1
|
Merle L, Rastelli M, Datiche F, Véjux A, Jacquin-Piques A, Bouret SG, Benani A. Maternal Diet and Vulnerability to Cognitive Impairment in Adulthood: Possible Link with Alzheimer's Disease? Neuroendocrinology 2025; 115:242-266. [PMID: 39799941 DOI: 10.1159/000543499] [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: 09/13/2024] [Accepted: 12/15/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND Aging is the main risk factor for developing cognitive impairments and associated neurodegenerative diseases. However, environmental factors, including nutritional health, are likely to promote or reduce cognitive impairments and neurodegenerative pathologies. An intricate relationship exists between maternal nutrition and adult eating behavior, metabolic phenotype, and cognitive abilities. SUMMARY The objective of the present review was to collect available data, suggesting a link between maternal overnutrition and the latter impairment of cognitive functions in the progeny, and to relate this relationship with Alzheimer's disease (AD). Indeed, cognitive impairments are major behavioral signs of AD. We first reviewed studies showing an association between unbalanced maternal diet and cognitive impairments in the progeny in humans and rodent models. Then we looked for cellular and molecular hallmarks which could constitute a breeding ground for AD in those models. With this end, we focused on synaptic dysfunction, altered neurogenesis, neuroinflammation, oxidative stress, and pathological protein aggregation. Finally, we proposed an indirect mechanism linking maternal unbalanced diet and progeny's vulnerability to cognitive impairments and neurodegeneration through promoting metabolic diseases. We also discussed the involvement of progeny's gut microbiota in the maternal diet-induced vulnerability to metabolic and neurodegenerative diseases. KEY MESSAGES Further investigations are needed to fully decipher how maternal diet programs the fetus and infant brain. Addressing this knowledge gap would pave the way to precise nutrition and personalized medicine to better handle cognitive impairments in adulthood.
Collapse
Affiliation(s)
- Laetitia Merle
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Marialetizia Rastelli
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, Inserm UMR-S1172, CHU Lille, University of Lille, Lille, France
| | - Frédérique Datiche
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Anne Véjux
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Agnès Jacquin-Piques
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Department of Clinical Neurophysiology, INRAE, Institut Agro, Université de Bourgogne, CHU Dijon, Dijon, France
| | - Sébastien G Bouret
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, Inserm UMR-S1172, CHU Lille, University of Lille, Lille, France
| | - Alexandre Benani
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| |
Collapse
|
2
|
Lee YJ, Kim J, Kwon YH. Long-Term Effects of Maternal Fat Consumption on the Brain Transcriptome of Obesogenic Diet-Fed Young Adult Mice Offspring. J Nutr 2024; 154:1532-1539. [PMID: 38484978 DOI: 10.1016/j.tjnut.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/17/2024] [Accepted: 03/11/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Substantial evidence has demonstrated that maternal high-fat (HF) consumption during gestation and lactation plays as a risk factor for neurodevelopmental alterations and subsequent neurological disorders. OBJECTIVE We investigated the regulatory mechanisms of maternal fat consumption on brain development and function in offspring at different ages. METHODS Mouse dams were fed either a control diet [low-fat (LF)] or an HF diet for 3 wk before mating and throughout pregnancy and lactation. Offspring were killed at postnatal day (PD) 21 (LF21 and HF21), and the rest were fed an HF diet for 12 wk until the killing at PD 105 (LF105 and HF105). The expression levels of genes and proteins in the brains of offspring were analyzed by microarray and immunoblotting, respectively. RESULTS Maternal dietary fat content, offspring age, and their interaction affected the expression levels of 1215, 10,453, and 2105 genes, respectively. The 67 differentially expressed genes (DEGs) between the HF21 and LF21 groups were enriched in several Gene Ontology terms related to nervous system development. Among 45 DEGs of the HF105/LF105 comparison, several genes associated with neurotransmitter action are detected. In addition, we observed increased activation of the AMP-dependent protein kinase-cAMP response element binding protein signaling pathway in HF105/LF105 comparison. However, maternal fat content did not change the protein levels of amyloid-β and tau hyperphosphorylation, the markers of neuropathogenesis. CONCLUSIONS Maternal HF feeding altered the expression of genes involved in the development and neurotransmitter system in the brains of PD 21 and HF diet-fed PD 105 offspring, respectively. Especially, the absence of overlap between DEGs at each comparison highlights the dynamic nature of alterations in gene expression in offspring of dams fed an HF diet. Further investigation on older adult offspring is necessary to elucidate the effects of maternal fat intake on the brain pathophysiology of offspring.
Collapse
Affiliation(s)
- Youn Ji Lee
- Department of Food and Nutrition, Seoul National University, Seoul, Korea
| | - Juhae Kim
- Department of Food and Nutrition, Seoul National University, Seoul, Korea; Research Institute of Human Ecology, Seoul National University, Seoul, Korea
| | - Young Hye Kwon
- Department of Food and Nutrition, Seoul National University, Seoul, Korea; Research Institute of Human Ecology, Seoul National University, Seoul, Korea.
| |
Collapse
|
3
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
4
|
Wen J, Wang Y, Wang C, Yuan M, Chen F, Zou Q, Cai Z, Zhao B. Dietary High-Fat Promotes Cognitive Impairment by Suppressing Mitophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:4822767. [PMID: 36718278 PMCID: PMC9884172 DOI: 10.1155/2023/4822767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/22/2023]
Abstract
Dietary habits contribute to the characteristics of Alzheimer's disease (AD) and cognitive impairment, which are partly induced by the accumulation of hyperphosphorylated Tau, a microtubule-associated protein. In mice, a fat-rich diet facilitates cognitive dysfunction. However, the mechanism by which dietary fat damages the brain remains unclear. In this study, 13-month-old C57BL/6 mice were fed a normal or high-fat diet (HFD) for 6 months. Neuro-2a cells were incubated with the normal medium or palmitic acid (200 μM). Spatial memory was assessed utilizing a behavioral test. Further, western blotting and immunofluorescence techniques were used to determine the levels of mitophagy-related proteins. The synaptic morphology and phosphorylation of Tau proteins were also evaluated. Administration of HFD decreased the expression of synaptophysin and brain-derived neurotrophic factor expression, leading to significant damage to neurons. Tau protein hyperphosphorylation was detected at different loci both in vivo and in vitro. Significantly impaired learning and memory abilities, accompanied by impaired mitophagy-related processes, were observed in mice fed with HFD as compared to mice fed with normal food. In conclusion, high fatty-acid intake hinders mitophagy and upregulates Tau protein phosphorylation, including age-related synaptic dysfunction, which leads to cognitive decline.
Collapse
Affiliation(s)
- Jie Wen
- Department and Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong 524001, China
- Guangdong Key Laboratory of Aging-Related Cardiac and Cerebral Diseases, Zhanjiang, Guangdong 524001, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Yuzhong District, Chongqing 400013, China
- Department of Neurology, Chongqing General Hospital, Yuzhong District, Chongqing 400013, China
| | - Yangyang Wang
- Chongqing Key Laboratory of Neurodegenerative Diseases, Yuzhong District, Chongqing 400013, China
- Department of Neurology, Chongqing General Hospital, Yuzhong District, Chongqing 400013, China
| | - Chuanling Wang
- Chongqing Key Laboratory of Neurodegenerative Diseases, Yuzhong District, Chongqing 400013, China
- Department of Neurology, Chongqing General Hospital, Yuzhong District, Chongqing 400013, China
- Chongqing Medical University, Yuzhong District, Chongqing 400016, China
| | - Minghao Yuan
- Chongqing Key Laboratory of Neurodegenerative Diseases, Yuzhong District, Chongqing 400013, China
- Department of Neurology, Chongqing General Hospital, Yuzhong District, Chongqing 400013, China
- Chongqing Medical University, Yuzhong District, Chongqing 400016, China
| | - Fei Chen
- Chongqing Key Laboratory of Neurodegenerative Diseases, Yuzhong District, Chongqing 400013, China
- Department of Neurology, Chongqing General Hospital, Yuzhong District, Chongqing 400013, China
- Chongqing Medical University, Yuzhong District, Chongqing 400016, China
| | - Qian Zou
- Chongqing Key Laboratory of Neurodegenerative Diseases, Yuzhong District, Chongqing 400013, China
- Department of Neurology, Chongqing General Hospital, Yuzhong District, Chongqing 400013, China
| | - Zhiyou Cai
- Chongqing Key Laboratory of Neurodegenerative Diseases, Yuzhong District, Chongqing 400013, China
- Department of Neurology, Chongqing General Hospital, Yuzhong District, Chongqing 400013, China
| | - Bin Zhao
- Department and Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong 524001, China
- Guangdong Key Laboratory of Aging-Related Cardiac and Cerebral Diseases, Zhanjiang, Guangdong 524001, China
| |
Collapse
|
5
|
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
|
6
|
Amelianchik A, Sweetland-Martin L, Norris EH. The effect of dietary fat consumption on Alzheimer's disease pathogenesis in mouse models. Transl Psychiatry 2022; 12:293. [PMID: 35869065 PMCID: PMC9307654 DOI: 10.1038/s41398-022-02067-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) is a fatal cognitive disorder with proteinaceous brain deposits, neuroinflammation, cerebrovascular dysfunction, and extensive neuronal loss over time. AD is a multifactorial disease, and lifestyle factors, including diet, are likely associated with the development of AD pathology. Since obesity and diabetes are recognized as risk factors for AD, it might be predicted that a high-fat diet (HFD) would worsen AD pathology. However, modeling HFD-induced obesity in AD animal models has yielded inconclusive results. Some studies report a deleterious effect of HFD on Aβ accumulation, neuroinflammation, and cognitive function, while others report that HFD worsens memory without affecting AD brain pathology. Moreover, several studies report no major effect of HFD on AD-related phenotypes in mice, while other studies show that HFD might, in fact, be protective. The lack of a clear association between dietary fat consumption and AD-related pathology and cognitive function in AD mouse models might be explained by experimental variations, including AD mouse model, sex and age of the animals, composition of the HFD, and timeline of HFD consumption. In this review, we summarize recent studies that aimed at elucidating the effect of HFD-induced obesity on AD-related pathology in mice and provide an overview of the factors that may have contributed to the results reported in these studies. Based on the heterogeneity of these animal model studies and given that the human population itself is quite disparate, it is likely that people will benefit most from individualized nutritional plans based on their medical history and clinical profiles.
Collapse
Affiliation(s)
- Anna Amelianchik
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA
| | - Lauren Sweetland-Martin
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA.
| |
Collapse
|
7
|
Gauvrit T, Benderradji H, Buée L, Blum D, Vieau D. Early-Life Environment Influence on Late-Onset Alzheimer's Disease. Front Cell Dev Biol 2022; 10:834661. [PMID: 35252195 PMCID: PMC8891536 DOI: 10.3389/fcell.2022.834661] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/27/2022] [Indexed: 12/30/2022] Open
Abstract
With the expand of the population's average age, the incidence of neurodegenerative disorders has dramatically increased over the last decades. Alzheimer disease (AD) which is the most prevalent neurodegenerative disease is mostly sporadic and primarily characterized by cognitive deficits and neuropathological lesions such as amyloid -β (Aβ) plaques and neurofibrillary tangles composed of hyper- and/or abnormally phosphorylated Tau protein. AD is considered a complex disease that arises from the interaction between environmental and genetic factors, modulated by epigenetic mechanisms. Besides the well-described cognitive decline, AD patients also exhibit metabolic impairments. Metabolic and cognitive perturbations are indeed frequently observed in the Developmental Origin of Health and Diseases (DOHaD) field of research which proposes that environmental perturbations during the perinatal period determine the susceptibility to pathological conditions later in life. In this review, we explored the potential influence of early environmental exposure to risk factors (maternal stress, malnutrition, xenobiotics, chemical factors … ) and the involvement of epigenetic mechanisms on the programming of late-onset AD. Animal models indicate that offspring exposed to early-life stress during gestation and/or lactation increase both AD lesions, lead to defects in synaptic plasticity and finally to cognitive impairments. This long-lasting epigenetic programming could be modulated by factors such as nutriceuticals, epigenetic modifiers or psychosocial behaviour, offering thus future therapeutic opportunity to protect from AD development.
Collapse
Affiliation(s)
- Thibaut Gauvrit
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Hamza Benderradji
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Luc Buée
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Didier Vieau
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| |
Collapse
|
8
|
Gestational high fat diet protects 3xTg offspring from memory impairments, synaptic dysfunction, and brain pathology. Mol Psychiatry 2021; 26:7006-7019. [PMID: 31451749 PMCID: PMC7044032 DOI: 10.1038/s41380-019-0489-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/29/2019] [Accepted: 05/24/2019] [Indexed: 01/25/2023]
Abstract
Maternal history for sporadic Alzheimer's disease (AD) predisposes the offspring to the disease later in life. However, the mechanisms behind this phenomenon are still unknown. Lifestyle and nutrition can directly modulate susceptibility to AD. Herein we investigated whether gestational high fat diet influences the offspring susceptibility to AD later in life. Triple transgenic dams were administered high fat diet or regular chow throughout 3 weeks gestation. Offspring were fed regular chow throughout their life and tested for spatial learning and memory, brain amyloidosis, tau pathology, and synaptic function. Gestational high fat diet attenuated memory decline, synaptic dysfunction, amyloid-β and tau neuropathology in the offspring by transcriptional regulation of BACE-1, CDK5, and tau gene expression via the upregulation of FOXP2 repressor. Gestational high fat diet protects offspring against the development of the AD phenotype. In utero dietary intervention could be implemented as preventative strategy against AD.
Collapse
|
9
|
Peleg-Raibstein D. Understanding the Link Between Maternal Overnutrition, Cardio-Metabolic Dysfunction and Cognitive Aging. Front Neurosci 2021; 15:645569. [PMID: 33716660 PMCID: PMC7953988 DOI: 10.3389/fnins.2021.645569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/10/2021] [Indexed: 12/16/2022] Open
Abstract
Obesity has long been identified as a global epidemic with major health implications such as diabetes and cardiovascular disease. Maternal overnutrition leads to significant health issues in industrial countries and is one of the risk factors for the development of obesity and related disorders in the progeny. The wide accessibility of junk food in recent years is one of the major causes of obesity, as it is low in nutrient content and usually high in salt, sugar, fat, and calories. An excess of nutrients during fetal life not only has immediate effects on the fetus, including increased growth and fat deposition in utero, but also has long-term health consequences. Based on human studies, it is difficult to discern between genetic and environmental contributions to the risk of disease in future generations. Consequently, animal models are essential for studying the impact of maternal overnutrition on the developing offspring. Recently, animal models provided some insight into the physiological mechanisms that underlie developmental programming. Most of the studies employed thus far have focused only on obesity and metabolic dysfunctions in the offspring. These studies have advanced our understanding of how maternal overnutrition in the form of high-fat diet exposure can lead to an increased risk of obesity in the offspring, but many questions remain open. How maternal overnutrition may increase the risk of developing brain pathology such as cognitive disabilities in the offspring and increase the risk to develop metabolic disorders later in life? Further, does maternal overnutrition exacerbate cognitive- and cardio-metabolic aging in the offspring?
Collapse
Affiliation(s)
- Daria Peleg-Raibstein
- Laboratory of Neurobehavioural Dynamics, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
10
|
Alrafiah A. Thymoquinone Protects Neurons in the Cerebellum of Rats through Mitigating Oxidative Stress and Inflammation Following High-Fat Diet Supplementation. Biomolecules 2021; 11:165. [PMID: 33513819 PMCID: PMC7911218 DOI: 10.3390/biom11020165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/21/2021] [Accepted: 01/24/2021] [Indexed: 12/30/2022] Open
Abstract
High-fat diet (HFD) is a major problem causing neuronal damage. Thymoquinone (TQ) could regulate oxidative stress and the inflammatory process. Hence, the present study elucidated the significant role of TQ on oxidative stress, inflammation, as well as morphological changes in the cerebellum of rats with HFD. Rats were divided into three groups as (1) control, (2) saturated HFD for eight weeks and (3) HFD supplementation (four weeks) followed by TQ 300 mg/kg/day treated (four weeks). After treatment, blood samples were collected to measure oxidative stress markers glutathione (GSH), malondialdehyde (MDA), superoxide dismutase (SOD), and inflammatory cytokines. Furthermore, neuronal morphological changes were also observed in the cerebellum of the rats. HFD rats show higher body weight (286.5 ± 7.4 g) as compared with the control group (224.67 ± 1.78 g). TQ treatment significantly (p < 0.05) lowered the body weight (225.83 ± 13.15 g). TQ produced a significant (p < 0.05) reduction in cholesterol, triglycerides, high-density lipoprotein (HDL), and low-density lipoprotein (LDL). The antioxidative enzymes significantly reduced in HFD rats (GSH, 1.46 ± 0.36 mol/L and SOD, 99.13 ± 5.41 µmol/mL) as compared with the control group (GSH, 6.25 ± 0.36 mol/L and SOD, 159.67 ± 10.67 µmol/mL). MDA was increased significantly in HFD rats (2.05 ± 0.25 nmol/L) compared to the control group (0.695 ± 0.11 nmol/L). Surprisingly, treatment with TQ could improve the level of GSH, MDA, and SOD. TQ treatment significantly (p < 0.05) reduced the inflammatory markers as compared with HFD alone. TQ treatment minimizes neuronal damage as well as reduces inflammation and improves antioxidant enzymes. TQ can be considered as a promising agent in preventing the neuronal morphological changes in the cerebellum of obese populations.
Collapse
Affiliation(s)
- Aziza Alrafiah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
11
|
Zhang X, de Oliveira Andrade F, Zhang H, Cruz I, Clarke R, Gaur P, Verma V, Hilakivi-Clarke L. Maternal obesity increases offspring's mammary cancer recurrence and impairs tumor immune response. Endocr Relat Cancer 2020; 27:469-482. [PMID: 32580156 PMCID: PMC7424355 DOI: 10.1530/erc-20-0065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
Over 50% of women at a childbearing age in the United States are overweight or obese, and this can adversely affect their offspring. We studied if maternal obesity-inducing high fat diet (HFD) not only increases offspring's mammary cancer risk but also impairs response to antiestrogen tamoxifen. Female rat offspring of HFD and control diet-fed dams, in which estrogen receptor-positive (ER+) mammary tumors were induced with the carcinogen 7,12-dimethylbenz[a]anthracene (DMBA), exhibited similar initial responses to antiestrogen tamoxifen. However, after tamoxifen therapy was completed, almost all (91%) tumors recurred in HFD offspring, compared with only 29% in control offspring. The increase in local mammary tumor recurrence in HFD offspring was linked to an increase in the markers of immunosuppression (Il17f, Tgfβ1, VEGFR2) in the tumor microenvironment (TME). Protein and mRNA levels of the major histocompatibility complex II (MHC-II), but not MHC-I, were reduced in the recurring DMBA tumors of HFD offspring. Further, infiltration of CD8+ effector T cells and granzyme B+ (GZMB+) cells were lower in their recurring tumors. To determine if maternal HFD can pre-program similar changes in the TME of allografted E0771 mammary tumors in offspring of syngeneic mice, flow cytometry analysis was performed. E0771 mammary tumor growth was significantly accelerated in the HFD offspring, and a reduction in the numbers of GZMB and non-significant reduction of interferon γ (IFNγ) secreting CD8+ T cells in the TME was seen. Thus, consumption of a HFD during pregnancy increases susceptibility of the female rat and mouse offspring to tumor immune suppression and mammary tumor growth and recurrence.
Collapse
Affiliation(s)
| | | | | | | | - Robert Clarke
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Pankaj Gaur
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Vivek Verma
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Leena Hilakivi-Clarke
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
- Correspondence should be addressed to L Hilakivi-Clarke:
| |
Collapse
|
12
|
Di Meco A, Praticò D. Early-life exposure to high-fat diet influences brain health in aging mice. Aging Cell 2019; 18:e13040. [PMID: 31560166 PMCID: PMC6826162 DOI: 10.1111/acel.13040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/11/2019] [Accepted: 08/28/2019] [Indexed: 12/16/2022] Open
Abstract
Epidemiological studies have suggested a link between exposure to environmental factors early in life and susceptibility to neurodegenerative diseases in adulthood. In the short term, maternal diet is important for the growth and development of the fetus; however, it may also have long-term effects on the health status of the offspring. Here, we investigate the effect that maternal high-fat diet during gestation has on brain health of the offspring later in life. B6129SF2/J dams were fed a high-fat diet during the 3 weeks' gestation, then switched to standard chow diet after delivery. Offspring were always fed regular diet for the entire study and assessed in learning, memory, and brain pathology when 18 months old. Compared with offspring from control mothers, the ones from mothers exposed to high-fat diet had significant better performance in learning and memory tests, which associated with an amelioration of synaptic integrity. Additionally, they had a significant reduction in total tau, a decrease in its pathological conformational changes and lower levels of caspase-3-cleaved isoforms. Our findings demonstrate that in utero exposure to high-fat diet plays a protective role for offspring brain health later in life. They support the novel hypothesis that targeted dietary intervention specifically restricted to the gestation period could be implemented as preventative strategy for the age-dependent decline in brain health.
Collapse
Affiliation(s)
- Antonio Di Meco
- Alzheimer’s Center at TempleLewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
| | - Domenico Praticò
- Alzheimer’s Center at TempleLewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
| |
Collapse
|
13
|
Contu L, Nizari S, Heath CJ, Hawkes CA. Pre- and Post-natal High Fat Feeding Differentially Affects the Structure and Integrity of the Neurovascular Unit of 16-Month Old Male and Female Mice. Front Neurosci 2019; 13:1045. [PMID: 31632236 PMCID: PMC6783577 DOI: 10.3389/fnins.2019.01045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/17/2019] [Indexed: 01/20/2023] Open
Abstract
Compelling experimental and clinical evidence supports a role for maternal obesity in offspring health. Adult children of obese mothers are at greater risk of obesity, diabetes, coronary heart disease and stroke. These offspring may also be at greater risk of age-related neurodegenerative diseases for which mid-life obesity is a risk factor. Rodent diet-induced obesity models have shown that high fat (HF) diet consumption damages the integrity of the blood–brain barrier (BBB) in the adult brain. However, there is currently little information about the effect of chronic HF feeding on the BBB of aged animals. Moreover, the long-term consequences of maternal obesity on the cerebrovasculature of aged offspring are not known. This study determined the impact of pre- and post-natal HF diet on the structure and integrity of cerebral blood vessels in aged male and female mice. Female C57Bl/6 mice were fed either a 10% fat control (C) or 45% HF diet before mating and during gestation and lactation. At weaning, male and female offspring were fed the C or HF diet until sacrifice at 16-months of age. Both dams and offspring fed the HF diet weighed significantly more than mice fed the C diet. Post-natal HF diet exposure increased hippocampal BBB leakiness in female offspring, in association with loss of astrocyte endfoot coverage of arteries. Markers of tight junctions, pericytes or smooth muscle cells were not altered by pre- or post-natal HF diet. Male offspring born to HF-fed mothers showed decreased parenchymal GFAP expression compared to offspring of mothers fed C diet, while microglial and macrophage markers were higher in the same female diet group. In addition, female offspring exposed to the HF diet for their entire lifespan showed more significant changes in vessel structure, BBB permeability and inflammation compared to male animals. These results suggest that the long-term impact of prenatal HF diet on the integrity of cerebral blood vessels differs between male and female offspring depending on the post-natal diet. This may have implications for the prevention and management of age- and obesity-related cerebrovascular diseases that differentially affect men and women.
Collapse
Affiliation(s)
- Laura Contu
- School of Life, Health and Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| | - Shereen Nizari
- School of Life, Health and Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| | - Christopher J Heath
- School of Life, Health and Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| | - Cheryl A Hawkes
- School of Life, Health and Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| |
Collapse
|
14
|
Rollins CPE, Gallino D, Kong V, Ayranci G, Devenyi GA, Germann J, Chakravarty MM. Contributions of a high-fat diet to Alzheimer's disease-related decline: A longitudinal behavioural and structural neuroimaging study in mouse models. NEUROIMAGE-CLINICAL 2018; 21:101606. [PMID: 30503215 PMCID: PMC6413478 DOI: 10.1016/j.nicl.2018.11.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/26/2018] [Accepted: 11/18/2018] [Indexed: 11/28/2022]
Abstract
Obesity is recognized as a significant risk factor for Alzheimer's disease (AD). Studies have supported that obesity accelerates AD-related pathophysiology and memory impairment in mouse models of AD. However, the nature of the brain structure-behaviour relationship mediating this acceleration remains unclear. In this manuscript we evaluated the impact of adolescent obesity on the brain morphology of the triple transgenic mouse model of AD (3xTg) and a non-transgenic control model of the same background strain (B6129s) using longitudinally acquired structural magnetic resonance imaging (MRI). At 8 weeks of age, animals were placed on a high-fat diet (HFD) or an ingredient-equivalent control diet (CD). Structural images were acquired at 8, 16, and 24 weeks. At 25 weeks, animals underwent the novel object recognition (NOR) task and the Morris water maze (MWM) to assess short-term non-associative memory and spatial memory, respectively. All analyses were carried out across four groups: B6129s-CD and -HFD and 3xTg-CD and -HFD. Neuroanatomical changes in MRI-derived brain morphology were assessed using volumetric and deformation-based analyses. HFD-induced obesity during adolescence exacerbated brain volume alterations by adult life in the 3xTg mouse model in comparison to control-fed mice and mediated volumetric alterations of select brain regions, such as the hippocampus. Further, HFD-induced obesity aggravated memory in all mice, lowering certain memory measures of B6129s control mice to the level of 3xTg mice maintained on a CD. Moreover, decline in the volumetric trajectories of hippocampal regions for all mice were associated with the degree of spatial memory impairments on the MWM. Our results suggest that obesity may interact with the brain changes associated with AD-related pathology in the 3xTg mouse model to aggravate brain atrophy and memory impairments and similarly impair brain structural integrity and memory capacity of non-transgenic mice. Further insight into this process may have significant implications in the development of lifestyle interventions for treatment of AD. Adolescent high-fat diet-induced obesity altered adult brain morphology and memory-related behaviours in a mouse model of AD High-fat feeding exacerbated brain volume changes in a mouse model of AD High-fat feeding mediated volumetric alterations of select brain regions, such as the hippocampus Degree of impairment on a spatial memory task showed linear trends with brain structural changes in AD-related regions High-fat feeding lowered certain memory measures of non-transgenic control mice
Collapse
Affiliation(s)
- Colleen P E Rollins
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada; Department of Psychiatry, University of Cambridge, Herchel Smith Building, Cambridge CB2 0SP, United Kingdom.
| | - Daniel Gallino
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - Vincent Kong
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Gülebru Ayranci
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - Gabriel A Devenyi
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Jürgen Germann
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 2B4, Canada; Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada.
| |
Collapse
|
15
|
Huber CM, Yee C, May T, Dhanala A, Mitchell CS. Cognitive Decline in Preclinical Alzheimer's Disease: Amyloid-Beta versus Tauopathy. J Alzheimers Dis 2018; 61:265-281. [PMID: 29154274 PMCID: PMC5734131 DOI: 10.3233/jad-170490] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We perform a large-scale meta-analysis of 51 peer-reviewed 3xTg-AD mouse publications to compare Alzheimer’s disease (AD) quantitative clinical outcome measures, including amyloid-β (Aβ), total tau, and phosphorylated tau (pTau), with cognitive performance in Morris water maze (MWM) and Novel Object Recognition (NOR). “High” levels of Aβ (Aβ40, Aβ42) showed significant but weak trends with cognitive decline (MWM: slope = 0.336, R2 = 0.149, n = 259, p < 0.001; NOR: slope = 0.156, R2 = 0.064, n = 116, p < 0.05); only soluble Aβ or directly measured Aβ meaningfully contribute. Tau expression in 3xTg-AD mice was within 10–20% of wild type and not associated with cognitive decline. In contrast, increased pTau is directly and significantly correlated with cognitive decline in MWM (slope = 0.408, R2 = 0.275, n = 371, p < < 0.01) and NOR (slope = 0.319, R2 = 0.176, n = 113, p < 0.05). While a variety of pTau epitopes (AT8, AT270, AT180, PHF-1) were examined, AT8 correlated most strongly with cognition (slope = 0.586, R2 = 0.521, n = 185, p < < 0.001). Multiple linear regression confirmed pTau is a stronger predictor of MWM performance than Aβ. Despite pTau’s lower physical concentration than Aβ, pTau levels more directly and quantitatively correlate with 3xTg-AD cognitive decline. pTau’s contribution to neurofibrillary tangles well after Aβ levels plateau makes pTau a viable treatment target even in late-stage clinical AD. Principal component analysis, which included hyperphosphorylation induced by kinases (pGSK3β, GSK3β, CDK5), identified phosphorylated ser9 GSK3β as the primary contributor to MWM variance. In summary, meta-analysis of cognitive decline in preclinical AD finds tauopathy more impactful than Aβ. Nonetheless, complex AD interactions dictate successful therapeutics harness synergy between Aβ and pTau, possibly through the GSK3 pathway.
Collapse
Affiliation(s)
- Colin M Huber
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA.,Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Sciences, Philadelphia, PA, USA
| | - Connor Yee
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Taylor May
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Apoorva Dhanala
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Cassie S Mitchell
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
16
|
Zhu C, Han TL, Zhao Y, Zhou X, Mao X, Qi H, Baker PN, Zhang H. A mouse model of pre-pregnancy maternal obesity combined with offspring exposure to a high-fat diet resulted in cognitive impairment in male offspring. Exp Cell Res 2018; 368:159-166. [PMID: 29698637 DOI: 10.1016/j.yexcr.2018.04.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Cognitive impairment is a brain dysfunction characterized by neuropsychological deficits in attention, working memory, and executive function. Maternal obesity and consumption of a high-fat diet (HFD) in the offspring has been suggested to have detrimental consequences for offspring cognitive function through its effect on the hippocampus and prefrontal cortex. Therefore, our study aimed to investigate the effects of maternal obesity and offspring HFD exposure on the brain metabolome of the offspring. METHODS In our pilot study, a LepRdb/+ mouse model was used to model pre-pregnancy maternal obesity and the c57bl/6 wildtype was used as a control group. Offspring were fed either a HFD or a low-fat control diet (LFD) after weaning (between 8 and 10 weeks). The Mirrors water maze was performed between 28 and 30 weeks to measure cognitive function. Fatty acid metabolomic profiles of the prefrontal cortex and hippocampus from the offspring at 30-32 weeks were analyzed using gas chromatography-mass spectrometry. RESULTS The memory of male offspring from obese maternal mice, consuming a HFD post-weaning, was significantly impaired when compared to the control offspring mice. No significant differences were observed in female offspring. In male mice, the fatty acid metabolites in the prefrontal cortex were most affected by maternal obesity, whereas, the fatty acid metabolites in the hippocampus were most affected by the offspring's diet. Hexadecanoic acid and octadecanoic acid were significantly affected in both the hippocampus and pre-frontal cortex, as a result of maternal obesity and a HFD in the offspring. CONCLUSION Our findings suggest that the combination of maternal obesity and HFD in the offspring can result in spatial cognitive deficiency in the male offspring, by influencing the fatty acid metabolite profiles in the prefrontal cortex and hippocampus. Further research is needed to validate the results of our pilot study.
Collapse
Affiliation(s)
- Chen Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Department of Obstetrics and Gynecology, Xin Qiao Hospital, The Second Medical College of Army Medical University, Chongqing, China
| | - Ting-Li Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Yalan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xiaobo Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xun Mao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Hongbo Qi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Philip N Baker
- Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Liggins Institute, University of Auckland, Auckland, New Zealand; College of Medicine, Biological Sciences and Psychology, University of Leicester, UK
| | - Hua Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Canada - China -New Zealand Joint Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
17
|
Effects of thymol on amyloid-β-induced impairments in hippocampal synaptic plasticity in rats fed a high-fat diet. Brain Res Bull 2018; 137:338-350. [PMID: 29339105 DOI: 10.1016/j.brainresbull.2018.01.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 01/01/2018] [Accepted: 01/08/2018] [Indexed: 12/11/2022]
Abstract
Obesity and a high-fat diet (HFD) are known to increase the incidence of Alzheimer's disease (AD). Oxidative stress, a major risk factor for AD, is increased with HFD consumption. Thymol (Thy) has antioxidant properties. Therefore, in the present study, we examined the protective and therapeutic effects of Thy on amyloid-β (Aβ)-induced impairments in the hippocampal synaptic plasticity of HFD-fed rats. In this study, 72 adult male Wistar rats were randomly assigned to 9 groups (n = 8 rats/group): Group 1 (control; standard diet); Group 2: Control + phosphate-buffered saline (PBS) + Oil (Thy vehicle); Group 3 (HFD + PBS); Group 4: (HFD + Aβ); Group 5: Control + PBS + Thy; Group 6: (HFD + Aβ + Oil); Group 7: Control + Aβ + Thy; Group 8: HFD + PBS + Thy; Group 9: (HFD + Aβ + Thy). After stereotaxic surgery, the field potentials were recorded after the implantation of the recording and stimulating electrodes in the dentate gyrus (DG) and perforant pathway, respectively. Following high-frequency stimulation, the long-term potentiation (LTP) of the population spike (PS) amplitude and the slope of the excitatory postsynaptic potentials (EPSPs) were measured in the DG. The HFD rats that received Aβ exhibited a significant decrease in their EPSP slope and PS amplitude as compared to the control group. In contrast, Thy administration in the HFD + Aβ rats reduced the decrease in the EPSP slope and PS amplitude. Thy decreased the Aβ-induced LTP impairments in HFD rats. The HFD significantly increased serum malondialdehyde levels and decreased total antioxidant capacity and total glutathione levels; whereas, Thy supplementation significantly reversed these parameters. Therefore, these results suggest that Thy, a natural antioxidant, can be therapeutic against high risk factors for AD, such as HFD.
Collapse
|
18
|
Kerdiles O, Layé S, Calon F. Omega-3 polyunsaturated fatty acids and brain health: Preclinical evidence for the prevention of neurodegenerative diseases. Trends Food Sci Technol 2017. [DOI: 10.1016/j.tifs.2017.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
19
|
Impact of enriched environment on production of tau, amyloid precursor protein and, amyloid-β peptide in high-fat and high-sucrose-fed rats. Acta Neuropsychiatr 2017; 29:291-298. [PMID: 27923413 DOI: 10.1017/neu.2016.63] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The Western-type diet is associated with an elevated risk of Alzheimer's disease and other milder forms of cognitive impairment. The aim of the present study was to investigate the effects of the environmental enrichment on amyloid and tau pathology in high-fat and high-sucrose-fed rats. METHODS In total, 40 adult male rats were categorised into two main groups according to their housing conditions: enriched environment (EE, n=16) and standard housing condition (n=24). The groups were further divided into five subgroups that received standard diet, high-fat diet, and high-sucrose diet. We performed the analysis of amyloid β-peptide (Aβ) (1-40), Aβ(1-42), amyloid precursor protein (APP), and tau levels in the hippocampus of rats that were maintained under standard housing conditions or exposed to an EE. RESULTS The EE decreased the Aβ(1-40), Aβ(1-42), APP, and tau levels in high-fat and high-sucrose-fed rats. CONCLUSION This observation shows that EE may rescue diet-induced amyloid and tau pathology.
Collapse
|
20
|
Sousa RALDE, Torres YS, Figueiredo CP, Passos GF, Clarke JR. Consequences of gestational diabetes to the brain and behavior of the offspring. AN ACAD BRAS CIENC 2017; 90:2279-2291. [PMID: 28813108 DOI: 10.1590/0001-3765201720170264] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 05/30/2017] [Indexed: 01/11/2023] Open
Abstract
Gestational diabetes mellitus (GD) is a form of insulin resistance triggered during the second/third trimesters of pregnancy in previously normoglycemic women. It is currently estimated that 10% of all pregnancies in the United States show this condition. For many years, the transient nature of GD has led researchers and physicians to assume that long-term consequences were absent. However, GD diagnosis leads to a six-fold increase in the risk of developing type 2 diabetes (T2D) in women and incidence of obesity and T2D is also higher among their infants. Recent and concerning evidences point to detrimental effects of GD on the behavior and cognition of the offspring, which often persist until adolescence or adulthood. Considering that the perinatal period is critical for determination of adult behavior, it is expected that the intra-uterine exposure to hyperglycemia, hyperinsulinemia and pro-inflammatory mediators, hallmark features of GD, might affect brain development. Here, we review early clinical and experimental evidence linking GD to consequences on the behavior of the offspring, focusing on memory and mood disorders. We also discuss initial evidence suggesting that downregulation of insulin signaling cascades are seen in the brains of GD offspring and could contribute to the consequences on their behavior.
Collapse
Affiliation(s)
- Ricardo A L DE Sousa
- School of Pharmacy, Carlos Chagas Filho Street, 373, Building A, Underground, Room 024, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Yasmin S Torres
- School of Pharmacy, Carlos Chagas Filho Street, 373, Building A, Underground, Room 024, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Claudia P Figueiredo
- School of Pharmacy, Carlos Chagas Filho Street, 373, Building A, Underground, Room 024, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Giselle F Passos
- School of Pharmacy, Carlos Chagas Filho Street, 373, Building A, Underground, Room 024, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Julia R Clarke
- School of Pharmacy, Carlos Chagas Filho Street, 373, Building A, Underground, Room 024, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
21
|
Intranasal insulin reverts central pathology and cognitive impairment in diabetic mother offspring. Mol Neurodegener 2017; 12:57. [PMID: 28768549 PMCID: PMC5541692 DOI: 10.1186/s13024-017-0198-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022] Open
Abstract
Background Adverse effects in diabetic mothers offspring (DMO) are a major concern of increasing incidence. Among these, chronic central complications in DMO remain poorly understood, and in extreme cases, diabetes can essentially function as a gestational brain insult. Nevertheless, therapeutic alternatives for DMO are limited. Methods Therefore, we have analyzed the central long-term complications in the offspring from CD1 diabetic mothers treated with streptozotozin, as well as the possible reversion of these alterations by insulin administration to neonates. Brain atrophy, neuronal morphology, tau phosphorylation, proliferation and neurogenesis were assessed in the short term (P7) and in the early adulthood (10 weeks) and cognitive function was also analyzed in the long-term. Results Central complications in DMO were still detected in the adulthood, including cortical and hippocampal thinning due to synaptic loss and neuronal simplification, increased tau hyperphosphorylation, and diminished cell proliferation and neurogenesis. Additionally, maternal diabetes increased the long-term susceptibility to spontaneous central bleeding, inflammation and cognition impairment in the offspring. On the other hand, intracerebroventricular insulin administration to neonates significantly reduced observed alterations. Moreover, non-invasive intranasal insulin reversed central atrophy and tau hyperphosphorylation, and rescued central proliferation and neurogenesis. Vascular damage, inflammation and cognitive alterations were also comparable to their counterparts born to nondiabetic mice, supporting the utility of this pathway to access the central nervous system. Conclusions Our data underlie the long-term effects of central complications in DMO. Moreover, observed improvement after insulin treatment opens the door to therapeutic alternatives for children who are exposed to poorly controlled gestational diabetes, and who may benefit from more individualized treatments.
Collapse
|
22
|
Asadbegi M, Yaghmaei P, Salehi I, Komaki A, Ebrahim-Habibi A. Investigation of thymol effect on learning and memory impairment induced by intrahippocampal injection of amyloid beta peptide in high fat diet- fed rats. Metab Brain Dis 2017; 32:827-839. [PMID: 28255862 DOI: 10.1007/s11011-017-9960-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/24/2017] [Indexed: 01/28/2023]
Abstract
Obesity and consumption of a high fat diet (HFD) are known to increase the risk of Alzheimer's disease (AD). In the present study, we have examined the protective and therapeutic effects of thymol (main monoterpene phenol found in thyme essential oil) on a HFD-fed rat model of AD. Fourty adult male Wistar rats were randomly assigned to 5 groups:(n = 8 rats/group): group 1, control, consumed an ordinary diet, group 2 consumed a HFD for 8 weeks, then received phosphate-buffered saline (PBS) via intrahippocampal (IHP) injection, group 3 consumed HFD for 8 weeks, then received beta-amyloid (Aβ)1-42 via IHP injections to induce AD, group 4 consumed HFD for 8 weeks, then received Aβ1-42, and was treated by thymol (30 mg/kg in sunflower oil) daily for 4 weeks, and group 5 consumed HFD for 8 week, then received Aβ1-42 after what sunflower oil was administered by oral gavage daily for 4 weeks. Biochemical tests showed an impaired lipid profile and higher glucose levels upon consumption of HFD, which was ameliorated by thymol treatment. In behavioral results, spatial memory in group 3 was significantly impaired, but groups treated with thymol showed better spatial memory compared to group 3 (p ≤ 0.01). In histological results, formation of Aβ plaque in hippocampus of group 3 increased significantly compared to group 1 and group 2 (p ≤ 0.05), but group 4 showed decreased Aβ plaques compared to group 3 (p ≤ 0.01). In conclusion, thymol decreased the effects of Aβ on memory and could be considered as neuroprotective.
Collapse
Affiliation(s)
- Masoumeh Asadbegi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Parichehreh Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Iraj Salehi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Azadeh Ebrahim-Habibi
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411413137, Iran.
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
A Review of the Impact of Maternal Obesity on the Cognitive Function and Mental Health of the Offspring. Int J Mol Sci 2017; 18:ijms18051093. [PMID: 28534818 PMCID: PMC5455002 DOI: 10.3390/ijms18051093] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/03/2017] [Accepted: 05/16/2017] [Indexed: 12/15/2022] Open
Abstract
Globally, more than 20% of women of reproductive age are currently estimated to be obese. Children born to obese mothers are at higher risk of developing obesity, coronary heart disease, diabetes, stroke, and asthma in adulthood. Increasing clinical and experimental evidence suggests that maternal obesity also affects the health and function of the offspring brain across the lifespan. This review summarizes the current findings from human and animal studies that detail the impact of maternal obesity on aspects of learning, memory, motivation, affective disorders, attention-deficit hyperactivity disorder, autism spectrum disorders, and neurodegeneration in the offspring. Epigenetic mechanisms that may contribute to this mother–child interaction are also discussed.
Collapse
|
24
|
McKee SE, Grissom NM, Herdt CT, Reyes TM. Methyl donor supplementation alters cognitive performance and motivation in female offspring from high-fat diet-fed dams. FASEB J 2017; 31:2352-2363. [PMID: 28209774 DOI: 10.1096/fj.201601172r] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/30/2017] [Indexed: 12/18/2022]
Abstract
During gestation, fetal nutrition is entirely dependent on maternal diet. Maternal consumption of excess fat during pregnancy has been linked to an increased risk of neurologic disorders in offspring, including attention deficit/hyperactivity disorder, autism, and schizophrenia. In a mouse model, high-fat diet (HFD)-fed offspring have cognitive and executive function deficits as well as whole-genome DNA and promoter-specific hypomethylation in multiple brain regions. Dietary methyl donor supplementation during pregnancy or adulthood has been used to alter DNA methylation and behavior. Given that extensive brain development occurs during early postnatal life-particularly within the prefrontal cortex (PFC), a brain region critical for executive function-we examined whether early life methyl donor supplementation (e.g., during adolescence) could ameliorate executive function deficits observed in offspring that were exposed to maternal HFD. By using operant testing, progressive ratio, and the PFC-dependent 5-choice serial reaction timed task (5-CSRTT), we determined that F1 female offspring (B6D2F1/J) from HFD-fed dams have decreased motivation (decreased progressive ratio breakpoint) and require a longer stimulus length to complete the 5-CSRTT task successfully, whereas early life methyl donor supplementation increased motivation and shortened the minimum stimulus length required for a correct response in the 5-CSRTT. Of interest, we found that expression of 2 chemokines, CCL2 and CXCL10, correlated with the median stimulus length in the 5-CSRTT. Furthermore, we found that acute adult supplementation of methyl donors increased motivation in HFD-fed offspring and those who previously received supplementation with methyl donors. These data point to early life as a sensitive time during which dietary methyl donor supplementation can alter PFC-dependent cognitive behaviors.-McKee, S. E., Grissom, N. M., Herdt, C. T., Reyes, T. M. Methyl donor supplementation alters cognitive performance and motivation in female offspring from high-fat diet-fed dams.
Collapse
Affiliation(s)
- Sarah E McKee
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicola M Grissom
- Department of Psychology, College of Liberal Arts, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christopher T Herdt
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa M Reyes
- Department of Psychiatry and Behavioral Neurosciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
25
|
Increased Aβ pathology in aged Tg2576 mice born to mothers fed a high fat diet. Sci Rep 2016; 6:21981. [PMID: 26911528 PMCID: PMC4766411 DOI: 10.1038/srep21981] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/03/2016] [Indexed: 01/24/2023] Open
Abstract
Maternal obesity is associated with increased risk of developing diabetes, obesity and premature death in adult offspring. Mid-life diabetes, hypertension and hypercholesterolaemia are risk factors for the development of sporadic Alzheimer’s disease (AD). A key pathogenic feature of AD is the accumulation of β-amyloid (Aβ) in the brain. The purpose of this study was to investigate the effect of high fat diet feeding during early life on Aβ pathology in the Tg2576 mouse model of AD. Female mice were fed a standard (C) or high fat (HF) diet before mating and during gestation and lactation. At weaning, male offspring were fed a C diet. Significantly higher levels of guanidine-soluble Aβ and plaque loads were observed in the hippocampi of 11-month old Tg2576 mice born to mothers fed a HF diet. Changes in the extracellular matrix led to increased retention of Aβ within the parenchyma. These data support a role for maternal and gestational health on the health of the aged brain and pathologies associated with AD and may provide a novel target for both the prevention and treatment of AD.
Collapse
|
26
|
Neuroprotective effects of metformin against Aβ-mediated inhibition of long-term potentiation in rats fed a high-fat diet. Brain Res Bull 2016; 121:178-85. [PMID: 26861514 DOI: 10.1016/j.brainresbull.2016.02.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/31/2016] [Accepted: 02/04/2016] [Indexed: 11/24/2022]
Abstract
Metformin (Met) is used to treat neurodegenerative disorders such as Alzheimer's disease (AD). Conversely, high-fat diets (HFD) have been shown to increase AD risk. In this study, we investigated the neuroprotective effects of Met on β-amyloid (Aβ)-induced impairments in hippocampal synaptic plasticity in AD model rats that were fed a HFD. In this study, 32 adult male Wistar rats were randomly assigned to four groups: group I (control group, regular diet); group II (HFD+vehicle); group III (HFD+Aβ); or group IV (Met+HFD+Aβ). Rats fed a HFD were injected with Aβ to induce AD, allowed to recover, and treated with Met for 8 weeks. The rats were then anesthetized with intraperitoneal injections of urethane and placed in a stereotaxic apparatus for surgery, electrode implantation, and field potential recording. In vivo electrophysiological recordings were then performed to measure population spike (PS) amplitude and excitatory postsynaptic potential (EPSP) slope in the hippocampal dentate gyrus. Long-term potentiation (LTP) was induced by high-frequency stimulation of the perforant pathway. Blood samples were then collected to measure plasma levels of triglycerides, low-density lipoproteins, very low-density lipoprotein, and cholesterol. After induction of LTP, PS amplitude and EPSP slope were significantly decreased in Aβ-injected rats fed a HFD compared to vehicle-injected animals or untreated animals that were fed a normal diet. Met treatment of Aβ-injected rats significantly attenuated these decreases, suggesting that Met decreased the effects of Aβ on LTP. These findings suggest that Met treatment is neuroprotective against the detrimental effects of Aβ and HFDs on hippocampal synaptic plasticity.
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Evidence for the benefit of nutrition in Alzheimer's disease continues to accumulate. Many studies with individual vitamins or supplements show marginal, if any, benefit. However, new findings with combinatorial formulations demonstrate improvement in cognitive performance and behavioral difficulties that accompany Alzheimer's disease. Herein, we review some of the most recent clinical advances and summarize supportive preclinical studies. RECENT FINDINGS We present novel positive effects on Alzheimer's disease derived from diet, trace elements, vitamins and supplements. We discuss the inherent difficulty in conducting nutritional studies because of the variance in participants' nutritional history, versus pharmacological interventions in which participants are naive to the intervention. We examine the evidence that epigenetics play a role in Alzheimer's disease and how nutritional intervention can modify the key epigenetic events to maintain or improve cognitive performance. SUMMARY Overall consideration of the most recent collective evidence suggests that the optimal approach for Alzheimer's disease would seem to combine early, multicomponent nutritional approaches (a Mediterranean-style diet, multivitamins and key combinatorial supplements), along with lifestyle modifications such as social activity and mental and physical exercise, with ultimate addition of pharmacological agents when warranted.
Collapse
|