1
|
Amaral A, Lister J, Rueckemann J, Wojnarowicz M, McGaughy J, Mokler D, Galler J, Rosene D, Rushmore R. Prenatal protein malnutrition decreases neuron numbers in the parahippocampal region but not prefrontal cortex in adult rats. Nutr Neurosci 2025; 28:333-346. [PMID: 39088448 PMCID: PMC11788924 DOI: 10.1080/1028415x.2024.2371256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
OBJECTIVE Prenatal protein malnutrition produces anatomical and functional changes in the developing brain that persist despite immediate postnatal nutritional rehabilitation. Brain networks of prenatally malnourished animals show diminished activation of prefrontal areas and an increased activation of hippocampal regions during an attentional task [1]. While a reduction in cell number has been documented in hippocampal subfield CA1, nothing is known about changes in neuron numbers in the prefrontal or parahippocampal cortices. METHODS In the present study, we used unbiased stereology to investigate the effect of prenatal protein malnutrition on the neuron numbers in the medial prefrontal cortex and the cortices of the parahippocampal region that comprise the larger functional network. RESULTS Results show that prenatal protein malnutrition does not cause changes in the neuronal population in the medial prefrontal cortex of adult rats, indicating that the decrease in functional activation during attentional tasks is not due to a reduction in the number of neurons. Results also show that prenatal protein malnutrition is associated with a reduction in neuron numbers in specific parahippocampal subregions: the medial entorhinal cortex and presubiculum. DISCUSSION The affected regions along with CA1 comprise a tightly interconnected circuit, suggesting that prenatal malnutrition confers a vulnerability to specific hippocampal circuits. These findings are consistent with the idea that prenatal protein malnutrition produces a reorganization of structural and functional networks, which may underlie observed alterations in attentional processes and capabilities.
Collapse
Affiliation(s)
- A.C. Amaral
- Department of Anatomy & Neurobiology,
Boston University Chobanian & Avedisian School of Medicine, Boston, MA
02118
| | - J.P. Lister
- Department of Anatomy & Neurobiology,
Boston University Chobanian & Avedisian School of Medicine, Boston, MA
02118
- Department of Pathology and Laboratory Medicine, University
of California Los Angeles, Los Angeles, CA 90095
| | - J.W. Rueckemann
- Department of Physiology and Biophysics, University of
Washington, Seattle, WA 98195
| | - M.W. Wojnarowicz
- Department of Pathology & Laboratory
Medicine, Boston University Chobanian & Avedisian School of
Medicine, Boston, MA 02118
| | - J.A. McGaughy
- Dept of Psychology, University of New Hampshire, Durham, NH
03824
| | - D.J. Mokler
- Dept of Biomedical Sciences, University of New England,
Biddeford, ME 04005
| | - J.R. Galler
- Department of Psychiatry, Harvard Medical School, Boston,
MA 02120
- Department of Pediatrics & Division of Gastroenterology
and Nutrition, MassGeneral Hospital for Children, Boston, MA
| | - D.L. Rosene
- Department of Anatomy & Neurobiology,
Boston University Chobanian & Avedisian School of Medicine, Boston, MA
02118
| | - R.J. Rushmore
- Department of Anatomy & Neurobiology,
Boston University Chobanian & Avedisian School of Medicine, Boston, MA
02118
- Psychiatry Neuroimaging Laboratory,
Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA
02120
- Department of Psychiatry, Massachusetts General Hospital,
Boston, MA 02120
| |
Collapse
|
2
|
Ruiz-Martínez SM, Guzmán-Gerónimo RI, Alvarado-Olivarez M, Santiago-Roque I, Palma-Jacinto JA. Effect of Blackberry Juice Consumption by Pregnant Rats on Brain Length and Cell Density of Dentate Gyrus in Male Wistar Pups. J Med Food 2024; 27:901-904. [PMID: 37792428 DOI: 10.1089/jmf.2023.0056] [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] [Indexed: 10/05/2023] Open
Abstract
The aim of this work was to evaluate the effect of blackberry juice consumption during pregnancy on the length of the brain, as well as on the cell density of the dentate gyrus in Wistar rat pups. Pregnant rats were divided into three groups: control (C), fed with standard diet and water ad libitum; BJ1, which received blackberry juice containing polyphenols (7.8 mg/kg) and anthocyanins (1.9 mg/kg); and BJ2, receiving blackberry juice containing polyphenols (9.3 mg/kg) and anthocyanins (3.54 mg/kg). On postnatal day 0, pups per litter, body weight, and length were measured, and cells in the dentate gyrus of male pups were quantified. Maternal body weight and pups per litter were statistically equal across experimental groups during pregnancy. Pups in BJ1 and BJ2 groups showed an increase in body weight (20%) and length (5%) when comparing to controls. An increase in brain length was observed in BJ2 group (8%) as compared to the control. A significant increase in the number of cells/mm2 was observed in the dentate gyrus of the offspring in BJ1 (21.8%) and BJ2 (23.7%) groups when compared to the control group. Given the above, blackberry juice may be considered a potential functional food during pregnancy, while further research on prenatal and postnatal development must be done.
Collapse
Affiliation(s)
| | - Rosa Isela Guzmán-Gerónimo
- Laboratorio de Innovación y Desarrollo de Alimentos, Instituto de Ciencias Básicas, Universidad Veracruzana, Xalapa, Veracruz, México
| | - Mayvi Alvarado-Olivarez
- Laboratorio de Neurofisiología, Instituto de Neuroetología, Universidad Veracruzana, Xalapa, Veracruz, México
| | - Isela Santiago-Roque
- Laboratorio de Bioquímica y Neurotoxicología, Facultad de Bioanálisis, Universidad Veracruzana, Xalapa, Veracruz, México
| | - José Antonio Palma-Jacinto
- Laboratorio de Bioquímica y Neurotoxicología, Facultad de Bioanálisis, Universidad Veracruzana, Xalapa, Veracruz, México
| |
Collapse
|
3
|
Vancamp P, Frapin M, Parnet P, Amarger V. Unraveling the Molecular Mechanisms of the Neurodevelopmental Consequences of Fetal Protein Deficiency: Insights From Rodent Models and Public Health Implications. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100339. [PMID: 39040432 PMCID: PMC11262180 DOI: 10.1016/j.bpsgos.2024.100339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/03/2024] [Accepted: 05/19/2024] [Indexed: 07/24/2024] Open
Abstract
Fetal brain development requires increased maternal protein intake to ensure that offspring reach their optimal cognitive potential in infancy and adulthood. While protein deficiency remains a prevalent issue in developing countries, it is also reemerging in Western societies due to the growing adoption of plant-based diets, some of which are monotonous and may fail to provide sufficient amino acids crucial for the brain's critical developmental phase. Confounding variables in human nutritional research have impeded our understanding of the precise impact of protein deficiency on fetal neurodevelopment, as well as its implications for childhood neurocognitive performance. Moreover, it remains unclear whether such deficiency could predispose to mental health problems in adulthood, mirroring observations in individuals exposed to prenatal famine. In this review, we sought to evaluate mechanistic data derived from rodent models, placing special emphasis on the involvement of neuroendocrine axes, the influence of sex and timing, epigenetic modifications, and cellular metabolism. Despite notable progress, critical knowledge gaps remain, including understanding the long-term reversibility of effects due to fetal protein restriction and the interplay between genetic predisposition and environmental factors. Enhancing our understanding of the precise mechanisms that connect prenatal nutrition to brain development in future research endeavors can be significantly advanced by integrating multiomics approaches and utilizing additional alternative models such as nonhuman primates. Furthermore, it is crucial to investigate potential interventions aimed at alleviating adverse outcomes. Ultimately, this research has profound implications for guiding public health strategies aimed at raising awareness about the crucial role of optimal maternal nutrition in supporting fetal neurodevelopment.
Collapse
Affiliation(s)
- Pieter Vancamp
- Nantes Université, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, UMR1280, Physiopathologie des Adaptations Nutritionnelles, l'Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - Morgane Frapin
- Organismal and Evolutionary Biology Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Patricia Parnet
- Nantes Université, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, UMR1280, Physiopathologie des Adaptations Nutritionnelles, l'Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - Valérie Amarger
- Nantes Université, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, UMR1280, Physiopathologie des Adaptations Nutritionnelles, l'Institut des Maladies de l'Appareil Digestif, Nantes, France
| |
Collapse
|
4
|
Nemati SS, Sadeghi L, Dehghan G, Sheibani N. Lateralization of the hippocampus: A review of molecular, functional, and physiological properties in health and disease. Behav Brain Res 2023; 454:114657. [PMID: 37683813 DOI: 10.1016/j.bbr.2023.114657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
The hippocampus is a part of the brain's medial temporal lobe that is located under the cortex. It belongs to the limbic system and helps to collect and transfer information from short-term to long-term memory, as well as spatial orientation in each mammalian brain hemisphere. After more than two centuries of research in brain asymmetry, the hippocampus has attracted much attention in the study of brain lateralization. The hippocampus is very important in cognitive disorders, related to seizures and dementia, such as epilepsy and Alzheimer's disease. In addition, the motivation to study the hippocampus has increased significantly due to the asymmetry in the activity of the left and right hippocampi in healthy people, and its disruption during some neurological diseases. After a general review of the hippocampal structure and its importance in related diseases, the asymmetry in the brain with a focus on the hippocampus during the growth and maturation of healthy people, as well as the differences created in patients at the molecular, functional, and physiological levels are discussed. Most previous work indicates that the hippocampus is lateralized in healthy people. Also, lateralization at different levels remarkably changes in patients, and it appears that the most complex cognitive disorder is caused by a new dominant asymmetric system.
Collapse
Affiliation(s)
- Seyed Saman Nemati
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, 51666-16471 Tabriz, Iran
| | - Leila Sadeghi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, 51666-16471 Tabriz, Iran.
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, 51666-16471 Tabriz, Iran.
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
5
|
Different Brain Phenotypes in Magnetic Resonance Imaging of Healthy Children after Prenatal Insults. Diagnostics (Basel) 2022; 12:diagnostics12112748. [PMID: 36359591 PMCID: PMC9689447 DOI: 10.3390/diagnostics12112748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/29/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
In this study, we used magnetic resonance imaging (MRI) to identify the different brain phenotypes within apparently healthy children and to evaluate whether these phenotypes had different prenatal characteristics. We included 65 healthy children (mean age, 10 years old) with normal neurological examinations and without structural abnormalities. We performed cluster analyses to identify the different brain phenotypes in the brain MRI images. We performed descriptive analyses, including demographic and perinatal characteristics, to assess the differences between the clusters. We identified two clusters: Cluster 1, or the “small brain phenotype” (n = 44), which was characterized by a global reduction in the brain volumes, with smaller total intracranial volumes (1044.53 ± 68.37 vs. 1200.87 ± 65.92 cm3 (p < 0.001)), total grey-matter volumes (644.65 ± 38.85 vs. 746.79 ± 39.37 cm3 (p < 0.001)), and total white-matter volumes (383.68 ± 40.17 vs. 443.55 ± 36.27 cm3 (p < 0.001)), compared with Cluster 2, or the “normal brain phenotype” (n = 21). Moreover, almost all the brain areas had decreased volumes, except for the ventricles, caudate nuclei, and pallidum areas. The risk of belonging to “the small phenotype” was 82% if the child was preterm, 76% if he/she was born small for his/her gestational age and up to 80% if the mother smoked during the pregnancy. However, preterm birth appears to be the only substantially significant risk factor associated with decreased brain volumes.
Collapse
|
6
|
Fang Q, Liu J, Chen L, Chen Q, Wang Y, Li Z, Fu W, Liu Y. Taurine supplementation improves hippocampal metabolism in immature rats with intrauterine growth restriction (IUGR) through protecting neurons and reducing gliosis. Metab Brain Dis 2022; 37:2077-2088. [PMID: 35048325 DOI: 10.1007/s11011-021-00896-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/10/2021] [Indexed: 02/05/2023]
Abstract
Taurine as an essential amino acid in the brain could play an important role in protecting the fetal brain of intrauterine growth restriction (IUGR). The hippocampus with IUGR showed neural metabolic disorder and structure changed that affected memory and learning ability. This study was aimed to identify the effect of taurine supplementation on the metabolism alterations and cellular composition changes of the hippocampus in IUGR immature rats. Metabolite concentrations were determined by magnetic resonance spectroscopy (MRS) in the hippocampus of juvenile rats with IUGR following taurine supplementation with antenatal or postnatal supply. The composition of neural cells in the hippocampus was observed by immunohistochemical staining (IHC) and western blotting (WB). Antenatal taurine supplementation increased the ratios of N-acetylaspartate (NAA) /creatine (Cr) and glutamate (Glu) /Cr of the hippocampus in the IUGR immature rats, but reduced the ratios of choline (Cho) /Cr and myoinositol (mI) /Cr. At the same time, the protein expression of NeuN in the IUGR rats was increased through intrauterine taurine supplementation, and the GFAP expression was reduced. Especially the effect of antenatal taurine was better than postpartum. Furthermore, there existed a positive correlation between the NAA/Cr ratio and the NeuN protein expression (R = 0.496 p < 0.001 IHC; R = 0.568 p < 0.001 WB), the same results existed in the relationship between the mI/Cr ratio and the GFAP protein expression (R = 0.338 p = 0.019 IHC; R = 0.440 p = 0.002 WB). Prenatal taurine supplementation can better improve hippocampal neuronal metabolism by increasing NAA / Cr ratio related to the number of neurons and reducing Cho / Cr ratio related to the number of glial cells.
Collapse
Affiliation(s)
- Qiong Fang
- Department of Pediatrics, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Jing Liu
- Department of Neonatology and Neonatal Intensive Care Unit, Beijing Chaoyang District Maternal and Child Healthcare Hospital, No. 25 Huaweili, Chaoyang District, Beijing, 100101, China.
- Department of Pediatrics, The Second School of Clinical Medicine, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun district, Guangzhou, 510515, Guangdong Province, China.
| | - Lang Chen
- Department of Pediatrics, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Qiaobin Chen
- Department of Pediatrics, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Yan Wang
- Neonatal Intensive Care Unit of Taian City Central Hospital, Taian, 271000, Shandong, China
| | - Zuanfang Li
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350001, Fujian Province, China
| | - Wei Fu
- Department of Neonatology and Neonatal Intensive Care Unit, Beijing Chaoyang District Maternal and Child Healthcare Hospital, No. 25 Huaweili, Chaoyang District, Beijing, 100101, China
| | - Ying Liu
- Department of Neonatology and Neonatal Intensive Care Unit, Beijing Chaoyang District Maternal and Child Healthcare Hospital, No. 25 Huaweili, Chaoyang District, Beijing, 100101, China
| |
Collapse
|
7
|
Grigoletti-Lima GB, Lopes MG, Franco ATB, Damico AM, Boer PA, Rocha Gontijo JA. Severe Gestational Low-Protein Intake Impacts Hippocampal Cellularity, Tau, and Amyloid-β Levels, and Memory Performance in Male Adult Offspring: An Alzheimer-Simile Disease Model? J Alzheimers Dis Rep 2022; 6:17-30. [PMID: 35243209 PMCID: PMC8842744 DOI: 10.3233/adr-210297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 11/25/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Maternal undernutrition has been associated with psychiatric and neurological disorders characterized by learning and memory impairment. OBJECTIVE Considering the lack of evidence, we aimed to analyze the effects of gestational protein restriction on learning and memory function associated with hippocampal cell numbers and neurodegenerative protein content later in life. METHODS Experiments were conducted in gestational low- (LP, 6% casein) or regular-protein (NP, 17% casein) diet intake offspring. Behavioral tests, isolated hippocampal isotropic fractionator cell studies, immunoblotting, and survival lifetime were observed. RESULTS The birthweight of LP males is significantly reduced relative to NP male progeny, and hippocampal mass increased in 88-week-old LP compared to age-matched NP offspring. The results showed an increased proximity measure in 87-week-old LP compared to NP offspring. Also, LP rats exhibited anxiety-like behaviors compared to NP rats at 48 and 86-wk of life. The estimated neuron number was unaltered in LP rats; however, non-neuron cell numbers increased compared to NP progeny. Here, we showed unprecedented hippocampal deposition of brain-derived neurotrophic factor, amyloid-β peptide (Aβ), and tau protein in 88-week-old LP relative to age-matched NP offspring. CONCLUSION To date, no predicted studies showed changes in hippocampal morphological structure in maternal protein-restricted elderly offspring. The current data suggest that gestational protein restriction may accelerate hippocampal function loss, impacting learning/memory performance, and supposedly developing diseases similar to Alzheimer's disease (AD) in elderly offspring. Thus, we propose that maternal protein restriction could be an elegant and novel method for constructing an AD-like model in adult male offspring.
Collapse
Affiliation(s)
- Gabriel Boer Grigoletti-Lima
- Fetal Programming and Hydroelectrolyte MetabolismLaboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, Faculty of Medical Sciences at StateUniversity of Campinas, Campinas, SP, Brazil
| | - Marcelo Gustavo Lopes
- Fetal Programming and Hydroelectrolyte MetabolismLaboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, Faculty of Medical Sciences at StateUniversity of Campinas, Campinas, SP, Brazil
| | - Ana Tereza Barufi Franco
- Fetal Programming and Hydroelectrolyte MetabolismLaboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, Faculty of Medical Sciences at StateUniversity of Campinas, Campinas, SP, Brazil
| | - Aparecida Marcela Damico
- Fetal Programming and Hydroelectrolyte MetabolismLaboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, Faculty of Medical Sciences at StateUniversity of Campinas, Campinas, SP, Brazil
| | - Patrìcia Aline Boer
- Fetal Programming and Hydroelectrolyte MetabolismLaboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, Faculty of Medical Sciences at StateUniversity of Campinas, Campinas, SP, Brazil
| | - José Antonio Rocha Gontijo
- Fetal Programming and Hydroelectrolyte MetabolismLaboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, Faculty of Medical Sciences at StateUniversity of Campinas, Campinas, SP, Brazil
| |
Collapse
|
8
|
Petrozziello T, Amaral AC, Dujardin S, Farhan SMK, Chan J, Trombetta BA, Kivisäkk P, Mills AN, Bordt EA, Kim SE, Dooley PM, Commins C, Connors TR, Oakley DH, Ghosal A, Gomez-Isla T, Hyman BT, Arnold SE, Spires-Jones T, Cudkowicz ME, Berry JD, Sadri-Vakili G. Novel genetic variants in MAPT and alterations in tau phosphorylation in amyotrophic lateral sclerosis post-mortem motor cortex and cerebrospinal fluid. Brain Pathol 2021; 32:e13035. [PMID: 34779076 PMCID: PMC8877756 DOI: 10.1111/bpa.13035] [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: 06/14/2021] [Revised: 09/22/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Although the molecular mechanisms underlying amyotrophic lateral sclerosis (ALS) are not yet fully understood, several studies report alterations in tau phosphorylation in both sporadic and familial ALS. Recently, we have demonstrated that phosphorylated tau at S396 (pTau‐S396) is mislocalized to synapses in ALS motor cortex (mCTX) and contributes to mitochondrial dysfunction. Here, we demonstrate that while there was no overall increase in total tau, pTau‐S396, and pTau‐S404 in ALS post‐mortem mCTX, total tau and pTau‐S396 were increased in C9ORF72‐ALS. Additionally, there was a significant decrease in pTau‐T181 in ALS mCTX compared controls. Furthermore, we leveraged the ALS Knowledge Portal and Project MinE data sets and identified ALS‐specific genetic variants across MAPT, the gene encoding tau. Lastly, assessment of cerebrospinal fluid (CSF) samples revealed a significant increase in total tau levels in bulbar‐onset ALS together with a decrease in CSF pTau‐T181:tau ratio in all ALS samples, as reported previously. While increases in CSF tau levels correlated with a faster disease progression as measured by the revised ALS functional rating scale (ALSFRS‐R), decreases in CSF pTau‐T181:tau ratio correlated with a slower disease progression, suggesting that CSF total tau and pTau‐T181 ratio may serve as biomarkers of disease in ALS. Our findings highlight the potential role of pTau‐T181 in ALS, as decreases in CSF pTau‐T181:tau ratio may reflect the significant decrease in pTau‐T181 in post‐mortem mCTX. Taken together, these results indicate that tau phosphorylation is altered in ALS post‐mortem mCTX as well as in CSF and, importantly, the newly described pathogenic or likely pathogenic variants identified in MAPT in this study are adjacent to T181 and S396 phosphorylation sites further highlighting the potential role of these tau functional domains in ALS. Although the molecular mechanisms underlying amyotrophic lateral sclerosis (ALS) are not yet fully understood, recent studies report alterations in tau phosphorylation in ALS. Our study builds on these findings and demonstrates that tau phosphorylation is altered in post‐mortem ALS motor cortex and highlights new and ALS‐specific variants in MAPT, the gene encoding tau. Lastly, we report alterations in phosphorylated tau in ALS cerebrospinal fluid that may function as a predictive biomarker for ALS.![]()
Collapse
Affiliation(s)
- Tiziana Petrozziello
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ana C Amaral
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Simon Dujardin
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sali M K Farhan
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - James Chan
- Biostatistics Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bianca A Trombetta
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pia Kivisäkk
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandra N Mills
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evan A Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Spencer E Kim
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Patrick M Dooley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Caitlin Commins
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Theresa R Connors
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Derek H Oakley
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anubrata Ghosal
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Teresa Gomez-Isla
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven E Arnold
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tara Spires-Jones
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, University of Edinburgh, UK
| | - Merit E Cudkowicz
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James D Berry
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ghazaleh Sadri-Vakili
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Fesser EA, Gianatiempo O, Berardino BG, Alberca CD, Urrutia L, Falasco G, Sonzogni SV, Chertoff M, Cánepa ET. Impaired social cognition caused by perinatal protein malnutrition evokes neurodevelopmental disorder symptoms and is intergenerationally transmitted. Exp Neurol 2021; 347:113911. [PMID: 34767796 DOI: 10.1016/j.expneurol.2021.113911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/05/2021] [Accepted: 11/01/2021] [Indexed: 12/26/2022]
Abstract
Nutritional inadequacy before birth and during postnatal life can seriously interfere with brain development and lead to persistent deficits in learning and behavior. In this work, we asked if protein malnutrition affects domains of social cognition and if these phenotypes can be transmitted to the next generation. Female mice were fed with a normal or hypoproteic diet during pregnancy and lactation. After weaning, offspring were fed with a standard chow. Social interaction, social recognition memory, and dominance were evaluated in both sexes of F1 offspring and in the subsequent F2 generation. Glucose metabolism in the whole brain was analyzed through preclinical positron emission tomography. Genome-wide transcriptional analysis was performed in the medial prefrontal cortex followed by gene-ontology enrichment analysis. Compared with control animals, malnourished mice exhibited a deficit in social motivation and recognition memory and displayed a dominant phenotype. These altered behaviors, except for dominance, were transmitted to the next generation. Positron emission tomography analysis revealed lower glucose metabolism in the medial prefrontal cortex of F1 malnourished offspring. This brain region showed genome-wide transcriptional dysregulation, including 21 transcripts that overlapped with autism-associated genes. Our study cannot exclude that the lower maternal care provided by mothers exposed to a low-protein diet caused an additional impact on social cognition. Our results showed that maternal protein malnutrition dysregulates gene expression in the medial prefrontal cortex, promoting altered offspring behavior that was intergenerationally transmitted. These results support the hypothesis that early nutritional deficiency represents a risk factor for the emergence of symptoms associated with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Estefanía A Fesser
- Grupo Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, Ciudad de Buenos Aires, Argentina
| | - Octavio Gianatiempo
- Grupo Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, Ciudad de Buenos Aires, Argentina
| | - Bruno G Berardino
- Grupo Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, Ciudad de Buenos Aires, Argentina
| | - Carolina D Alberca
- Grupo Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, Ciudad de Buenos Aires, Argentina
| | - Leandro Urrutia
- Centro de Imágenes Moleculares, Fleni, Escobar, Buenos Aires, Argentina
| | - Germán Falasco
- Centro de Imágenes Moleculares, Fleni, Escobar, Buenos Aires, Argentina
| | - Silvina V Sonzogni
- Grupo Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, Ciudad de Buenos Aires, Argentina
| | - Mariela Chertoff
- Grupo Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, Ciudad de Buenos Aires, Argentina
| | - Eduardo T Cánepa
- Grupo Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
10
|
Intrauterine Growth Restriction Causes Abnormal Embryonic Dentate Gyrus Neurogenesis in Mouse Offspring That Leads to Adult Learning and Memory Deficits. eNeuro 2021; 8:ENEURO.0062-21.2021. [PMID: 34544755 PMCID: PMC8503959 DOI: 10.1523/eneuro.0062-21.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 11/25/2022] Open
Abstract
Human infants who suffer from intrauterine growth restriction (IUGR), which is a failure to attain their genetically predetermined weight, are at increased risk for postnatal learning and memory deficits. Hippocampal dentate gyrus (DG) granule neurons play an important role in memory formation; however, it is unknown whether IUGR affects embryonic DG neurogenesis, which could provide a potential mechanism underlying abnormal postnatal learning and memory function. Using a mouse model of the most common cause of IUGR, induced by hypertensive disease of pregnancy, we first assessed adult learning and memory function. We quantified the percentages of embryonic hippocampal DG neural stem cells (NSCs) and progenitor cells and developing glutamatergic granule neurons, as well as hippocampal volumes and neuron cell count and morphology 18 and 40 d after delivery. We characterized the differential embryonic hippocampal transcriptomic pathways between appropriately grown and IUGR mouse offspring. We found that IUGR offspring of both sexes had short-term adult learning and memory deficits. Prenatally, we found that IUGR caused accelerated embryonic DG neurogenesis and Sox2+ neural stem cell depletion. IUGR mice were marked by decreased hippocampal volumes and decreased doublecortin+ neuronal progenitors with increased mean dendritic lengths at postnatal day 18. Consistent with its known molecular role in embryonic DG neurogenesis, we also found evidence for decreased Wnt pathway activity during IUGR. In conclusion, we have discovered that postnatal memory deficits are associated with accelerated NSC differentiation and maturation into glutamatergic granule neurons following IUGR, a phenotype that could be explained by decreased embryonic Wnt signaling.
Collapse
|
11
|
Differential longitudinal changes of neuronal and glial damage markers in anorexia nervosa after partial weight restoration. Transl Psychiatry 2021; 11:86. [PMID: 33558486 PMCID: PMC7870648 DOI: 10.1038/s41398-021-01209-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022] Open
Abstract
Atrophic brain changes in acute anorexia nervosa (AN) are often visible to the naked eye on computed tomography or magnetic resonance imaging scans, but it remains unclear what is driving these effects. In neurological diseases, neurofilament light (NF-L) and tau protein have been linked to axonal damage. Glial fibrillary acidic protein (GFAP) has been associated with astroglial injury. In an attempt to shed new light on factors potentially underlying past findings of structural brain alterations in AN, the current study investigated serum NF-L, tau protein, and GFAP levels longitudinally in AN patients undergoing weight restoration. Blood samples were obtained from 54 acutely underweight, predominantly adolescent female AN patients and 54 age-matched healthy control participants. AN patients were studied in the severely underweight state and again after short-term partial weight restoration. Group comparisons revealed higher levels of NF-L, tau protein, and GFAP in acutely underweight patients with AN compared to healthy control participants. Longitudinally, a decrease in NF-L and GFAP but not in tau protein levels was observed in AN patients upon short-term partial weight restoration. These results may be indicative of ongoing neuronal and astroglial injury during the underweight phase of AN. Normalization of NF-L and GFAP but not tau protein levels may indicate an only partial restoration of neuronal and astroglial integrity upon weight gain after initial AN-associated cell damage processes.
Collapse
|
12
|
Rushmore RJ, McGaughy JA, Amaral AC, Mokler DJ, Morgane PJ, Galler JR, Rosene DL. The neural basis of attentional alterations in prenatally protein malnourished rats. Cereb Cortex 2021; 31:497-512. [PMID: 33099611 PMCID: PMC7947171 DOI: 10.1093/cercor/bhaa239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/29/2020] [Accepted: 08/01/2020] [Indexed: 11/13/2022] Open
Abstract
Protein malnutrition during gestation alters brain development and produces specific behavioral and cognitive changes that persist into adulthood and increase the risks of neuropsychiatric disorders. Given evidence for the role of the prefrontal cortex in such diseases, it is significant that studies in humans and animal models have shown that prenatal protein malnutrition specifically affects functions associated with prefrontal cortex. However, the neural basis underlying these changes is unclear. In the current study, prenatally malnourished and control rats performed a sustained attention task with an unpredictable distractor, a task that depends on intact prefrontal cortical function. Radiolabeled 2-deoxyglucose was used to measure neural and brain network activity during the task. Results confirmed that adult prenatally malnourished rats were more distractible than controls and exhibited lower functional activity in prefrontal cortices. Thus, prefrontal activity was a predictor of task performance in controls but not prenatally malnourished animals. Instead, prenatally malnourished animals relied on different brain networks involving limbic structures such as the hippocampus. These results provide evidence that protein reduction during brain development has more wide-reaching effects on brain networks than previously appreciated, resulting in the formation of brain networks that may reflect compensatory responses in prenatally malnourished brains.
Collapse
Affiliation(s)
- R J Rushmore
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston MA
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Boston, MA
- Department of Psychiatry, Harvard Medical School, Boston, MA
| | - J A McGaughy
- Department of Psychology, University of New Hampshire, Durham, NH
| | - A C Amaral
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston MA
| | - D J Mokler
- Department of Biomedical Sciences, University of New England, Biddeford ME
| | - P J Morgane
- Department of Biomedical Sciences, University of New England, Biddeford ME
| | - J R Galler
- Department of Psychiatry, Harvard Medical School, Boston, MA
- Division of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | - D L Rosene
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston MA
| |
Collapse
|
13
|
Rushmore RJ, McGaughy JA, Mokler DJ, Rosene DL. The enduring effect of prenatal protein malnutrition on brain anatomy, physiology and behavior. Nutr Neurosci 2020; 25:1392-1399. [PMID: 33314995 DOI: 10.1080/1028415x.2020.1859730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
There is increasing evidence that the maternal environment exerts enduring influences on the fetal brain. In response to certain environmental stimuli such as reduced protein content, the fetus changes the course of its brain development, which leads to specific and programed changes in brain anatomy and physiology. These alterations produce a brain with a fundamentally altered organization, which then translates to alterations in adult cognitive function. The effects on brain and behavior may be linked, such that a prenatal stimulus relays a signal to alter brain development and encourage the selection and development of brain circuits and behaviors that would be beneficial for the environment in which the animal was anticipated to emerge. At the same time, the signal would deselect behaviors unlikely to be adaptive. We draw on evidence from rodent models to suggest that the brain that develops after a reduction in protein during the prenatal phase is not uniformly dysfunctional, but simply different. This perspective has implications for the role of prenatal factors in the production and expression of behavior, and may account for the elevation of risk factors for neurological and psychiatric illnesses.
Collapse
Affiliation(s)
- R J Rushmore
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA.,Psychiatric Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA.,Center for Morphometric Analysis, Massachusetts General Hospital, Boston, MA, USA
| | - J A McGaughy
- Department of Psychology, University of New Hampshire, Durham, NH, USA
| | - D J Mokler
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, USA
| | - D L Rosene
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
14
|
Sarkar T, Patro N, Patro IK. Cumulative multiple early life hits- a potent threat leading to neurological disorders. Brain Res Bull 2019; 147:58-68. [DOI: 10.1016/j.brainresbull.2019.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/31/2019] [Accepted: 02/08/2019] [Indexed: 12/11/2022]
|
15
|
Perinatal Nutrition and Programmed Risk for Neuropsychiatric Disorders: A Focus on Animal Models. Biol Psychiatry 2019; 85:122-134. [PMID: 30293647 PMCID: PMC6309477 DOI: 10.1016/j.biopsych.2018.08.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 08/07/2018] [Accepted: 08/08/2018] [Indexed: 01/02/2023]
Abstract
Maternal nutrition is critically important for fetal development. Recent human studies demonstrate a strong connection between diet during pregnancy and offspring risk for neuropsychiatric disorders including depression, anxiety, and attention-deficit/hyperactivity disorder. Animal models have emerged as a crucial tool for understanding maternal nutrition's contribution to prenatal programming and the later development of neuropsychiatric disorders. This review highlights preclinical studies examining how maternal consumption of the three macronutrients (protein, fats, and carbohydrates) influence offspring negative-valence behaviors relevant to neuropsychiatric disorders. We highlight the translational aspects of animal models and so examine exposure periods that mirror the neurodevelopmental stages of human gestation. Because of our emphasis on programmed changes in neurobehavioral development, studies that continue diet exposure until assessment in adulthood are not discussed. The presented research provides a strong foundation of preclinical evidence of nutritional programming of neurobehavioral impairments. Alterations in risk assessment and response were observed alongside neurodevelopmental impairments related to neurogenesis, synaptogenesis, and synaptic plasticity. To date, the large majority of studies utilized rodent models, and the field could benefit from additional study of large-animal models. Additional future directions are discussed, including the need for further studies examining how sex as a biological variable affects the contribution of maternal nutrition to prenatal programming.
Collapse
|
16
|
Sinha S, Patro N, Patro IK. Maternal Protein Malnutrition: Current and Future Perspectives of Spirulina Supplementation in Neuroprotection. Front Neurosci 2018; 12:966. [PMID: 30618587 PMCID: PMC6305321 DOI: 10.3389/fnins.2018.00966] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/03/2018] [Indexed: 12/30/2022] Open
Abstract
Malnutrition has been widely recognized as a grave burden restricting the progress of underdeveloped and developing countries. Maternal, neonatal and postnatal nutritional immunity provides an effective approach to decrease the risk of malnutrition associated stress in adulthood. Particularly, maternal nutritional status is a critical contributor for determining the long-term health aspects of an offspring. Maternal malnutrition leads to increased risk of life, poor immune system, delayed motor development and cognitive dysfunction in the children. An effective immunomodulatory intervention using nutraceutical could be used to enhance immunity against infections. The immune system in early life possesses enormous dynamic capacity to manage both genetic and environment driven processes and can adapt to rapidly changing environmental exposures. These immunomodulatory stimuli or potent nutraceutical strategy can make use of early life plasticity to target pathways of immune ontogeny, which in turn could increase the immunity against infectious diseases arising from malnutrition. This review provides appreciable human and animal data showing enduring effects of protein deprivation on CNS development, oxidative stress and inflammation and associated behavioral and cognitive impairments. Relevant studies on nutritional supplementation and rehabilitation using Spirulina as a potent protein source and neuroprotectant against protein malnutrition (PMN) induced deleterious changes have also been discussed. However, there are many futuristic issues that need to be resolved for proper modulation of these therapeutic interventions to prevent malnutrition.
Collapse
Affiliation(s)
- Shrstha Sinha
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India.,School of Studies in Zoology, Jiwaji University, Gwalior, India
| | - Nisha Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India
| | - Ishan K Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India.,School of Studies in Zoology, Jiwaji University, Gwalior, India
| |
Collapse
|
17
|
Damodara Gowda KM, Suchetha Kumari N, Ullal H. Role of astaxanthin in the modulation of brain-derived neurotrophic factor and spatial learning behavior in perinatally undernourished Wistar rats. Nutr Neurosci 2018; 23:422-431. [PMID: 30200858 DOI: 10.1080/1028415x.2018.1515301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Objective: Maternal health and nutrition during the perinatal period is the predominant factor influencing the functional development of the brain. Maternal malnutrition during the perinatal period causes retardation of brain development. The current study investigates the role of Astaxanthin (AsX) in spatial learning and memory and BDNF in perinatally undernourished Wistar rats.Methods: The albino wistar rats were perinatally undernourished and administered with different dosages of AsX. The spatial learning and memory performance and BDNF level were assessed. Data were collected and analysed.Results: The % Correct choice during the acquisition phase, performance at the end of the acquisition phase and the mean BDNF level at the Hippocampus, Cerebellum, and Cerebral cortex showed significant decline (P<0.001) in the PUN group and significantly high (P<0.001) in the PUNA2 group compared to the control. However, the mean RME and mean WME during different days of the acquisition phase were significantly high (P<0.001) in the PUN group and insignificant (P>0.05) in PUNA2 compared to the control.Discussion: The results showed that AsX effectively modulated the cognitive deficit that occurred in perinatally undernourished rats. This can be attributed to BDNF upregulation as evidenced by the significant increase of the BDNF level.
Collapse
Key Words
- AsX: Astaxanthin, BDNF: Brain-Derived Neurotropic Factor, ELISA: Enzyme-Linked Immuno Sorbent Assay, FDA: Food and Drug Administration, NA1: Normal rats supplemented with AsX (6 mg/kg bw), NA2: Normal rats supplemented with AsX (12 mg/kg bw), PUN: Perinatally Undernourished, PUNA1: Perinatally Undernourished rats but supplemented with AsX (6 mg/kg bw), PUNA2: Perinatally Undernourished but supplemented with AsX (12 mg/kg bw), RME: Reference Memory Error, WME: Working Memory Error, RM-ANOVA: Repeated Measures of ANOVA
- Astaxanthin
- Brain-derived neurotrophic factor
- Perinatal undernutrition
- Reference memory error and working memory error
- Spatial learning
Collapse
Affiliation(s)
- K M Damodara Gowda
- Department of Physiology, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, Karnataka 575018, India
| | - N Suchetha Kumari
- Department of Biochemistry, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Harshini Ullal
- Central Research Laboratory, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, Karnataka 575018, India
| |
Collapse
|
18
|
Effects of early-life malnutrition on neurodevelopment and neuropsychiatric disorders and the potential mechanisms. Prog Neuropsychopharmacol Biol Psychiatry 2018; 83:64-75. [PMID: 29287829 DOI: 10.1016/j.pnpbp.2017.12.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 12/21/2017] [Accepted: 12/24/2017] [Indexed: 02/08/2023]
Abstract
Lines of evidence have demonstrated that early-life malnutrition is highly correlated with neurodevelopment and adulthood neuropsychiatric disorders, while some findings are conflicting with each other. In addition, the biological mechanisms are less investigated. We systematically reviewed the evidence linking early-life nutrition status with neurodevelopment and clinical observations in human and animal models. We summarized the effects of special nutritious on neuropsychiatric disorders and explored the underlying potential mechanisms. The further understanding of the biological regulation of early-life nutritional status on neurodevelopment might shed light on precision nutrition at an integrative systems biology framework.
Collapse
|
19
|
Reyes-Castro LA, Padilla-Gómez E, Parga-Martínez NJ, Castro-Rodríguez DC, Quirarte GL, Díaz-Cintra S, Nathanielsz PW, Zambrano E. Hippocampal mechanisms in impaired spatial learning and memory in male offspring of rats fed a low-protein isocaloric diet in pregnancy and/or lactation. Hippocampus 2017; 28:18-30. [PMID: 28843045 DOI: 10.1002/hipo.22798] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 08/15/2017] [Accepted: 08/22/2017] [Indexed: 01/08/2023]
Abstract
Maternal nutritional challenges during fetal and neonatal development result in developmental programming of multiple offspring organ systems including brain maturation and function. A maternal low-protein diet during pregnancy and lactation impairs associative learning and motivation. We evaluated effects of a maternal low-protein diet during gestation and/or lactation on male offspring spatial learning and hippocampal neural structure. Control mothers (C) ate 20% casein and restricted mothers (R) 10% casein, providing four groups: CC, RR, CR, and RC (first letter pregnancy, second lactation diet). We evaluated the behavior of young adult male offspring around postnatal day 110. Corticosterone and ACTH were measured. Males were tested for 2 days in the Morris water maze (MWM). Stratum lucidum mossy fiber (MF) area, total and spine type in basal dendrites of stratum oriens in the hippocampal CA3 field were measured. Corticosterone and ACTH were higher in RR vs. CC. In the MWM acquisition test CC offspring required two, RC three, and CR seven sessions to learn the maze. RR did not learn in eight trials. In a retention test 24 h later, RR, CR, and RC spent more time locating the platform and performed fewer target zone entries than CC. RR and RC offspring spent less time in the target zone than CC. MF area, total, and thin spines were lower in RR, CR, and RC than CC. Mushroom spines were lower in RR and RC than CC. Stubby spines were higher in RR, CR, and RC than CC. We conclude that maternal low-protein diet impairs spatial acquisition and memory retention in male offspring, and that alterations in hippocampal presynaptic (MF), postsynaptic (spines) elements and higher glucocorticoid levels are potential mechanisms to explain these learning and memory deficits.
Collapse
Affiliation(s)
- L A Reyes-Castro
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición SZ, México 14080, México
| | - E Padilla-Gómez
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro 76230, México
| | - N J Parga-Martínez
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro 76230, México
| | - D C Castro-Rodríguez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición SZ, México 14080, México
| | - G L Quirarte
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro 76230, México
| | - S Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro 76230, México
| | - P W Nathanielsz
- Department of Animal Science, University of Wyoming, Laramie, Wyoming 82071-3684
| | - E Zambrano
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición SZ, México 14080, México
| |
Collapse
|
20
|
Influence of catch up growth on spatial learning and memory in a mouse model of intrauterine growth restriction. PLoS One 2017; 12:e0177468. [PMID: 28542302 PMCID: PMC5443512 DOI: 10.1371/journal.pone.0177468] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 04/27/2017] [Indexed: 01/21/2023] Open
Abstract
Background Intrauterine growth restriction (IUGR) and rapid postnatal weight gain or catch up growth (CUG) increase the susceptibility to metabolic syndrome during adult life. Longitudinal studies have also revealed a high incidence of learning difficulties in children with IUGR. The aim of the present study was to investigate the effect of nutrition and CUG on learning memory in an IUGR animal model. We hypothesized that synaptic protein expression and transcription, an essential mechanism for memory consolidation, might be affected by intrauterine undernutrition. Methods IUGR was induced by 50% maternal caloric undernutrition throughout late gestation. During the suckling period, dams were either fed ad libitum or food restricted. The pups were divided into: Normal prenatal diet-Normal postnatal diet (NN), Restricted prenatal diet- Normal postnatal diet + catch up growth (RN+), Normal prenatal diet-Restricted postnatal diet (NR) and Restricted prenatal diet-Restricted postnatal diet (RR). At 4 weeks of age, memory was assessed via a water maze test. To evaluate synaptic function, 2 specific synaptic proteins (postsynaptic density-95 [PSD95], synaptophysin) as well as insulin receptors (IR) were tested by Western Blot and quantitative polymerase chain reaction (qPCR). Brain-derived neurotrophic factor and serum insulin levels were also studied. Results and conclusions The RN+ group presented a learning curve similar to the NN animals. The RR animals without CUG showed learning disabilities. PSD95 was lower in the RR group than in the NN and RN+ mice. In contrast, synaptophysin was similar in all groups. IR showed an inverse expression pattern to that of the PSD95. In conclusion, perinatal nutrition plays an important role in learning. CUG after a period of prenatal malnutrition seems to improve learning skills. The functional alterations observed might be related to lower PSD95 activity and a possible dysfunction in the hormone regulation of synaptic plasticity.
Collapse
|
21
|
Camprubí Camprubí M, Balada Caballé R, Ortega Cano JA, Ortega de la Torre MDLA, Duran Fernández-Feijoo C, Girabent-Farrés M, Figueras-Aloy J, Krauel X, Alcántara S. Learning and memory disabilities in IUGR babies: Functional and molecular analysis in a rat model. Brain Behav 2017; 7:e00631. [PMID: 28293472 PMCID: PMC5346519 DOI: 10.1002/brb3.631] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 07/22/2016] [Accepted: 11/30/2016] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION 1Intrauterine growth restriction (IUGR) is the failure of the fetus to achieve its inherent growth potential, and it has frequently been associated with neurodevelopmental problems in childhood. Neurological disorders are mostly associated with IUGR babies with an abnormally high cephalization index (CI) and a brain sparing effect. However, a similar correlation has never been demonstrated in an animal model. The aim of this study was to determine the correlations between CI, functional deficits in learning and memory and alterations in synaptic proteins in a rat model of IUGR. METHODS 2Utero-placental insufficiency was induced by meso-ovarian vessel cauterization (CMO) in pregnant rats at embryonic day 17 (E17). Learning performance in an aquatic learning test was evaluated 25 days after birth and during 10 days. Some synaptic proteins were analyzed (PSD95, Synaptophysin) by Western blot and immunohistochemistry. RESULTS 3Placental insufficiency in CMO pups was associated with spatial memory deficits, which are correlated with a CI above the normal range. CMO pups presented altered levels of synaptic proteins PSD95 and synaptophysin in the hippocampus. CONCLUSIONS 4The results of this study suggest that learning disabilities may be associated with altered development of excitatory neurotransmission and synaptic plasticity. Although interspecific differences in fetal response to placental insufficiency should be taken into account, the translation of these data to humans suggest that both IUGR babies and babies with a normal birth weight but with intrauterine Doppler alterations and abnormal CI should be closely followed to detect neurodevelopmental alterations during the postnatal period.
Collapse
Affiliation(s)
- Marta Camprubí Camprubí
- Neonatology Service Sant Joan de Déu BCNatal Hospital Sant Joan de Déu i Clínic University of Barcelona Barcelona Spain
| | - Rafel Balada Caballé
- Department of Pathology and Experimental Therapeutics School of Medicine University of Barcelona Barcelona Spain
| | - Juan A Ortega Cano
- Department of Pathology and Experimental Therapeutics School of Medicine University of Barcelona Barcelona Spain; Present address: Department of Neurology Feinberg School of Medicine Northwestern University Chicago IL 60611 USA
| | | | | | | | - Josep Figueras-Aloy
- Neonatology Service Sant Joan de Déu BCNatal Hospital Sant Joan de Déu i Clínic University of Barcelona Barcelona Spain
| | - Xavier Krauel
- Neonatology Service Sant Joan de Déu BCNatal Hospital Sant Joan de Déu i Clínic University of Barcelona Barcelona Spain
| | - Soledad Alcántara
- Department of Pathology and Experimental Therapeutics School of Medicine University of Barcelona Barcelona Spain
| |
Collapse
|
22
|
Crossland RF, Balasa A, Ramakrishnan R, Mahadevan SK, Fiorotto ML, Van den Veyver IB. Chronic Maternal Low-Protein Diet in Mice Affects Anxiety, Night-Time Energy Expenditure and Sleep Patterns, but Not Circadian Rhythm in Male Offspring. PLoS One 2017; 12:e0170127. [PMID: 28099477 PMCID: PMC5242516 DOI: 10.1371/journal.pone.0170127] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/29/2016] [Indexed: 12/14/2022] Open
Abstract
Offspring of murine dams chronically fed a protein-restricted diet have an increased risk for metabolic and neurobehavioral disorders. Previously we showed that adult offspring, developmentally exposed to a chronic maternal low-protein (MLP) diet, had lower body and hind-leg muscle weights and decreased liver enzyme serum levels. We conducted energy expenditure, neurobehavioral and circadian rhythm assays in male offspring to examine mechanisms for the body-weight phenotype and assess neurodevelopmental implications of MLP exposure. C57BL/6J dams were fed a protein restricted (8%protein, MLP) or a control protein (20% protein, C) diet from four weeks before mating until weaning of offspring. Male offspring were weaned to standard rodent diet (20% protein) and single-housed until 8–12 weeks of age. We examined body composition, food intake, energy expenditure, spontaneous rearing activity and sleep patterns and performed behavioral assays for anxiety (open field activity, elevated plus maze [EPM], light/dark exploration), depression (tail suspension and forced swim test), sociability (three-chamber), repetitive (marble burying), learning and memory (fear conditioning), and circadian behavior (wheel-running activity during light-dark and constant dark cycles). We also measured circadian gene expression in hypothalamus and liver at different Zeitgeber times (ZT). Male offspring from separate MLP exposed dams had significantly greater body fat (P = 0.03), less energy expenditure (P = 0.004), less rearing activity (P = 0.04) and a greater number of night-time rest/sleep bouts (P = 0.03) compared to control. MLP offspring displayed greater anxiety-like behavior in the EPM (P<0.01) but had no learning and memory deficit in fear-conditioning assay (P = 0.02). There was an effect of time on Per1, Per 2 and Clock circadian gene expression in the hypothalamus but not on circadian behavior. Thus, transplacental and early developmental exposure of dams to chronic MLP reduces food intake and energy expenditure, increases anxiety like behavior and disturbs sleep patterns but not circadian rhythm in adult male offspring.
Collapse
Affiliation(s)
- Randy F. Crossland
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States of America
| | - Alfred Balasa
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States of America
| | - Rajesh Ramakrishnan
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States of America
| | - Sangeetha K. Mahadevan
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States of America
- Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Marta L. Fiorotto
- USDA/Agricultural Research Service Children’s Nutrition Research Center, Houston, TX, United States of America
| | - Ignatia B. Van den Veyver
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States of America
- Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States of America
- * E-mail:
| |
Collapse
|
23
|
Chen B, Deng X, Wang B, Liu H. Etanercept, an inhibitor of TNF-a, prevents propofol-induced neurotoxicity in the developing brain. Int J Dev Neurosci 2016; 55:91-100. [PMID: 27756568 DOI: 10.1016/j.ijdevneu.2016.10.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/18/2016] [Accepted: 10/10/2016] [Indexed: 11/19/2022] Open
Abstract
Propofol can induce acute neuronal apoptosis, neuronal loss or long-term cognitive impairment when exposed in neonatal rodents, but the mechanisms by which propofol induces developmental neurotoxicity are unclear. Recent studies have demonstrated that propofol can increase the TNF-α level in the developing brain, but there is a lack of direct evidence to show whether TNF-α is partially or fully involved in propofol-induced neurotoxicity. The present study shows that propofol exposure in neonatal rats induces an increase of TNF-α in the cerebral spinal fluid, hippocampus and prefrontal cortex (PFC). Etanercept, a TNF-α inhibitor, prevents propofol-induced short- or long-term neuronal apoptosis, neuronal loss, synaptic loss and long-term cognitive impairment. Furthermore, mTNF-α (precursor of TNF-α) expression in microglia cells is increased after propofol anaesthesia in either the hippocampus or PFC, but mTNF-α expression in neurons is only increased in the PFC. These findings suggest that TNF-α may mediate propofol-induced developmental neurotoxicity, and etanercept can provide neural protection. Microglia are the main cellular source of TNF-α after propofol exposure, while the synthesis of TNF-α in neurons is brain-region selective.
Collapse
Affiliation(s)
- Bo Chen
- Department of Anesthesiology, Chongqing Cancer Institute, Chongqing 40030, PR China; Guangxi Medical University, Nanning, Guangxi 530021, PR China
| | - Xiaoyuan Deng
- Department of Anesthesiology, Chongqing Cancer Institute, Chongqing 40030, PR China
| | - Bin Wang
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Hongliang Liu
- Department of Anesthesiology, Chongqing Cancer Institute, Chongqing 40030, PR China.
| |
Collapse
|
24
|
Rideau Batista Novais A, Pham H, Van de Looij Y, Bernal M, Mairesse J, Zana-Taieb E, Colella M, Jarreau PH, Pansiot J, Dumont F, Sizonenko S, Gressens P, Charriaut-Marlangue C, Tanter M, Demene C, Vaiman D, Baud O. Transcriptomic regulations in oligodendroglial and microglial cells related to brain damage following fetal growth restriction. Glia 2016; 64:2306-2320. [PMID: 27687291 DOI: 10.1002/glia.23079] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 11/06/2022]
Abstract
Fetal growth restriction (FGR) is a major complication of human pregnancy, frequently resulting from placental vascular diseases and prenatal malnutrition, and is associated with adverse neurocognitive outcomes throughout life. However, the mechanisms linking poor fetal growth and neurocognitive impairment are unclear. Here, we aimed to correlate changes in gene expression induced by FGR in rats and abnormal cerebral white matter maturation, brain microstructure, and cortical connectivity in vivo. We investigated a model of FGR induced by low-protein-diet malnutrition between embryonic day 0 and birth using an interdisciplinary approach combining advanced brain imaging, in vivo connectivity, microarray analysis of sorted oligodendroglial and microglial cells and histology. We show that myelination and brain function are both significantly altered in our model of FGR. These alterations, detected first in the white matter on magnetic resonance imaging significantly reduced cortical connectivity as assessed by ultrafast ultrasound imaging. Fetal growth retardation was found associated with white matter dysmaturation as shown by the immunohistochemical profiles and microarrays analyses. Strikingly, transcriptomic and gene network analyses reveal not only a myelination deficit in growth-restricted pups, but also the extensive deregulation of genes controlling neuroinflammation and the cell cycle in both oligodendrocytes and microglia. Our findings shed new light on the cellular and gene regulatory mechanisms mediating brain structural and functional defects in malnutrition-induced FGR, and suggest, for the first time, a neuroinflammatory basis for the poor neurocognitive outcome observed in growth-restricted human infants. GLIA 2016;64:2306-2320.
Collapse
Affiliation(s)
- Aline Rideau Batista Novais
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service de Réanimation et Pédiatrie Néonatales, Groupe Hospitalier Robert Debré, Paris, France.,Université Paris Diderot, Paris, France.,Fondation PremUp, Paris, France
| | - Hoa Pham
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Fondation PremUp, Paris, France
| | - Yohan Van de Looij
- Laboratory for Functional and Metabolic Imaging (LIFMET), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Division of Development and Growth, Department of Child and Adolescent Medicine, Geneva University Hospital and School of Medicine, Geneva, Switzerland
| | - Miguel Bernal
- Institut Langevin, CNRS UMR 7587, Inserm U979, ESPCI ParisTech, PSL Research University, Paris, France
| | - Jerome Mairesse
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Fondation PremUp, Paris, France
| | - Elodie Zana-Taieb
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Fondation PremUp, Paris, France.,Université Paris-Descartes, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service de Médecine et Réanimation Néonatales de Port-Royal, Groupe Hospitalier Cochin, Broca, Hôtel-Dieu, Paris, France
| | - Marina Colella
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Fondation PremUp, Paris, France
| | - Pierre-Henri Jarreau
- Fondation PremUp, Paris, France.,Université Paris-Descartes, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service de Médecine et Réanimation Néonatales de Port-Royal, Groupe Hospitalier Cochin, Broca, Hôtel-Dieu, Paris, France
| | - Julien Pansiot
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Fondation PremUp, Paris, France
| | - Florent Dumont
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Paris, France
| | - Stéphane Sizonenko
- Division of Development and Growth, Department of Child and Adolescent Medicine, Geneva University Hospital and School of Medicine, Geneva, Switzerland
| | - Pierre Gressens
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Université Paris Diderot, Paris, France.,Fondation PremUp, Paris, France
| | - Christiane Charriaut-Marlangue
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France.,Université Paris Diderot, Paris, France.,Fondation PremUp, Paris, France
| | - Mickael Tanter
- Institut Langevin, CNRS UMR 7587, Inserm U979, ESPCI ParisTech, PSL Research University, Paris, France
| | - Charlie Demene
- Institut Langevin, CNRS UMR 7587, Inserm U979, ESPCI ParisTech, PSL Research University, Paris, France
| | - Daniel Vaiman
- Institut Cochin, Inserm U1016, UMR8104 CNRS, Paris, France
| | - Olivier Baud
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1141, Paris, France. .,Assistance Publique - Hôpitaux de Paris, Service de Réanimation et Pédiatrie Néonatales, Groupe Hospitalier Robert Debré, Paris, France. .,Université Paris Diderot, Paris, France. .,Fondation PremUp, Paris, France.
| |
Collapse
|
25
|
Veena SR, Gale CR, Krishnaveni GV, Kehoe SH, Srinivasan K, Fall CH. Association between maternal nutritional status in pregnancy and offspring cognitive function during childhood and adolescence; a systematic review. BMC Pregnancy Childbirth 2016; 16:220. [PMID: 27520466 PMCID: PMC4982007 DOI: 10.1186/s12884-016-1011-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 08/04/2016] [Indexed: 11/30/2022] Open
Abstract
Background The mother is the only source of nutrition for fetal growth including brain development. Maternal nutritional status (anthropometry, macro- and micro-nutrients) before and/or during pregnancy is therefore a potential predictor of offspring cognitive function. The relationship of maternal nutrition to offspring cognitive function is unclear. This review aims to assess existing evidence linking maternal nutritional status with offspring cognitive function. Methods Exposures considered were maternal BMI, height and weight, micronutrient status (vitamins D, B12, folate and iron) and macronutrient intakes (carbohydrate, protein and fat). The outcome was any measure of cognitive function in children aged <18 years. We considered observational studies and trials with allocation groups that differed by single nutrients. We searched Medline/PubMed and the Cochrane Library databases and reference lists of retrieved literature. Two reviewers independently extracted data from relevant articles. We used methods recommended by the Centre for Reviews and Dissemination, University of York and the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. Results Of 16,143 articles identified, 38 met inclusion criteria. Most studies were observational, and from high-income settings. There were few randomized controlled trials. There was consistent evidence linking maternal obesity with lower cognitive function in children; low maternal BMI has been inadequately studied. Among three studies of maternal vitamin D status, two showed lower cognitive function in children of deficient mothers. One trial of folic acid supplementation showed no effects on the children’s cognitive function and evidence from 13 observational studies was mixed. Among seven studies of maternal vitamin B12 status, most showed no association, though two studies in highly deficient populations suggested a possible effect. Four out of six observational studies and two trials (including one in an Iron deficient population) found no association of maternal iron status with offspring cognitive function. One trial of maternal carbohydrate/protein supplementation showed no effects on offspring cognitive function. Conclusions Current evidence that maternal nutritional status during pregnancy as defined by BMI, single micronutrient studies, or macronutrient intakes influences offspring cognitive function is inconclusive. There is a need for more trials especially in populations with high rates of maternal undernutrition. Systematic review registration Registered in PROSPERO CRD42013005702. Electronic supplementary material The online version of this article (doi:10.1186/s12884-016-1011-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sargoor R Veena
- Epidemiology Research Unit, CSI Holdsworth Memorial Hospital, Mysore, India.
| | - Catharine R Gale
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK.,Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | | | - Sarah H Kehoe
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | | | - Caroline Hd Fall
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| |
Collapse
|
26
|
Buschdorf J, Ong M, Ong S, MacIsaac J, Chng K, Kobor M, Meaney M, Holbrook J. Low birth weight associates with hippocampal gene expression. Neuroscience 2016; 318:190-205. [DOI: 10.1016/j.neuroscience.2016.01.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 01/05/2016] [Accepted: 01/05/2016] [Indexed: 02/06/2023]
|
27
|
Chen B, Deng X, Wang B, Liu H. Persistent neuronal apoptosis and synaptic loss induced by multiple but not single exposure of propofol contribute to long-term cognitive dysfunction in neonatal rats. J Toxicol Sci 2016; 41:627-36. [DOI: 10.2131/jts.41.627] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Bo Chen
- Department of Anesthesiology, Chongqing Cancer Institute, China
- Guangxi Medical University, China
| | - Xiaoyuan Deng
- Department of Anesthesiology, Chongqing Cancer Institute, China
| | - Bin Wang
- Institute of Life Science, Chongqing Medical University, China
| | - Hongliang Liu
- Department of Anesthesiology, Chongqing Cancer Institute, China
| |
Collapse
|
28
|
Erbs E, Faget L, Ceredig RA, Matifas A, Vonesch JL, Kieffer BL, Massotte D. Impact of chronic morphine on delta opioid receptor-expressing neurons in the mouse hippocampus. Neuroscience 2015; 313:46-56. [PMID: 26480813 DOI: 10.1016/j.neuroscience.2015.10.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/08/2015] [Accepted: 10/12/2015] [Indexed: 11/18/2022]
Abstract
Delta opioid (DOP) receptors participate to the control of chronic pain and emotional responses. Recent data also identified their implication in spatial memory and drug-context associations pointing to a critical role of hippocampal delta receptors. To better appreciate the impact of repeated drug exposure on their modulatory activity, we used fluorescent knock-in mice that express a functional delta receptor fused at its carboxy-terminus with the green fluorescent protein in place of the native receptor. We then tested the impact of chronic morphine treatment on the density and distribution of delta receptor-expressing cells in the hippocampus. A decrease in delta receptor-positive cell density was observed in the CA1, CA3 and dentate gyrus without alteration of the distribution across the different GABAergic populations that mainly express delta receptors. This effect partly persisted after four weeks of morphine abstinence. In addition, we observed increased DOP receptor expression at the cell surface compared to saline-treated animals. In the hippocampus, chronic morphine administration thus induces DOP receptor cellular redistribution and durably decreases delta receptor-expressing cell density. Such modifications are likely to alter hippocampal physiology, and to contribute to long-term cognitive deficits.
Collapse
Affiliation(s)
- E Erbs
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France
| | - L Faget
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France
| | - R A Ceredig
- Institut des Neurosciences Cellulaires et Intégratives, UPR 3212, 5 Rue Blaise Pascal, F-67084 Strasbourg, France
| | - A Matifas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France
| | - J-L Vonesch
- Imaging Center IGBMC, CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France
| | - B L Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France; Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875, Boulevard LaSalle, Montreal (Quebec) H4H 1R3, Canada
| | - D Massotte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France; Institut des Neurosciences Cellulaires et Intégratives, UPR 3212, 5 Rue Blaise Pascal, F-67084 Strasbourg, France.
| |
Collapse
|
29
|
Besson AA, Lagisz M, Senior AM, Hector KL, Nakagawa S. Effect of maternal diet on offspring coping styles in rodents: a systematic review and meta-analysis. Biol Rev Camb Philos Soc 2015; 91:1065-1080. [DOI: 10.1111/brv.12210] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 06/07/2015] [Accepted: 06/18/2015] [Indexed: 11/29/2022]
Affiliation(s)
- Anne A. Besson
- Department of Zoology; University of Otago; PO Box 56 Dunedin 9054 New Zealand
| | - Malgorzata Lagisz
- Department of Zoology; University of Otago; PO Box 56 Dunedin 9054 New Zealand
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental Sciences, Biological Science Building; University of New South Wales; Sydney 2052 New South Wales Australia
| | - Alistair M. Senior
- Department of Zoology; University of Otago; PO Box 56 Dunedin 9054 New Zealand
- Charles Perkins Centre, The University of Sydney; Johns Hopkins Drive, Sydney 2009 New South Wales Australia
| | - Katie L. Hector
- Department of Zoology; University of Otago; PO Box 56 Dunedin 9054 New Zealand
| | - Shinichi Nakagawa
- Department of Zoology; University of Otago; PO Box 56 Dunedin 9054 New Zealand
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental Sciences, Biological Science Building; University of New South Wales; Sydney 2052 New South Wales Australia
| |
Collapse
|
30
|
de Bie HMA, de Ruiter MB, Ouwendijk M, Oostrom KJ, Wilke M, Boersma M, Veltman DJ, Delemarre-van de Waal HA. Using fMRI to Investigate Memory in Young Children Born Small for Gestational Age. PLoS One 2015; 10:e0129721. [PMID: 26132815 PMCID: PMC4488594 DOI: 10.1371/journal.pone.0129721] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 05/12/2015] [Indexed: 12/13/2022] Open
Abstract
Objectives Intrauterine growth restriction (IUGR) can lead to infants being born small for gestational age (SGA). SGA is associated with differences in brain anatomy and impaired cognition. We investigated learning and memory in children born SGA using neuropsychological testing and functional Magnetic Resonance Imaging (fMRI). Study Design 18 children born appropriate for gestational age (AGA) and 34 SGA born children (18 with and 16 without postnatal catch-up growth) participated in this study. All children were between 4 and 7 years old. Cognitive functioning was assessed by IQ and memory testing (Digit/Word Span and Location Learning). A newly developed fMRI picture encoding task was completed by all children in order to assess brain regions involved in memory processes. Results Neuropsychological testing demonstrated that SGA children had IQ’s within the normal range but lower than in AGA and poorer performances across measures of memory. Using fMRI, we observed memory related activity in posterior parahippocampal gyrus as well as the hippocampus proper. Additionally, activation was seen bilaterally in the prefrontal gyrus. Children born SGA showed less activation in the left parahippocampal region compared to AGA. Conclusions This is the first fMRI study demonstrating different brain activation patterns in 4-7 year old children born SGA, suggesting that intrauterine growth restriction continues to affect neural functioning in children later-on.
Collapse
Affiliation(s)
- Henrica M. A. de Bie
- Department of Pediatrics, VU University Medical Center, Amsterdam, The Netherlands
| | - Michiel B. de Ruiter
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mieke Ouwendijk
- Department of Pediatric Psychology, VU University Medical Center, Amsterdam, The Netherlands
- * E-mail:
| | - Kim J. Oostrom
- Department of Pediatric Psychology, VU University Medical Center, Amsterdam, The Netherlands
| | - Marko Wilke
- Department of Pediatric Neurology and Developmental Medicine and Experimental Pediatric Neuroimaging Neuroimaging Group, Children’s Hospital, University of Tübingen, Tübingen, Germany
| | - Maria Boersma
- Department of Clinical Neurophysiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Dick J. Veltman
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Henriette A. Delemarre-van de Waal
- Department of Pediatrics, VU University Medical Center, Amsterdam, The Netherlands
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
31
|
Venkareddy LK, Muralidhara. Potential of casein as a nutrient intervention to alleviate lead (Pb) acetate-mediated oxidative stress and neurotoxicity: First evidence in Drosophila melanogaster. Neurotoxicology 2015; 48:142-51. [DOI: 10.1016/j.neuro.2015.03.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 02/27/2015] [Accepted: 03/06/2015] [Indexed: 12/25/2022]
|
32
|
Heggland I, Storkaas IS, Soligard HT, Kobro-Flatmoen A, Witter MP. Stereological estimation of neuron number and plaque load in the hippocampal region of a transgenic rat model of Alzheimer's disease. Eur J Neurosci 2015; 41:1245-62. [PMID: 25808554 DOI: 10.1111/ejn.12876] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 02/08/2015] [Indexed: 11/29/2022]
Abstract
The main hallmarks of Alzheimer's disease (AD) are senile plaques, neurofibrillary tangles and neuronal death. The McGill-R-Thy1-APP rat is one of the few transgenic rat models of AD that displays progressive amyloid pathology. This study aimed to further characterise this rat model, focusing on the pathological changes in the hippocampal formation and the parahippocampal region. These structures, that are important for episodic memory and spatial navigation, are affected in the early stages of the disease. This study used unbiased stereology to investigate possible neuronal loss in the CA1, subiculum and entorhinal cortex of 18-month-old homozygous McGill-R-Thy1-APP rats, and also quantified the plaque load in all the areas of the hippocampal formation and parahippocampal region from 9 to 18 months old. A significant reduction of neurons at 18 months was only seen in the subiculum. The first plaque pathology was seen at 9 months in the subiculum. Although the quantified plaque load was variable between animals, the pattern of spatiotemporal progression was similar for all animals. The spread of plaque pathology mainly affected anatomically connected regions. Overall, the plaque pathology observed in the transgenic rats was similar to the early phases of amyloid beta (Aβ)-deposition described in human patients. The findings here thus indicate that the McGill-R-Thy1-APP rat could be a good model of the Aβ pathology in AD, but less so with respect to neuron loss.
Collapse
Affiliation(s)
- Ingrid Heggland
- Kavli Institute for Systems Neuroscience & Centre for Neural Computation, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Postboks 8905, 7491, Trondheim, Norway
| | | | | | | | | |
Collapse
|
33
|
Fischer LK, McGaughy JA, Bradshaw SE, Weissner WJ, Amaral AC, Rosene DL, Mokler DJ, Fitzmaurice GM, Galler JR. Prenatal protein level impacts homing behavior in Long-Evans rat pups. Nutr Neurosci 2015; 19:187-95. [PMID: 25603489 DOI: 10.1179/1476830515y.0000000001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study assessed the effect of varying prenatal protein levels on the development of homing behavior in rat pups. METHODS Long-Evans rats were fed one of the four isocaloric diets containing 6% (n = 7 litters), 12% (n = 9), 18% (n = 9), or 25% (n = 10) casein prior to mating and throughout pregnancy. At birth, litters were fostered to well-nourished control mothers fed a 25% casein diet during pregnancy, and an adequate protein diet (25% casein) was provided to weaning. On postnatal days 5, 7, 9, 11, and 13, homing behaviors, including activity levels, rate of successful returns to the nest quadrant and latencies to reach the nest over a 3-minute test period were recorded from two starting positions in the home cage. Adult body and brain weights were obtained at sacrifice (postnatal day 130 or 200). RESULTS Growth was impaired in pups whose mothers were fed a 6% or, to a lesser extent, a 12% casein diet relative to pups whose mothers were fed the 18 and 25% casein diets. The 6 and 12% prenatal protein levels resulted in lower activity levels, with the greatest reduction on postnatal day 13. However, only the 6% pups had reduced success and higher latencies in reaching the nest quadrant when compared with pups from the three other nutrition groups. Latency in reaching the nest quadrant was significantly and negatively associated with adult brain weight. DISCUSSION Home orientation is a sensitive measure of developmental deficits associated with variations in prenatal protein levels, including levels of protein deficiency that do not lead to overt growth failure.
Collapse
Affiliation(s)
- L K Fischer
- a Judge Baker Children's Center and Department of Psychiatry , Harvard Medical School , Boston , MA , USA
| | - J A McGaughy
- b Department of Psychology , University of New Hampshire , Durham , NH , USA
| | - S E Bradshaw
- a Judge Baker Children's Center and Department of Psychiatry , Harvard Medical School , Boston , MA , USA
| | - W J Weissner
- c Department of Biological Sciences , University of New England , Biddeford , ME , USA
| | - A C Amaral
- d Department of Anatomy & Neurobiology , Boston University Medical Campus , Boston , MA , USA
| | - D L Rosene
- d Department of Anatomy & Neurobiology , Boston University Medical Campus , Boston , MA , USA
| | - D J Mokler
- c Department of Biological Sciences , University of New England , Biddeford , ME , USA
| | - G M Fitzmaurice
- e Department of Biostatistics , Harvard School of Public Health , Boston , MA , USA.,f Laboratory for Psychiatric Biostatistics , McLean Hospital , Belmont , MA , USA
| | - J R Galler
- a Judge Baker Children's Center and Department of Psychiatry , Harvard Medical School , Boston , MA , USA
| |
Collapse
|
34
|
The effects of black garlic ethanol extract on the spatial memory and estimated total number of pyramidal cells of the hippocampus of monosodium glutamate-exposed adolescent male Wistar rats. Anat Sci Int 2014; 90:275-86. [DOI: 10.1007/s12565-014-0262-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 11/06/2014] [Indexed: 10/24/2022]
|
35
|
Akitake Y, Katsuragi S, Hosokawa M, Mishima K, Ikeda T, Miyazato M, Hosoda H. Moderate maternal food restriction in mice impairs physical growth, behavior, and neurodevelopment of offspring. Nutr Res 2014; 35:76-87. [PMID: 25433908 DOI: 10.1016/j.nutres.2014.10.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 10/07/2014] [Accepted: 10/31/2014] [Indexed: 01/03/2023]
Abstract
Intrauterine growth retardation (IUGR) occurs in 3% to 7% of all pregnancies. Recent human studies have indicated that neurodevelopmental disabilities, learning disorders, memory impairment, and mood disturbance are common in IUGR offspring. However, the interactions between IUGR and neurodevelopmental disorders are unclear because of the wide range of causes of IUGR, such as maternal malnutrition, placental insufficiency, pregnancy toxemia, and fetal malformations. Meanwhile, many studies have shown that moderate food restriction enhances spatial learning and improves mood disturbance in adult humans and animals. To date, the effects of maternal moderate food restriction on fetal brain remain largely unknown. In this study, we hypothesized that IUGR would be caused by even moderate food restriction in pregnant females and that the offspring would have neurodevelopmental disabilities. Mid-pregnant mice received moderate food restriction through the early lactation period. The offspring were tested for aspects of physical development, behavior, and neurodevelopment. The results showed that moderate maternal food restriction induced IUGR. Offspring had low birth weight and delayed development of physical and coordinated movement. Moreover, IUGR offspring exhibited mental disabilities such as anxiety and poor cognitive function. In particular, male offspring exhibited significantly impaired cognitive function at 3 weeks of age. These results suggested that a restricted maternal diet could be a risk factor for developmental disability in IUGR offspring and that male offspring might be especially susceptible.
Collapse
Affiliation(s)
- Yoshiharu Akitake
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, 565-8565, Japan.
| | - Shinji Katsuragi
- Department of Perinatology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, 565-8565, Japan
| | - Masato Hosokawa
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Kenichi Mishima
- Department of Physiology and Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Mie University School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, 565-8565, Japan
| | - Hiroshi Hosoda
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, 565-8565, Japan.
| |
Collapse
|
36
|
McGaughy JA, Amaral AC, Rushmore RJ, Mokler DJ, Morgane PJ, Rosene DL, Galler JR. Prenatal malnutrition leads to deficits in attentional set shifting and decreases metabolic activity in prefrontal subregions that control executive function. Dev Neurosci 2014; 36:532-41. [PMID: 25342495 DOI: 10.1159/000366057] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/22/2014] [Indexed: 11/19/2022] Open
Abstract
Globally, over 25% of all children under the age of 5 years experience malnutrition leading to cognitive and emotional impairments that can persist into adulthood and beyond. We use a rodent model to determine the impact of prenatal protein malnutrition on executive functions in an attentional set-shifting task and metabolic activity in prefrontal cortex (PFC) subregions critical to these behaviors. Long-Evans dams were provided with a low (6% casein) or adequate (25% casein) protein diet 5 weeks before mating and during pregnancy. At birth, the litters were culled to 8 pups and fostered to control dams on the 25% casein diet. At postnatal day 90, prenatally malnourished rats were less able to shift attentional set and reverse reward contingencies than controls, demonstrating cognitive rigidity. Naive same-sexed littermates were assessed for regional brain activity using the metabolic marker (14)C-2-deoxyglucose (2DG). The prenatally malnourished rats had lower metabolic activity than controls in prelimbic, infralimbic, anterior cingulate, and orbitofrontal cortices, but had comparable activity in the nearby piriform cortex and superior colliculus. This study demonstrates that prenatal protein malnutrition in a well-described animal model produces cognitive deficits in tests of attentional set shifting and reversal learning, similar to findings of cognitive inflexibility reported in humans exposed to early childhood malnutrition.
Collapse
Affiliation(s)
- Jill A McGaughy
- Department of Psychology, University of New Hampshire, Durham, N.H., USA
| | | | | | | | | | | | | |
Collapse
|
37
|
TIAN X, XIANG Y, FAN Y, BU H, YANG H, MANYANDE A, GAO F, TIAN Y. Impact of malnutrition on propofol consumption and recovery time among patients undergoing laparoscopic gastrointestinal surgery. Acta Anaesthesiol Scand 2014; 58:942-7. [PMID: 25060045 DOI: 10.1111/aas.12373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND Malnutrition is a major health problem, especially in hospitalized patients as it can be closely related to many post-operative complications. However, research on malnutrition and its effect on the outcome of general anesthesia have been largely neglected. Here we investigated malnutrition status on propofol consumption and recovery time among patients undergoing laparoscopic gastrointestinal surgery under general anesthesia. METHODS One hundred and one patients were recruited between January and June 2012 at Tongji Hospital and assigned into three groups according to Nutritional Risk Screening Tool 2002 score. A standard combined general anesthesia procedure was performed under regular monitoring. The dosage of propofol needed for induction, consumption during maintenance and recovery time were recorded. RESULTS When compared with normal nutritional status individuals, the propofol dosage at induction was significantly decreased about 4.3% in moderate malnutritional status patients (P < 0.01) and about 16.8% in severely malnutritional status patients (P < 0.01). The average consumption of propofol was also significantly lower in malnourished individuals; for moderate malnutritional, the decrease was about 20% (P < 0.01) while for the severely malnutritional, it was 30% (P < 0.01) when compared with normal nutritional status individuals. For the recovery time of propofol anesthesia, the patients with severe malnutritional status awoke average 6.8 min later than those normally nourished (P < 0.01), but those patients with moderate malnutrition status did not (P = 0.885). CONCLUSION The present results indicate that the dosage and recovery time of propofol does change in malnourished individuals. Therefore, malnutrition may somehow affect the outcome of general anesthesia.
Collapse
Affiliation(s)
- X. TIAN
- Department of Anesthesiology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - Y. XIANG
- Department of Ophthalmology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - Y. FAN
- Department of Nutrition and Food Hygiene; School of Public Health; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - H. BU
- Department of Anesthesiology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - H. YANG
- Department of Anesthesiology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - A. MANYANDE
- School of Psychology; Social Work and Human Sciences; University of West London; London UK
| | - F. GAO
- Department of Anesthesiology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - Y. TIAN
- Department of Anesthesiology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| |
Collapse
|
38
|
Waber DP, Bryce CP, Fitzmaurice GM, Zichlin ML, McGaughy J, Girard JM, Galler JR. Neuropsychological outcomes at midlife following moderate to severe malnutrition in infancy. Neuropsychology 2014; 28:530-40. [PMID: 24635710 DOI: 10.1037/neu0000058] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE To compare neuropsychological profiles of adults who had experienced an episode of moderate to severe protein-energy malnutrition confined to the first year of life with that of a healthy community comparison group. METHOD We assessed neuropsychological functioning in a cohort of Barbadian adults, all of whom had birth weight >2268 g. The previously malnourished group (N = 77, mean age = 38 years, 53% male) had been hospitalized during the first year of life for moderate to severe protein energy malnutrition and subsequently enrolled in a program providing nutrition education, home visits and subsidized foods to 12 years of age. They also had documented, adequate nutrition throughout childhood and complete catch-up in growth by the end of adolescence. The healthy comparison group (N = 59, mean age = 38 years, 54% male) were recruited as children from the same classrooms and neighborhoods. RESULTS Adjusted for effects of standard of living during childhood and adolescence and current intellectual ability level, there were nutrition group differences on measures of cognitive flexibility and concept formation, as well as initiation, verbal fluency, working memory, processing speed, and visuospatial integration. Behavioral and cognitive regulation were not affected. CONCLUSIONS Postnatal malnutrition confined to the first year of life is associated with neurocognitive compromise persisting into midlife. Early malnutrition may have a specific neuropsychological signature, affecting response initiation to a somewhat greater extent than response inhibition.
Collapse
Affiliation(s)
| | | | | | | | - Jill McGaughy
- Department of Psychology, University of New Hampshire
| | | | | |
Collapse
|
39
|
Oros D, Altermir I, Elia N, Tuquet H, Pablo LE, Fabre E, Pueyo V. Pathways of neuronal and cognitive development in children born small-for-gestational age or late preterm. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2014; 43:41-47. [PMID: 23836499 DOI: 10.1002/uog.12556] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 06/19/2013] [Accepted: 06/28/2013] [Indexed: 06/02/2023]
Abstract
OBJECTIVE To assess the effects of late small-for-gestational-age (SGA) birth and late prematurity on cognitive outcomes and structural changes in the central nervous system at primary school age, using a novel approach to examine changes in neuronal integrity of the retina. METHODS We conducted a cross-sectional study of 347 children aged 6-13 years, including in the analysis only infants born after 34 weeks' gestation. We recorded all perinatal outcomes through a survey of parents. Neuronal damage was evaluated using optical coherence tomography of the retina. In a subgroup of 112 children aged 6-8 years, visuospatial perception was evaluated with the Children's Bender Visual Motor Gestalt Test. RESULTS The proportions of SGA and late preterm children were 11.8 and 6.3%, respectively. Prematurity and SGA were simultaneously present in five children. When compared with controls, SGA children showed significantly lower than average retinal nerve fiber layer (RNFL) thickness (94.1 vs 98.8 μm; P = 0.007) and an increased percentage of abnormal Bender scores (27.3 vs 6.2%; P = 0.017) (odds ratio 5.6 (95% CI, 1.2-26.8)). These differences increased when late SGA infants with a birth weight below the 3(rd) percentile were compared with SGA infants with a birth weight between the 3(rd) and 10(th) percentiles and with controls, for RNFL thickness (92.5 vs 94.6 and 98.8 μm, respectively; P = 0.021) and abnormal Bender tests (33.3 vs 25.0 and 6.2%, respectively; P = 0.036). However, no differences were found in retinal structure and visuomotor performance between late preterm and term infants. CONCLUSIONS These data suggest that late SGA and late prematurity induce a distinct neuronal pattern of structural changes that persist at school age. Late-onset SGA infants are at increased risk for axonal loss in the retina and present specific visuomotor difficulties.
Collapse
Affiliation(s)
- D Oros
- Obstetrics Department, Hospital Clínico Universitario Lozano Blesa Zaragoza, Instituto Aragonés de Ciencias de la Salud, University of Zaragoza, Zaragoza, Spain
| | | | | | | | | | | | | |
Collapse
|
40
|
Hipólito-Reis J, Pereira PA, Andrade JP, Cardoso A. Prolonged protein deprivation differentially affects calretinin- and parvalbumin-containing interneurons in the hippocampal dentate gyrus of adult rats. Neurosci Lett 2013; 555:154-8. [DOI: 10.1016/j.neulet.2013.09.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/03/2013] [Accepted: 09/16/2013] [Indexed: 01/29/2023]
|
41
|
Françolin-Silva AL, Brandão ML, Almeida SS. Early postnatal protein malnutrition causes resistance to the anxiolytic effects of diazepam as assessed by the fear-potentiated startle test. Nutr Neurosci 2013; 10:23-9. [PMID: 17539480 DOI: 10.1080/10284150601168346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Given that protein malnutrition induces structural, neurochemical and functional changes in the CNS, the present study aimed to investigate the effects of different early periods of protein malnutrition on the behavior and reactivity to diazepam (DZ) in a model of anxiety: the fear-potentiated startle (FPS). Male Wistar rats (n = 110) from well-nourished (16 %-protein) or malnourished (6%-protein) litters were distributed in five different groups: W (well-nourished), M7 (malnourished for 7-days, since day 0), M14 (14-days), M21 (21-days) and M28 (28-days). The results obtained in FPS revealed that malnourished-animals acquired the startle response, irrespective of the time they were exposed to the diet. Besides, DZ reduced the startle amplitude in the noise-alone and light-noise trials. The data concerning the total freezing time showed that the expression of this response was affected by malnutrition and varied in accordance with the findings of previous studies in which malnutrition procedures was imposed for long periods (more than 50 days). Therefore, we suggest that early protein malnutrition: (a) did not produce deficits in the associative learning process of these animals in the FPS, and (b) decreased freezing time in the FPS and produce hyporeactivity to the effects of DZ in rats malnourished for 21 days or more, indicating alterations in the GABAergic neurotransmitter system.
Collapse
Affiliation(s)
- A L Françolin-Silva
- Laboratory of Nutrition and Behavior, Department of Psychology, FFCLRP, University of São Paulo, Avenida dos Bandeirantes, 3900, 14040-901 Ribeirão Preto SP, Brazil
| | | | | |
Collapse
|
42
|
Zhang Y, Wei J, Yang Z. Perinatal undernutrition attenuates field excitatory postsynaptic potentials and influences dendritic spine density and morphology in hippocampus of male rat offspring. Neuroscience 2013; 244:31-41. [DOI: 10.1016/j.neuroscience.2013.03.061] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/26/2013] [Accepted: 03/30/2013] [Indexed: 01/22/2023]
|
43
|
Valadares C, Fukuda M, Françolin-Silva A, Hernandes A, Almeida S. Effects of postnatal protein malnutrition on learning and memory procedures. Nutr Neurosci 2013; 13:274-82. [DOI: 10.1179/147683010x12611460764769] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
44
|
Partadiredja G, Bedi K. Undernutrition during the gestation and suckling periods does not cause any loss of pyramidal neurons in the CA2–CA3 region of the rat hippocampus. Nutr Neurosci 2013; 13:102-8. [DOI: 10.1179/147683010x12611460764165] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
45
|
Cardoso A, Castro JP, Pereira PA, Andrade JP. Prolonged protein deprivation, but not food restriction, affects parvalbumin-containing interneurons in the dentate gyrus of adult rats. Brain Res 2013; 1522:22-30. [DOI: 10.1016/j.brainres.2013.05.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/22/2013] [Indexed: 10/26/2022]
|
46
|
da Silva AAM, Borba TKF, de Almeida Lira L, Cavalcante TCF, de Freitas MFL, Leandro CG, do Nascimento E, de Souza SL. Perinatal undernutrition stimulates seeking food reward. Int J Dev Neurosci 2013; 31:334-41. [PMID: 23669181 DOI: 10.1016/j.ijdevneu.2013.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 04/06/2013] [Accepted: 05/02/2013] [Indexed: 01/07/2023] Open
Abstract
Experiments in animals have revealed that perinatal nutritional restriction, which manifests in adulthood, increases food intake and preference for palatable foods. Considering this, we aimed to evaluate the effects of perinatal malnutrition on hedonic control of feeding behavior. In this study, we divided Wistar rats into two groups according to the diet provided to their mothers during pregnancy and lactation: the control group (diet with 17% casein) and low-protein group (diet with 8% casein). We assessed the animals' motivational behavior in adulthood by giving them a stimulus of food reward. We also assessed their neuronal activation triggered by the stimulus of palatable food using FOS protein labeling of neurons activated in the caudate putamen, paraventricular, dorsomedial, ventromedial, and lateral hypothalamic nuclei and amygdala. Evaluation of body weight in malnourished animals showed reduction from the 6th day of life until adulthood. Analysis of feeding behavior revealed that these animals were more motivated by food reward, but they had delays during learning of the task. This finding correlated with the number of c-FOS-immunoreactive neurons, which indicated that malnourished animals had an increase in the number of neurons activated in response to the palatable diet, especially in the amygdala and caudate putamen. The study therefore confirmed our hypothesis that early nutritional insults promote changes in encephalic control mechanisms, especially those related to food intake and search for reward.
Collapse
|
47
|
Buchanan KL, Grindstaff JL, Pravosudov VV. Condition dependence, developmental plasticity, and cognition: implications for ecology and evolution. Trends Ecol Evol 2013; 28:290-6. [PMID: 23518414 DOI: 10.1016/j.tree.2013.02.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Revised: 02/11/2013] [Accepted: 02/13/2013] [Indexed: 12/29/2022]
Abstract
Across taxa, both neural growth and cognitive function show considerable developmental plasticity. Data from studies of decision making, learning, and discrimination demonstrate that early life conditions have an impact on subsequent neural growth, maintenance, and cognition, with important ecological and evolutionary implications. Here, we provide a synthesis of the evidence that spatial and vocal learning are condition dependent, addressing what is known about their physiological control and the functional explanations. Neural investment is predicted to be affected by environmental conditions, but the shape of the response should depend on the fitness benefits of the cognitive traits under control. From an evolutionary perspective, traits promoting resistance to environmental perturbations should be favored when the cognitive trait is a crucial determinant of fitness.
Collapse
Affiliation(s)
- Katherine L Buchanan
- School of Life and Environmental Sciences, Deakin University, Geelong, VIC 3216, Australia.
| | | | | |
Collapse
|
48
|
Effects of protein restriction during gestation and lactation on cell proliferation in the hippocampus and subventricular zone: Functional implications. Protein restriction alters hippocampal/SVZ cell proliferation. Brain Res 2013; 1496:10-27. [DOI: 10.1016/j.brainres.2012.10.047] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 10/24/2012] [Accepted: 10/25/2012] [Indexed: 12/11/2022]
|
49
|
Ong SX, Chng K, Meaney MJ, Buschdorf JP. Decreased hippocampal mineralocorticoid:glucocorticoid receptor ratio is associated with low birth weight in female cynomolgus macaque neonates. J Mol Endocrinol 2013; 51:59-67. [PMID: 23592886 DOI: 10.1530/jme-12-0218] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
During pregnancy, glucocorticoids transfer environmental signals to the growing brain and its associated neuroendocrine system to modulate their maturation and function during adolescence and adulthood. Increased in utero exposure to glucocorticoids is associated with impaired fetal growth resulting in low birth weight (LBW) and compromised neural development. The underlying molecular changes affecting brain development, however, are largely unknown. Here, we compared the relative mRNA expression of genes directly involved in glucocorticoid signaling in the hippocampus, amygdala, and cortex of female non-human primate neonates (Macaca fascicularis) of naturally occurring normal birth weight and LBW. We focused on the glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) genes as well as that for 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) and found a significantly decreased MR:GR mRNA ratio in the hippocampus and lower expression of 11β-HSD1 in the amygdala associated with LBW. The MR:GR mRNA ratio in the amygdala and cortex was not associated with birth weight, reflecting tissue-specific effects. Protein quantification in the hippocampus confirmed our finding of a decreased hippocampal MR:GR ratio. Our data suggest that the MR:GR ratio in the hippocampus and the expression of 11β-HSD1 in the amygdala are associated with intrauterine growth restriction in non-human primates during early perinatal development.
Collapse
Affiliation(s)
- Shirlene X Ong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, Singapore 117609, Singapore
| | | | | | | |
Collapse
|
50
|
Lopes A, Torres DB, Rodrigues AJ, Cerqueira JJ, Pêgo JM, Sousa N, Gontijo JAR, Boer PA. Gestational protein restriction induces CA3 dendritic atrophy in dorsal hippocampal neurons but does not alter learning and memory performance in adult offspring. Int J Dev Neurosci 2012; 31:151-6. [PMID: 23280060 DOI: 10.1016/j.ijdevneu.2012.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 12/14/2012] [Indexed: 11/29/2022] Open
Abstract
Studies have demonstrated that nutrient deficiency during pregnancy or in early postnatal life results in structural abnormalities in the offspring hippocampus and in cognitive impairment. In an attempt to analyze whether gestational protein restriction might induce learning and memory impairments associated with structural changes in the hippocampus, we carried out a detailed morphometric analysis of the hippocampus of male adult rats together with the behavioral characterization of these animals in the Morris water maze (MWM). Our results demonstrate that gestational protein restriction leads to a decrease in total basal dendritic length and in the number of intersections of CA3 pyramidal neurons whereas the cytoarchitecture of CA1 and dentate gyrus remained unchanged. Despite presenting significant structural rearrangements, we did not observe impairments in the MWM test. Considering the clear dissociation between the behavioral profile and the hippocampus neuronal changes, the functional significance of dendritic remodeling in fetal processing remains undisclosed.
Collapse
Affiliation(s)
- A Lopes
- Fetal Programming Laboratory, Department of Morphology of Biosciences Institute, São Paulo State University, Botucatu, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|