022; 12:306. [PMID: 35915071 PMCID: PMC9343623 DOI: 10.1038/s41398-022-02092-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
Early life adversity (ELA) is a major risk factor for mental illness, but the neurobiological mechanisms by which ELA increases the risk for future psychopathology are still poorly understood. Brain development is particularly malleable during prenatal and early postnatal life, when complex neural circuits are being formed and refined through an interplay of excitatory and inhibitory neural input, synaptogenesis, synaptic pruning, myelination, and neurogenesis. Adversity that influences these processes during sensitive periods of development can thus have long-lasting and pervasive effects on neural circuit maturation. In this review, we will discuss clinical and preclinical evidence for the impact of ELA on neural circuit formation with a focus on the early postnatal period, and how long-lasting impairments in these circuits can affect future behavior. We provide converging evidence from human and animal studies on how ELA alters the functional development of brain regions, neural circuits, and neurotransmitter systems that are crucial for cognition and affective behavior, including the hippocampus, the hypothalamus-pituitary-adrenal (HPA) axis, neural networks of fear responses and cognition, and the serotonin (5-HT) system. We also discuss how gene-by-environment (GxE) interactions can determine individual differences in susceptibility and resilience to ELA, as well as molecular pathways by which ELA regulates neural circuit development, for which we emphasize epigenetic mechanisms. Understanding the molecular and neurobiological mechanisms underlying ELA effects on brain function and psychopathology during early postnatal sensitive periods may have great potential to advance strategies to better treat or prevent psychiatric disorders that have their origin early in life.
Collapse
|
13
|
Willekens J, Runnels LW. Impact of Zinc Transport Mechanisms on Embryonic and Brain Development. Nutrients 2022; 14:2526. [PMID: 35745255 PMCID: PMC9231024 DOI: 10.3390/nu14122526] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 12/04/2022] Open
Abstract
The trace element zinc (Zn) binds to over ten percent of proteins in eukaryotic cells. Zn flexible chemistry allows it to regulate the activity of hundreds of enzymes and influence scores of metabolic processes in cells throughout the body. Deficiency of Zn in humans has a profound effect on development and in adults later in life, particularly in the brain, where Zn deficiency is linked to several neurological disorders. In this review, we will summarize the importance of Zn during development through a description of the outcomes of both genetic and early dietary Zn deficiency, focusing on the pathological consequences on the whole body and brain. The epidemiology and the symptomology of Zn deficiency in humans will be described, including the most studied inherited Zn deficiency disease, Acrodermatitis enteropathica. In addition, we will give an overview of the different forms and animal models of Zn deficiency, as well as the 24 Zn transporters, distributed into two families: the ZIPs and the ZnTs, which control the balance of Zn throughout the body. Lastly, we will describe the TRPM7 ion channel, which was recently shown to contribute to intestinal Zn absorption and has its own significant impact on early embryonic development.
Collapse
Affiliation(s)
| | - Loren W. Runnels
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| |
Collapse
|
14
|
Becker M, Abaev K, Pinhasov A, Ornoy A. S-Adenosyl-Methionine alleviates sociability aversion and reduces changes in gene expression in a mouse model of social hierarchy. Behav Brain Res 2022; 427:113866. [DOI: 10.1016/j.bbr.2022.113866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 02/06/2023]
|
15
|
Triplett RL, Lean RE, Parikh A, Miller JP, Alexopoulos D, Kaplan S, Meyer D, Adamson C, Smyser TA, Rogers CE, Barch DM, Warner B, Luby JL, Smyser CD. Association of Prenatal Exposure to Early-Life Adversity With Neonatal Brain Volumes at Birth. JAMA Netw Open 2022; 5:e227045. [PMID: 35412624 PMCID: PMC9006107 DOI: 10.1001/jamanetworkopen.2022.7045] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/24/2022] [Indexed: 12/21/2022] Open
Abstract
Importance Exposure to early-life adversity alters the structural development of key brain regions underlying neurodevelopmental impairments. The association between prenatal exposure to adversity and brain structure at birth remains poorly understood. Objective To examine whether prenatal exposure to maternal social disadvantage and psychosocial stress is associated with neonatal global and regional brain volumes and cortical folding. Design, Setting, and Participants This prospective, longitudinal cohort study included 399 mother-infant dyads of sociodemographically diverse mothers recruited in the first or early second trimester of pregnancy and their infants, who underwent brain magnetic resonance imaging in the first weeks of life. Mothers were recruited from local obstetric clinics in St Louis, Missouri from September 1, 2017, to February 28, 2020. Exposures Maternal social disadvantage and psychosocial stress in pregnancy. Main Outcomes and Measures Confirmatory factor analyses were used to create latent constructs of maternal social disadvantage (income-to-needs ratio, Area Deprivation Index, Healthy Eating Index, educational level, and insurance status) and psychosocial stress (Perceived Stress Scale, Edinburgh Postnatal Depression Scale, Everyday Discrimination Scale, and Stress and Adversity Inventory). Neonatal cortical and subcortical gray matter, white matter, cerebellum, hippocampus, and amygdala volumes were generated using semiautomated, age-specific, segmentation pipelines. Results A total of 280 mothers (mean [SD] age, 29.1 [5.3] years; 170 [60.7%] Black or African American, 100 [35.7%] White, and 10 [3.6%] other race or ethnicity) and their healthy, term-born infants (149 [53.2%] male; mean [SD] infant gestational age, 38.6 [1.0] weeks) were included in the analysis. After covariate adjustment and multiple comparisons correction, greater social disadvantage was associated with reduced cortical gray matter (unstandardized β = -2.0; 95% CI, -3.5 to -0.5; P = .01), subcortical gray matter (unstandardized β = -0.4; 95% CI, -0.7 to -0.2; P = .003), and white matter (unstandardized β = -5.5; 95% CI, -7.8 to -3.3; P < .001) volumes and cortical folding (unstandardized β = -0.03; 95% CI, -0.04 to -0.01; P < .001). Psychosocial stress showed no association with brain metrics. Although social disadvantage accounted for an additional 2.3% of the variance of the left hippocampus (unstandardized β = -0.03; 95% CI, -0.05 to -0.01), 2.3% of the right hippocampus (unstandardized β = -0.03; 95% CI, -0.05 to -0.01), 3.1% of the left amygdala (unstandardized β = -0.02; 95% CI, -0.03 to -0.01), and 2.9% of the right amygdala (unstandardized β = -0.02; 95% CI, -0.03 to -0.01), no regional effects were found after accounting for total brain volume. Conclusions and Relevance In this baseline assessment of an ongoing cohort study, prenatal social disadvantage was associated with global reductions in brain volumes and cortical folding at birth. No regional specificity for the hippocampus or amygdala was detected. Results highlight that associations between poverty and brain development begin in utero and are evident early in life. These findings emphasize that preventive interventions that support fetal brain development should address parental socioeconomic hardships.
Collapse
Affiliation(s)
- Regina L. Triplett
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| | - Rachel E. Lean
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Amisha Parikh
- School of Medicine, Washington University in St Louis, St Louis, Missouri
| | - J. Philip Miller
- Department of Biostatistics, Washington University in St Louis, St Louis, Missouri
| | | | - Sydney Kaplan
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| | - Dominique Meyer
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| | - Christopher Adamson
- Developmental Imaging, Murdoch Children’s Institute, Melbourne, Australia
- Electrical and Electronic Engineering, University of Melbourne, Melbourne, Australia
| | - Tara A. Smyser
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Cynthia E. Rogers
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
- Department of Pediatrics, Washington University in St Louis, St Louis, Missouri
| | - Deanna M. Barch
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
- Department of Psychological and Brain Sciences, Washington University in St Louis, St Louis, Missouri
- Department of Radiology, Washington University in St Louis, St Louis, Missouri
| | - Barbara Warner
- Department of Pediatrics, Washington University in St Louis, St Louis, Missouri
| | - Joan L. Luby
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Christopher D. Smyser
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
- Department of Pediatrics, Washington University in St Louis, St Louis, Missouri
- Department of Radiology, Washington University in St Louis, St Louis, Missouri
| |
Collapse
|
16
|
Ornoy A, Weinstein-Fudim L, Becker M. SAMe, Choline, and Valproic Acid as Possible Epigenetic Drugs: Their Effects in Pregnancy with a Special Emphasis on Animal Studies. Pharmaceuticals (Basel) 2022; 15:192. [PMID: 35215304 PMCID: PMC8879727 DOI: 10.3390/ph15020192] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 11/29/2022] Open
Abstract
In this review, we discuss the functions and main effects on pregnancy outcomes of three agents that have the ability to induce epigenetic modifications: valproic acid (VPA), a well-known teratogen that is a histone deacetylase inhibitor; S-adenosylmethionine (SAMe), the most effective methyl donor; and choline, an important micronutrient involved in the one methyl group cycle and in the synthesis of SAMe. Our aim was to describe the possible effects of these compounds when administered during pregnancy on the developing embryo and fetus or, if administered postnatally, their effects on the developing child. These substances are able to modify gene expression and possibly alleviate neurobehavioral changes in disturbances that have epigenetic origins, such as autism spectrum disorder (ASD), depression, Rett syndrome, and fetal alcohol spectrum disorder (FASD). Valproic acid and SAMe are antagonistic epigenetic modulators whether administered in utero or postnatally. However, VPA is a major human teratogen and, whenever possible, should not be used by pregnant women. Most currently relevant data come from experimental animal studies that aimed to explore the possibility of using these substances as epigenetic modifiers and possible therapeutic agents. In experimental animals, each of these substances was able to alleviate the severity of several well-known diseases by inducing changes in the expression of affected genes or by other yet unknown mechanisms. We believe that additional studies are needed to further explore the possibility of using these substances, and similar compounds, for the treatment of "epigenetic human diseases".
Collapse
Affiliation(s)
- Asher Ornoy
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel;
| | - Liza Weinstein-Fudim
- Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel;
| | - Maria Becker
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| |
Collapse
|
17
|
Bastianini S, Lo Martire V, Alvente S, Berteotti C, Matteoli G, Rullo L, Stamatakos S, Silvani A, Candeletti S, Romualdi P, Cohen G, Zoccoli G. Early-life nicotine or cotinine exposure produces long-lasting sleep alterations and downregulation of hippocampal corticosteroid receptors in adult mice. Sci Rep 2021; 11:23897. [PMID: 34903845 PMCID: PMC8668915 DOI: 10.1038/s41598-021-03468-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/30/2021] [Indexed: 11/24/2022] Open
Abstract
Early-life exposure to environmental toxins like tobacco can permanently re-program body structure and function. Here, we investigated the long-term effects on mouse adult sleep phenotype exerted by early-life exposure to nicotine or to its principal metabolite, cotinine. Moreover, we investigated whether these effects occurred together with a reprogramming of the activity of the hippocampus, a key structure to coordinate the hormonal stress response. Adult male mice born from dams subjected to nicotine (NIC), cotinine (COT) or vehicle (CTRL) treatment in drinking water were implanted with electrodes for sleep recordings. NIC and COT mice spent significantly more time awake than CTRL mice at the transition between the rest (light) and the activity (dark) period. NIC and COT mice showed hippocampal glucocorticoid receptor (GR) downregulation compared to CTRL mice, and NIC mice also showed hippocampal mineralocorticoid receptor downregulation. Hippocampal GR expression significantly and inversely correlated with the amount of wakefulness at the light-to-dark transition, while no changes in DNA methylation were found. We demonstrated that early-life exposure to nicotine (and cotinine) concomitantly entails long-lasting reprogramming of hippocampal activity and sleep phenotype suggesting that the adult sleep phenotype may be modulated by events that occurred during that critical period of life.
Collapse
Affiliation(s)
- Stefano Bastianini
- grid.6292.f0000 0004 1757 1758PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Piazza di Porta San Donato 2, 40126 Bologna, Italy
| | - Viviana Lo Martire
- grid.6292.f0000 0004 1757 1758PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Piazza di Porta San Donato 2, 40126 Bologna, Italy
| | - Sara Alvente
- grid.6292.f0000 0004 1757 1758PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Piazza di Porta San Donato 2, 40126 Bologna, Italy
| | - Chiara Berteotti
- grid.6292.f0000 0004 1757 1758PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Piazza di Porta San Donato 2, 40126 Bologna, Italy
| | - Gabriele Matteoli
- grid.6292.f0000 0004 1757 1758PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Piazza di Porta San Donato 2, 40126 Bologna, Italy
| | - Laura Rullo
- grid.6292.f0000 0004 1757 1758Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Serena Stamatakos
- grid.6292.f0000 0004 1757 1758Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Alessandro Silvani
- grid.6292.f0000 0004 1757 1758PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Piazza di Porta San Donato 2, 40126 Bologna, Italy
| | - Sanzio Candeletti
- grid.6292.f0000 0004 1757 1758Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Patrizia Romualdi
- grid.6292.f0000 0004 1757 1758Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Gary Cohen
- grid.4714.60000 0004 1937 0626Department of Women and Child Health, Karolinska Institutet, Stockholm, Sweden ,grid.412703.30000 0004 0587 9093Centre for Sleep Health and Research, Sleep Investigation Laboratory, Royal North Shore Hospital, Sydney, Australia
| | - Giovanna Zoccoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Piazza di Porta San Donato 2, 40126, Bologna, Italy.
| |
Collapse
|
18
|
Ruiz-Raya F. Ecophysiology of egg rejection in hosts of avian brood parasites: new insights and perspectives. Curr Zool 2021; 67:631-638. [PMID: 34805540 PMCID: PMC8599070 DOI: 10.1093/cz/zoab042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/09/2021] [Indexed: 12/11/2022] Open
Abstract
Egg rejection is the most effective and widespread defense used by host species to counteract the extreme fitness costs frequently imposed by obligate avian brood parasites. Yet, the proximate mechanisms underlying between- and within-individual variation in host responses remain poorly explored. Emerging evidence suggests that egg rejection is dependent on individual physiological states, and draws attention to the role of hormones as mediators of flexible antiparasitic responses. In this perspective article, I outline recent advances in our understanding of the proximate factors that mediate egg rejection. I also point out some areas where knowledge remains still lacking, especially those related to the development and maintenance of effective cognitive functions, the potential role of oxidative stress, immunological state, and developmental stressors. I propose new hypotheses that stimulate future research on behavioral host responses toward brood parasitism.
Collapse
Affiliation(s)
- Francisco Ruiz-Raya
- Centro de Investigación Mariña, Universidade de Vigo, GEA, Vigo 36310, Spain
| |
Collapse
|
19
|
Wendel KM, Short AK, Noarbe BP, Haddad E, Palma AM, Yassa MA, Baram TZ, Obenaus A. Early life adversity in male mice sculpts reward circuits. Neurobiol Stress 2021; 15:100409. [PMID: 34746338 PMCID: PMC8554344 DOI: 10.1016/j.ynstr.2021.100409] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/24/2021] [Accepted: 10/12/2021] [Indexed: 01/08/2023] Open
Abstract
Early life adversity (ELA) comprises a wide variety of negative experiences during early life and has been linked to cognitive impairments, reduced experiences of pleasure (anhedonia), and other long-term consequences implying that ELA impacts the reward circuitry. In this study, we focused on the projections from the dorsal raphe (DR) to the ventral tegmental area (VTA) and on to the nucleus accumbens (NAcc), an important pathway within the reward circuit. We hypothesized that ELA alters connectivity within the DR-VTA-NAcc pathway, associated with deficient reward seeking behaviors in adulthood. We used the limited bedding and nesting model to induce ELA in mice and measured reward-related behaviors in adulthood using the three-chamber social interaction and sucrose preference tests. High resolution ex vivo diffusion tensor imaging (DTI) was acquired and processed for regional DTI metrics, including tractography to assess circuit organization. We found brain-wide changes in radial diffusivity (RD) and altered connectivity of the reward circuit in the ELA group. DR-VTA-NAcc circuit tractography and axial diffusivity (AD) along this tract exhibited dispersed organization where AD was increased in the VTA segment. Behaviorally, ELA elicited a social anhedonia-like phenotype in adulthood with decreased direct social approach and time spent with peers in the three-chamber task, and no overt differences in sucrose preference. Our findings suggest that reward circuits, assessed using DTI, are altered following ELA and that these changes may reflect enduring reward deficits.
Collapse
Affiliation(s)
- Kara M. Wendel
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Annabel K. Short
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Brenda P. Noarbe
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Elizabeth Haddad
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Anton M. Palma
- Institute for Clinical and Translational Science, University of California, Irvine, CA, USA
| | - Michael A. Yassa
- Department of Neurobiology and Behavior, University of California, Irvine School of Biological Sciences, Irvine, CA, USA
| | - Tallie Z. Baram
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine, Irvine, CA, USA
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Andre Obenaus
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine, Irvine, CA, USA
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, USA
| |
Collapse
|
20
|
Ruigrok SR, Yim K, Emmerzaal TL, Geenen B, Stöberl N, den Blaauwen JL, Abbink MR, Kiliaan AJ, van Schothorst EM, Kozicz T, Korosi A. Effects of early-life stress on peripheral and central mitochondria in male mice across ages. Psychoneuroendocrinology 2021; 132:105346. [PMID: 34274734 DOI: 10.1016/j.psyneuen.2021.105346] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/11/2021] [Accepted: 06/25/2021] [Indexed: 01/06/2023]
Abstract
Exposure to early-life stress (ES) increases the vulnerability to develop metabolic diseases as well as cognitive dysfunction, but the specific biological underpinning of the ES-induced programming is unknown. Metabolic and cognitive disorders are often comorbid, suggesting possible converging underlying pathways. Mitochondrial dysfunction is implicated in both metabolic diseases and cognitive dysfunction and chronic stress impairs mitochondrial functioning. However, if and how mitochondria are impacted by ES and whether they are implicated in the ES-induced programming remains to be determined. ES was applied by providing mice with limited nesting and bedding material from postnatal day (P)2-P9, and metabolic parameters, cognitive functions and multiple aspects of mitochondria biology (i.e. mitochondrial electron transport chain (ETC) complex activity, mitochondrial DNA copy number, expression of genes relevant for mitochondrial function, and the antioxidant capacity) were studied in muscle, hypothalamus and hippocampus at P9 and late adulthood (10-12 months of age). We show that ES altered bodyweight (gain), adiposity and glucose levels at P9, but not in late adulthood. At this age, however, ES exposure led to cognitive impairments. ES affected peripheral and central mitochondria in an age-dependent manner. At P9, both muscle and hypothalamic ETC activity were affected by ES, while in hippocampus, ES altered the expression of genes involved in fission and antioxidant defence. In adulthood, alterations in ETC complex activity were observed in the hypothalamus specifically, whereas in muscle and hippocampus ES affected the expression of genes involved in mitophagy and fission, respectively. Our study demonstrates that ES affects peripheral and central mitochondria biology throughout life, thereby uncovering a converging mechanism that might contribute to the ES-induced vulnerability for both metabolic diseases and cognitive dysfunction, which could serve as a novel target for intervention.
Collapse
Affiliation(s)
- S R Ruigrok
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - K Yim
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - T L Emmerzaal
- Department of Medical Imaging - Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands; Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - B Geenen
- Department of Medical Imaging - Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - N Stöberl
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - J L den Blaauwen
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - M R Abbink
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - A J Kiliaan
- Department of Medical Imaging - Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - E M van Schothorst
- Human and Animal Physiology, Wageningen University, 6700AH Wageningen, The Netherlands
| | - T Kozicz
- Department of Medical Imaging - Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands; Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - A Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Parker KN, Donovan MH, Smith K, Noble-Haeusslein LJ. Traumatic Injury to the Developing Brain: Emerging Relationship to Early Life Stress. Front Neurol 2021; 12:708800. [PMID: 34484104 PMCID: PMC8416304 DOI: 10.3389/fneur.2021.708800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/22/2021] [Indexed: 12/01/2022] Open
Abstract
Despite the high incidence of brain injuries in children, we have yet to fully understand the unique vulnerability of a young brain to an injury and key determinants of long-term recovery. Here we consider how early life stress may influence recovery after an early age brain injury. Studies of early life stress alone reveal persistent structural and functional impairments at adulthood. We consider the interacting pathologies imposed by early life stress and subsequent brain injuries during early brain development as well as at adulthood. This review outlines how early life stress primes the immune cells of the brain and periphery to elicit a heightened response to injury. While the focus of this review is on early age traumatic brain injuries, there is also a consideration of preclinical models of neonatal hypoxia and stroke, as each further speaks to the vulnerability of the brain and reinforces those characteristics that are common across each of these injuries. Lastly, we identify a common mechanistic trend; namely, early life stress worsens outcomes independent of its temporal proximity to a brain injury.
Collapse
Affiliation(s)
- Kaila N. Parker
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Michael H. Donovan
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Kylee Smith
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Linda J. Noble-Haeusslein
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
22
|
Leschik J, Lutz B, Gentile A. Stress-Related Dysfunction of Adult Hippocampal Neurogenesis-An Attempt for Understanding Resilience? Int J Mol Sci 2021; 22:7339. [PMID: 34298958 PMCID: PMC8305135 DOI: 10.3390/ijms22147339] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
Newborn neurons in the adult hippocampus are regulated by many intrinsic and extrinsic cues. It is well accepted that elevated glucocorticoid levels lead to downregulation of adult neurogenesis, which this review discusses as one reason why psychiatric diseases, such as major depression, develop after long-term stress exposure. In reverse, adult neurogenesis has been suggested to protect against stress-induced major depression, and hence, could serve as a resilience mechanism. In this review, we will summarize current knowledge about the functional relation of adult neurogenesis and stress in health and disease. A special focus will lie on the mechanisms underlying the cascades of events from prolonged high glucocorticoid concentrations to reduced numbers of newborn neurons. In addition to neurotransmitter and neurotrophic factor dysregulation, these mechanisms include immunomodulatory pathways, as well as microbiota changes influencing the gut-brain axis. Finally, we discuss recent findings delineating the role of adult neurogenesis in stress resilience.
Collapse
Affiliation(s)
- Julia Leschik
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy;
| |
Collapse
|
23
|
Swaab DF, Kreier F, Lucassen PJ, Salehi A, Buijs RM. Introduction: The middle and posterior hypothalamus. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:1-4. [PMID: 34225923 DOI: 10.1016/b978-0-12-820107-7.00001-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Dick F Swaab
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
| | - Felix Kreier
- Department Pediatrics, OLVG Hospitals, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Ahmad Salehi
- Department of Psychiatry and Behavioral Sciences, Stanford Medical School, Palo Alto, CA, United States
| | - Ruud M Buijs
- Hypothalamic Integration Mechanisms Laboratory, Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| |
Collapse
|
24
|
Hunsche C, Hernandez O, Mela V, Viveros MP, De la Fuente M. The Postnatal Leptin Surge Supports Immune Cell Function in Rats. Immunol Invest 2021; 51:1347-1363. [PMID: 34121590 DOI: 10.1080/08820139.2021.1940199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Background: Leptin plays an important role in the regulation of the immune response. There is a physiological surge of leptin in rodents during the neonatal period, which has mainly been studied in the context of brain development. However, little is known about the effects of this neonatal leptin surge on immunity. Therefore, we investigated whether blocking this leptin surge could affect several immune functions.Methods: Male and female rats were injected subcutaneously with 5 mg/Kg/day of rat pegylated super leptin antagonist during the neonatal period (PND5-9). On the peripubertal period, relevant functions as well as cytokine release by spleen leukocytes were studied in these animals.Results: The results showed that the animals significantly display an impaired anti-tumor NK activity and chemotactic and proliferation capacity of lymphocytes in response to mitogens. In addition, several cytokine concentrations, released under mitogen-stimulated conditions, were also altered.Conclusion: In conclusion, the neonatal leptin surge seems to be involved in the establishment of an adequate immune response and cytokine profile, which are crucial for the maintenance of a healthy life.
Collapse
Affiliation(s)
- Caroline Hunsche
- Department of Genetics, Physiology and Microbiology (Unit of Animal Physiology). Faculty of Biology, Complutense University of Madrid, Madrid, Spain.,Research Institute of Hospital 12 de Octubre (I+12), Madrid, Spain
| | - Oskarina Hernandez
- Department of Genetics, Physiology and Microbiology (Unit of Animal Physiology). Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Virginia Mela
- Department of Genetics, Physiology and Microbiology (Unit of Animal Physiology). Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - M Paz Viveros
- Department of Genetics, Physiology and Microbiology (Unit of Animal Physiology). Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Mónica De la Fuente
- Department of Genetics, Physiology and Microbiology (Unit of Animal Physiology). Faculty of Biology, Complutense University of Madrid, Madrid, Spain.,Research Institute of Hospital 12 de Octubre (I+12), Madrid, Spain
| |
Collapse
|
25
|
Siomek-Gorecka A, Dlugosz A, Czarnecki D. The Molecular Basis of Alcohol Use Disorder (AUD). Genetics, Epigenetics, and Nutrition in AUD: An Amazing Triangle. Int J Mol Sci 2021; 22:ijms22084262. [PMID: 33924016 PMCID: PMC8072802 DOI: 10.3390/ijms22084262] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol use disorder (AUD) is a very common and complex disease, as alcohol is the most widely used addictive drug in the world. This disorder has an enormous impact on public health and social and private life, and it generates a huge number of social costs. Alcohol use stimulates hypothalamic-pituitary-adrenal (HPA) axis responses and is the cause of many physical and social problems (especially liver disease and cancer), accidental injury, and risky sexual behavior. For years, researchers have been trying to identify the genetic basis of alcohol use disorder, the molecular mechanisms responsible for its development, and an effective form of therapy. Genetic and environmental factors are known to contribute to the development of AUD, and the expression of genes is a complicated process that depends on epigenetic modulations. Dietary nutrients, such as vitamins, may serve as one these modulators, as they have a direct impact on epigenomes. In this review, we connect gathered knowledge from three emerging fields-genetics, epigenetics, and nutrition-to form an amazing triangle relating to alcohol use disorder.
Collapse
Affiliation(s)
- Agnieszka Siomek-Gorecka
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-095 Bydgoszcz, Poland
- Correspondence: ; Tel.: +48-52-585-37-48
| | - Anna Dlugosz
- Department of Engineering and Chemical and Food Analytics, Faculty of Chemical Technology and Engineering, UTP University of Science and Technology, 85-326 Bydgoszcz, Poland;
| | - Damian Czarnecki
- Department of Preventive Nursing, Faculty of Health Sciences, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-821 Bydgoszcz, Poland;
| |
Collapse
|
26
|
Bielefeld P, Abbink MR, Davidson AR, Reijner N, Abiega O, Lucassen PJ, Korosi A, Fitzsimons CP. Early life stress decreases cell proliferation and the number of putative adult neural stem cells in the adult hypothalamus. Stress 2021; 24:189-195. [PMID: 33494651 DOI: 10.1080/10253890.2021.1879787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Stress is a potent environmental factor that can confer potent and enduring effects on brain structure and function. Exposure to stress during early life (ELS) has been linked to a wide range of consequences later in life. In particular, ELS exerts lasting effects on neurogenesis in the adult hippocampus, suggesting that ELS is a significant regulator of adult neural stem cell numbers and function. Here, we investigated the effect of ELS on cell proliferation and the numbers of neural stem/precursor cells in another neurogenic region: the hypothalamus of adult mice. We show that ELS has long-term suppressive effects on cell proliferation in the hypothalamic parenchyma and reduces the numbers of putative hypothalamic neural stem/precursor cells at 4 months of age. Specifically, ELS reduced the number of PCNA + cells present in hypothalamic areas surrounding the 3rd ventricle with a specific reduction in the proliferation of Sox2+/Nestin-GFP + putative stem cells present in the median eminence at the base of the 3rd ventricle. Furthermore, ELS reduced the total numbers of β-tanycytes lining the ventral 3rd ventricle, without affecting α-tanycyte numbers in more dorsal areas. These results are the first to indicate that ELS significantly reduces proliferation and β-tanycyte numbers in the adult hypothalamus, and may have (patho)physiological consequences for metabolic regulation or other hypothalamic functions in which β-tanycytes are involved.
Collapse
Affiliation(s)
- Pascal Bielefeld
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Maralinde R Abbink
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Anna R Davidson
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Niels Reijner
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Oihane Abiega
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlos P Fitzsimons
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
27
|
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: 8.0] [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
|
28
|
D'avila LF, Dias VT, Milanesi LH, Roversi K, Trevizol F, Maurer LH, Emanuelli T, Burger ME, Segat HJ. Interesterified fat consumption since gestation decreases striatal dopaminergic targets levels and gdnf impairing locomotion of adult offspring. Toxicol Lett 2020; 339:23-31. [PMID: 33359558 DOI: 10.1016/j.toxlet.2020.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/29/2020] [Accepted: 12/19/2020] [Indexed: 12/25/2022]
Abstract
Interesterified fat (IF) currently substitutes the hydrogenated vegetable fat (HVF) in processed foods. However, the IF consumption impact on the central nervous system (CNS) has been poorly studied. The current study investigated connections between IF chronic consumption and locomotor impairments in early life period and adulthood of rats and access brain molecular targets related to behavior changes in adulthood offspring. During pregnancy and lactation, female rats received soybean oil (SO) or IF and their male pups received the same maternal supplementation from weaning until adulthood. Pups' motor ability and locomotor activity in adulthood were evaluated. In the adult offspring striatum, dopaminergic targets, glial cell line-derived neurotrophic factor (GDFN) and lipid profile were quantified. Pups from IF supplementation group presented impaired learning concerning complex motor skill and sensorimotor behavior. The same animals showed decreased locomotion in adulthood. Moreover, IF group showed decreased immunoreactivity of all dopaminergic targets evaluated and GDNF, along with important changes in FA composition in striatum. This study shows that the brain modifications induce by IF consumption resulted in impaired motor control in pups and decreased locomotion in adult animals. Other studies about health damages induced by IF consumption may have a contribution from our current outcomes.
Collapse
Affiliation(s)
- Lívia Ferraz D'avila
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil.
| | - Verônica Tironi Dias
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil.
| | - Laura Hautrive Milanesi
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil.
| | - Karine Roversi
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil.
| | - Fabíola Trevizol
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil.
| | - Luana Haselein Maurer
- Programa de Pós-graduação em Ciência e Tecnologia dos Alimentos, Universidade Federal de Santa Maria (UFSM), RS, Brazil.
| | - Tatiana Emanuelli
- Programa de Pós-graduação em Ciência e Tecnologia dos Alimentos, Universidade Federal de Santa Maria (UFSM), RS, Brazil.
| | - Marilise Escobar Burger
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil; Departamento de Fisiologia e Farmacologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil.
| | - Hecson Jesser Segat
- Departamento de Patologia, Universidade Federal de Santa Maria (UFSM), RS, Brazil.
| |
Collapse
|
29
|
Bolton JL, Schulmann A, Garcia-Curran MM, Regev L, Chen Y, Kamei N, Shao M, Singh-Taylor A, Jiang S, Noam Y, Molet J, Mortazavi A, Baram TZ. Unexpected Transcriptional Programs Contribute to Hippocampal Memory Deficits and Neuronal Stunting after Early-Life Adversity. Cell Rep 2020; 33:108511. [PMID: 33326786 PMCID: PMC7817243 DOI: 10.1016/j.celrep.2020.108511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 07/08/2020] [Accepted: 11/19/2020] [Indexed: 01/23/2023] Open
Abstract
Early-life adversity (ELA) is associated with lifelong memory deficits, yet the responsible mechanisms remain unclear. We impose ELA by rearing rat pups in simulated poverty, assess hippocampal memory, and probe changes in gene expression, their transcriptional regulation, and the consequent changes in hippocampal neuronal structure. ELA rats have poor hippocampal memory and stunted hippocampal pyramidal neurons associated with ~140 differentially expressed genes. Upstream regulators of the altered genes include glucocorticoid receptor and, unexpectedly, the transcription factor neuron-restrictive silencer factor (NRSF/REST). NRSF contributes critically to the memory deficits because blocking its function transiently following ELA rescues spatial memory and restores the dendritic arborization of hippocampal pyramidal neurons in ELA rats. Blocking NRSF function in vitro augments dendritic complexity of developing hippocampal neurons, suggesting that NRSF represses genes involved in neuronal maturation. These findings establish important, surprising contributions of NRSF to ELA-induced transcriptional programming that disrupts hippocampal maturation and memory function.
Collapse
Affiliation(s)
- Jessica L Bolton
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Anton Schulmann
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Megan M Garcia-Curran
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Limor Regev
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Yuncai Chen
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Noriko Kamei
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Manlin Shao
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Akanksha Singh-Taylor
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Shan Jiang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Yoav Noam
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Jenny Molet
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-4475, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA 92697-4475, USA; Department of Neurology, University of California, Irvine, Irvine, CA 92697-4475, USA.
| |
Collapse
|
30
|
Cattaneo A, Suderman M, Cattane N, Mazzelli M, Begni V, Maj C, D'Aprile I, Pariante CM, Luoni A, Berry A, Wurst K, Hommers L, Domschke K, Cirulli F, Szyf M, Menke A, Riva MA. Long-term effects of stress early in life on microRNA-30a and its network: Preventive effects of lurasidone and potential implications for depression vulnerability. Neurobiol Stress 2020; 13:100271. [PMID: 33344724 PMCID: PMC7739180 DOI: 10.1016/j.ynstr.2020.100271] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023] Open
Abstract
Exposure to early life stress can interfere with neurodevelopmental trajectories to increase the vulnerability for psychiatric disorders later in life. With this respect, epigenetic mechanisms play a key role for the long-lasting changes in brain functions that may elicit and sustain psychopathologic outcomes. Here, we investigated DNA methylation changes as possible epigenetic mechanism mediating the effect of prenatal stress (PNS), an experimental paradigm associated with behavioral and molecular alterations relevant for psychiatric disorders. We identified 138 genes as being differentially methylated in the prefrontal cortex (PFC) and in the hippocampus (HIP) of male and female adult rats exposed to PNS. Among these genes, miR-30a and Neurod1 emerged as potential players for the negative outcomes associated with PNS exposure. Indeed, in addition to showing consistent methylation differences in both brain regions and in both sexes, and interacting with each other, they are both involved in Axon guidance and Neurotrophin signaling, which are important to neurodevelopmental disorders. We also found a significant reduction in the expression of a panel of genes (CAMK2A, c-JUN, LIMK1, MAP2K1, MAP2K2, PIK3CA and PLCG1) that belong to these two biological pathways and are also validated targets of miR-30a, pointing to a down-regulation of these pathways as a consequence of PNS exposure. Interestingly, we also found that miR-30a levels were significantly upregulated in depressed patients exposed to childhood trauma, as compared to control individuals. Importantly, we also found that a sub-chronic treatment with the atypical antipsychotic drug, lurasidone, during adolescence was able to prevent the up-regulation of miR-30a and normalized the expression of its target genes in response to PNS exposure. Our results demonstrate that miR-30a undergoes epigenetic changes following early life stress exposure and suggest that this miRNA could play a key role in producing broad and long-lasting alterations in neuroplasticity-related pathways, contributing to the etiology of psychiatric disorders. MiR-30a and Neurod1 undergo epigenetic changes following PNS exposure. MiR-30 and Neurod1 are involved in Axon guidance and Neurotrophin signaling, two important pathways for neurodevelopment. We found lower expression levels of a panel of genes targeted by miR-30a. MiR-30a was significantly up-regulated in depressed patients exposed to childhood trauma. A chronic treatment with lurasidone during adolescence prevented the up-regulation of miR-30a following PNS exposure.
Collapse
Affiliation(s)
- Annamaria Cattaneo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133, Milan, Italy.,Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Matthew Suderman
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, BSB 1TH, UK
| | - Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Monica Mazzelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133, Milan, Italy.,Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133, Milan, Italy
| | - Carlo Maj
- Institute for Genomic Statistics and Bioinformatics, University Hospital, Bonn, Germany
| | - Ilari D'Aprile
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom
| | - Alessia Luoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133, Milan, Italy
| | - Alessandra Berry
- Section of Behavioural Neurosciences, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Katharina Wurst
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Würzburg, Germany
| | - Leif Hommers
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Würzburg, Germany.,Interdisciplinary Center for Clinical Research, University Hospital of Würzburg, Germany
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Francesca Cirulli
- Section of Behavioural Neurosciences, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Moshe Szyf
- Department of Pharmacology and Therapeutics, McGill University, 3655 Sir William Osler Promenade #1311, Montreal, Quebec, Canada, H3G 1Y6
| | - Andreas Menke
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Würzburg, Germany.,Interdisciplinary Center for Clinical Research, University Hospital of Würzburg, Germany.,Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133, Milan, Italy
| |
Collapse
|
31
|
Luby JL, Baram TZ, Rogers CE, Barch DM. Neurodevelopmental Optimization after Early-Life Adversity: Cross-Species Studies to Elucidate Sensitive Periods and Brain Mechanisms to Inform Early Intervention. Trends Neurosci 2020; 43:744-751. [PMID: 32863044 PMCID: PMC7530018 DOI: 10.1016/j.tins.2020.08.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 01/01/2023]
Abstract
Human brain development is influenced by early-life experiences, particularly during sensitive periods, with impact on cognitive and emotional outcomes. Understanding how the timing and nature of such experiences (including adversity, trauma, and enrichment) govern their influence on brain organization is crucial for harnessing key environmental factors early in life to enhance brain development. Here we synthesize findings from human and animal studies focusing on sensitive periods and their regional and circuit specificity and highlight the challenge and power of such cross-species approaches in informing the 'next steps' to optimize cognitive and emotional health in developing children. We propose designs for neurodevelopmental optimization research programs utilizing randomized enhancement trials in early childhood to inform public health strategies on prevention and early intervention.
Collapse
Affiliation(s)
- Joan L Luby
- Department of Psychiatry, Washington University School of Medicine, Early Emotional Development Program, 4444 Forest Park Avenue, St. Louis, MO, USA.
| | - Tallie Z Baram
- Departments of Pediatrics, Anatomy/Neurobiology, and Neurology, University of California, Irvine, Irvine, CA 92697, USA
| | - Cynthia E Rogers
- Department of Psychiatry, Washington University School of Medicine, Early Emotional Development Program, 4444 Forest Park Avenue, St. Louis, MO, USA
| | - Deanna M Barch
- Department of Psychiatry, Washington University School of Medicine, Early Emotional Development Program, 4444 Forest Park Avenue, St. Louis, MO, USA; Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
32
|
Stress & sleep: A relationship lasting a lifetime. Neurosci Biobehav Rev 2020; 117:65-77. [DOI: 10.1016/j.neubiorev.2019.08.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/28/2019] [Accepted: 08/31/2019] [Indexed: 12/29/2022]
|
33
|
McKenna BG, Hendrix CL, Brennan PA, Smith AK, Stowe ZN, Newport DJ, Knight AK. Maternal prenatal depression and epigenetic age deceleration: testing potentially confounding effects of prenatal stress and SSRI use. Epigenetics 2020; 16:327-337. [PMID: 32660321 DOI: 10.1080/15592294.2020.1795604] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Previous studies suggest epigenetic alterations may contribute to the association between maternal prenatal depression and adverse offspring outcomes. Developmental researchers have recently begun to examine these associations in relation to epigenetic age acceleration/deceleration, a biomarker of developmental risk that reflects the deviation between epigenetic age and chronological age. In the perinatal period, preliminary studies indicate that maternal prenatal depression may lead to epigenetic age deceleration in newborns, which may predict adverse developmental outcomes. The present study examined the relationship between maternal prenatal exposures (i.e., depression, stress, and SSRI use) and offspring epigenetic age deceleration in 303 mother-offspring dyads. Women were recruited in the first trimester of pregnancy and followed longitudinally until delivery. Maternal depression, perceived stress, and SSRI use were assessed at each prenatal visit. Newborn epigenetic age was determined via cord blood samples. Results indicated maternal prenatal stress was not associated with newborn epigenetic age deceleration (ΔR2 = 0.002; p = 0.37). Maternal prenatal depression was associated with decelerated epigenetic age (ΔR2 = 0.01, p = 0.04), but this relationship did not hold when accounting for maternal use of SSRIs (ΔR2 = 0.002, p = 0.43). Conversely, maternal SSRI use significantly predicted newborn epigenetic age deceleration over and above the influence of maternal depression (ΔR2 = 0.03, p = 0.001). These findings suggest maternal prenatal SSRI use may significantly contribute to the previously documented association between maternal prenatal depression and epigenetic age deceleration. Further studies are needed to examine how these epigenetic differences at birth may contribute to adverse outcomes in later development.
Collapse
Affiliation(s)
| | | | | | - Alicia K Smith
- Department of Gynecology and Obstetrics, Emory University , Atlanta, GA, USA.,Department of Psychiatry and Behavioral Sciences, Emory University , Atlanta, GA, USA
| | - Zachary N Stowe
- Department of Psychiatry, University of Wisconsin , Madison, WI, USA
| | - D Jeffrey Newport
- Department of Psychiatry, University of Texas at Austin , Austin, TX, USA
| | - Anna K Knight
- Department of Gynecology and Obstetrics, Emory University , Atlanta, GA, USA
| |
Collapse
|
34
|
Mao ZF, Ouyang SH, Zhang QY, Wu YP, Wang GE, Tu LF, Luo Z, Li WX, Kurihara H, Li YF, He RR. New insights into the effects of caffeine on adult hippocampal neurogenesis in stressed mice: Inhibition of CORT-induced microglia activation. FASEB J 2020; 34:10998-11014. [PMID: 32619083 DOI: 10.1096/fj.202000146rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022]
Abstract
Chronic stress-evoked depression has been implied to associate with the decline of adult hippocampal neurogenesis. Caffeine has been known to combat stress-evoked depression. Herein, we aim to investigate whether the protective effect of caffeine on depression is related with improving adult hippocampus neurogenesis and explore the mechanisms. Mouse chronic water immersion restraint stress (CWIRS) model, corticosterone (CORT)-established cell stress model, a coculture system containing CORT-treated BV-2 cells and hippocampal neural stem cells (NSCs) were utilized. Results showed that CWIRS caused obvious depressive-like disorders, abnormal 5-HT signaling, and elevated-plasma CORT levels. Notably, microglia activation-evoked brain inflammation and inhibited neurogenesis were also observed in the hippocampus of stressed mice. In comparison, intragastric administration of caffeine (10 and 20 mg/kg, 28 days) significantly reverted CWIRS-induced depressive behaviors, neurogenesis recession and microglia activation in the hippocampus. Further evidences from both in vivo and in vitro mechanistic experiments demonstrated that caffeine treatment significantly suppressed microglia activation via the A2AR/MEK/ERK/NF-κB signaling pathway. The results suggested that CORT-induced microglia activation contributes to stress-mediated neurogenesis recession. The antidepression effect of caffeine was associated with unlocking microglia activation-induced neurogenesis inhibition.
Collapse
Affiliation(s)
- Zhong-Fu Mao
- Guangdong Engineering Research Centre of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| | - Shu-Hua Ouyang
- Guangdong Engineering Research Centre of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| | - Qiong-Yi Zhang
- Guangdong Engineering Research Centre of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| | - Yan-Ping Wu
- Guangdong Engineering Research Centre of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| | - Guo-En Wang
- Guangdong Engineering Research Centre of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| | - Long-Fang Tu
- Guangdong Engineering Research Centre of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhuo Luo
- Guangdong Engineering Research Centre of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| | - Wei-Xi Li
- School of Traditional Chinese Pharmacy, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Hiroshi Kurihara
- Guangdong Engineering Research Centre of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| | - Yi-Fang Li
- Guangdong Engineering Research Centre of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China.,School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Rong-Rong He
- Guangdong Engineering Research Centre of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, China
| |
Collapse
|
35
|
Grace JK, Parenteau C, Angelier F. Post-natal corticosterone exposure downregulates the HPA axis through adulthood in a common passerine. Gen Comp Endocrinol 2020; 292:113421. [PMID: 32032605 DOI: 10.1016/j.ygcen.2020.113421] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 01/29/2020] [Accepted: 02/03/2020] [Indexed: 10/25/2022]
Abstract
The hypothalamic-pituitaryadrenal (HPA) axis is one of the most important physiological mechanisms for mediating life-history trade-offs by reallocating resources to immediate survival from other life-history components during a perturbation. Early-life stressor experience and associated upregulation of glucocorticoids can induce short- and long-term changes to the HPA axis in ways that may optimize survival and/or reproduction for the expected adult environment. Although short-term changes to the HPA axis following perinatal stress are well documented, we know less about the long-term effects of early-life stress especially for non-mammalian wild species. Here, we determined long-term effects of experimental post-natal increases in a circulating glucocorticoid on the HPA axis in a common passerine bird, the house sparrow (Passer domesticus). We manipulated circulating corticosterone in wild, free-living nestlings, transferred fledglings to captivity and assessed corticosterone response to a standardized capture-restraint protocol at the pre-fledging, juvenile, and adult stages. Early-life corticosterone manipulation was associated with depressed baseline and stress-induced concentrations of corticosterone at all stages of life, through adulthood. These results provide rare evidence for the effects of early-life stressor experiences through adulthood, with important implications for understanding developmental programming of an endocrine mediator of life history trade-offs.
Collapse
Affiliation(s)
- Jacquelyn K Grace
- Dept. of Ecology and Conservation Biology, Texas A&M University, College Station, TX 77843, USA.
| | - Charline Parenteau
- Centre d'Etudes Biologiques de Chizé, Centre National de la Recherche Scientifique-Université de la Rochelle, UMR 7372, F-79360 Villiers en Bois, France
| | - Frédéric Angelier
- Centre d'Etudes Biologiques de Chizé, Centre National de la Recherche Scientifique-Université de la Rochelle, UMR 7372, F-79360 Villiers en Bois, France
| |
Collapse
|
36
|
Argyraki M, Damdimopoulou P, Chatzimeletiou K, Grimbizis GF, Tarlatzis BC, Syrrou M, Lambropoulos A. In-utero stress and mode of conception: impact on regulation of imprinted genes, fetal development and future health. Hum Reprod Update 2020; 25:777-801. [PMID: 31633761 DOI: 10.1093/humupd/dmz025] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/04/2019] [Accepted: 07/12/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Genomic imprinting is an epigenetic gene regulatory mechanism; disruption of this process during early embryonic development can have major consequences on both fetal and placental development. The periconceptional period and intrauterine life are crucial for determining long-term susceptibility to diseases. Treatments and procedures in assisted reproductive technologies (ART) and adverse in-utero environments may modify the methylation levels of genomic imprinting regions, including insulin-like growth factor 2 (IGF2)/H19, mesoderm-specific transcript (MEST), and paternally expressed gene 10 (PEG10), affecting the development of the fetus. ART, maternal psychological stress, and gestational exposures to chemicals are common stressors suspected to alter global epigenetic patterns including imprinted genes. OBJECTIVE AND RATIONALE Our objective is to highlight the effect of conception mode and maternal psychological stress on fetal development. Specifically, we monitor fetal programming, regulation of imprinted genes, fetal growth, and long-term disease risk, using the imprinted genes IGF2/H19, MEST, and PEG10 as examples. The possible role of environmental chemicals in genomic imprinting is also discussed. SEARCH METHODS A PubMed search of articles published mostly from 2005 to 2019 was conducted using search terms IGF2/H19, MEST, PEG10, imprinted genes, DNA methylation, gene expression, and imprinting disorders (IDs). Studies focusing on maternal prenatal stress, psychological well-being, environmental chemicals, ART, and placental/fetal development were evaluated and included in this review. OUTCOMES IGF2/H19, MEST, and PEG10 imprinted genes have a broad developmental effect on fetal growth and birth weight variation. Their disruption is linked to pregnancy complications, metabolic disorders, cognitive impairment, and cancer. Adverse early environment has a major impact on the developing fetus, affecting mostly growth, the structure, and subsequent function of the hypothalamic-pituitary-adrenal axis and neurodevelopment. Extensive evidence suggests that the gestational environment has an impact on epigenetic patterns including imprinting, which can lead to adverse long-term outcomes in the offspring. Environmental stressors such as maternal prenatal psychological stress have been found to associate with altered DNA methylation patterns in placenta and to affect fetal development. Studies conducted during the past decades have suggested that ART pregnancies are at a higher risk for a number of complications such as birth defects and IDs. ART procedures involve multiple steps that are conducted during critical windows for imprinting establishment and maintenance, necessitating long-term evaluation of children conceived through ART. Exposure to environmental chemicals can affect placental imprinting and fetal growth both in humans and in experimental animals. Therefore, their role in imprinting should be better elucidated, considering the ubiquitous exposure to these chemicals. WIDER IMPLICATIONS Dysregulation of imprinted genes is a plausible mechanism linking stressors such as maternal psychological stress, conception using ART, and chemical exposures with fetal growth. It is expected that a greater understanding of the role of imprinted genes and their regulation in fetal development will provide insights for clinical prevention and management of growth and IDs. In a broader context, evidence connecting impaired imprinted gene function to common diseases such as cancer is increasing. This implies early regulation of imprinting may enable control of long-term human health, reducing the burden of disease in the population in years to come.
Collapse
Affiliation(s)
- Maria Argyraki
- First Department of Obstetrics and Gynecology, Laboratory of Genetics, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece
| | - Pauliina Damdimopoulou
- Karolinska Institutet, Department of Clinical Sciences, Intervention and Technology, Unit of Obstetrics and Gynecology, K57 Karolinska University Hospital Huddinge, SE-14186 Stockholm, Sweden
| | - Katerina Chatzimeletiou
- First Department of Obstetrics and Gynecology, Unit for Human Reproduction, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece
| | - Grigoris F Grimbizis
- First Department of Obstetrics and Gynecology, Unit for Human Reproduction, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece
| | - Basil C Tarlatzis
- First Department of Obstetrics and Gynecology, Unit for Human Reproduction, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece
| | - Maria Syrrou
- Department of Biology, Laboratory of Biology, School of Health Sciences, University of Ioannina, Dourouti University Campus, 45110, Ioannina, Greece
| | - Alexandros Lambropoulos
- First Department of Obstetrics and Gynecology, Laboratory of Genetics, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece
| |
Collapse
|
37
|
Yang S, Zhao N, Sun B, Yang Y, Hu Y, Zhao R. Grandmaternal betaine supplementation enhances hepatic IGF2 expression in F2 rat offspring through modification of promoter DNA methylation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:1486-1494. [PMID: 31756772 DOI: 10.1002/jsfa.10156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 10/07/2019] [Accepted: 11/18/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND We reported previously that maternal betaine promotes hepatic insulin-like growth factor (IGF2) expression in F1 offspring rats through hypermethylation of the IGF2/H19 imprinting control region (ICR). It remains unknown whether this acquired trait can be transmitted to the F2 generation. This study aimed to determine whether dietary betaine supplementation to grand dams affects the hepatic IGF2 expression in F2 rat offspring and how it is related to alterations in DNA methylation. F2 rat offspring derived from grand dams fed basal or betaine-supplemented diet (10 g kg-1 ) were examined at weaning. Serum IGF2 concentration was measured with enzyme-linked immunosorbent assay (ELISA). Hepatic expression of IGF2, together with other proliferation and apoptosis markers, was determined by using quantitative polymerase chain reaction (qPCR), western blot, and immunohistochemistry. The methylation status of the IGF2/H19 ICR and the promoters of IGF2 gene were detected by methylated DNA immunoprecipitation quantitative polymerase chain reaction (MeDIP-qPCR). RESULTS The maternal betaine-induced up-regulation of hepatic IGF2 expression in F1 rat offspring was transmitted to the F2 generation. The F2 rats from the betaine group demonstrated enhanced hepatic IGF2 expression at both mRNA and protein levels, in association with higher serum IGF2 concentration. No alterations were observed in the ICR methylation of the IGF2/H19 locus, and hypomethylation was detected in promoters of IGF2 gene in betaine group. CONCLUSION These results indicate that maternal betaine enhances hepatic IGF2 expression in F2 rat offspring through modification of DNA methylation on IGF2 promoters. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Shu Yang
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, P. R. China
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, P. R. China
| | - Nannan Zhao
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, P. R. China
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, P. R. China
| | - Bo Sun
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, P. R. China
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, P. R. China
| | - Yang Yang
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, P. R. China
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, P. R. China
| | - Yun Hu
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, P. R. China
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, P. R. China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, P. R. China
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, P. R. China
| |
Collapse
|
38
|
van Keulen BJ, Dolan CV, Andrew R, Walker BR, Hulshoff Pol HE, Boomsma DI, Rotteveel J, Finken MJJ. Heritability of Cortisol Production and Metabolism Throughout Adolescence. J Clin Endocrinol Metab 2020; 105:5586817. [PMID: 31608377 PMCID: PMC7046020 DOI: 10.1210/clinem/dgz016] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022]
Abstract
CONTEXT Inter-individual differences in cortisol production and metabolism emerge with age and may be explained by genetic factors. OBJECTIVE To estimate the relative contributions of genetic and environmental factors to inter-individual differences in cortisol production and metabolism throughout adolescence. DESIGN Prospective follow-up study of twins. SETTING Nationwide register. PARTICIPANTS 218 mono- and dizygotic twins (N = 109 pairs) born between 1995 amd 1996, recruited from the Netherlands Twin Register. Cortisol metabolites were determined in 213, 169, and 160 urine samples at the ages of 9, 12, and 17, respectively. MAIN OUTCOME MEASURES The total contribution of genetic factors (broad-sense heritability) and shared and unshared environmental influences to inter-individual differences in cortisol production and activities of 5α-reductase, 5β-reductase, and 11β-hydroxysteroid dehydrogenases and cytochrome P450 3A4. RESULTS For cortisol production rate at the ages of 9, 12, and 17, broad-sense heritability was estimated as 42%, 30%, and 0%, respectively, and the remainder of the variance was explained by unshared environmental factors. For cortisol metabolism indices, the following heritability was observed: for the A-ring reductases (5α-and 5β-reductases), broad-sense heritability increased with age (to >50%), while for the other indices (renal 11β-HSD2, global 11β-HSD, and CYP3A4), the contribution of genetic factors was highest (68%, 18%, and 67%, respectively) at age 12. CONCLUSIONS The contribution of genetic factors to inter-individual differences in cortisol production decreased between 12 and 17y, indicative of a predominant role of individual circumstances. For cortisol metabolism, distinct patterns of genetic and environmental influences were observed, with heritability that either increased with age or peaked at age 12y.
Collapse
Affiliation(s)
- Britt J van Keulen
- Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Endocrinology, Amsterdam, The Netherlands
- Correspondence and Requests: Britt J van Keulen, MD, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric endocrinology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands. E-mail:
| | - Conor V Dolan
- Department of Biological Psychology, Vrije Universiteit Amsterdam, The Netherlands
| | - Ruth Andrew
- Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, UK
| | - Brian R Walker
- Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, UK
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Hilleke E Hulshoff Pol
- Department of Psychiatry, Brian Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, The Netherlands
| | - Joost Rotteveel
- Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Endocrinology, Amsterdam, The Netherlands
| | - Martijn J J Finken
- Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Endocrinology, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Short AK, Baram TZ. Early-life adversity and neurological disease: age-old questions and novel answers. Nat Rev Neurol 2019; 15:657-669. [PMID: 31530940 PMCID: PMC7261498 DOI: 10.1038/s41582-019-0246-5] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
Neurological illnesses, including cognitive impairment, memory decline and dementia, affect over 50 million people worldwide, imposing a substantial burden on individuals and society. These disorders arise from a combination of genetic, environmental and experiential factors, with the latter two factors having the greatest impact during sensitive periods in development. In this Review, we focus on the contribution of adverse early-life experiences to aberrant brain maturation, which might underlie vulnerability to cognitive brain disorders. Specifically, we draw on recent robust discoveries from diverse disciplines, encompassing human studies and experimental models. These discoveries suggest that early-life adversity, especially in the perinatal period, influences the maturation of brain circuits involved in cognition. Importantly, new findings suggest that fragmented and unpredictable environmental and parental signals comprise a novel potent type of adversity, which contributes to subsequent vulnerabilities to cognitive illnesses via mechanisms involving disordered maturation of brain 'wiring'.
Collapse
Affiliation(s)
- Annabel K Short
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA.
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA.
- Departments of Neurology, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
40
|
Fusco S, Spinelli M, Cocco S, Ripoli C, Mastrodonato A, Natale F, Rinaudo M, Livrizzi G, Grassi C. Maternal insulin resistance multigenerationally impairs synaptic plasticity and memory via gametic mechanisms. Nat Commun 2019; 10:4799. [PMID: 31641124 PMCID: PMC6805915 DOI: 10.1038/s41467-019-12793-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
Metabolic diseases harm brain health and cognitive functions, but whether maternal metabolic unbalance may affect brain plasticity of next generations is still unclear. Here, we demonstrate that maternal high fat diet (HFD)-dependent insulin resistance multigenerationally impairs synaptic plasticity, learning and memory. HFD downregulates BDNF and insulin signaling in maternal tissues and epigenetically inhibits BDNF expression in both germline and hippocampus of progeny. Notably, exposure of the HFD offspring to novel enriched environment restores Bdnf epigenetic activation in the male germline and counteracts the transmission of cognitive impairment to the next generations. BDNF administration to HFD-fed mothers or preserved insulin sensitivity in HFD-fed p66Shc KO mice also prevents the intergenerational transmission of brain damage to the progeny. Collectively, our data suggest that maternal diet multigenerationally impacts on descendants' brain health via gametic mechanisms susceptible to lifestyle.
Collapse
Affiliation(s)
- Salvatore Fusco
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy. .,Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
| | - Matteo Spinelli
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Sara Cocco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Cristian Ripoli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy.,Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Alessia Mastrodonato
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Francesca Natale
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Marco Rinaudo
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Giulia Livrizzi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy. .,Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
| |
Collapse
|
41
|
Arika WM, Kibiti CM, Njagi JM, Ngugi MP. Modulation of Cognition: The Role of Gnidia glauca on Spatial Learning and Memory Retention in High-Fat Diet-Induced Obese Rats. Neural Plast 2019; 2019:2867058. [PMID: 31565046 PMCID: PMC6745098 DOI: 10.1155/2019/2867058] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/22/2019] [Accepted: 08/13/2019] [Indexed: 11/26/2022] Open
Abstract
Chronic exposures to high-fat diets are linked to neuropathological changes that culminate in obesity-related cognitive dysfunction and brain alteration. Learning, memory performance, and executive function are the main domains affected by an obesogenic diet. There are limited effective therapies for addressing cognitive deficits. Thus, it is important to identify additional and alternative therapies. In African traditional medicine, Gnidia glauca has putative efficacy in the management of obesity and associated complications. The use of Gnidia glauca is largely based on its long-term traditional use. Its therapeutic application has not been accompanied by sufficient scientific evaluation to validate its use. Therefore, the current study sought to explore the modulatory effects of dichloromethane leaf extracts of Gnidia glauca on cognitive function in the high-fat diet- (HFD-) induced obese rats. Obesity was induced by feeding the rats with prepared HFD and water ad libitum for 6 weeks. The in vivo antiobesity effects were determined by oral administration of G. glauca at dosage levels of 200, 250, and 300 mg/kg body weight in HFD-induced obese rats from the 6th to the 12th weeks. The Lee obesity index was used as a diagnostic criterion of obesity. The Morris water maze was employed to test spatial learning and memory retention in rats. The results indicated that Gnidia glauca showed potent antiobesity effects as indicated in the reduction of body weight and obesity index in extract-treated rats. Moreover, Gnidia glauca exhibited cognitive-enhancing effects in obese rats. The positive influences on cognitive functions might be attributed to the extracts' phytochemicals that have been suggested to confer protection against obesity-induced oxidative damage, reduction of central inflammation, and increased neurogenesis. The therapeutic effects observed suggest that Gnidia glauca might be an alternative to current medications for the symptomatic complications of obesity, such as learning and memory loss. Further studies are therefore needed to establish its toxicity profiles.
Collapse
Affiliation(s)
- Wycliffe Makori Arika
- Department of Biochemistry, Microbiology and Biotechnology, School of Pure and Applied Sciences, Kenyatta University, P.O. Box 43844-00100, Nairobi, Kenya
| | - Cromwell Mwiti Kibiti
- Department of Pure and Applied Sciences, Technical University of Mombasa, P.O. Box 90420-80100, Mombasa, Kenya
| | - Joan Murugi Njagi
- Department of Environmental and Occupational Health, School of Environmental Sciences, Kenyatta University, P.O. Box 43844-00100, Nairobi, Kenya
| | - Mathew Piero Ngugi
- Department of Biochemistry, Microbiology and Biotechnology, School of Pure and Applied Sciences, Kenyatta University, P.O. Box 43844-00100, Nairobi, Kenya
| |
Collapse
|
42
|
Probiotic treatment restores normal developmental trajectories of fear memory retention in maternally separated infant rats. Neuropharmacology 2019; 153:53-62. [DOI: 10.1016/j.neuropharm.2019.04.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 01/01/2023]
|
43
|
Abbink MR, van Deijk ALF, Heine VM, Verheijen MH, Korosi A. The involvement of astrocytes in early-life adversity induced programming of the brain. Glia 2019; 67:1637-1653. [PMID: 31038797 PMCID: PMC6767561 DOI: 10.1002/glia.23625] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 12/13/2022]
Abstract
Early‐life adversity (ELA) in the form of stress, inflammation, or malnutrition, can increase the risk of developing psychopathology or cognitive problems in adulthood. The neurobiological substrates underlying this process remain unclear. While neuronal dysfunction and microglial contribution have been studied in this context, only recently the role of astrocytes in early‐life programming of the brain has been appreciated. Astrocytes serve many basic roles for brain functioning (e.g., synaptogenesis, glutamate recycling), and are unique in their capacity of sensing and integrating environmental signals, as they are the first cells to encounter signals from the blood, including hormonal changes (e.g., glucocorticoids), immune signals, and nutritional information. Integration of these signals is especially important during early development, and therefore we propose that astrocytes contribute to ELA induced changes in the brain by sensing and integrating environmental signals and by modulating neuronal development and function. Studies in rodents have already shown that ELA can impact astrocytes on the short and long term, however, a critical review of these results is currently lacking. Here, we will discuss the developmental trajectory of astrocytes, their ability to integrate stress, immune, and nutritional signals from the early environment, and we will review how different types of early adversity impact astrocytes.
Collapse
Affiliation(s)
- Maralinde R Abbink
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne-Lieke F van Deijk
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Vivi M Heine
- Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Mark H Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Umeoka EHL, Robinson EJ, Turimella SL, van Campen JS, Motta-Teixeira LC, Sarabdjitsingh RA, Garcia-Cairasco N, Braun K, de Graan PN, Joëls M. Hyperthermia-induced seizures followed by repetitive stress are associated with age-dependent changes in specific aspects of the mouse stress system. J Neuroendocrinol 2019; 31:e12697. [PMID: 30773738 DOI: 10.1111/jne.12697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/04/2019] [Accepted: 02/13/2019] [Indexed: 11/28/2022]
Abstract
Stress is among the most frequently self-reported factors provoking epileptic seizures in children and adults. It is still unclear, however, why some people display stress-sensitive seizures and others do not. Recently, we showed that young epilepsy patients with stress-sensitive seizures exhibit a dysregulated hypothalamic-pituitary-adrenal (HPA)-axis. Most likely, this dysregulation gradually develops, and is triggered by stressors occurring early in life (early-life stress [ELS]). ELS may be particularly impactful when overlapping with the period of epileptogenesis. To examine this in a controlled and prospective manner, the present study investigated the effect of repetitive variable stressors or control treatment between postnatal day (PND) 12 and 24 in male mice exposed on PND10 to hyperthermia (HT)-induced prolonged seizures (control: normothermia). A number of peripheral and central indices of HPA-axis activity were evaluated at pre-adolescent and young adult age (ie, at PND25 and 90, respectively). At PND25 but not at PND90, body weight gain and absolute as well as relative (to body weight) thymus weight were reduced by ELS (vs control), whereas relative adrenal weight was enhanced, confirming the effectiveness of the stress treatment. Basal and stress-induced corticosterone levels were unaffected, though, by ELS at both ages. HT by itself did not affect any of these peripheral markers of HPA-axis activity, nor did it interact with ELS. However, centrally we did observe age-specific interaction effects of HT and ELS with regard to hippocampal glucocorticoid receptor mRNA expression, neurogenesis with the immature neurone marker doublecortin and the number of hilar (ectopic) granule cells using Prox1 staining. This lends some support to the notion that exposure to repetitive stress after HT-induced seizures may dysregulate central components of the stress system in an age-dependent manner. Such dysregulation could be one of the mechanisms conferring higher vulnerability of individuals with epilepsy to develop seizures in the face of stress.
Collapse
Affiliation(s)
- Eduardo H L Umeoka
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Neuroscience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Edward J Robinson
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Sada Lakshmi Turimella
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Jolien S van Campen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lívia C Motta-Teixeira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - R Angela Sarabdjitsingh
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Norberto Garcia-Cairasco
- Neuroscience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Kees Braun
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pierre N de Graan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Marian Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
45
|
Cheng W, Han F, Shi Y. Neonatal isolation modulates glucocorticoid-receptor function and synaptic plasticity of hippocampal and amygdala neurons in a rat model of single prolonged stress. J Affect Disord 2019; 246:682-694. [PMID: 30611912 DOI: 10.1016/j.jad.2018.12.084] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/23/2018] [Accepted: 12/24/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Early life and stressful experiences affect hippocampal and amygdala structure and function. They also increase the incidence of mental and nervous system disorders in adults. However, prospective studies have yet to show if early-life experiences affect the risk/severity of post-traumatic stress disorder (PTSD). METHODS We applied neonatal isolation (NI) alone, single prolonged stress (SPS) alone and NI + SPS to rats. We evaluated anxiety-like behavior and spatial memory of behavior using open field, elevated plus maze, and Morris water maze tests. Then, we measured expression of glucocorticoid receptors (GRs) and synaptic-related proteins by immunofluorescence, immunohistochemistry and western blotting in the hippocampus and amygdala. RESULTS NI + SPS exacerbated the increased anxiety levels and impaired spatial memory induced by NI alone or SPS alone. NI alone or SPS alone induced varying degrees of change in expression of GRs and synaptic proteins (synapsin I and postsynaptic density protein-95) in the hippocampus and amygdala. There were opposite changes in GR expression in the hippocampal dentate gyrus and basolateral amygdala. The degree of such change was exacerbated considerably by NI + SPS. In addition, neuroligin (NLG)-1 and NLG-2 were distributed in postsynaptic sites of excitatory and inhibitory synapses, respectively. NI, SPS, and NI + SPS altered the patterns of NLG-1 and NLG-2 colocalization as well as their intensity. NI + SPS strengthened the increased ratio of NLG-1/NLG-2 in the hippocampus, but decreased this ratio in the amygdala. CONCLUSIONS NI and SPS together induced greater degrees of change in anxiety and spatial memory, as well as GR and synaptic protein levels, in the hippocampus and amygdala than the changes induced by NI alone or SPS alone.
Collapse
Affiliation(s)
- Wei Cheng
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 77, Puhe Road, Shenbei New District, 110001 Shenyang, China; Neonatal Department, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Fan Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 77, Puhe Road, Shenbei New District, 110001 Shenyang, China
| | - Yuxiu Shi
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 77, Puhe Road, Shenbei New District, 110001 Shenyang, China.
| |
Collapse
|
46
|
Crouse MS, Caton JS, Cushman RA, McLean KJ, Dahlen CR, Borowicz PP, Reynolds LP, Ward AK. Moderate nutrient restriction of beef heifers alters expression of genes associated with tissue metabolism, accretion, and function in fetal liver, muscle, and cerebrum by day 50 of gestation. Transl Anim Sci 2019; 3:855-866. [PMID: 32704851 PMCID: PMC7200894 DOI: 10.1093/tas/txz026] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/22/2019] [Indexed: 01/05/2023] Open
Abstract
We hypothesized that a moderate maternal nutrient restriction during the first 50 d of gestation in beef heifers would affect transcript abundance of genes associated with tissue metabolism, accretion, and function in fetal liver, muscle, and cerebrum. Angus-cross heifers were estrus synchronized and assigned at breeding to one of two dietary treatments (CON- 100% of nutrient requirements to gain 0.45 kg/d; RES- 60% of CON). At day 50 of gestation, 14 heifers were ovariohysterectomized, and fetal liver, muscle, and cerebrum were collected. Transcriptome analysis via RNA-seq was conducted on the Illumina HiSeq 2500 platform using 50-bp paired-end reads at a depth of 2 × 10.4M reads/sample. Bioinformatic analysis was performed using the Tuxedo Suite and ontological analysis with DAVID 6.8. For fetal liver, muscle, and cerebrum, a total of 548, 317, and 151 genes, respectively (P < 0.01) were differentially expressed, of which 201, 144, and 28 genes, respectively were false discovery rate protected (FDR; q < 0.10). Differentially expressed genes were screened for fit into functional categories of pathways or ontologies associated with known impacts on tissue metabolism, accretion, and function. In fetal liver, five functional categories of interest (n = 125 genes) were affected by nutritional treatment: metabolic pathways, protein kinase, nucleosome core, mRNA splicing, and complement/coagulation cascades, of which 105 genes were upregulated in RES. In fetal muscle, three functional categories of interest (n = 106 genes) were affected by nutritional treatment: skeletal muscle, embryogenesis, and signaling cascades, of which 64 genes were upregulated in RES. In fetal cerebrum, three functional categories of interest (n = 60 genes) were affected by nutritional treatment: hippocampus and neurogenesis, metal-binding, and cytoskeleton, of which 58 genes were upregulated in RES. These results demonstrate that a moderate maternal nutrient restriction during the first 50 d of gestation in beef heifers alters transcript abundance of genes potentially impacting tissue metabolism, accretion, and function in fetal liver, muscle, and cerebrum. Furthermore, these results indicate that affected categories are tissue-specific and moderate maternal nutrient restriction generally increases expression of genes in fetuses from RES fed dams. Finally, these data lay the foundation upon which further research that identifies phenotypic responses to changes in these pathways may be elucidated.
Collapse
Affiliation(s)
- Matthew S Crouse
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND
| | - Joel S Caton
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND
| | | | - Kyle J McLean
- Department of Animal Science, University of Tennessee, Knoxville, TN
| | - Carl R Dahlen
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND
| | - Pawel P Borowicz
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND
| | - Lawrence P Reynolds
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND
| | - Alison K Ward
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND
| |
Collapse
|
47
|
Bolton JL, Short AK, Simeone KA, Daglian J, Baram TZ. Programming of Stress-Sensitive Neurons and Circuits by Early-Life Experiences. Front Behav Neurosci 2019; 13:30. [PMID: 30833892 PMCID: PMC6387907 DOI: 10.3389/fnbeh.2019.00030] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/04/2019] [Indexed: 12/22/2022] Open
Abstract
Early-life experiences influence brain structure and function long-term, contributing to resilience or vulnerability to stress and stress-related disorders. Therefore, understanding the mechanisms by which early-life experiences program specific brain cells and circuits to shape life-long cognitive and emotional functions is crucial. We identify the population of corticotropin-releasing hormone (CRH)-expressing neurons in the hypothalamic paraventricular nucleus (PVN) as a key, early target of early-life experiences. Adverse experiences increase excitatory neurotransmission onto PVN CRH cells, whereas optimal experiences, such as augmented and predictable maternal care, reduce the number and function of glutamatergic inputs onto this cell population. Altered synaptic neurotransmission is sufficient to initiate large-scale, enduring epigenetic re-programming within CRH-expressing neurons, associated with stress resilience and additional cognitive and emotional outcomes. Thus, the mechanisms by which early-life experiences influence the brain provide tractable targets for intervention.
Collapse
Affiliation(s)
- Jessica L Bolton
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Annabel Katherine Short
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Kristina A Simeone
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Jennifer Daglian
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Tallie Z Baram
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
48
|
Yam K, Schipper L, Reemst K, Ruigrok SR, Abbink MR, Hoeijmakers L, Naninck EFG, Zarekiani P, Oosting A, Van Der Beek EM, Lucassen PJ, Korosi A. Increasing availability of ω‐3 fatty acid in the early‐life diet prevents the early‐life stress‐induced cognitive impairments without affecting metabolic alterations. FASEB J 2019; 33:5729-5740. [DOI: 10.1096/fj.201802297r] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Kit‐Yi Yam
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | | | - Kitty Reemst
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Silvie R. Ruigrok
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Maralinde R. Abbink
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Lianne Hoeijmakers
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Eva F. G. Naninck
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Parand Zarekiani
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | | | - Eline M. Van Der Beek
- Danone Nutricia Research Utrecht The Netherlands
- Department of PediatricsUniversity Medical Centre GroningenUniversity of Groningen Groningen The Netherlands
| | - Paul J. Lucassen
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Aniko Korosi
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| |
Collapse
|
49
|
Lindsay KL, Buss C, Wadhwa PD, Entringer S. The Interplay Between Nutrition and Stress in Pregnancy: Implications for Fetal Programming of Brain Development. Biol Psychiatry 2019; 85:135-149. [PMID: 30057177 PMCID: PMC6389360 DOI: 10.1016/j.biopsych.2018.06.021] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/04/2018] [Accepted: 06/15/2018] [Indexed: 12/17/2022]
Abstract
Growing evidence supports an important role for the intrauterine environment in shaping fetal development and subsequent child health and disease risk. The fetal brain is particularly plastic, whereby even subtle changes in structure and function produced by in utero conditions can have long-term implications. Based on the consideration that conditions related to energy substrate and likelihood of survival to reproductive age are particularly salient drivers of fetal programming, maternal nutrition and stress represent the most commonly, but independently, studied factors in this context. However, the effects of maternal nutrition and stress are context dependent and may be moderated by one another. Studies examining the effects of the bidirectional nutrition-stress interplay in pregnancy on fetal programming of brain development are beginning to emerge in the literature. This review incorporates all currently available animal and human studies of this interplay and provides a synthesis and critical discussion of findings. Nine of the 10 studies included here assessed nutrition-stress interactions and offspring neurodevelopmental or brain development outcomes. Despite significant heterogeneity in study design and methodology, two broad patterns of results emerge to suggest that the effects of prenatal stress on various aspects of brain development may be mitigated by 1) higher fat diets or increased intake and/or status of specific dietary fats and 2) higher dietary intake or supplementation of targeted nutrients. The limitations of these studies are discussed, and recommendations are provided for future research to expand on this important area of fetal programming of brain development.
Collapse
Affiliation(s)
- Karen L Lindsay
- Department of Pediatrics, University of California, Irvine, Irvine, California; UC Irvine Development, Health and Disease Research Program, University of California, Irvine, Irvine, California
| | - Claudia Buss
- Department of Pediatrics, University of California, Irvine, Irvine, California; UC Irvine Development, Health and Disease Research Program, University of California, Irvine, Irvine, California; Institute of Medical Psychology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Pathik D Wadhwa
- Department of Pediatrics, University of California, Irvine, Irvine, California; Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, California; Department of Obstetrics and Gynecology, University of California, Irvine, Irvine, California; UC Irvine Development, Health and Disease Research Program, University of California, Irvine, Irvine, California
| | - Sonja Entringer
- Department of Pediatrics, University of California, Irvine, Irvine, California; UC Irvine Development, Health and Disease Research Program, University of California, Irvine, Irvine, California; Institute of Medical Psychology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
50
|
Sandgren AM, Brummer RJ. ADHD-originating in the gut? The emergence of a new explanatory model. Med Hypotheses 2018; 120:135-145. [DOI: 10.1016/j.mehy.2018.08.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 08/25/2018] [Indexed: 12/12/2022]
|